Laboratory Models For Foodborne Infections
Laboratory Models For Foodborne Infections
Foodborne Infections
Series Editor
Dongyou Liu
Edited by
Dongyou Liu
Royal College of Pathologists of Australasia Quality Assurance Programs
New South Wales, Australia
CRC Press
Taylor & Francis Group
6000 Broken Sound Parkway NW, Suite 300
Boca Raton, FL 33487-2742
This book contains information obtained from authentic and highly regarded sources. Reasonable efforts have been made to publish
reliable data and information, but the author and publisher cannot assume responsibility for the validity of all materials or the con-
sequences of their use. The authors and publishers have attempted to trace the copyright holders of all material reproduced in this
publication and apologize to copyright holders if permission to publish in this form has not been obtained. If any copyright material
has not been acknowledged, please write and let us know so we may rectify in any future reprint.
Except as permitted under U.S. Copyright Law, no part of this book may be reprinted, reproduced, transmitted, or utilized in any form
by any electronic, mechanical, or other means, now known or hereafter invented, including photocopying, microfilming, and record-
ing, or in any information storage or retrieval system, without written permission from the publishers.
For permission to photocopy or use material electronically from this work, please access www.copyright.com (http://www.copyright.
com/) or contact the Copyright Clearance Center, Inc. (CCC), 222 Rosewood Drive, Danvers, MA 01923, 978-750-8400. CCC is a
not-for-profit organization that provides licenses and registration for a variety of users. For organizations that have been granted a
photocopy license by the CCC, a separate system of payment has been arranged.
Trademark Notice: Product or corporate names may be trademarks or registered trademarks, and are used only for identification and
explanation without intent to infringe.
1. Introductory Remarks...................................................................................................................... 1
Dongyou Liu
3. Astrovirus......................................................................................................................................... 29
Matthew D. Koci and Stacey L. Schultz-Cherry
4. Hepatitis E Virus..............................................................................................................................41
Kavita Lole, Prudhvi Lal Bhukya, and Subhashis Chatterjee
6. Rotavirus.......................................................................................................................................... 95
Lijuan Yuan and Ke Wen
7. Prions...............................................................................................................................................117
Akikazu Sakudo and Takashi Onodera
9. Clostridium......................................................................................................................................155
Emilio Aranda, María G. Córdoba, María J. Benito, and Juan José Córdoba
10. Enterococcus...................................................................................................................................175
Dongyou Liu
v
vi Contents
21. Escherichia......................................................................................................................................317
Dongyou Liu
22. Helicobacter....................................................................................................................................331
Tetsuya Tsukamoto, Yuka Kiriyama, and Masae Tatematsu
24. Proteus.............................................................................................................................................355
Paola Scavone, Victoria Iribarnegaray, and Pablo Zunino
26. Salmonella.......................................................................................................................................391
Dongyou Liu
31. Aspergillus.......................................................................................................................................455
László Kredics, János Varga, Rajagopalaboopathi Jayasudha, Sándor Kocsubé,
Nikolett Baranyi, Coimbatore Subramanian Shobana, Muthusamy Chandrasekaran,
Shine Kadaikunnan, Venkatapathy Narendran, Csaba Vágvölgyi, and
Palanisamy Manikandan
46. Metagonimus...................................................................................................................................743
Jong-Yil Chai
Index....................................................................................................................................................... 809
Preface for Food Microbiology Series
Microorganisms (including viruses, bacteria, molds, yeasts, protozoa, and helminths) represent abundant
and diverse forms of life that occupy various ecological niches of earth. Those utilizing food and food
products for growth and maintenance are important to human society due not only to their positive and
negative impacts on food supply, but also to their potential pathogenicity to human and animal hosts.
On one hand, foodborne microorganisms are known to play a critical role in fermentation and modi-
fication of foods, leading to a variety of nutritious food products (e.g., bread, beverage, yogurt, cheese,
etc.) that have helped sustain the human civilization from time immemorial. On the other hand, food-
borne microorganisms may be responsible for food spoilage, which, albeit a necessary step in keeping
up ecological balance, reduces the quality and quantity of foods for human and animal consumption.
Furthermore, some foodborne microorganisms are pathogenic to humans and animals, which, besides
creating havoc on human health and animal welfare, decrease the availability of meat and other animal-
related products.
Food microbiology is a continuously evolving field of biological sciences that addresses issues arising
from the interactions between food-/waterborne microorganisms and foods. Topics of relevance to food
microbiology include, but are not limited to, adoption of innovative fermentation and other techniques
to improve food production; optimization of effective preservation procedures to reduce food spoilage;
development of rapid, sensitive, and specific methods to identify and monitor foodborne microbes and
toxins, helping alleviate food safety concerns among consumers; use of -omic approaches to unravel
the pathogenicity of foodborne microbes and toxins; selection of nonpathogenic foodborne microbes as
probiotics to inhibit and eliminate pathogenic viruses, bacteria, fungi, and parasites; design and imple-
mentation of novel control and prevention strategies against foodborne diseases in human and animal
populations.
The Food Microbiology Series aims to present a state-of-art coverage on topics central to the under-
standing of the interactions between food-/waterborne microorganisms and foods. The series consists
of individual volumes, each of which focuses on a particular aspect/group of foodborne microbes and
toxins, in relation to their biology, ecology, epidemiology, immunology, clinical features, pathogenesis,
diagnosis, antibiotic resistance, stress responses, treatment and prevention, etc. The volume editors and
the authors are professionals with expertise in their respective fields of food microbiology, and the chap-
ter contributors are scientists directly involved in foodborne microbe and toxin research.
Extending the contents of classical textbooks on food microbiology, this series serves as an indispens-
able tool for food microbiology researchers, industry food microbiologists, and food regulation authori-
ties wishing to keep abreast with latest developments in food microbiology. In addition, the series offers
a reliable reference for undergraduate and graduate students in their pursuit to becoming competent and
consummate future food microbiologists. Moreover, the series provides a trustworthy source of informa-
tion to the general public interested in food safety and other related issues.
ix
Preface
Foodborne infections result from the ingestion of foods and beverages (including drinking water) that are
contaminated by pathogenic microorganisms (including viruses, bacteria, fungi, and parasites). While
some microbial pathogens stay in the gastrointestinal system and produce toxins (e.g., enterotoxins, exo-
toxins, and mycotoxins) that are absorbed into the bloodstream, others may directly invade deeper body
tissues. Although foodborne infections generally tend to induce mild clinical symptoms (e.g., nausea,
vomiting, fever, abdominal cramps, and diarrhea) in immunocompetent individuals, they may have seri-
ous consequences in young children and people with suppressed immune functions.
With the increasing consumption of manufactured foods and beverages, foodborne infections are
becoming a common and expensive public health problem worldwide. The World Health Organization
(WHO) estimates that food-/waterborne diarrheal diseases kill about 2.2 million people (mostly chil-
dren) annually. Based on FoodNet data collected between 2000 and 2007 by the Centers for Disease
Control and Prevention (CDC), 48 million foodborne illness cases (16,000 cases for 100,000 inhabitants)
occur in the United States every year, including 128,000 hospitalizations and 3,000 deaths. Interestingly,
31 foodborne pathogens have been implicated in 9.4 of the 48 million foodborne illness cases, with
7 (Salmonella, norovirus, Campylobacter, Toxoplasma, Escherichia coli O157:H7, Listeria, and
Clostridium perfringens) accounting for 90% of these illnesses alone. Similarly, an estimated 4.1 mil-
lion cases of foodborne gastroenteritis were documented in Australia in 2010, with norovirus, pathogenic
E. coli, Campylobacter spp., and nontyphoidal Salmonella spp. being the main culprits.
Although proper storage and refrigeration of food play a vital role in the prevention of foodborne infec-
tions, other good food safety practices (handwashing, preventing cross-contamination, and maintaining
cooking temperatures in the kitchen) are also valuable. In addition, accurate diagnosis and prompt medi-
cal intervention are crucial in reducing the mortality due to foodborne infections. However, thorough
understanding of host–pathogen interactions and elucidation of molecular mechanisms of pathogenesis
are critical for the development of effective vaccines that will lead to ultimate elimination of foodborne
infections in human population. Toward this goal, application of laboratory models (including both in
vivo and in vitro models) is essential.
As a part of the Food Microbiology Series, this book focuses on the value and utility of various
animal and cellular systems (ranging from mice, rats, hamsters, guinea pigs, rabbits, nonhuman pri-
mates, birds, zebrafish, frogs, chicken embryo, fruit fly, nematode, and waxworm to established and
nonestablished cell lines) in the study of foodborne infections. Written by experts involved in foodborne
pathogen research, each chapter presents a state-of-the-art review of laboratory models in the study of a
particular foodborne pathogen (of viral, bacterial, fungal, or parasitic origin) in relation to its life cycle,
host–pathogen interaction, pathogenesis, immunity, and other related aspects. Besides providing a reli-
able reference for undergraduates and postgraduates of food microbiology, this book is a valuable guide
for scientists using laboratory models in their investigation of foodborne infections.
Given the diversity of foodborne pathogens, a comprehensive book such as this is clearly beyond an
individual’s capacity. I am fortunate and honored to have a large group of scientists as chapter contribu-
tors, whose in-depth knowledge and technical insights on foodborne pathogens have greatly enriched
this book. Additionally, the professionalism and dedication of the senior editor, Stephen Zollo, have
enhanced its presentation. Finally, the understanding and support from my family—Liling Ma, Brenda,
and Cathy—have helped me keep focused during the compilation of this all-inclusive volume.
Dongyou Liu
xi
Editor
Dongyou Liu, PhD,studied veterinary science at Hunan Agricultural University, China, and completed
his postgraduate training at the University of Melbourne, Victoria, Australia. Over the past two decades,
he has worked at several research and clinical laboratories in Australia and the United States of America,
focusing on molecular characterization and virulence determination of microbial pathogens such as
ovine footrot bacterium (Dichelobacter nodosus), dermatophyte fungi (Trichophyton, Microsporum,
and Epidermophyton), and listeriae (Listeria spp.), as well as development of nucleic-acid-based quality
assurance models for security-sensitive and emerging viral pathogens. He is the author of over 50 origi-
nal research and review articles in various international journals, a contributor of 165 book chapters,
and the editor of Handbook of Listeria monocytogenes (2008), Handbook of Nucleic Acid Purification
(2009), Molecular Detection of Foodborne Pathogens (2009), Molecular Detection of Human Viral
Pathogens (2010), Molecular Detection of Human Bacterial Pathogens (2011), Molecular Detection
of Human Fungal Pathogens (2011), Molecular Detection of Human Parasitic Pathogens (2012),
Manual of Security Sensitive Microbes and Toxins (2014), and Molecular Detection of Animal Viral
Pathogens (2016), all of which are published by CRC Press. He is also a coeditor of Molecular Medical
Microbiology, 2nd edition (2014), which was published by Elsevier.
xiii
Contributors
xv
xvi Contributors
Bruce A. Vallance
Division of Gastroenterology
Child and Family Research Institute and
University of British Columbia
Vancouver, British Columbia, Canada
1
Introductory Remarks
Dongyou Liu
CONTENTS
1.1 Preamble............................................................................................................................................ 1
1.2 Foodborne Pathogens and Diseases.................................................................................................. 1
1.2.1 Foodborne Pathogens........................................................................................................... 1
1.2.2 Foodborne Diseases.............................................................................................................. 3
1.3 Laboratory Models............................................................................................................................ 3
1.3.1 Rationales for Using Laboratory Models............................................................................. 3
1.3.2 Milestones in the Use of Laboratory Models....................................................................... 4
1.3.3 Characteristics of Laboratory Models.................................................................................. 4
1.4 Future Perspective............................................................................................................................. 9
References................................................................................................................................................... 9
1.1 P reamble
Foodborne disease (also known as foodborne illness or colloquially as foodborne poisoning) is largely attrib-
utable to microbial pathogens and their toxins contained in food and food products that are inappropriately
prepared or stored before consumption. Once inside the host, these pathogens establish in their predilection
sites and cause damages to the host either through direct physical/mechanical destruction or through secre-
tion of toxins and antigens that provoke host innate and acquired immune responses, leading to a range of
clinical symptoms (e.g., diarrhea, abdominal cramps, nausea, fever, joint/back aches, and fatigue).
Although foodborne disease is a current buzzword that appears in various popular media outlets with
alarming frequency, it has a long and tortuous history. Our awareness of as well as our struggle against
foodborne disease goes hand in hand with our attempts to survive and prosper in a constantly chang-
ing, and challenging, world, with significant milestones marked by the use of fire, the development of
crop cultivation, the luxury of food storage, the evolution of culinary art, the sophistication of sewage
system, the observation of disease-causing microbes, the application of refrigeration, and the discovery
of antibiotics [1,2].
From scavengers who searched for the scraps left by other predators for survival, humans have
made enormous technological advances that overcome the barrier of seasonality for food supply, that
reduce the proliferation of foodborne disease, and that enable rapid identification and tracking of food-
borne pathogens implicated in any food-related disease outbreaks. Nonetheless, it is still a long way
before we can call foodborne pathogens the genie in the bottle, foodborne disease a memory of the past,
and foodborne outbreak an absolute nonevent.
1
2 Laboratory Models for Foodborne Infections
canned chili sauce, hot peppers, white and black pepper, raw cookie dough, hazelnuts, fenugreek sprouts,
papayas, pine nuts, raw frozen scraped ground tuna, etc.), the consumption of which by humans leads to
infections and diseases. In a broader sense, foodborne pathogens include microbes that occur in animals
(in farm/zoo animals and pets), the environment (soil, water, and air), and foods, the ingestion, inhalation,
and contact of which by humans result in discomfort and illness. Based on the latter premise, this book
covers not only microbial pathogens that come along with foods and food products (foodborne), but also
those that may occasionally enter into human host via water (water-borne), air (airborne), or direct contact
(skin wound), as well as those that cause diseases not through infection, but through production of toxins
and antigens that disturb/upset/confuse the gut, neurological, and immune systems of the human host.
As steadfast survivors, microbes (e.g., viruses, bacteria, fungi, and parasites) utilize animals (includ-
ing humans), foods, beverages, and water as growth or maintenance media. Some microbes remain in
humans (e.g., Salmonella Typhi and norovirus) or animal reservoirs and contaminate the food supply
via excreta, meat, milk, or eggs. Others persist in the environment and contaminate the ecosystems that
are fundamental to food production. Some microbes demonstrate the unusual ability to endure extreme
temperature, pH, and osmolarity, to sustain for long periods on dry surfaces, food processing plants,
and to exploit any temporary weakness in human innate and acquired immune defense networks (as
seen in pregnant women, infants, the elderly, and individuals under immune suppressing therapies).
Although a large number of foodborne microbes are known to infect humans and cause diseases
of varying severity, those having the most significant impact on human health in terms of prevalence,
morbidity, and mortality include Escherichia coli O157:H7, Campylobacter jejuni, Salmonella enteriti-
dis (e.g., serotypes Typhi and Typhimurium), Staphylococcus aureus, Listeria monocytogenes, Bacillus
cereus, Clostridium botulinum, Clostridium perfringens, Streptococcus pyogenes, Vibrio cholerae,
Vibrio parahaemolyticus, Vibrio vulnificus, Shigella, hepatitis A virus, hepatitis E virus, norovirus,
rotavirus, Cryptosporidium, Cyclospora cayetanensis, and Toxoplasma gondii [3]. It is notable that a
majority of these high-impact foodborne pathogens are bacteria, and the remainder are viruses and
parasites. Interestingly, some of these pathogens have emerged only in the past 30 years presumably due
to the increased consumption of processed food products, globalization of food trade, and population
ageing. For instance, E. coli O157:H7 is a shiga-toxin-producing bacterial strain that was first recognized
as a human pathogen in foodborne outbreaks associated with ground beef in 1982, producing symptoms
ranging from simple diarrhea and hemorrhagic colitis to hemolytic-uremic syndrome (which is charac-
terized by hemolytic anemia, thrombocytopenia, and renal injury) [4,5]. Subsequently, lettuce grown
in close proximity to a dairy farm from which wastewater contaminated with animal feces was used to
irrigate the plant was linked to a 2006 outbreak of E. coli O157:H7 infection in Iowa and Minnesota (see
Chapter 21 in this book). Another recently emerged foodborne pathogen of note is Aeromonas (mainly
A. hydrophila, A. caviae, and A. veronii), which is responsible for both intestinal and extraintestinal
diseases in humans (see Chapter 15 in this book). There is no doubt that new foodborne pathogens will
likely emerge or reemerge in the future.
Apart from infections with foodborne viruses, bacteria, fungi, and parasites, another important cause
of foodborne diseases is toxins or toxic chemicals produced by foodborne bacteria and fungi as well as
those associated with shellfish and plants [6]. Toxins originated from foodborne bacteria can be sepa-
rated into exotoxins (which remain part of the bacteria, and are secreted, or, similar to endotoxins,
released during bacterial lysis) and endotoxins (which form part of the bacterial outer membrane, and are
released during bacterial lysis). Some well-known foodborne bacterial exotoxins include superantigens
from S. aureus and Streptococcus pyogenes; pore-forming toxins (PFTs) from E. coli, L. monocyto-
genes, and Streptococcus pneumoniae; heat-stable enterotoxins (ST, exotoxins targeting the intestine)
from pathogenic strains of E. coli; and botulinum neurotoxin (BoNT) from C. botulinum. A notable
foodborne bacterial endotoxin is lipopolysaccharide (LPS, which is made up of O antigen, core oligosac-
charide, and lipid A) from Gram-negative bacteria. As water-soluble proteins, PFTs induce host mem-
brane damages as amphiphilic surfactants and phospholipases. On the other hand, endotoxins (e.g., LPS)
cause severe inflammation, endotoxemia (septic shock), and autoimmune disease. Being the by-products
of foodborne fungi, mycotoxins are responsible for alimentary mycotoxicoses in humans through food
consumption. The most common foodborne mycotoxins consist of aflatoxins (from Aspergillus para-
siticus and Aspergillus flavus), altertoxins (from Alternaria), fumonisins (from Fusarium moniliforme),
Introductory Remarks 3
to examine host immune responses to foodborne pathogens; to uncover the pathological and histological
details resulting from foodborne infections; to validate the accuracy of clinical diagnostic techniques;
and to evaluate the efficacy of newly developed antimicrobial and vaccine preparations against food-
borne pathogens without harming human hosts. This is made possible by the common ancestry of all
living organisms, the similarity of anatomical structures and functions (e.g., breathing, digestion, move-
ment, sight, hearing, reproduction, immunity, etc.), the homology of genetic materials, the sharing of
hundreds of illnesses, and the conservation of cell biological and developmental pathways among verte-
brates as well as between vertebrates and invertebrates [8].
on the use various established and non-established cell lines (derived from epithelia, endothelia, macro-
phage, etc.), embryonated eggs, and organs and tissues from hosts (Table 1.1).
Among various in vivo animal models, nonhuman primates (NHPs, with genomes of 2.85–3.09 Gb dis-
persed in 21–24 chromosome pairs) are the closest relatives to humans (with a genome of 3.23 Gb dispersed
in 23 chromosome pairs), and represent ideal models for investigation of foodborne infections and other
human diseases, on the basis of biological, physiological, immunological, and genetic similarities. However,
because of limited availability, prohibitive cost, and ethical concerns, NHPs are rarely used nowadays [15].
TABLE 1.1
Characteristics of Laboratory Models for Foodborne Infections
Common Species/
Modela Cell Type Characteristics Exemplary Application
In Vivo
Nonhuman primates Chimpanzee (Pan Chimpanzee has a genome of 3.02 Gb, Helicobacter pylori,
(family Hominidae, troglodytes), rhesus rhesus monkey 3.09 Gb, cynomolgus L. monocytogenes,
order Primates) monkey (Macaca monkey (crab-eating macaque, Mycobacterium,
mulatta), cynomolgus long-tailed macaque, or Java macaque) hepatitis E virus
monkey (Macaca 2.85 Gb, olive baboon 2.94 Gb. Ideal
fascicularis), olive models for foodborne infections and
baboon (Papio anubis) other human diseases, but limited by
availability, cost, and ethical concerns
Mice (family Muridae, House mouse (Mus Mice (house mice) possess a genome L. monocytogenes,
order Rodentia) musculus) strains: of 2.67 Gb, are small, readily S. aureus, Salmonella
BALB/c/(inbred, available, easy to handle, amenable
albino), C57BL/6/ to genetic manipulation, and
(inbred, dark brown), reproduce quickly, representing an
athymic nude mice efficient, cost-effective, and widely
(outbred) applicable animal model for
experimentation on foodborne
infections and other human diseases
Rats (family Muridae, Norway rat (Rattus Norway rat (brown rat) has a genome Salmonella, S. aureus,
order Rodentia) norvegicus) (inbred), of 2.61 Gb. Developed in 1906, Yersinia,
Wistar rat (outbred, Wistar rat (outbred albino) is the Acanthamoeba
albino), Lewis rat ancestor of most laboratory rats used
(inbred) today, including the Lewis rat. Wistar
rat shows albino coloring, a docile
behavior, and low fertility, and
tolerates crowding
Gerbils (family Mongolian gerbil Mongolian gerbil (Mongolian jird) is H. pylori,
Muridae, order (Meriones easy to keep as it adapts to a new L. monocytogenes,
Rodentia) unguiculatus) (outbred) setting well Giardia
Hamsters (family Syrian hamster Syrian hamster (golden hamster) Mycobacterium,
Cricetidae, order (Mesocricetus auratus) possesses a genome of 2.50 Gb, Acanthamoeba
Rodentia) (outbred), Chinese Chinese hamster 2.36 Gb. Hamsters
hamster (Cricetulus have a short life cycle and breed well
griseus) in captivity; being relatively free from
natural diseases, hamsters are
susceptible to experimental pathogens
Chinchillas (family Long-tailed chinchilla Chinchilla has a genome of 2.39 Gb. L. monocytogenes,
Chinchillidae, order (Chinchilla lanigera) Being crepuscular rodents, chinchilla Yesinia
Rodentia) is a robust host for experimental study
Guinea pigs (family Hartley Guinea pig Guinea pig has a genome of 2.72 Gb, and L. monocytogenes,
Caviidae, order (Cavia porcellus) shows similarity to humans in disease S. aureus
Rodentia) (outbred, albino) symptoms, immune response, and
pathogenesis
(Continued)
6 Laboratory Models for Foodborne Infections
On the other hand, as the next in the order of closeness to humans, rodents (especially mice) are
increasingly employed as preferred animal models for foodborne infections and other human diseases.
Mice possess a genome of 2.67 Gb dispersed in 20 chromosome pairs with ∼25,000 genes, 99% of
which have human counterparts. Having a relatively small body size, 18-day gestation, 10–15 pups per
litter, 7 weeks to sexual maturity, and a 2–3-year lifespan (1 mouse year equals about 30 human years),
mice provide an efficient and cost-effective model for human disease research including foodborne infec-
tions. It should be noted that mice practice coprophagy, an aspect that may be considered in experimental
design for certain disease types.
Mice are highly amenable to manipulation, and can be inbred to yield genetically identical strains,
which allows for more accurate and repeatable experiments. Through practice of cesarian birth, flexible-
film isolator cages, and irradiated food, mice (and other animal species) can be maintained in completely
germ-free conditions or colonized with one or more defined bacterial species (gnotobiotics). In addition,
use of genetic selection and manipulation technologies enables insertion of extra genetic materials into
genome, creating a variety of transgenic mice (including knockout, knock-in, and humanized mice as
well as mice with conditional gene modifications or chromosomal rearrangement) [8].
For example, athymic nude mice are selected for the nude spontaneous mutation (Foxn1nu, formerly
Hfh11nu) (which results in abnormal hair growth) and in the defective development of the thymic epithe-
lium (which abrogates a cell-mediated immunity, despite the presence of T-cell precursors). Homozygous
nude mice show partial defect in B cell development probably due to the absence of functional T cells,
and their responses to thymus-dependent antigens are primarily limited to IgM due to a defect in helper
T-cell activity.
Knockout mice are created by inserting a specific mutation into the endogenous gene. This leads to
inactivation/silencing of the gene of interest, suppressing its expression and function. Knock-in mice are
created by inserting a transgene into an exact location for overexpression. Both knockout and knock-in
animals rely on the use of embryonic stem (ES) cells containing null or point mutations and complex
chromosomal rearrangements (e.g., large deletions, translocations, or inversions), which are injected into
the host mouse embryo, and subsequently implanted into a foster mother.
Humanized mice are created by inserting human genes (more recently entire human systems) into
mice for subsequent expression. For instance, mice with human “immune systems” were generated by
implanting either fetal lymphoid tissue or peripheral blood leukocytes into mice with spontaneous severe
combined immunodeficiency. Humanized mice are capable of accepting a variety of human cells (blood,
immune, cancer, etc.) without rejection.
Mice with conditional gene modifications are created with two different types of genetic alterations:
one contains a conditional vector [through inserting recognition sequences for the bacterial Cre recom-
binase (loxP sites) using homologous recombination in ES cells], which functions as an “on switch”
for the mutation, and the other contains specific sites (called loxP) inserted on either side of a whole
gene, or part of a gene, that encodes a certain component of a protein that will be deleted. Similarly,
mice with chromosomal rearrangement are created using the Cre/loxP recombination system to induce
site-specific mutations that display defects resembling those caused by human chromosomal rearrange-
ments (e.g., chromosome deletions, duplications, inversions, translocations, and nested chromosome
deletions) [8].
Depending on the levels of simulation to human disease, animal models may be separated into three
types: homologous, isomorphic, and predictive. Homologous animals demonstrate identical causes,
symptoms, and treatments relative to human diseases; isomorphic animals have identical symptoms
and treatments; predictive models share only a couple of aspects of human disease with humans, but
nevertheless provide useful predictions about mechanisms of particular disease features. Similarly,
depending on the way in which animal disease is induced, animal models may be divided into four
categories: experimental, spontaneous, negative, and orphan. Experimental disease models resemble
human disease conditions in phenotype or response to treatment but are induced artificially in the labo-
ratory. Spontaneous disease models are analogous to human disease conditions that occur naturally.
Negative disease models are essentially control animals, and are used to validate an experimental result.
Orphan disease models have no human analog and occur exclusively in the species studied. Furthermore,
to examine a particular disease, various approaches may be used. For example, inflammation may be
Introductory Remarks 9
studied via Carrageenan footpad edema (CFE) model, collagen-induced arthritis (CIA) model, pristane-
induced arthritis (PIA) model, adjuvant-induced arthritis (AIA) model, ovalbumin-induced arthritis (OIA)
model, air pouch model, and delayed-type hypersensitivity (DTH) model [8].
The in vitro culture models provide an alternative to the in vivo animals for mechanistic studies, by
preserving the physiology of the living cell, without the need to sacrifice an animal. The advantages
of the in vitro culture models include low cost, easy maintenance, relatively high efficiency, and little
ethical concern. For instance, Caco-2 cells (of human colonic origin) can differentiate in culture, form
brush border membranes, demonstrate transport properties (similar to intestinal epithelia), and express
abundant intestinal microvilli, enzymes, and differentiation markers (typical of human small intestinal
enterocytes), offering a valuable model for investigation of vectorial epithelial passage by para- and
transcellular routes. Apart from the established cell lines (of epithelial, endothelial, and macrophage
origins), other cells, organs, and tissues may be obtained from animal and human hosts for in vitro mod-
eling. These include enterocyte suspensions, brush border membranes and vesicles, perfused duodenal
segment, everted gut sacs, lymphocytes, etc.
When selecting an animal model for research, considerations should include: (1) appropriateness as
an analog, (2) transferability of information, (3) genetic uniformity of organisms, (4) background knowl-
edge of biological properties, (5) cost and availability, (6) generalizability of the results, (7) ease of and
adaptability to experimental manipulation, (8) ecological consequences, and (9) ethical implications. If
possible, three basic principles should be applied: replacement, reduction, and refinement. Replacement
aims to use alternatives [e.g., computer models, tissues and cells, “lower-order” animals (cold-blooded
animals, invertebrates, bacteria) instead of “higher-order” animals (primates and mammals) for experi-
mentation]. Reduction employs mathematical calculations of statistical power to minimize the number of
animals used. For example, by using an alternative way to LD50 for result interpretation, the number of
experimental mouse groups for assessing L. monocytogenes virulence may be reduced from four to two,
with further advantage of obviating the necessity to perform colony forming unit (CFU) estimation [16].
Refinement aims to minimize the suffering of each animal subject through experimental design that is
as painless and efficient as possible [8].
REFERENCES
1.
Bell A. Foodborne illness: the history of an invisible enemy. Available at https://antiquitynow.
org/2014/06/05/foodborne-illness-the-history-of-an-invisible-enemy/.
2.
Satin M. History of foodborne disease—Part 1—ancient history. Encyclopedia of Food Safety
(Internet). 2014. Available at http://tinyurl.com/kmzt56u.
3.
Behravesh CB, Williams IT, Tauxe RV. Emerging foodborne pathogens and problems: expanding pre-
vention efforts before slaughter and harvest. In: Institute of Medicine (US). Improving Food Safety
through a One Health Approach: Workshop Summary. Washington, DC: National Academies Press
(US); 2012. A14. Available at http://www.ncbi.nlm.nih.gov/books/NBK114501/.
10 Laboratory Models for Foodborne Infections
4. Riley LW, et al. Hemorrhagic colitis associated with a rare Escherichia coli serotype. New Engl J Med.
1983;308(12):681, 684–685.
5. Rangel JM, et al. Epidemiology of Escherichia coli O157:H7 outbreaks, United States, 1982–2002.
Emerg Infect Dis. 2005;11(4):603.
6. Henkel JS, Baldwin MR, Barbieri JT. Toxins from bacteria. Experienta Suppl. 2010;100:1–29.
7. Walker MJ, et al. Disease manifestations and pathogenic mechanisms of group A Streptococcus. Clin
Microbiol Rev. 2014;27(2):264–301.
8. McGonigle P, Ruggeri B. Animal models of human disease: challenges in enabling translation. Biochem
Pharmacol. 2014;87(1):162–171.
9. Ericsson AC, Crim MJ, Franklin CL. A brief history of animal modeling. Mo Med. 2013;110(3):201–205.
10. Wolfgang MJ, Golos TG. Nonhuman primate transgenesis: progress and prospects. Trends Biotechnol.
2002;20:479–484.
11. Padilla-Carlin DJ, McMurray DN, Hickey AJ. The guinea pig as a model of infectious diseases. Comp
Med. 2008;58(4):324–340.
12. Meurens F, Summerfield A, Nauwynck H, Saif L, Gerdts V. The pig: a model for human infectious
diseases. Trends Microbiol. 2012;20(1):50–57.
13. Bou Ghanem EN, Myers-Morales T, D’Orazio SE. A mouse model of foodborne Listeria monocyto-
genes infection. Curr Protoc Microbiol. 2013;31:9B.3.1–9B.3.16.
14. Disson O, Lecuit M. In vitro and in vivo models to study human listeriosis: mind the gap. Microbes
Infect. 2013;15(14–15):971–980.
15. García Y, Díaz-Castro J. Advantages and disadvantages of the animal models v. in vitro studies in iron
metabolism: a review. Animal. 2013;7(10):1651–1658.
16. Liu D. Listeria monocytogenes: comparative interpretation of mouse virulence assay. FEMS Microbiol
Lett. 2004;233(1):159–164.
Section I
CONTENTS
2.1 Introduction..................................................................................................................................... 13
2.2 Classification and Morphology........................................................................................................14
2.3 Genome Organization and Conserved Features............................................................................. 15
2.4 Protein Function...............................................................................................................................16
2.5 Life Cycle........................................................................................................................................ 20
2.5.1 Cell Entry and Replication.................................................................................................... 20
2.5.2 DNA Packaging and Virion Assembly.................................................................................. 21
2.6 Pathogenesis.................................................................................................................................... 21
2.7 Host–Pathogen Interaction.............................................................................................................. 22
2.8 Immunity......................................................................................................................................... 22
2.9 Conclusions and Future Perspectives.............................................................................................. 23
References................................................................................................................................................. 23
2.1 I ntroduction
Adenovirus (AdV) is an important human pathogen and is estimated to account for 8% of clini-
cally relevant viral diseases globally.1 First identified in 1953 as the cause for acute febrile respira-
tory disease, AdVs are endemic in the pediatric population worldwide, affecting children younger
than 5 years old with mild symptoms and generally self-limiting illnesses.2–4 Self-limiting infections
may also occur in adults, but some serotypes have been associated with severe respiratory illness
and potentially fatal outbreaks of pneumonia in residential facilities and military bases. The main
etiologic agents for these outbreaks are serotype 4 and occasionally serotypes 3, 7, 14, and 21. It is
possible that stress and crowding may contribute to AdV transmission and susceptibility.5 AdVs have
serious complications, impacting morbidity and mortality in immunocompromised individuals of
any age.6–8
With more AdV serotypes being identified, it becomes clear that AdVs cause an array of clinical
diseases, including epidemic keratoconjunctivitis (EKC), acute hemorrhagic cystitis, hepatitis, gastroen-
teritis, myocarditis, and pneumonia. Being one of the most prevalent enteropathogens causing infantile
gastroenteritis, enteric AdVs are implicated in sporadic cases as well as in outbreaks of food-borne
illness in kindergartens, schools, and hospitals.9 Gastroenteritis due to AdVs often occurs in children
younger than 5 years of age, accounting for ∼12% of all enteropathogenic viruses identified, and is
most commonly associated with serotypes 40 and 41; however, other types including 1, 2, 7, 9, 10, 18,
19, 22, 31, 42, 52, 58, 65, and 67 have also been reported as etiologic agents of viral gastroenteritis.9–20
Serotypes 40 and 41 account for 5%–20% of hospital-admitted diarrhea cases in children under 2 years
old. As children age, the incidence of AdV gastroenteritis decreases due to increasing levels of popula-
tion immunity to AdV infection.
AdVs can be easily propagated in cell culture, and there are several cell lines that can be used as
laboratory models for AdVs. The primary human embryonic kidney (HEK) cells are the best host for
13
14 Laboratory Models for Foodborne Infections
replicating various serotypes of AdVs. The lung epithelial cell line A549 and other epithelial cell lines,
such as HEP-2, HeLa, and KB, are also good hosts for AdVs. For enteric AdVs, such as AdV40 and
AdV41, the HEK 293 cell offers a convenient laboratory model.2 In addition, Sigmodon hispidus cotton
rats and mice, such as C57BL/6N, C57BL/lOScN, CBA/N, and C3H/N strains, were used as animal
models to investigate the molecular pathogenesis of pneumonia caused by AdV infection.21 AdVs have
been used as models of virus–cell interaction. Decades of studies have contributed to the extensive
understanding of the molecular biology, including life cycles, the host–pathogen interaction, genet-
ics, epidemiology, and pathogenesis of AdVs, which are discussed in this chapter. AdVs continue to be
studied as delivery vehicles for gene therapy, vaccination, and cancer treatment, which underscores the
importance of understanding these viruses.
2.2 C
lassification and Morphology
AdVs constitute the Adenoviridae family, which is divided into the genera Mastadenovirus and
Aviadenovirus. The genus Mastadenovirus covers viruses of several different animals, including
bat, bovine, canine, equine, human, murine, ovine, porcine, simian, and so on, whereas the genus
Aviadenovirus is limited to viruses of birds.2 Currently, there are 68 reported human AdVs according
to the National Center for Biotechnology Information Taxonomy Browser, representing seven different
species or subgroups (A–G). The classification of AdVs was originally based on their hemagglutina-
tion patterns and serologic profiles. Recent advancements in sequencing capability have allowed the
discovery and classification of new AdVs (types 52–68), where the differentiation of strains is based
on bioinformatics analysis of their genomic sequencing (Table 2.1).2,4,11–15,22–37 The majority of these
newly discovered AdVs are products of homologous recombination, a common evolutionary adaptation
of AdVs. Among the seven different species, species B can be further divided into B1 and B2 based on
their organ tropisms.22 There is a correlation between the species and their tissue tropisms, which deter-
mines the clinical manifestation of AdV infection. Species A, F, and G show tissue tropisms toward the
gastrointestinal tract and induce gastroenteritis. Species B1, C, and E mainly cause respiratory illness;
species B1, B2, D, and E produce ocular infection, whereas B2 AdVs cause kidney and urinary tract
infection.8,37
AdVs are nonenveloped double-stranded DNA (ds DNA) viruses with icosahedral shells and nucleo-
protein cores, ranging in size from 65 to 100 nm in diameter. The capsid of the viral particle is composed
of seven proteins: hexon, three hexon-associated proteins, penton, a penton-associated protein, and fiber.
The proteins form 252 capsomeres, which consist of 240 trimers of the major capsid protein hexon and
12 pentons. The fiber protein, which has a length that varies among the different serotypes, embeds in
the penton base and projects out from the capsid. These 12 extensions out of the particle serve crucial
TABLE 2.1
Classification of Human Adenoviruses
Species Hemagglutination Serotype References
A IV 12, 18, 31, 61 2,12
B1 I 3, 7, 16, 21, 50, 66, 68 22–25
B2 I 11, 14, 34, 35, 55 22,26,27
C III 1, 2, 5, 6, 57 2,28
D II 8–10, 13, 15, 17, 19, 20, 22–30, 32, 33, 36–39, 2,13–15,25,29–36,120
42–49, 51, 53, 54, 56, 58, 59, 60a, 62–65, 67
E III 4 2
F III 40, 41 2
G 52 11
I, complete agglutination of monkey erythrocytes; II, complete agglutination of rat erythrocytes; III, partial
agglutination of rat erythrocytes; IV, little or no agglutination.
Adenoviruses 15
(A) (B)
Core-V
Terminal protein
Hexon
Penton base
Fiber
FIGURE 2.1 Image and schematic depiction of the structure of adenovirus. (A) Transmission electron micrograph (TEM)
of adenovirus virions. Visible at this high magnification are the capsomeres, which are hexagonally shaped, also called
hexons. Hexons comprised the outer shell of the adenovirus known as a capsid. These adenoviruses displayed an icosahe-
dral symmetry, characterized by 12 vertices and 20 facets. Each virion was 70–80 nm and exhibited no spikes. Image by
Dr. G. William Gary, Jr. was downloaded from the public health image library (ID #237) of Centers for Disease Control
and Prevention, USA. (B) Schematic depiction of the icosahedral capsid of human adenovirus (HAdV) showing the major
structural proteins, hexon, penton based, and fiber in the virion. The ds DNA genome is shown as a dark line inside the
capsid. (Modified from Russell, W.C., J. Gen. Virol., 90, 1–20, 2009.)
functions in cell attachment and entry (Figure 2.1).2,6 All the reported AdVs encode a single fiber protein
except AdV40, AdV41, and AdV52, which encode two fiber proteins. Fiber proteins mediate viral entry
by interacting with receptors on susceptible cells; the expression of two fiber proteins might expand the
range of susceptible cells for these AdVs.2,11 The nucleoprotein core of AdVs contains the viral genome
and five structural proteins, V, VII, Mu (μ), IVa2, and a terminal protein (TP).2,6
2.3 G
enome Organization and Conserved Features
The AdV genome consists of a linear ds DNA of ∼35 kb encoding more than 30 structural and nonstruc-
tural proteins in most serotypes. All AdV serotypes have the same general organization; that is, the genes
encoding specific functions are located at the same position on the viral chromosome. However, DNA
sequence homology among the strains within a human AdV species ranges from 48% to 99%, with the
highest homology among strains of species C. The homology between species is <20%.2,4 The 5′ and
3′ ends of the human AdV genome consist of inverted terminal repeats (ITRs) ranging from 40 to 212
nucleotides, which contain the origin of DNA replication and are hallmark motifs of the HAdV genome.
All HAdV genomes contain five early (E) transcribed genes (E1A, E1B, E2–E4), two intermediate genes
(IX, IVa2), and five late (L) genes (L1–L5) (Figure 2.2). The early genes are mainly involved in the
control of DNA replication, transcription, transportation of Ad mRNAs (messenger ribonucleic acids) to
the cytoplasm, and immunoregulation; the intermediate genes control DNA packing and also function as
transcription factors, whereas the late genes are coded for viral structural proteins.38
The E1A gene is the first to be transcribed and expressed after infection, and multiple E1A proteins
are generated through alternative splicing, which regulate the transcription of viral and cellular genes.
The E1B gene encodes proteins that block apoptosis and potentiate viral replication. The E2 region is
composed of two transcripts, E2A and E2B, and produces proteins required for viral replications, which
include DNA-binding protein (DBP), terminal protein precursor (pTP), and DNA polymerase (pol).29,39–41
Proteins encoded by E3 gene are not essential for viral growth, but function as modulators of the host
immune response.38,42 The E4 gene encodes proteins with various functions, including promoting viral
gene expression and replication, regulating cellular transcription factor E2F, and controlling protein
phosphorylation.43 The two intermediate genes produce proteins, IX and IVa2, which control DNA
16 Laboratory Models for Foodborne Infections
1 2K 4K 6K 8K 10 K 12 K 14 K 16 K 18 K 20 K 22 K 24 K 26 K 28 K 30 K 32 K 32, 214
ITR ITR
L3 L4A L5A
E2B E2A Fiber-2
E1A
ssDBP E4
IVa2 L1 E2A-L U
L2 L5
E1B VAI Fiber
E3B
IX
E3A
FIGURE 2.2 Genome structure of human AdV-type 40. The genome structure was constructed based on the sequence
deposited in GenBank (NC_001454). The half-filled rectangle indicates a gene, while the fully filled rectangle indicates
CDS. The arrows indicate the direction of the gene that is transcribed. VAI, virus-associated RNA I; ssDBP, single-
stranded DNA-binding protein.
packing and also function as transcription factors. The five late genes encode viral proteins involved in
capsid production for mature virions, assembly, and viral maturation, such as 52-kDa protein, IIIa, pen-
ton base (III), V, VII, X, minor capsid protein (VI), hexon, 23-kDa protease, 100-kDa protein, 33-kDa
protein, VIII, and fiber.2,39,40
There are 16 genus-common proteins; that is, there is homology in all genera in AdV genomes, which
are probably rooted in a common ancestral AdV. These proteins primarily function in DNA replication,
DNA encapsidation, and viral particle structural formation. The genus-common proteins involved in
DNA replication are DBP, pTP, and pol, and two proteins are involved in DNA encapsidation, 52-kDa
protein and IVa2. Viral proteins, such as IIIa, III, VII, X, VI, hexon, protease, 100-kDa protein, 33-kDa
protein, pVIII, and fiber, necessary for virion formation are also genus common.41 Additionally, U exon
is also considered as a genus-common feature, even though it has been lost in certain members of the
Mastadenovirus genus (PAdV-5 and MAdV-1). U exon locates between the E3 region and the fiber gene,
encodes the U exon protein (UXP, ∼24 kDa), which initiates in U exon, continues in frame in the DBP
leader, and extends throughout the DBP coding region but with a different reading frame from DBP that
has functions relevant to DNA replication or RNA transcription.41,44 For HAdVs, most genus-specific
genes are located within the E1 and E4 regions.
in addition to stabilizing hexon capsomeres and bridging the core to the icosahedral shell. In the early
phase of viral infection, the N-terminal amphipathic α-helix is responsible for inducing curvature of the
endosome membrane, which leads to membrane disruption, thereby allowing the virus to escape into
the cell’s cytosol. pVI also contains a ubiquitin ligase-interacting motif (PPxY), which not only facili-
tates microtubule-dependent trafficking toward the nucleus, but also activates E1A promoter and leads
to initiation of adenoviral gene expression. In later phases of AdV infection, the nuclear export signals
of pVI interact with importin α/β, which mediates the nuclear import of hexon and the subsequent pro-
teolytic removal of C-terminal nuclear export signal transition pVI to support infectious viral particle
assembly.45,49–51 Minor coat protein pVIII is not as well characterized, and its only known function is
stabilizing capsomeres. pIX, located on the outer part of the capsid, also has multiple functions in the life
cycle of AdV infection. Upon viral entry and escaping from endosome, pIX is responsible for interacting
with kinesin-1, which results in capsid dismantling and promotion of transport of the viral genome into
nucleus. Subsequently, the C-terminal leucine repeat is important for enhancing gene expression from
the E1A promoter in the early stage of infection, and the N-terminal region is required for incorporation
into viral capsid at the later stage of infection. pIX also plays a role in virus-induced nuclear reorganiza-
tion, that is, the formation of specific nuclear structures, which appear as dispersed nuclear globules on
immunofluorescence staining and as clear amorphous spherical inclusions on the electron microscopy
of cells that are in the later stages of infection. It is suggested that the reorganization of nuclear proteins
provides a favorable environment for viral replication. Evidence also suggests that pIX may play a role in
modulating the viral tropism and/or interfering with the immune response.45,52–54
In addition to hexon capsomeres, the penton base and fiber proteins are also the main building blocks
of AdV capsid and form penton capsomeres, which locate at 12 vertices of the icosahedral facets. The
penton capsomere consists of a homopentameric penton base and homotrimeric fibers, with the fiber
molecules projecting from the penton base at each vertex. Penton and fiber molecules are the key play-
ers in AdV attachment and cell entry.45,55 The C-terminal knob domain of fiber molecules initiates the
interaction with cell receptors. Once the cell binding is initiated, the integrins (coreceptor) αvβ3 and αvβ5
bind to conserved RGD domain at penton base, which in turn starts the receptor-mediated endocytosis
with the exception of species F enteric AdVs, which neither have the RGD motif nor use integrin for
cell infection.56 The size of the penton protein varies with the serotype, ranging from 493 to 594 amino
acids. The most significant size variation of the major capsid proteins of AdVs is observed in fiber pro-
tein, which ranges from 319 to 587 amino acids (aa). Serotypes within species A and C have long fibers
(>500 aa), serotypes within B and D species have short fibers (<400 aa), and species E has intermediate
length of fibers (425 aa). Interestingly, serotypes belonging to species F and G have two fibers (long and
short fibers) and all cause gastroenteritis. For species F, the long fiber has been shown to bind to the AdV
receptor, while the function of the short fiber remains unknown, but it likely plays a role in enteric AdV
tropism and cell infection.57
Capsid core proteins include pV, pVII, Mu, TP, pIVa2, and the protease, and all but the protease are
associated with the viral genome. The minor core protein pV, containing a basic arginine-rich domain,
interacts with viral DNA. Functional studies indicated that pV is not essential for viral replication and
infectious viral particle assembly in cancerous cells, but it mediates the translocation of nucleophosmin
1/NPM1/B23.1 from the nucleoli to the nucleoplasm that is essential for productive infection in normal
cells.37,58,59 The major core protein pVII, with four basic lysine- and arginine-rich domains, serves as a
histone-like structure that viral DNA wraps around. pVII is synthesized as a precursor protein (preVII)
and is matured by the removal of the N-terminal 24 amino acids via the viral protease. Interestingly,
preVII is involved in condensing the viral DNA during particle assembly, but only mature pVII is pres-
ent in the viral nucleoprotein core. In addition, pVII also mediates virus DNA import into the nucleus
through the transportin pathway during infection, potentially through its nuclear and nucleolar local-
ization signals. Furthermore, pVII interacts with pIVa2 and L1 52/55 kDa during viral particle assem-
bly.37,60,61 pIVa2 performs multiple tasks during the life cycle of an AdV. Encapsidation of viral DNA
requires the binding of pIVa2 to the packaging domain as a multimeric complex with the L4 22-kDa
protein and the L1 52/55-kDa protein, as well as pVII. The binding of pIVa2 to the packaging domain
initiates genome recognition and recruits the L4 22-kDa protein to the packaging domain. In addi-
tion, pIVa2 forms a dimer with the L4 33-kDa protein and functions as a transcription activator of the
18 Laboratory Models for Foodborne Infections
viral major late promoter and regulates the synthesis of the majority of late structural proteins of the
virus.37,62–64 Core protein Mu (μ), generated from a precursor protein (79 aa) by the removal of the N- and
C-terminal amino acids via the viral protease, is a function partner of pV and pVII in condensing the
viral genome and DNA encapsidation.65,66 TP is a 55-kDa protein, covalently linked to the 5′ end of ITR,
which allows the circularization of viral DNA and enhances in vitro DNA replication.67–69 The TP is
synthesized as an 80-kDa pTP, which forms a heterodimer with pol that plays a key role in DNA replica-
tion by serving as a primer for initiating viral DNA replication. Furthermore, pTP is a key participant
in the AdV replication complex consisting of pTP, pol, and DBP as well as cellular factors NFI, Oct-1,
and type I topoisomerase NFII. This replication complex is tightly bound to the nuclear matrix through
interaction with the pTP, thereby anchoring the viral genome to the nuclear matrix.68,70–72 Protease plays
a key role in several steps of the viral life cycle, including uncoating the viral particles upon viral entry
into cells and enabling the cleavage of core precursor proteins that facilitates the formation of infec-
tious viral particles and the cleavage of cytokeratins leading to host cell lysis. It is synthesized in an
inactive form and becomes partially activated by binding to the viral DNA, which then cleaves out an
11-amino-acid viral peptide (pVIc, GVQSLKRRRCF) from the C-terminal of the precursor of pVI. pVIc
binds to the protease and fully activates its function.73–78
The majority of nonstructural proteins of AdVs are encoded by the early-transcribed genes. E1A is
the first gene expressed upon viral infection and plays an essential role in regulating the viral transcrip-
tion of genes necessary for replication and reprograming the cellular transcription to facilitate viral rep-
lication. Two major E1A proteins, 12S and 13S E1A, are synthesized via alternative splicing of mRNA
transcripts, which results in a 46 aa conserved region (CR3) that is unique in 13S E1A. Despite their
similarity, 13S E1A is primarily responsible for activating the viral gene expression via CR3, while
12S E1A represses transcription, which is achieved via temporal regulation.79–82 E1A proteins activate
transcription through a TATA motif, YY1 recognition sites, by interacting with a variety of cellular
transcription factors and relieving transcription repression.2 In addition to modulating transcription,
E1A proteins also regulate signaling pathways and 26S proteasome, interfere with pathways involved
in immune regulation, as well as involve the evasion of T-cell immunity to ensure viral survival and
infectivity.82
E1B gene products are required for efficient viral replication during AdV infection. At least five E1B
gene products were identified in Ad5, which were generated through alternative splicing. One of these
proteins, E1B-55-kDa protein, in conjunction with E4 open reading frame (ORF) 6 protein (E4ORF6)
forms an E3 ubiquitin ligase complex consisting of cellular proteins cullin 5 (Cul5), RING-box 1 (Rbx1),
and elongins B and C. This viral ubiquitin ligase regulates the degradation of cellular proteins, such as
p53, the MRE11-Rad50-NBS1 (MRN) DNA damage recognition/repair complex, and DNA ligase IV,
that have a detrimental effect on viral replication during the early stage of viral infection. In addi-
tion, the E1B-55-kDa/E4ORF6 ubiquitin ligase complex also prevents DNA damage repair mechanisms
by interfering with the MRN complex. During AdV infection, promyelocytic leukemia (PML) bodies,
consisting of several critical DNA response proteins including components of the MRN complex, are
disrupted and relocated to nuclear track-like structures. The MRN complex is initially relocalized into
PML-containing nuclear tracks by another E4 protein, E4ORF3, where it subsequently binds to the E1B-
55-kDa protein. The interaction of E1B-55 kDa and E4ORF3 enables the localization of E1B-55 kDa to
the PML-containing nuclear track, and forms complexes containing MRN, E1B-55 kDa, E4ORF3, and
E4ORF6 proteins, which are then transported out of the nucleus into cytoplasmic aggresomes to further
MRN proteasomal degradation by the E1B-55K/E4orf6 ubiquitin ligase complex. E1B-55K protein can
also contribute to the deregulation of the cell cycle during lytic infection. During the later stages of
viral infection, the E1B-55-kDa/E4orf6 ubiquitin ligase complex inhibits the export of host cell mRNA
while promoting the export of late viral mRNA from the nucleus and its translation.83,84 Another E1B
gene product, E1B-19-kDa protein (small T antigen), is the first viral BCL-2 homolog discovered, and it
inhibits apoptosis triggered by AdV infection by blocking the BIK/NBK pathway to ensure successful
virus production.85,86
E2 encodes three replication proteins, DBP, pol, and pTP. These proteins along with three cellu-
lar transcription factors, NFI, Oct-1, and type I topoisomerase, constitute the AdV DNA replication
system.2 DBP, a product of the E2A gene, binds dsDNA, ssDNA, and RNA without apparent sequence
Adenoviruses 19
specificity. It is involved in initiating DNA replication and is also essential for elongation by enhancing
the activity of pol, unwinding dsDNA, removing secondary structures, and protecting ssDNA. DBP is
a multifunctional protein involved in DNA replication, transcription regulation, mRNA stability, trans-
formation, virion assembly, and host range determination. During DNA replication, DBP first stimulates
the initiation by increasing the binding of NFI and recruiting the pTP–pol complex to the replication
origin. Then, DBP unwinds the parental strand and enhances the progress of the polymerase by binding
to the displaced strand and protecting it from nuclease digestion, which enhances strand displacement
activity during the elongation phase. Finally, DBP enhances the rewinding of complementary displaced
strands.87–89 AdV pol is a DNA-dependent DNA-pol encoded by E2B transcript with a molecular weight
of 140 kDa and belongs to a distinct family of protein priming pol, eukaryotic pol α family, that uses a
protein primer for the initiation of replication. It also shows 3′→5′ exonuclease activity for proofread-
ing. AdV pol forms a stable heterodimer with pTP, protein primer, which initiates the viral DNA rep-
lication by binding to NFI and Oct-1 and results in a pre-initiation complex which then binds the viral
core origin.90–92
The E3 region is distinct for each species varying in length and number of ORFs representing areas
of major sequence divergence.93 Common to all species, E3 encodes three proteins, gp19K, receptor
internalization and degradation (RIDα/β, 10.4 kDa/14.5 kDa), and 14.7-kDa protein, which function in
modulating host immune response and AdV death protein (ADP, 11.6 kDa), which is expressed in the
later stages of infection.38,94 The main function of gp19K is to suppress the cell surface expression of
class I major histocompatibility complex (MHC) by inhibiting the transport of class I MHC from the
endoplasmic reticulum (ER) to the plasma membrane and the processing of peptides by tapasin, thus
decreasing the amount of peptide presented by class I MHC in infected cells. The E3 14.7-kDa protein
inhibits apoptosis induced by TNF-α via reducing the secretion of arachidonic acid, while RIDα/β inhib-
its apoptosis induced by Fas-L and TRAIL. RIDα/β not only destroys the receptors for Fas-L and TRAIL
to prevent cell death caused by these factors but also inhibits apoptosis induced by TNF-α through the
same mechanism as the E3 14.7-kDa protein.38 In contrast, ADP is proapoptotic and mediates the release
of AdV by lysing the infected cells at very late stages of infection.94
E4 is predicted to encode seven different polypeptides that are required for lytic growth, and all but
one are expressed. Both ORF3 and ORF6 are involved in viral DNA replication by interacting with
E1B-55-kDa protein, as described earlier (see section above).83 Both ORF3 and ORF6 promote viral
gene expression by facilitating the accumulation of relevant mRNAs at a posttranscriptional level in
the cytoplasm and stabilizing the unprocessed late RNA in the nucleus. Evidence suggests that the
E1B-55-kDa–E4ORF6 complex causes the redistribution of cellular factors necessary for RNA biogen-
esis and that ORF3 directly affects the distribution of essential transcription/replication factors in the
nucleus. In addition, ORF6 interacts with p53 to inactivate its function, which is crucial for successful
infection by AdV.43 Besides ORF3 and ORF6, studies suggested that ORF1 is a transforming protein and
may stimulate quiescent cells to promote lytic infection and oncogenesis. E4ORF6/7 forms dimers to
interact and modulate the activity of the cellular transcription factor E2F and subsequently activate cel-
lular factors that are important for the S phase of cell cycle.43,95 The function of E4ORF4 is to negatively
regulate E1A and E4 transcription via protein phosphorylation by interacting with protein phosphatase
(PP)2A, which creates a regulatory loop limiting cytotoxicity in the early stages of infection. The func-
tional information for E4ORF2 and E4ORF3/4 still awaits discovery.43
AdVs possess two intermediate (also called delay early) genes IVa2 and IX, which encode pIVa2 and
pIX, respectively. pIVa2 interacts with L1 52/55-kDa protein to activate a major late promoter which is
required for viral DNA packaging.96 pIX is a minor coat protein and its function has been described in
the previous section. In addition, pIX also stimulates an AdV major late promoter containing TATA box
elements and contributes to the efficient induction of the late transcription unit.97
One of the nonstructural proteins is encoded by L4 gene. The L4-100-kDa protein inhibits the transla-
tion of cellular mRNA by eliminating the cap-dependent translation pathway via binding to eukaryotic
initiation factor 4G (eIF4G) and displacing Mnk1 from eIF4G. It also promotes translation of late viral
mRNAs through ribosome shunting, which leads to nuclear accumulation of viral products for capsid
assembly. The underlying mechanism for these functions is the interaction of L4-100 kDa with the tri-
partite leader (TL) sequence present in all the late viral transcripts and eIF4G, which is the scaffolding
20 Laboratory Models for Foodborne Infections
element of the cap-dependent translation initiation complex. In addition, L4-100-kDa protein not only
acts as a chaperone facilitating hexon trimerization but also assists in nuclear accumulation of hexon
trimers and in the scaffolding process of AdV capsid.98–101 Another function of L4-100 kDa is to prevent
apoptosis induced by granzyme B in infected cells. Granzyme B, one of the lymphocyte granule serine
proteases, catalyzes the cleavage and activation of several caspases which induce apoptotic events in
infected cells. By interacting with granzyme B, the L4-100-kDa protein prevents access of substrates
to the proteinase catalytic site through steric hindrance and inhibits its activity.102 A unique feature of
L4-100 kDa is its posttranslational modification by arginine methyltransferase 1 (RPMT1). The methyla-
tion of L4-100 kDa has regulatory effects on modulating its interaction with hexon and TL mRNA as
well as on promoting the capsid assembly.101
A conserved ORF in Mastadenovirus genus, U exon, located between E3 and L5, was first identified in
Ad40 genome sequence, and later identified to be a 24-kDa protein, UXP. UXP is a late protein encoded
in an overlapping reading frame with DBP and part of the 100-kDa protein and has been found in all
species C serotypes so far. The characterization of UXP indicated that it localizes within the nucleoli,
nucleus, and the periphery of the replication center, suggesting its involvement in DNA replication or
RNA transcription.44,103
Endocytosis
Endosomal lysis
Structural L1–L5 mRNAs
proteins
Vesicle escape
Assembly
DNA replication
E1–E4 mRNAs
Nonstructural
proteins
FIGURE 2.3 The life cycle of HAdV. First, the virus attaches to a primary receptor on the cell surface via fiber. This is
followed by penton base interaction with αvβ3 and αvβ5 integrins, which facilitates clathrin-mediated endocytosis. Inside
the endosome, the virion begins to dissociate in the low pH environment and releases the vertex proteins including pVI,
which has been implicated in the disruption of the endosomal membrane, allowing the virion to escape from the endo-
some. The partially disassembled virion is transported to the nuclear pore complex. At the nuclear pore, the viral DNA is
imported into the nucleus allowing the transcription and replication of the virion. At the final stage, the viral proteinase
cleaves its multiple substrates and an icosahedral structure is formed, which contains the full-length DNA. (Modified from
Nemerow, G.R. et al., Virology 384, 380–388, 2009; Shenk, T.E., Adenoviridae. in Fields Virology, Vol. 2. eds. Knipe,
D.M., Howley, P.M., Lippincott Williams & Wilkins, Philadelphia, 2265–2300, 2001.)
2.6 Pathogenesis
While AdVs generally infect the mucosal epithelium, individual serotypes differ in their tissue specific-
ity and cause a number of distinct clinical syndromes, including gastroenteritis, respiratory disease, con-
junctivitis, hemorrhagic cystitis, and exanthema. Thus, the pathogenicity of AdVs varies depending on
22 Laboratory Models for Foodborne Infections
the species, serotype, and organ specificity. In general, AdV-infected cells degenerate, and the infected
respiratory epithelial cells have enlarged nuclei containing inclusion bodies.3 Most of our understanding
on AdV pathogenesis comes from studies using S. hispidus cotton rats and mice for respiratory illness.
For gastroenteritis, it is likely that AdV infection causes destruction of functional mature cells, which in
turn disrupts the water readsorption from the gut and consequently diarrhea ensues. In response to the
damage, the villi retract and the crypt cells undergo rapid division to repopulate the villi with young and
undifferentiated cells which are resistant to virus infection. However, the malabsorption continues until
these undifferentiated cells mature and develop the necessary functional capabilities.106,107
2.8 I mmunity
Murine pneumonia models (nonpermissive infection with HAdVs) have been used to study the cytokine
response induced by AdV infections. During early-phase infiltration, the infected mouse lungs showed
high titers of TNF-α, IL-1, and IL-6, but only IL-6 was found in the peripheral blood. IL-6 reached max-
imum titers 6–24 h after infection, whereas maximum levels of TNF-α and IL-1 were attained 2–3 days
Adenoviruses 23
after infection.21 IL-8, a proinflammatory cytokine, secreted by airway macrophages and the epithelial
cells within the lung epithelium, was also induced by AdV infections. The higher titer of IL-8 increases
the protein synthesis and apical localization of CAR in polarized cells, which in turn tethers the infiltrat-
ing neutrophils on the apical surface of polarized epithelia. The adherence of neutrophils on the apical
surface provides for amplification of AdV infection.115 IL-12 is also induced by AdV infections, which
may be important in innate immune activation and the subsequent adaptive immune response to AdV
infection; however, the exact mechanism remains unclear.116 AdV infections also induce the produc-
tion of high levels of type I IFN. The increased levels of type I IFN enhance the transcription of IFN-
stimulated genes; however, the E1A gene products specifically suppressed this transcription.70,109 Recent
studies also indicated that antimicrobial peptides, such as defensins and cathelicidins, play a role during
AdV infection. Defensins inhibit the infectivity of wild-type AdV via blocking the uncoating events of
AdVs, thus preventing genome exposure and nuclear entry by α-defensin HD5 and HNP1, while catheli-
cidins are upregulated by cytokines induced by AdV, which are chemotactic for neutrophils, monocytes,
and T cells, thus causing the inflammatory infiltrate to be seen during AdV infection.117
REFERENCES
1.
Rubin, B.A. Clinical picture and epidemiology of adenovirus infections (a review). Acta Microbiol Hung
40, 303–323 (1993).
2.
Shenk, T.E. Adenoviridae. in Fields Virology, Vol. 2 (eds. Knipe, D.M. & Howley, P.M.) 2265–2300
(Lippincott Williams & Wilkins, Philadelphia, 2001).
3.
Horwitz, M.S. Adenoviruses. in Fields Virology, Vol. 2 (eds. Knipe, D.M. & Howley, P.M.) 2301–2326
(Lippincott Williams & Wilkins, Philadelphia, 2001).
4.
Ghebremedhin, B. Human adenovirus: viral pathogen with increasing importance. Eur J Microbiol
Immunol (Bp) 4, 26–33 (2014).
5.
Kajon, A.E. et al. Molecular epidemiology of adenovirus type 4 infections in US military recruits in the
postvaccination era (1997–2003). J Infect Dis 196, 67–75 (2007).
6.
Nemerow, G.R., Pache, L., Reddy, V. & Stewart, P.L. Insights into adenovirus host cell interactions from
structural studies. Virology 384, 380–388 (2009).
7.
Hoffman, J.A. Adenoviral disease in pediatric solid organ transplant recipients. Pediatr Transplant 10,
17–25 (2006).
24 Laboratory Models for Foodborne Infections
8. Leen, A.M. & Rooney, C.M. Adenovirus as an emerging pathogen in immunocompromised patients. Br
J Haematol 128, 135–144 (2005).
9. Dey, S.K. et al. Molecular epidemiology of adenovirus infection among infants and children with acute
gastroenteritis in Dhaka City, Bangladesh. Infect Genet Evol 9, 518–522 (2009).
10. Ushijima, H. et al. A study of adenovirus gastroenteritis in the Tokyo area. Eur J Pediatr 147, 90–92 (1988).
11. Jones, M.S., II et al. New adenovirus species found in a patient presenting with gastroenteritis. J Virol
81, 5978–5984 (2007).
12. Matsushima, Y., Shimizu, H., Phan, T.G. & Ushijima, H. Genomic characterization of a novel human
adenovirus type 31 recombinant in the hexon gene. J Gen Virol 92, 2770–2775 (2011).
13. Matsushima, Y. et al. Novel human adenovirus strain, Bangladesh. Emerg Infect Dis 18, 846–848 (2012).
14. Matsushima, Y. et al. Genome sequence of a novel virus of the species human adenovirus D associated
with acute gastroenteritis. Genome Announc 1, e00068-12 (2013).
15. Liu, E.B. et al. Genetic analysis of a novel human adenovirus with a serologically unique hexon and a
recombinant fiber gene. PLoS One 6, e24491 (2011).
16. Wood, D.J. Adenovirus gastroenteritis. Br Med J (Clin Res Ed) 296, 229–230 (1988).
17. Adrian, T., Wigand, R. & Richter, J. Gastroenteritis in infants, associated with a genome type of adeno-
virus 31 and with combined rotavirus and adenovirus 31 infection. Eur J Pediatr 146, 38–40 (1987).
18. Filho, E.P. et al. Adenoviruses associated with acute gastroenteritis in hospitalized and community
children up to 5 years old in Rio de Janeiro and Salvador, Brazil. J Med Microbiol 56, 313–319 (2007).
19. Moyo, S.J. et al. Prevalence and molecular characterisation of human adenovirus in diarrhoeic children
in Tanzania; a case control study. BMC Infect Dis 14, 666 (2014).
20. Magwalivha, M. et al. High prevalence of species D human adenoviruses in fecal specimens from urban
Kenyan children with diarrhea. J Med Virol 82, 77–84 (2010).
21. Ginsberg, H.S. et al. A mouse model for investigating the molecular pathogenesis of adenovirus pneu-
monia. Proc Natl Acad Sci USA 88, 1651–1655 (1991).
22. Segerman, A., Arnberg, N., Erikson, A., Lindman, K. & Wadell, G. There are two different species B
adenovirus receptors: sBAR, common to species B1 and B2 adenoviruses, and sB2AR, exclusively used
by species B2 adenoviruses. J Virol 77, 1157–1162 (2003).
23. Dehghan, S. et al. Five genome sequences of subspecies B1 human adenoviruses associated with acute
respiratory disease. J Virol 86, 635–636 (2012).
24. Dehghan, S. et al. Genomics and bioinformatics of human adenovirus type 68 strain Arg 827/04.
November 15, 2012 edn (National Center for Biotechnology Information, U.S. National Library of
Medicine, Bethesda, MD, 2012).
25. De Jong, J.C. et al. Adenoviruses from human immunodeficiency virus-infected individuals, including
two strains that represent new candidate serotypes Ad50 and Ad51 of species B1 and D, respectively.
J Clin Microbiol 37, 3940–3945 (1999).
26. Yang, Z. et al. Genomic analyses of recombinant adenovirus type 11a in China. J Clin Microbiol 47,
3082–3090 (2009).
27. Lu, Q.B. et al. Epidemiology of human adenovirus and molecular characterization of human adenovirus
55 in China, 2009–2012. Influenza Other Respir Viruses 8, 302–308 (2014).
28. Walsh, M.P. et al. Computational analysis of two species C human adenoviruses provides evidence of a
novel virus. J Clin Microbiol 49, 3482–3490 (2011).
29. Robinson, C.M. et al. Computational analysis and identification of an emergent human adenovirus
pathogen implicated in a respiratory fatality. Virology 409, 141–147 (2011).
30. Singh, G. et al. Homologous recombination in E3 genes of human adenovirus species D. J Virol 87,
12481–12488 (2013).
31. Kaneko, H. et al. Recombination analysis of intermediate human adenovirus type 53 in Japan by com-
plete genome sequence. J Gen Virol 92, 1251–1259 (2011).
32. Alissa Alkhalaf, M., Al Qurashi, Y.M., Guiver, M. & Cooper, R.J. Genome sequences of three species
D adenoviruses isolated from AIDS patients. Genome Announc 2(1), e01267-13 (2014).
33. Singh, G. et al. Overreliance on the hexon gene, leading to misclassification of human adenoviruses. J
Virol 86, 4693–4695 (2012).
34. Robinson, C.M. et al. Genomics and bioinformatics of human adenovirus type 19 Pittsburgh strain
88. February 19, 2012 edn (National Center for Biotechnology Information, U.S. National Library of
Medicine, Bethesda, MD, 2012).
Adenoviruses 25
35. Robinson, C.M. et al. Predicting the next eye pathogen: analysis of a novel adenovirus. mBio 4,
e00595–12 (2013).
36. Liu, E.B. et al. Computational and serologic analysis of novel and known viruses in species human
adenovirus D in which serology and genomics do not correlate. PLoS One 7, e33212 (2012).
37. Russell, W.C. Adenoviruses: update on structure and function. J Gen Virol 90, 1–20 (2009).
38. Horwitz, M.S. Function of adenovirus E3 proteins and their interactions with immunoregulatory cell
proteins. J Gene Med 6 (Suppl 1), S172–S183 (2004).
39. Robinson, C.M., Shariati, F., Gillaspy, A.F., Dyer, D.W. & Chodosh, J. Genomic and bioinformatics
analysis of human adenovirus type 37: new insights into corneal tropism. BMC Genomics 9, 213 (2008).
40. Lauer, K.P. et al. Natural variation among human adenoviruses: genome sequence and annotation of
human adenovirus serotype 1. J Gen Virol 85, 2615–2625 (2004).
41. Davison, A.J., Benko, M. & Harrach, B. Genetic content and evolution of adenoviruses. J Gen Virol 84,
2895–2908 (2003).
42. Windheim, M., Hilgendorf, A. & Burgert, H.G. Immune evasion by adenovirus E3 proteins: exploitation
of intracellular trafficking pathways. Curr Top Microbiol Immunol 273, 29–85 (2004).
43. Leppard, K.N. E4 gene function in adenovirus, adenovirus vector and adeno-associated virus infec-
tions. J Gen Virol 78 ( Pt 9), 2131–2138 (1997).
44. Tollefson, A.E., Ying, B., Doronin, K., Sidor, P.D. & Wold, W.S. Identification of a new human adeno-
virus protein encoded by a novel late l-strand transcription unit. J Virol 81, 12918–12926 (2007).
45. San Martin, C. Latest insights on adenovirus structure and assembly. Viruses 4, 847–877 (2012).
46. Roberts, M.M., White, J.L., Grutter, M.G. & Burnett, R.M. Three-dimensional structure of the adeno-
virus major coat protein hexon. Science 232, 1148–1151 (1986).
47. Liu, H. et al. Atomic structure of human adenovirus by cryo-EM reveals interactions among protein
networks. Science 329, 1038–1043 (2010).
48. Ma, H.C. & Hearing, P. Adenovirus structural protein IIIa is involved in the serotype specificity of viral
DNA packaging. J Virol 85, 7849–7855 (2011).
49. Wodrich, H. et al. Switch from capsid protein import to adenovirus assembly by cleavage of nuclear
transport signals. EMBO J 22, 6245–6255 (2003).
50. Maier, O., Galan, D.L., Wodrich, H. & Wiethoff, C.M. An N-terminal domain of adenovirus protein VI
fragments membranes by inducing positive membrane curvature. Virology 402, 11–19 (2010).
51. Schreiner, S. et al. Transcriptional activation of the adenoviral genome is mediated by capsid protein VI.
PLoS Pathog 8, e1002549 (2012).
52. Parks, R.J. Adenovirus protein IX: a new look at an old protein. Mol Ther 11, 19–25 (2005).
53. Rosa-Calatrava, M., Grave, L., Puvion-Dutilleul, F., Chatton, B. & Kedinger, C. Functional analysis
of adenovirus protein IX identifies domains involved in capsid stability, transcriptional activity, and
nuclear reorganization. J Virol 75, 7131–7141 (2001).
54. Strunze, S. et al. Kinesin-1-mediated capsid disassembly and disruption of the nuclear pore complex
promote virus infection. Cell Host Microbe 10, 210–223 (2011).
55. Veesler, D. et al. Single-particle EM reveals plasticity of interactions between the adenovirus penton
base and integrin αVβ3. Proc Natl Acad Sci USA 111, 8815–8819 (2014).
56. Zubieta, C., Schoehn, G., Chroboczek, J. & Cusack, S. The structure of the human adenovirus 2 penton.
Mol Cell 17, 121–135 (2005).
57. Seiradake, E. & Cusack, S. Crystal structure of enteric adenovirus serotype 41 short fiber head. J Virol
79, 14088–14094 (2005).
58. Ugai, H. et al. Adenoviral protein V promotes a process of viral assembly through nucleophosmin 1.
Virology 432, 283–295 (2012).
59. Matthews, D.A. Adenovirus protein V induces redistribution of nucleolin and B23 from nucleolus to
cytoplasm. J Virol 75, 1031–1038 (2001).
60. Hindley, C.E., Lawrence, F.J. & Matthews, D.A. A role for transportin in the nuclear import of adeno-
virus core proteins and DNA. Traffic 8, 1313–1322 (2007).
61. Lee, T.W., Blair, G.E. & Matthews, D.A. Adenovirus core protein VII contains distinct sequences that
mediate targeting to the nucleus and nucleolus, and colocalization with human chromosomes. J Gen
Virol 84, 3423–3428 (2003).
62. Ahi, Y.S., Vemula, S.V. & Mittal, S.K. Adenoviral E2 IVa2 protein interacts with L4 33K protein and
E2 DNA-binding protein. J Gen Virol 94, 1325–1334 (2013).
26 Laboratory Models for Foodborne Infections
63. Yang, T.C. & Maluf, N.K. Characterization of the non-specific DNA binding properties of the adenovi-
ral IVa2 protein. Biophys Chem 193–194, 1–8 (2014).
64. Ewing, S.G., Byrd, S.A., Christensen, J.B., Tyler, R.E. & Imperiale, M.J. Ternary complex formation on the
adenovirus packaging sequence by the IVa2 and L4 22-kilodalton proteins. J Virol 81, 12450–12457 (2007).
65. Lee, T.W. et al. Precursor of human adenovirus core polypeptide Mu targets the nucleolus and modu-
lates the expression of E2 proteins. J Gen Virol 85, 185–196 (2004).
66. Ugai, H., Borovjagin, A.V., Le, L.P., Wang, M. & Curiel, D.T. Thermostability/infectivity defect caused
by deletion of the core protein V gene in human adenovirus type 5 is rescued by thermo-selectable muta-
tions in the core protein X precursor. J Mol Biol 366, 1142–1160 (2007).
67. Ruben, M., Bacchetti, S. & Graham, F. Covalently closed circles of adenovirus 5 DNA. Nature 301,
172–174 (1983).
68. Webster, A., Leith, I.R., Nicholson, J., Hounsell, J. & Hay, R.T. Role of preterminal protein processing
in adenovirus replication. J Virol 71, 6381–6389 (1997).
69. Pronk, R. & van der Vliet, P.C. The adenovirus terminal protein influences binding of replication pro-
teins and changes the origin structure. Nucleic Acids Res 21, 2293–2300 (1993).
70. Joung, I., Angeletti, P.C. & Engler, J.A. Functional implications in apoptosis by interferon inducible gene
product 1-8D, the binding protein to adenovirus preterminal protein. J Microbiol 41, 295–299 (2003).
71. de Jong, R.N., Meijer, L.A. & van der Vliet, P.C. DNA binding properties of the adenovirus DNA repli-
cation priming protein pTP. Nucleic Acids Res 31, 3274–3286 (2003).
72. Botting, C.H. & Hay, R.T. Role of conserved residues in the activity of adenovirus preterminal protein.
J Gen Virol 82, 1917–1927 (2001).
73. Grosche, P. et al. Structure-based design and optimization of potent inhibitors of the adenoviral prote-
ase. Bioorg Med Chem Lett 25, 438–443 (2015).
74. Mangel, W.F., Baniecki, M.L. & McGrath, W.J. Specific interactions of the adenovirus proteinase with
the viral DNA, an 11-amino-acid viral peptide, and the cellular protein actin. Cell Mol Life Sci 60,
2347–2355 (2003).
75. Baniecki, M.L., McGrath, W.J. & Mangel, W.F. Regulation of a viral proteinase by a peptide and DNA
in one-dimensional space: III. Atomic resolution structure of the nascent form of the adenovirus pro-
teinase. J Biol Chem 288, 2081–2091 (2013).
76. Blainey, P.C. et al. Regulation of a viral proteinase by a peptide and DNA in one-dimensional space: IV.
Viral proteinase slides along DNA to locate and process its substrates. J Biol Chem 288, 2092–2102 (2013).
77. Graziano, V. et al. Regulation of a viral proteinase by a peptide and DNA in one-dimensional space: II.
Adenovirus proteinase is activated in an unusual one-dimensional biochemical reaction. J Biol Chem
288, 2068–2080 (2013).
78. Graziano, V. et al. Regulation of a viral proteinase by a peptide and DNA in one-dimensional space: I.
Binding to DNA AND to hexon of the precursor to protein VI, pVI, of human adenovirus. J Biol Chem
288, 2059–2067 (2013).
79. Radko, S. et al. Adenovirus E1A targets the DREF nuclear factor to regulate virus gene expression,
DNA replication, and growth. J Virol 88, 13469–13481 (2014).
80. Padmanabhan, R., Tanimoto, A. & Sasaguri, Y. Transactivation of human cdc2 promoter by adenovirus
E1A. Curr Top Microbiol Immunol 272, 365–397 (2003).
81. Berscheminski, J., Groitl, P., Dobner, T., Wimmer, P. & Schreiner, S. The adenoviral oncogene E1A-13S
interacts with a specific isoform of the tumor suppressor PML to enhance viral transcription. J Virol 87,
965–977 (2013).
82. Gallimore, P.H. & Turnell, A.S. Adenovirus E1A: remodelling the host cell, a life or death experience.
Oncogene 20, 7824–7835 (2001).
83. Blackford, A.N. & Grand, R.J. Adenovirus E1B 55-kilodalton protein: multiple roles in viral infection
and cell transformation. J Virol 83, 4000–4012 (2009).
84. Cheng, C.Y. et al. Role of E1B55K in E4orf6/E1B55K E3 ligase complexes formed by different human
adenovirus serotypes. J Virol 87, 6232–6245 (2013).
85. Tarakanova, V.L. & Wold, W.S. Adenovirus E1A and E1B-19K proteins protect human hepatoma cells
from transforming growth factor β1-induced apoptosis. Virus Res 147, 67–76 (2010).
86. Chiou, S.K., Tseng, C.C., Rao, L. & White, E. Functional complementation of the adenovirus E1B
19-kilodalton protein with Bcl-2 in the inhibition of apoptosis in infected cells. J Virol 68, 6553–6566
(1994).
Adenoviruses 27
87. van Breukelen, B., Kanellopoulos, P.N., Tucker, P.A. & van der Vliet, P.C. The formation of a flexible
DNA-binding protein chain is required for efficient DNA unwinding and adenovirus DNA chain elonga-
tion. J Biol Chem 275, 40897–40903 (2000).
88. Dekker, J. et al. Multimerization of the adenovirus DNA-binding protein is the driving force for ATP-
independent DNA unwinding during strand displacement synthesis. EMBO J 16, 1455–1463 (1997).
89. de Jong, R.N., van der Vliet, P.C. & Brenkman, A.B. Adenovirus DNA replication: protein priming,
jumping back and the role of the DNA binding protein DBP. Curr Top Microbiol Immunol 272,
187–211 (2003).
90. de Jong, R.N. & van der Vliet, P.C. Mechanism of DNA replication in eukaryotic cells: cellular host
factors stimulating adenovirus DNA replication. Gene 236, 1–12 (1999).
91. Brenkman, A.B., Breure, E.C. & van der Vliet, P.C. Molecular architecture of adenovirus DNA poly-
merase and location of the protein primer. J Virol 76, 8200–8207 (2002).
92. Hoeben, R.C. & Uil, T.G. Adenovirus DNA replication. Cold Spring Harb Perspect Biol 5, a013003 (2013).
93. Signas, C., Akusjarvi, G. & Pettersson, U. Region E3 of human adenoviruses: differences between the
oncogenic adenovirus-3 and the non-oncogenic adenovirus-2. Gene 50, 173–184 (1986).
94. Tollefson, A.E. et al. The adenovirus death protein (E3–11.6K) is required at very late stages of infection
for efficient cell lysis and release of adenovirus from infected cells. J Virol 70, 2296–2306 (1996).
95. Johnson, D.G., Ohtani, K. & Nevins, J.R. Autoregulatory control of E2F1 expression in response to posi-
tive and negative regulators of cell cycle progression. Genes Dev 8, 1514–1525 (1994).
96. Zhang, W., Low, J.A., Christensen, J.B. & Imperiale, M.J. Role for the adenovirus IVa2 protein in pack-
aging of viral DNA. J Virol 75, 10446–10454 (2001).
97. Lutz, P., Rosa-Calatrava, M. & Kedinger, C. The product of the adenovirus intermediate gene IX is a
transcriptional activator. J Virol 71, 5102–5109 (1997).
98. Hayes, B.W., Telling, G.C., Myat, M.M., Williams, J.F. & Flint, S.J. The adenovirus L4 100-kilodalton
protein is necessary for efficient translation of viral late mRNA species. J Virol 64, 2732–2742 (1990).
99. Riley, D. & Flint, S.J. RNA-binding properties of a translational activator, the adenovirus L4
100-kilodalton protein. J Virol 67, 3586–3595 (1993).
100. Hong, S.S. et al. The 100K-chaperone protein from adenovirus serotype 2 (subgroup C) assists in
trimerization and nuclear localization of hexons from subgroups C and B adenoviruses. J Mol Biol 352,
125–138 (2005).
101. Koyuncu, O.O. & Dobner, T. Arginine methylation of human adenovirus type 5 L4 100-kilodalton pro-
tein is required for efficient virus production. J Virol 83, 4778–4790 (2009).
102. Andrade, F., Casciola-Rosen, L.A. & Rosen, A. A novel domain in adenovirus L4-100K is required for
stable binding and efficient inhibition of human granzyme B: possible interaction with a species-specific
exosite. Mol Cell Biol 23, 6315–6326 (2003).
103. Ying, B., Tollefson, A.E. & Wold, W.S. Identification of a previously unrecognized promoter that drives
expression of the UXP transcription unit in the human adenovirus type 5 genome. J Virol 84, 11470–
11478 (2010).
104. Cupelli, K. & Stehle, T. Viral attachment strategies: the many faces of adenoviruses. Curr Opin Virol 1,
84–91 (2011).
105. Ostapchuk, P. & Hearing, P. Control of adenovirus packaging. J Cell Biochem 96, 25–35 (2005).
106. Carter, M.J. Enterically infecting viruses: pathogenicity, transmission and significance for food and
waterborne infection. J Appl Microbiol 98, 1354–1380 (2005).
107. Mavromichalis, J. et al. Intestinal damage in rotavirus and adenovirus gastroenteritis assessed by
d-xylose malabsorption. Arch Dis Child 52, 589–591 (1977).
108. Hayder, H. & Mullbacher, A. Molecular basis of immune evasion strategies by adenoviruses. Immunol
Cell Biol 74, 504–512 (1996).
109. Gutch, M.J. & Reich, N.C. Repression of the interferon signal transduction pathway by the adenovirus
E1A oncogene. Proc Natl Acad Sci USA 88, 7913–7917 (1991).
110. Arany, Z., Newsome, D., Oldread, E., Livingston, D.M. & Eckner, R. A family of transcriptional adaptor
proteins targeted by the E1A oncoprotein. Nature 374, 81–84 (1995).
111. Janaswami, P.M., Kalvakolanu, D.V., Zhang, Y. & Sen, G.C. Transcriptional repression of interleukin-6
gene by adenoviral E1A proteins. J Biol Chem 267, 24886–24891 (1992).
112. White, E. et al. The 19-kilodalton adenovirus E1B transforming protein inhibits programmed cell death
and prevents cytolysis by tumor necrosis factor α. Mol Cell Biol 12, 2570–2580 (1992).
28 Laboratory Models for Foodborne Infections
113. Lázaro, S., Gamarra, D. & Del Val, M. Proteolytic enzymes involved in MHC class I antigen processing:
a guerrilla army that partners with the proteasome. Mol Immunol 68, 72–76 (2015).
114. Schouten, G.J., van der Eb, A.J. & Zantema, A. Downregulation of MHC class I expression due to inter-
ference with p105-NFκB1 processing by Ad12E1A. EMBO J 14, 1498–1507 (1995).
115. Kotha, P.L. et al. Adenovirus entry from the apical surface of polarized epithelia is facilitated by the
host innate immune response. PLoS Pathog 11, e1004696 (2015).
116. Coutelier, J.P., Van Broeck, J. & Wolf, S.F. Interleukin-12 gene expression after viral infection in the
mouse. J Virol 69, 1955–1958 (1995).
117. Gregory, S.M., Nazir, S.A. & Metcalf, J.P. Implications of the innate immune response to adenovirus
and adenoviral vectors. Future Virol 6, 357–374 (2011).
118. Johansson, M.E., Wirgart, B.Z., Grillner, L. & Bjork, O. Severe gastroenteritis in an immunocompro-
mised child caused by adenovirus type 5. Pediatr Infect Dis J 9, 449–450 (1990).
119. Schofield, K.P., Morris, D.J., Bailey, A.S., de Jong, J.C. & Corbitt, G. Gastroenteritis due to adenovirus
type 41 in an adult with chronic lymphocytic leukemia. Clin Infect Dis 19, 311–312 (1994).
120. Ishiko, H. et al. Novel human adenovirus causing nosocomial epidemic keratoconjunctivitis. J Clin
Microbiol 46, 2002–2008 (2008).
3
Astrovirus
CONTENTS
3.1 Introduction..................................................................................................................................... 29
3.2 Life Cycle.........................................................................................................................................31
3.3 Pathogenesis.................................................................................................................................... 32
3.4 Immunity......................................................................................................................................... 34
3.5 Future Perspectives......................................................................................................................... 35
References................................................................................................................................................. 35
3.1 I ntroduction
Astroviruses are small, round, nonenveloped, positive-sense single-stranded RNA viruses. They were
named for their “star-like” appearance when visualized by negative staining electron microscopy
in the feces of infants suffering from gastroenteritis.1,2 Since their original discovery, astroviruses
have been identified in almost all animal species examined.3 In the vast majority of these species,
astrovirus infection is transmitted through the fecal–oral route and causes acute gastroenteritis. In
humans, the classical astroviruses are associated with mild to moderate diarrhea and are recognized
as one of the leading causes of enteritis in children and the immunocompromised.4,5 More recently,
astroviruses have been associated with encephalitis.6,7 In other species, especially poultry, diarrhea
can be severe and can involve other organ systems including the liver and kidneys.8,9 Historically,
astroviruses have been thought to be species-specific, with only limited evidence of interspecies
transmission.10 However, there is increasing evidence that astroviruses can infect multiple species.
Convincing evidence for this was first found for avian astroviruses,11–17 but recent reports suggest the
same may be true for mammalian astroviruses,18–20 and that there may even be transmission between
birds and mammals.21 More recently, astrovirus genotypes associated with human infections were
detected in nonhuman primates, strongly supporting the suggestion that astroviruses can cross spe-
cies barriers.21a
Since the Astroviridae family was established by the ICTV in 1995, there have been considerable
changes in our understanding of the diversity, molecular evolution, and host range of the family.
In just the last decade, the numbers of host species detected with astroviruses have increased more
than fourfold.22 Additionally our appreciation of the diversity of astrovirus genotypes capable of
infecting a given host has led to a reevaluation of their classification system. Currently, Astroviridae
is divided into two genera (Table 3.1), Mamastrovirus (MAstV) and Avastrovirus (AAstV), repre-
senting viruses that affect mammals and avian species, respectively. Within each of these two gen-
era, viruses are classified by distinct genotypic differences within their polymerase and/or capsid
genomic sequences.23
Based on this nomenclature system, there are 19 species of MAstV recognized to infect humans
(MAstV 1–19). Of these species, MAstV 1 encompasses the human astroviruses (HAstVs) first rec-
ognized and most extensively studied.22 This group of classical HAstVs contains eight distinct serotypes
(HAstV-1 to HAstV-8), which are known to have a worldwide distribution and are recognized as one of
29
30 Laboratory Models for Foodborne Infections
TABLE 3.1
Astroviridae
Genus Species Host(s)
Mamastrovirus (MAstV) MAstV 1 Human
MAstV 2 Cat, cheetah
MAstV 3 Pig
MAstV 4 California sea lion
MAstV 5 Dog
MAstV 6 Human
MAstV 7 Bottlenose dolphin
MAstV 8 Human
MAstV 9 Human
MAstV 10 Mink
MAstV 11 California sea lion
MAstV 12 Bat
MAstV 13 Sheep
MAstV 14 Bat
MAstV 15 Bat
MAstV 16 Bat
MAstV 17 Bat
MAstV 18 Bat
MAstV 19 Bat
Unassigned Pig, sheep, mouse, rat, cattle, deer
Avastrovirus (AAstV) AAvstV 1 Turkey, chicken
AAvstV 2 Turkey, guinea fowl, duck
AAvstV 3 Chicken, turkey
Unassigned Dove, pigeon, heron, pintail
Environmental
Swimming/bathing
Fresh or saltwater
Fomites
Direct fecal–oral
Contaminated
Drinking water
Fruits and vegetables
Bivalves
Food Water systems
Freshwater
Groundwater
Water Saltwater
FIGURE 3.1 Direct and indirect routes of astrovirus transmission. As the cause of gastroenteritis, the direct fecal–oral
transmission of astroviruses is well established, as is its ability to be transmitted by fomites; however, astroviruses are
environmentally very stable, allowing them to be transmitted indirectly to new hosts via water and the environment.
Astrovirus 31
the top three causes of diarrhea in children under 2 years of age.24–26 The other HAstVs found in MAstV
were identified through pathogen discovery surveys of fecal material.27–32
Astroviruses are transmitted through the fecal–oral route (Figure 3.1), as demonstrated by several
volunteer studies.33,34 This direct transmission of fecal material from an infected person to a new host, or
through fomites is thought to be the major mechanism for the spread of astroviruses through a popula-
tion; however, astroviruses are highly stable in the environment.35,36 This stability allows them to spread
through wastewater, groundwater, saltwater, and even drinking water.37–45 These astrovirus-contaminated
waters can then infect a new patient following drinking, eating fruits and vegetables treated with these
waters,46 consuming bivalves harvested from these waters,38,39,47 or even bathing, swimming, or surfing
in astrovirus-contaminated waters.48,49 Given this link between astrovirus stability and water, it is espe-
cially important to recognize that most municipal water treatment systems do not filter out, or inactivate
astroviruses as part of their process, as studies have demonstrated by isolating astroviruses from house-
hold tap water.4,50,51
(A)
(B)
ORF1a ORF1b ORF2
FIGURE 3.2 Model of astrovirus virion (A) and diagram of astrovirus genome (B). The astrovirus virion is a nonenvel-
oped capsid ranging in size from 28 to 41 nm depending on the species and propagation conditions. Studies of HAstV-8 by
Dryden et al. suggest that astroviruses are released as immature virions, which become highly infectious mature virions
following digestion by trypsin.112 The mature virion surface is comprised of multiple spike-like projections (dark gray,
VP27 and VP25), extending from the shell (light gray, VP34), giving the virus its distinctive five- or six-pointed star-like
appearance when visualized under negative staining electron microscopy. Inside the capsid, the astrovirus genome is com-
prised of single-stranded, positive-sense RNA ranging from 6.4 to 7.7 kb in size. The genome contains three open reading
frames: ORF1a encodes the nonstructural proteins; ORF1b encodes the RNA-dependent RNA polymerase; and ORF2
encodes the capsid protein. The retrovirus-like frameshift structure formed between ORF1a and ORF1b allows these two
ORFs to be translated as one polyprotein and is one of the molecular hallmarks that distinguishes the astrovirus replication
strategy from other small, round RNA viruses.
32 Laboratory Models for Foodborne Infections
establish replication complex vesicles.59 As part of this process, the virus also produces a subgenomic
RNA encoding the viral capsid protein (ORF2) (Figure 3.2).60 The subgenomic message is then used
to produce ample copies of the virus structural protein, while the genomic RNA is used to produce
more nsPs and RdRp, as well as a template for the progeny genome.58 Once enough capsid and progeny
genomes have been produced, the daughter viruses are assembled. This initial assembly step involves the
full 90-kDa capsid protein, which is then digested by host caspases to yield progeny viruses made up of
70-kDa capsid proteins.55,61–63 These immature progeny viruses are then released from the infected cell,
where they will be further digested by host trypsin-like proteases, activating them and allowing them to
infect new cells.52–56
3.3 Pathogenesis
Classical HAstVs cause gastrointestinal disease in young children, aged adults, and the immunocom-
promised,5,64–68 but are only rarely associated with the disease in normal healthy adults.33,69,70 Molecular
epidemiology and serology studies suggest that humans are exposed to HAstVs regularly and antibodies
against HAstVs develop early in life.22 In fact, preexisting anti-HAstV antibody levels seem to explain the
biphasic age distribution of HAstV infections, as well as the sporadic development of HAstV-mediated
diarrhea during adult volunteer studies.54,71,72
In susceptible people, following an incubation period of 4–5 days,73 HAstV infection leads to mild,
watery diarrhea, lasting 48–72 h.5,22 Clinical symptoms can also include vomiting, fever, and abdominal
pain. Overall, astrovirus-induced disease is generally less severe than that caused by rotavirus or calici-
virus infections, and rarely requires hospitalization in otherwise healthy people.5,22
While HAstVs are regarded as a disease of the intestinal tract, recent evidence suggests that they
can induce viremia, potentially causing more severe infections in immunocompromised patients.74–77
This concept of extraintestinal astrovirus was first reported in studies examining the pathology of avian
astroviruses,12,74,78–80 and was more recently demonstrated in a newly established immunodeficient mouse
model of astrovirus infection81 as well as in immunocompetent mice orally inoculated with murine astro-
virus (unpublished observations).
Despite the well-established disease burden caused by HAstVs, the mechanisms involved in astrovirus-
mediated disease in humans are still poorly understood. Much of what has been reported regarding the
pathophysiological changes induced by astrovirus infection has come from animal models; however, the
changes noted and the cell types involved appear to vary considerably between models.
Studies using experimentally infected lambs reported atrophy of the villi of the jejunum and ileum,
with astroviruses found in the mature enterocytes.82 In astrovirus-infected calves, the virus was found
in the M cells and enterocytes associated with Peyer’s patches in the jejunum and ileum.83 In ducks and
chickens, astroviruses are more commonly associated with other tissues, causing fetal hepatitis in ducks
and mild nephritis in chickens.12,78,84,85 The young turkey model, however, has been the most extensively
used to understand astrovirus pathogenesis.74,79,86–92 These studies demonstrate that astrovirus infection
leads to very few, if any, remarkable changes in the intestinal histology. No significant inflammation, flux
of immune cells, or changes in the villus height, width, or surface area were observed.74,79,88,91 While not
wholly unique to astroviruses, the absence of histological changes correlating with the onset of diarrhea
is uncommon among diarrhea-inducing agents. These observations in turkeys have been supported by
histological analysis of small intestine biopsy samples from an immunosuppressed child suffering from
persistent diarrhea.93
The lack of histological changes consistent with the severity of diarrhea observed has led to a more
specific examination of the changes in intestinal function to determine how astroviruses cause diseases.
These studies have demonstrated decreased expression of digestive enzymes, specifically maltase and
sucrose, in the brush border of the small intestine, suggesting the development of malabsorption of
carbohydrates, leading to osmotic diarrhea (Figures 3.3 and 3.4).91,92 More recent studies suggest that
astrovirus infection leads to a rearrangement of actin in intestinal epithelial cells.88,94 These changes
were associated with decreases in the electrical resistance (decreased barrier function) in the jejunum
that allowed for increased movement of ions but not larger macromolecules (Figure 3.3).88 In addition,
Astrovirus 33
+ +
- + -
+- +
- + -
-
+
-
+
-
+
+
-
+
-
-
+
+
+
-
-
+
-
+
-
FIGURE 3.3 Astrovirus infection and exposure to astrovirus capsid protein lead to changes in intestinal barrier function.
Under normal conditions, the tight junctions (gray boxes) between neighboring cells form water-tight barriers, are held
in place and regulated by actin (red lines). Following astrovirus replication, or even just on exposure to astrovirus capsid
protein, the actin at the apical ends of epithelial cells is rearranged. The more significant this rearrangement is, the greater
the change in barrier function. Smaller changes can allow for the diffusion of solutes and ions across the epithelium, while
still larger disruptions allow diffusion of macromolecules.
FIGURE 3.4 Astrovirus-induced changes in the apical expression of digestive enzymes and exchangers. Following infec-
tion or exposure to astrovirus capsid protein, the apical expression of digestive enzymes such as maltase and the NHE-3
have been reported to be significantly decreased, resulting in malabsorption and osmotic diarrhea. While the specific
mechanism(s) involved is not known, it is likely that the astrovirus-induced disruption to actin (red lines) prevents the
normal recycling of vesicles and proteins whose function is regulated by a near-constant shuttling in and out of the brush
border, and they are more dramatically affected by astrovirus-induced actin rearrangement.
34 Laboratory Models for Foodborne Infections
there was a decrease in the absorption of sodium ions that correlated with decreased apical expression
of the sodium/hydrogen exchanger-3 (NHE-3) molecule in the jejunum.88 Collectively, these findings
suggested an infection established by sodium malabsorption, leading to osmotic diarrhea. The changes
in the turkey jejunum were similar, but not completely consistent with studies of HAstV in vitro where
viral infection as well as exposure to inactivated HAstV capsid was sufficient to induce actin rearrange-
ment leading to tight-junction disruption, allowing macromolecules to diffuse across the barrier.88,94 The
observation that the capsid protein alone is capable of inducing barrier changes in vitro suggests that it
possesses enterotoxin-like activity and could be capable of inducing even diarrhea. Current experiments
using turkey,94a as well as other animal models (unpublished observation), have confirmed this hypoth-
esis, but they also suggest that the potential impact a nonhuman astrovirus may have on a human disease
could be greatly underappreciated if replication is not required to induce clinical signs.95
3.4 I mmunity
Epidemiological evidence suggests that immunity to HAstV is largely mediated through antibodies.
Clinical signs of astrovirus infection are most pronounced, if evident at all, in those with no preexisting
antibodies or those who are immunocompromised due to age, HIV status, or on immunosuppressive
therapy.67,71,72 The levels of anti-HAstV antibodies observed in normal healthy adults, in some studies,
suggest that we may be under frequent exposure to these pathogens.33,34,96 In addition to antibodies,
anti-HAstV-specific T cells have been recovered from intestinal biopsy samples from a normal healthy
adult97,98 suggesting that both the cellular and humoral arms of the adaptive immune system respond to
HAstV infections. The relative importance of each in clearing the pathogen and preventing subsequent
infection is still unknown.
Given that astrovirus infection is most severe when the adaptive immune system is least active, many
researchers have focused on the role of the innate immune response to understand how the body fights
the infection under these conditions. Most interestingly, recent studies by Guix et al. have demonstrated
that HAstVs can inhibit the production of type I interferon in the early phase of replication allowing for
greater viral production. This effect seems to correlate with mutations in one of the mature nsP proteins
(nsP1a/4), suggesting that some strains and some genotypes may have different abilities to modulate the
interferon response.99
This is not the only evidence that astroviruses are able to circumvent the innate immune response. The
astrovirus capsid protein can inhibit activation of the lectin and classical complement pathways.100–103
Amino acids in the N-terminal region of the capsid interact with the C1 and MBL molecules, prevent-
ing the formation of C3 convertase and subsequent cleavage of C3 into C3a and C3b. The blockage of
these complement activation pathways prevents the formation of a very powerful pro-inflammatory and
immune-activating signal,71 one that host cells use to detect the presence of numerous RNA, DNA, and
bacterial infections.104 The specific role the inhibition of the complement pathway serves in astrovirus
pathology is unclear, but it is easy to speculate that it helps to mute the inflammatory signal and is at least
partially responsible for the astrovirus’s immune evasion in vivo.71
As with astrovirus pathogenesis, most of our understanding of the immune response to astroviruses
in vivo comes from the turkey model. In this model, however, the adaptive immune response seems to play
little to no role in controlling the infection,79,86 and in fact some researchers have suggested that it induces
an immune dysfunction that can persist long after the virus has cleared.105–107 Macrophages isolated from
astrovirus-infected turkeys demonstrate reduced phagocytosis and the ability to kill engulfed bacteria
in ex vivo assays, along with the reduced production of pro-inflammatory cytokines.108–111 Lymphocytes
from astrovirus-infected turkeys also demonstrated reduced proliferation in response to mitogenic stim-
ulation. The apparent inhibition of these immune cells could be a response to increased levels of active
TGFβ in the serum of infected turkeys.79 The mechanism leading to the activation of TGFβ in these
animals is unclear but given its role as a potent anti-inflammatory cytokine, it is likely key to the limited
role the adaptive system plays in response to astrovirus infection.
In spite of numerous immune factors being downregulated following astrovirus infection, the treat-
ment of avian macrophages with astrovirus capsid was seen to stimulate the expression of inducible
Astrovirus 35
nitric oxide synthase (iNOS) and its antimicrobial mediator NO.86 The modulation of this response in an
in ovo culture system demonstrated that the iNOS response could modulate virus replication; however,
there was no evidence that macrophages responded to the sites of infection.79,86,88 Subsequent studies
demonstrated that the intestinal epithelial cells can and do express iNOS following astrovirus infection
suggesting that in the absence of professional immune cells the intestinal cells are capable of mounting
their own response.87
REFERENCES
1. Appleton, H. & Higgins, P.G. Letter: viruses and gastroenteritis in infants. Lancet 1, 1297 (1975).
2. Madeley, C.R. & Cosgrove, B.P. Letter: 28 nm particles in faeces in infantile gastroenteritis. Lancet 2,
451–452 (1975).
3. De Benedictis, P., Schultz-Cherry, S., Burnham, A. & Cattoli, G. Astrovirus infections in humans and
animals—molecular biology, genetic diversity, and interspecies transmissions. Infect Genet Evol 11,
1529–1544 (2011).
4. Bosch, A., Guix, S. & Pintó, R. Epidemiology of human astroviruses. in Astrovirus Research (ed.
Schultz-Cherry, S.) 1–18 (Springer, New York, 2013).
5. Meliopoulos, V. & Schultz-Cherry, S. Astrovirus pathogenesis. in Astrovirus Research (ed. Schultz-
Cherry, S.) 65–77 (Springer, New York, 2013).
6. Naccache, S.N. et al. Diagnosis of neuroinvasive astrovirus infection in an immunocompromised adult
with encephalitis by unbiased next-generation sequencing. Clin Infect Dis 60, 919–923 (2015).
7. Quan, P.L. et al. Astrovirus encephalitis in boy with X-linked agammaglobulinemia. Emerg Infect Dis
16, 918–925 (2010).
8. Cattoli, G., Chu, D. & Peiris, M. Astrovirus infections in animal mammalian species. in Astrovirus
Research (ed. Schultz-Cherry, S.) 135–149 (Springer, New York, 2013).
9. Pantin-Jackwood, M., Todd, D. & Koci, M. Avian astroviruses. in Astrovirus Research (ed. Schultz-
Cherry, S.) 151–180 (Springer, New York, 2013).
10. Lukashov, V.V. & Goudsmit, J. Evolutionary relationships among Astroviridae. J Gen Virol 83, 1397–
1405 (2002).
11. Cattoli, G. et al. Co-circulation of distinct genetic lineages of astroviruses in turkeys and guinea fowl.
Arch Virol 152, 595–602 (2007).
12. Imada, T. et al. Avian nephritis virus (ANV) as a new member of the family Astroviridae and construc-
tion of infectious ANV cDNA. J Virol 74, 8487–8493 (2000).
13. Todd, D. et al. Capsid protein sequence diversity of avian nephritis virus. Avian Pathol 40, 249–259
(2011).
14. Toffan, A. et al. Experimental infection of poults and guinea fowl with genetically distinct avian astro-
viruses. Avian Pathol 41, 429–435 (2012).
36 Laboratory Models for Foodborne Infections
15. Zhao, W. et al. Sequence analyses of the representative Chinese-prevalent strain of avian nephritis virus
in healthy chicken flocks. Avian Dis 55, 65–69 (2011).
16. Zhao, W. et al. Complete sequence and genetic characterization of pigeon avian nephritis virus, a mem-
ber of the family Astroviridae. Arch Virol 156, 1559–1565 (2011).
17. Zhao, W. et al. Detection of astrovirus infection in pigeons (Columbia livia) during an outbreak of diar-
rhoea. Avian Pathol 40, 361–365 (2011).
18. Mendenhall, I.H., Smith, G.J. & Vijaykrishna, D. Ecological drivers of virus evolution: astrovirus as a
case study. J Virol 89, 6978–6981 (2015).
19. Monini, M. et al. Detection and molecular characterization of zoonotic viruses in swine fecal samples
in Italian pig herds. Arch Virol 160, 2547–2556 (2015).
20. Nagai, M. et al. Full genome analysis of bovine astrovirus from fecal samples of cattle in Japan: identi-
fication of possible interspecies transmission of bovine astrovirus. Arch Virol 160, 2491–2501 (2015).
21. Pankovics, P., Boros, A., Kiss, T., Delwart, E. & Reuter, G. Detection of a mammalian-like astrovirus in
bird, European roller (Coracias garrulus). Infect Genet Evol 34, 114–121 (2015).
21a. Karlsson, E.A. et al. Non-human primates harbor diverse mammalian and avian astroviruses includ-
ing those associated with human infections. PLoS Pathog 11(11), e1005225 (2015). doi:10.1371/journal.
ppat.1005225.
22. Bosch, A., Pinto, R.M. & Guix, S. Human astroviruses. Clin Microbiol Rev 27, 1048–1074 (2014).
23. Guix, S., Bosch, A. & Pintó, R. Astrovirus taxonomy. in Astrovirus Research (ed. Schultz-Cherry, S.)
97–118 (Springer, New York, 2013).
24. Ham, H. et al. Prevalence of human astrovirus in patients with acute gastroenteritis. Ann Lab Med 34,
145–147 (2014).
25. Koopmans, M.P., Bijen, M.H., Monroe, S.S. & Vinje, J. Age-stratified seroprevalence of neutralizing
antibodies to astrovirus types 1 to 7 in humans in the Netherlands. Clin Diagn Lab Immunol 5, 33–37
(1998).
26. Willcocks, M.M., Kurtz, J.B., Lee, T.W. & Carter, M.J. Prevalence of human astrovirus serotype 4:
capsid protein sequence and comparison with other strains. Epidemiol Infect 114, 385–391 (1995).
27. Finkbeiner, S.R. et al. Human stool contains a previously unrecognized diversity of novel astroviruses.
Virol J 6, 161 (2009).
28. Hu, B. et al. Detection of diverse novel astroviruses from small mammals in China. J Gen Virol 95,
2442–2449 (2014).
29. Jiang, H. et al. Comparison of novel MLB-clade, VA-clade and classic human astroviruses highlights
constrained evolution of the classic human astrovirus nonstructural genes. Virology 436, 8–14 (2013).
30. Kapoor, A. et al. Multiple novel astrovirus species in human stool. J Gen Virol 90, 2965–2972 (2009).
31. Pativada, M., Bhattacharya, R. & Krishnan, T. Novel human astrovirus strains showing multiple recom-
binations within highly conserved ORF1b detected from hospitalized acute watery diarrhea cases in
Kolkata, India. Infect Genet Evol 20, 284–291 (2013).
32. Finkbeiner, S. R. & Holtz, L. R. New human astroviruses. in Astrovirus Research (ed. Schultz-Cherry,
S.) 119–133 (Springer, New York, 2013).
33. Kurtz, J.B., Lee, T.W., Craig, J.W. & Reed, S.E. Astrovirus infection in volunteers. J Med Virol 3,
221–230 (1979).
34. Midthun, K. et al. Characterization and seroepidemiology of a type 5 astrovirus associated with an
outbreak of gastroenteritis in Marin County, California. J Clin Microbiol 31, 955–962 (1993).
35. Espinosa, A.C. et al. Infectivity and genome persistence of rotavirus and astrovirus in groundwater and
surface water. Water Res 42, 2618–2628 (2008).
36. Schultz-Cherry, S., King, D.J. & Koci, M.D. Inactivation of an astrovirus associated with poult enteritis
mortality syndrome. Avian Dis 45, 76–82 (2001).
37. Le Cann, P., Ranarijaona, S., Monpoeho, S., Le Guyader, F. & Ferre, V. Quantification of human astro-
viruses in sewage using real-time RT-PCR. Res Microbiol 155, 11–15 (2004).
38. Le Guyader, F., Haugarreau, L., Miossec, L., Dubois, E. & Pommepuy, M. Three-year study to assess
human enteric viruses in shellfish. Appl Environ Microbiol 66, 3241–3248 (2000).
39. Le Guyader, F.S. et al. Aichi virus, norovirus, astrovirus, enterovirus, and rotavirus involved in clinical
cases from a French oyster-related gastroenteritis outbreak. J Clin Microbiol 46, 4011–4017 (2008).
Astrovirus 37
40. Morsy El-Senousy, W., Guix, S., Abid, I., Pinto, R.M. & Bosch, A. Removal of astrovirus from water and
sewage treatment plants, evaluated by a competitive reverse transcription-PCR. Appl Environ Microbiol
73, 164–167 (2007).
41. Nadan, S., Walter, J.E., Grabow, W.O., Mitchell, D.K. & Taylor, M.B. Molecular characterization of
astroviruses by reverse transcriptase PCR and sequence analysis: comparison of clinical and environ-
mental isolates from South Africa. Appl Environ Microbiol 69, 747–753 (2003).
42. Pinto, R.M. et al. Astrovirus detection in wastewater samples. Water Sci Technol 43, 73–76 (2001).
43. Prevost, B. et al. Deciphering the diversities of astroviruses and noroviruses in wastewater treatment
plant effluents by a high-throughput sequencing method. Appl Environ Microbiol 81, 7215–7222 (2015).
44. Prevost, B. et al. Large scale survey of enteric viruses in river and waste water underlines the health
status of the local population. Environ Int 79, 42–50 (2015).
45. Steyer, A., Torkar, K.G., Gutierrez-Aguirre, I. & Poljsak-Prijatelj, M. High prevalence of enteric viruses
in untreated individual drinking water sources and surface water in Slovenia. Int J Hyg Environ Health
214, 392–398 (2011).
46. Pintó, R.M. & Bosch, A. Rethinking virus detection in food. in Food-Borne Viruses: Progress and
Challenges (eds. Koopmans, M., Cliver, D.O. & Bosch, A.) (American Society of Microbiology,
Washington, DC, 2008).
47. Gallimore, C.I., Cheesbrough, J.S., Lamden, K., Bingham, C. & Gray, J.J. Multiple norovirus genotypes
characterised from an oyster-associated outbreak of gastroenteritis. Int J Food Microbiol 103, 323–330
(2005).
48. Maunula, L., Kalso, S., Von Bonsdorff, C.H. & Ponka, A. Wading pool water contaminated with both
noroviruses and astroviruses as the source of a gastroenteritis outbreak. Epidemiol Infect 132, 737–743
(2004).
49. Myint, S., Manley, R. & Cubitt, D. Viruses in bathing waters. Lancet 343, 1640–1641 (1994).
50. Abad, F.X., Pinto, R.M., Villena, C., Gajardo, R. & Bosch, A. Astrovirus survival in drinking water.
Appl Environ Microbiol 63, 3119–3122 (1997).
51. Superti, F. et al. In vitro effect of synthetic flavanoids on astrovirus infection. Antiviral Res 13, 201–208
(1990).
52. Bass, D.M. & Qiu, S. Proteolytic processing of the astrovirus capsid. J Virol 74, 1810–1814 (2000).
53. Lee, T.W. & Kurtz, J.B. Prevalence of human astrovirus serotypes in the Oxford region 1976–92, with
evidence for two new serotypes. Epidemiol Infect 112, 187–193 (1994).
54. Mendez, E. & Arias, C.F. Astroviruses. in Fields Virology, Vol. 1 (eds. Knipe, D.M. & Howley, P.M.)
981–1000 (Lippincott Williams & Wilkins, Baltimore, 2007).
55. Mendez, E., Fernandez-Luna, T., Lopez, S., Mendez-Toss, M. & Arias, C.F. Proteolytic processing of a
serotype 8 human astrovirus ORF2 polyprotein. J Virol 76, 7996–8002 (2002).
56. Sanchez-Fauquier, A. et al. Characterization of a human astrovirus serotype 2 structural protein (VP26)
that contains an epitope involved in virus neutralization. Virology 201, 312–320 (1994).
57. Donelli, G., Superti, F., Tinari, A. & Marziano, M.L. Mechanism of astrovirus entry into Graham 293
cells. J Med Virol 38, 271–277 (1992).
58. Méndez, E., Murillo, A., Velázquez, R., Burnham, A. & Arias, C. Replication cycle of astroviruses. in
Astrovirus Research (ed. Schultz-Cherry, S.) 19–45 (Springer, New York, 2013).
59. Guix, S., Caballero, S., Bosch, A. & Pinto, R.M. C-terminal nsP1a protein of human astrovirus colocal-
izes with the endoplasmic reticulum and viral RNA. J Virol 78, 13627–13636 (2004).
60. Monroe, S.S., Jiang, B., Stine, S.E., Koopmans, M. & Glass, R.I. Subgenomic RNA sequence of human
astrovirus supports classification of Astroviridae as a new family of RNA viruses. J Virol 67, 3611–3614
(1993).
61. Banos-Lara Mdel, R. & Mendez, E. Role of individual caspases induced by astrovirus on the process-
ing of its structural protein and its release from the cell through a non-lytic mechanism. Virology 401,
322–332 (2010).
62. Guix, S., Bosch, A., Ribes, E., Dora Martinez, L. & Pinto, R.M. Apoptosis in astrovirus-infected CaCo-2
cells. Virology 319, 249–261 (2004).
63. Mendez, E., Salas-Ocampo, E. & Arias, C.F. Caspases mediate processing of the capsid precursor and
cell release of human astroviruses. J Virol 78, 8601–8608 (2004).
38 Laboratory Models for Foodborne Infections
64. Cubitt, W.D., Mitchell, D.K., Carter, M.J., Willcocks, M.M. & Holzel, H. Application of electronmicros-
copy, enzyme immunoassay, and RT-PCR to monitor an outbreak of astrovirus type 1 in a paediatric
bone marrow transplant unit. J Med Virol 57, 313–321 (1999).
65. Gallimore, C.I. et al. Use of a heminested reverse transcriptase PCR assay for detection of astrovirus in
environmental swabs from an outbreak of gastroenteritis in a pediatric primary immunodeficiency unit.
J Clin Microbiol 43, 3890–3894 (2005).
66. Khamrin, P. et al. Multiple astrovirus MLB1, MLB2, VA2 clades and classic human astrovirus in
children with acute gastroenteritis in Japan. J Med Virol 88, 356–360 (2015).
67. Liste, M.B. et al. Enteric virus infections and diarrhea in healthy and human immunodeficiency virus-
infected children. J Clin Microbiol 38, 2873–2877 (2000).
68. Walter, J.E. & Mitchell, D.K. Role of astroviruses in childhood diarrhea. Curr Opin Pediatr 12, 275–
279 (2000).
69. Belliot, G., Laveran, H. & Monroe, S.S. Capsid protein composition of reference strains and wild
isolates of human astroviruses. Virus Res 49, 49–57 (1997).
70. Pager, C.T. & Steele, A.D. Astrovirus-associated diarrhea in South African adults. Clin Infect Dis 35,
1452–1453 (2002).
71. Krishna, N. K., Koci, M. D. & Guix, S. Immune responses. in Astrovirus Research (ed. Schultz-Cherry,
S.) 79–95 (Springer, New York, 2013).
72. Kurtz, J. & Lee, T. Astrovirus gastroenteritis age distribution of antibody. Med Microbiol Immunol
(Berl) 166, 227–230 (1978).
73. Lee, R.M. et al. Incubation periods of viral gastroenteritis: a systematic review. BMC Infect Dis 13, 446
(2013).
74. Behling-Kelly, E. et al. Localization of astrovirus in experimentally infected turkeys as determined by
in situ hybridization. Vet Pathol 39, 595–598 (2002).
75. Brnic, D. et al. Porcine astrovirus viremia and high genetic variability in pigs on large holdings in
Croatia. Infect Genet Evol 14, 258–264 (2013).
76. Holtz, L.R. et al. Astrovirus MLB2 viremia in febrile child. Emerg Infect Dis 17, 2050–2052 (2011).
77. Wunderli, W. et al. Astrovirus infection in hospitalized infants with severe combined immunodeficiency
after allogeneic hematopoietic stem cell transplantation. PLoS One 6, e27483 (2011).
78. Gough, R.E., Collins, M.S., Borland, E. & Keymer, L.F. Astrovirus-like particles associated with hepa-
titis in ducklings. Vet Rec 114, 279 (1984).
79. Koci, M.D. et al. Astrovirus induces diarrhea in the absence of inflammation and cell death. J Virol 77,
11798–11808 (2003).
80. Schultz-Cherry, S. et al. Identifying agent(s) associated with poult enteritis mortality syndrome: impor-
tance of the thymus. Avian Dis 44, 256–265 (2000).
81. Yokoyama, C.C. et al. Adaptive immunity restricts replication of novel murine astroviruses. J Virol 86,
12262–12270 (2012).
82. Gray, E.W., Angus, K.W. & Snodgrass, D.R. Ultrastructure of the small intestine in astrovirus-infected
lambs. J Gen Virol 49, 71–82 (1980).
83. Woode, G.N., Pohlenz, J.F., Gourley, N.E. & Fagerland, J.A. Astrovirus and Breda virus infections of
dome cell epithelium of bovine ileum. J Clin Microbiol 19, 623–630 (1984).
84. Fu, Y. et al. The complete sequence of a duck astrovirus associated with fatal hepatitis in ducklings. J
Gen Virol 90 (Pt 5), 1104–1108 (2009).
85. Todd, D. et al. Identification of chicken enterovirus-like viruses, duck hepatitis virus type 2 and duck
hepatitis virus type 3 as astroviruses. Avian Pathol 38, 21–30 (2009).
86. Koci, M.D., Kelley, L.A., Larsen, D. & Schultz-Cherry, S. Astrovirus-induced synthesis of nitric oxide
contributes to virus control during infection. J Virol 78, 1564–1574 (2004).
87. Meyerhoff, R.R., Nighot, P.K., Ali, R.A., Blikslager, A.T. & Koci, M.D. Characterization of turkey
inducible nitric oxide synthase and identification of its expression in the intestinal epithelium following
astrovirus infection. Comp Immunol Microbiol Infect Dis 35, 63–69 (2012).
88. Nighot, P.K., Moeser, A., Ali, R.A., Blikslager, A.T. & Koci, M.D. Astrovirus infection induces sodium
malabsorption and redistributes sodium hydrogen exchanger expression. Virology 401, 146–154 (2010).
89. Pantin-Jackwood, M.J., Spackman, E. & Day, J.M. Pathogenesis of type 2 turkey astroviruses with vari-
ant capsid genes in 2-day-old specific pathogen free poults. Avian Pathol 37, 193–201 (2008).
Astrovirus 39
90. Tang, Y., Murgia, M.V., Ward, L. & Saif, Y.M. Pathogenicity of turkey astroviruses in turkey embryos
and poults. Avian Dis 50, 526–531 (2006).
91. Thouvenelle, M.L., Haynes, J.S. & Reynolds, D.L. Astrovirus infection in hatchling turkeys: histologic,
morphometric, and ultrastructural findings. Avian Dis 39, 328–336 (1995).
92. Thouvenelle, M.L., Haynes, J.S., Sell, J.L. & Reynolds, D.L. Astrovirus infection in hatchling turkeys:
alterations in intestinal maltase activity. Avian Dis 39, 343–348 (1995).
93. Sebire, N.J. et al. Pathology of astrovirus associated diarrhoea in a paediatric bone marrow transplant
recipient. J Clin Pathol 57, 1001–1003 (2004).
94. Moser, L.A., Carter, M. & Schultz-Cherry, S. Astrovirus increases epithelial barrier permeability inde-
pendently of viral replication. J Virol 81, 11937–11945 (2007).
94a. Meliopoulos, V.A. et al. Oral administration of astrovirus capsid protein is sufficient to induce acute
diarrhea in vivo. mBio 7(6), e01494–16 (2016). doi:10.1128/mBio.01494-16.
95. Meliopoulos, V.A. et al. Detection of antibodies against Turkey astrovirus in humans. PLoS One 9,
e96934 (2014).
96. Burbelo, P.D. et al. Serological studies confirm the novel astrovirus HMOAstV-C as a highly prevalent
human infectious agent. PLoS One 6, e22576 (2011).
97. Molberg, O. et al. HLA restriction patterns of gliadin- and astrovirus-specific CD4+ T cells isolated in
parallel from the small intestine of celiac disease patients. Tissue Antigens 52, 407–415 (1998).
98. Molberg, O. et al. CD4+ T cells with specific reactivity against astrovirus isolated from normal human
small intestine. Gastroenterology 114, 115–122 (1998).
99. Guix, S. et al. Type I interferon response is delayed in human astrovirus infections. PLoS One 10,
e0123087 (2015).
100. Bonaparte, R.S. et al. Human astrovirus coat protein inhibits serum complement activation via C1, the
first component of the classical pathway. J Virol 82, 817–827 (2008).
101. Gronemus, J.Q. et al. Potent inhibition of the classical pathway of complement by a novel C1q-binding
peptide derived from the human astrovirus coat protein. Mol Immunol 48, 305–313 (2010).
102. Hair, P.S. et al. Human astrovirus coat protein binds C1q and MBL and inhibits the classical and lectin
pathways of complement activation. Mol Immunol 47, 792–798 (2010).
103. Krishna, N.K. & Cunnion, K.M. Human astrovirus coat protein: a novel C1 inhibitor. Adv Exp Med Biol
632, 237–251 (2008).
104. Tam, J.C., Bidgood, S.R., McEwan, W.A. & James, L.C. Intracellular sensing of complement C3 acti-
vates cell autonomous immunity. Science 345, 1256070 (2014).
105. Koci, M.D. Immunity and resistance to astrovirus infection. Viral Immunol 18, 11–16 (2005).
106. Qureshi, M.A., Edens, F.W. & Havenstein, G.B. Immune system dysfunction during exposure to poult
enteritis and mortality syndrome agents. Poult Sci 76, 564–569 (1997).
107. Qureshi, M.A., Yu, M. & Saif, Y.M. A novel “small round virus” inducing poult enteritis and mortality
syndrome and associated immune alterations. Avian Dis 44, 275–283 (2000).
108. Heggen, C.L., Qureshi, M.A., Edens, F.W. & Barnes, H.J. Alterations in macrophage-produced cyto-
kines and nitrite associated with poult enteritis and mortality syndrome. Avian Dis 44, 59–65 (2000).
109. Heggen, C.L., Qureshi, M.A., Edens, F.W., Barnes, H.J. & Havenstein, G.B. Alterations in the lympho-
cytic and mononuclear phagocytic systems of turkey poults associated with exposure to poult enteritis
and mortality syndrome. Avian Dis 42, 711–720 (1998).
110. Qureshi, M.A., Edens, F.W., Ali, R.A. & Saif, Y.M. Alteration in macrophage-mediated cytokines/
metabolite production after challenge with a “small round virus” of turkeys. in Poultry Science
Association 89th Annual Meeting 64 (Montreal, Canada, 2000).
111. Qureshi, M.A., Saif, Y.M., Heggen-Peay, C.L., Edens, F.W. & Havenstein, G.B. Induction of functional
defects in macrophages by a poult enteritis and mortality syndrome-associated turkey astrovirus. Avian
Dis 45, 853–861 (2001).
112. Dryden, K.A. et al. Immature and mature human astrovirus: structure, conformational changes, and
similarities to hepatitis E virus. J Mol Biol 422, 650–658 (2012).
4
Hepatitis E Virus
CONTENTS
4.1 Introduction......................................................................................................................................41
4.1.1 HEV Transmission............................................................................................................. 43
4.1.2 HEV Infection in Humans.................................................................................................. 44
4.1.3 Clinical Course/Manifestations of HEV Infection............................................................ 46
4.1.4 Immune Response.............................................................................................................. 46
4.1.4.1 Innate Immune Response................................................................................... 46
4.1.4.2 HEV-Induced Adaptive Immune Response........................................................ 47
4.1.5 Diagnosis............................................................................................................................ 47
4.1.6 HEV Vaccine...................................................................................................................... 48
4.1.7 Antiviral Therapy against HEV......................................................................................... 48
4.1.8 HEV Biology...................................................................................................................... 49
4.1.8.1 Proteins Encoded by the HEV Genome............................................................. 50
4.1.8.2 cis-Regulatory Genomic Regions....................................................................... 52
4.1.9 HEV Life Cycle.................................................................................................................. 52
4.2 Tools That Can Be Used for Studying HEV................................................................................... 53
4.2.1 Use of Infectious cDNA Clones of HEV............................................................................ 53
4.2.2 HEV Cell-Culture Models.................................................................................................. 54
4.2.2.1 Genotype 3 Virus Culture Systems.................................................................... 55
4.2.2.2 Kernow C1 Virus Strain..................................................................................... 55
4.2.2.3 3D-Cell-Culture System..................................................................................... 55
4.2.2.4 Primary Hepatocytes as the Culture System...................................................... 56
4.2.2.5 Genotype 4 Virus Culture System...................................................................... 56
4.2.2.6 Propagation of HEV Strains Circulating in Blood............................................. 56
4.2.3 HEV Animal Models.......................................................................................................... 58
4.2.3.1 Nonhuman Primates........................................................................................... 58
4.2.3.2 Pigs...................................................................................................................... 59
4.2.3.3 Rodents............................................................................................................... 59
4.2.3.4 Rabbits................................................................................................................ 60
4.2.3.5 Ferrets................................................................................................................. 60
4.2.3.6 Chickens.............................................................................................................. 60
4.3 Conclusions......................................................................................................................................61
References..................................................................................................................................................61
4.1 Introduction
Hepatitis E virus (HEV) is a major causative agent of acute viral hepatitis in developing countries.
HEV is primarily transmitted via the fecal–oral route through contaminated water and is endemic in
several countries in Asia, Africa, and Central America1–3 due to poor sanitary conditions. It is also
41
42 Laboratory Models for Foodborne Infections
highly endemic in India, Bangladesh, Egypt, Mexico, and China, where hepatitis E can occur either
in the form of sporadic cases or occasional large outbreaks. Hepatitis E represents the major cause of
waterborne outbreaks in these countries, and contamination of water with sewage has been a common
feature preceding such outbreaks.4–10 HEV is believed to be the most common cause of sporadic acute
hepatitis (>50%–60% cases) in adults in regions where the virus is endemic.11–13 There have also been
large outbreaks among displaced persons in Sudan, Chad, and Uganda.14–17
Although, previously, sporadic cases of hepatitis E in developed countries were attributed to travel to
hyperendemic areas, in recent years, a surprisingly higher number of cases of autochthonous transmis-
sion of hepatitis E with high seroprevalence have been documented from the United States, Canada,
Europe (including UK, France, the Netherlands, Austria, Denmark, Spain, Italy, Greece, and Germany),
and Asia–Pacific countries (Japan, Taiwan, Korea, Hong Kong, Australia, and New Zealand).18,19 It is
estimated that about 2 billion people live in HEV-endemic areas. A global disease burden study esti-
mated that HEV genotypes 1 and 2 account for approximately 20.1 million incidents of HEV infections,
3.4 million cases of symptomatic disease, 70,000 deaths, and 3,000 stillbirths. Clinical HEV infection is
most common in adolescents and young adults, but also occurs to a lesser extent in children.20–22 During
epidemics of HEV, the ratio of clinical to subclinical or anicteric infection was shown to be 1:26 in
pregnant women, 1:4 in children, and a pproximately 1:10 in adults.20,23 In developing countries, although
HEV is endemic, seroprevalence usually remains below 40% as against hepatitis A virus, which shows
almost 100% seropositivity in adults.1
Nucleotide sequence analysis of HEV genomes from different geographical isolates has revealed wide
genetic diversity among HEV strains forming four distinct phylogenetic clusters. Of these, genotype 1
and 2 strains are restricted to humans. Genotype 1 strains are associated with large outbreaks of acute
hepatitis E in humans in Asia. Genotype 2 includes one Mexican strain and several African strains.
Genotype 3 is distributed worldwide. Distribution of genotype 2 virus is restricted to few countries and is
not a major problem in comparison with the more widely distributed genotype 1 virus. Genotype 3 is also
associated with sporadic, cluster, and chronic cases of hepatitis E in humans, mostly in industrialized
countries. Genotype 3 strains have also been isolated from domestic pigs, wild pigs, deer, mongoose, and
rabbits.24–26 Wang et al.27 identified an additional HEV strain in the sera of Chinese patients with acute
hepatitis, classified as genotype 4. Genotype 4 virus can also infect pigs.28,29 Genotype 3 and 4 strains
have been found to infect a broader range of hosts including deer and boars.26,30–32 Intra- and intergeno-
type recombination has been recently documented.33
HEV is enzootic in domestic pigs even though the virus may not be endemic in human population or
belongs to a different genotype than that is circulating in humans in endemic settings. Detection of HEV
RNA and antibodies in wild boar populations has been reported from Japan, Germany, Italy, Spain, the
Netherlands, and Australia.34–39 In addition, several divergent strains of HEV have been isolated from
animals such as bats,40 rats,31 wild boar,41 ferrets,42 mongoose,25 fish, and cutthroat trout.43
HEV strains within genotypes 1 and 2 are less divergent as compared with HEV strains from
genotypes 3 and 4. These HEV genotypes have been classified further into 24 subgenotypes.44,45
Currently, four putative genera have been proposed in the family Hepeviridae, one comprising human
HEV genotypes and closely related animal viruses and the other three including viruses of rodent (rat),
chiropteran (bat), and avian (chicken) origins.40 HEV isolates from rats share approximately 59.9% and
49.9% sequence identities with human and avian HEVs, respectively, while the ferret HEV sequences
share 72.3% identity with the rat HEV and have been proposed to be grouped in the genus Ortho
hepevirus.42,46 Bat HEV strains from Africa, Central America, and Europe have a very high sequence
divergence (53% at amino acid level) from known HEV isolates and are suggested to be classified in
a separate genus Chiropteranhepevirus.47,48 Avian HEV strains share 50%–60% nucleotide sequence
identity with swine or human HEV strains, and recently it was proposed that all three known genotypes
of avian HEV in chickens (genotype 1 in Australia and Korea, genotype 2 in the United States, and
genotype 3 in Europe and China) should be grouped into a new genus Avihepevirus.47,49,50 The cutthroat
trout HEV from the United States exhibits only 13%–27% sequence homology leading to a proposal
of another genus Piscihepevirus.43,47 Interestingly, the range of different animal species that appear
to harbor HEV/HEV-like agents is continuously extending, indicating the ubiquity of HEV/HEV-like
agents in different species from many regions. We expect many more additions in this family in the
Hepatitis E Virus 43
e2
Genotype 4
typ
uman
Human
an
A F151963 C hina - H u m an
an
AY7237
no
um
Hum
Ge
AF32
ina - H
-H
A F5
EV
AF134812 China -
159 hina -
AJ3
wan
45 Indi
-H
4503
058
AY
2
179 Ch
441
Tai
an
59
AJ
60 I
AJ
e
C
India
um
41
42
92
34
typ
AF 44171
a - Swin e
88
41
99
ndia
Genotype 1
AB
oH
Ch
an
81
an
53
AB
AJ344
08
Ch a - S
um
296
- Swin
in a
09
um
Ch
xic
25
Ch
AJ3
no
- Sw
AB 781
ina
-H
58 in
H
Me
in
-H
099 2 J a-
e
J an
- S ine
a-
a
an
347 apa
ine
pa H
Ge
um
um
06
th
m
AB n um
wi
097 Japan - H n-
ur
kis
-H
45
ne
an
AB0 811 u H an
8B
- m u
Pa
ia
M7
749 H m
15 J Japan umaan an nd
21
22
I n
AB69 apa - S n ma an
73
58
8654 n - H win 67 Hu um
12
18
e
M
u
AB1085 Ja
37 Chipan - Hum an
m
06 0 ia - - H
AF
45 IndIndia
na
AJ428855 Ch - Human an LC 17
0 2 man
ina - Swine AF
3
25 20 - Hu
AB124818 Indonesia - Swine U2 4437 6 India uman
JF 4372 dia- H
JF4 3725 In Human
JF44 30 Nepal -
18
AF05
a - Human
Burm
M73218 an
X98292 India - Hum
AY697428 Kyrgyzstan - Human
Avian strains D11093 China - Hum
AY23020
AY 2 Mor
an
AF02504877 C occo- Human
1352 had -
Rabbit strains Egyp Huma
t-H
uma
n
n
JQ0
GU 65070
186 C
uman 165 hina R
USA - H inean Ch abb
JN837481herlands -- Sw um e ina it
62 99 Net Japan H - Swin man -R
abb
AF33 B08982 4 ea u AB AY3
Kor - H winean it
A
6 403 rea S AF 09 623
FJ4 2
72
K o A - um os
e 50 3 5 57
US - H go 35 35 J Un
ine
142 0
EU
Sw ine
7 8 A an 12 ap it
AY 66 US an ed K
e
6
11
y- w
in
4 M Un
GQ 1950 0 Neth
-S
AF 9544 ing
an s - S
-
an
-H
63
ite
- Hum an
n do
pa
da
um
39
40
38
um
d
AFF3326
m
nd
Hum
Ja
A
Ki
HQ
80
-H
na
an
an
Fr
rla
-H
34 ng
A B 189071 Japan - D eer
um
24
an
JF71 5182 France
Ca
Swine
he
7 do
rm
an
ce
1
BU55
Fr
et
59 m
an -
zil
88
Ge
6
N
-H
an
8800
AB -S
3S
54
2
Bra
0
ce
59
um
wi
29
1 Jap
0 Japan -
11
pa
Japan
ne
36
53
an
884
Sw
AY
in
Fran
F3
70
in
8256
-S
A
FJ
152
e
rlan
wi
9072
ce -
KF
ne
AB18907
AB0
Genotype 3
ds -
Swin
AB18
- Hum
Swin
e
e
an
FIGURE 4.1 HEV phylogenetic tree: Full genome/capsid region nucleotide sequence analysis of global hepatitis E virus
isolates. Genotypes 1 and 2 (HEV-1 and HEV-2) circulate among humans, primarily in Africa and Asia, while genotypes 3
and 4 (HEV-3 and HEV-4) have animal reservoirs. Recently discovered rabbit strains appear to form a closely related clade.
Alignment was done using MEGA 7 and tree by using software available on www.itol.embl.de.
coming years. A phylogenetic tree based on a portion of the nucleotide sequence encoding the capsid
protein is shown in Figure 4.1.
4.1.1 HEV Transmission
The virus is relatively stable in the environment51 and is sensitive to heat, chlorination, and ultraviolet
light.52,53 In endemic regions, HEV remains in circulation due to the excretion of the virus by humans and
other animals in the acute phase of infection and environment acts as the major reservoir of the virus.54
Contamination of drinking and irrigation water mainly occurs due to improper sewage disposal, leading to
epidemics in developing countries. Water scarcity in summers or floods during rainy season can increase
the possibility of the virus spread. Surface water bodies can easily get contaminated with human and ani-
mal wastes. Inadequate treatment and release of sewage in rivers has been documented, with 4.9% HEV
RNA positivity in 403 water samples from different locations of a river in one Indian city (Mutha River,
Pune).55 Waterborne route could also be important for genotype 3 and 4 HEVs. Genotype 3 virus has
been detected in waste/runoff waters from pig slaughterhouses, swine manure, and swine slurry storage
facilities.56,57 An increased risk of hepatitis E in sewage treatment plant workers has been documented.58,59
Person-to-person transmission of HEV is rare in both epidemic60 and sporadic settings.61 Occasionally,
nosocomial spread has also been reported.62 Vertical transmission from mother to infant is also known
to occur.63 In endemic as well as nonendemic settings, transfusion-associated hepatitis E infection is
possible64–69; however, this route of transmission is not so common.70,71 Plasma products have been
44 Laboratory Models for Foodborne Infections
documented as the source of HEV infections.72,73 Easy and rapid HEV infectivity laboratory assays to
evaluate virus inactivation processes would be useful for monitoring plasma-derived products.
Close phylogenetic relationships between human and animal viruses prove that HEV is transmitted
from animals to humans. Domestic pigs are the major animal reservoirs of HEV. Hepatitis E is now
a recognized zoonotic disease, with swine and other likely animals serving as the reservoir for these
viruses.74,75 Naturally acquired anti-HEV antibodies have been shown in many animal species including
swine, sheep, cattle, goats, horses, macaques, cats, dogs, rats, and mice, raising the possibility of these
animals being the reservoirs and causing zoonotic infections.10,76,77 Occupational exposure to infected
pigs poses the risk of acquiring HEV infection.56,78–80
Consumption of undercooked or raw pig liver, pork, and game meat (wild boar, deer, or rabbit) plays
a significant role in HEV transmission in industrialized countries. Studies from Japan have documented
foodborne HEV infections that occurred due to the consumption of undercooked pork81,82 and wild boar
or deer meat.83–86 HEV infections due to wild boar meat have also been reported from Germany86 and
South Korea.32 Consumption of pig liver figatellu sausages has been documented to cause human HEV
infections in France, as seen from the identical HEV sequences from the infected individuals and sau-
sages from the local grocery stores.87,88 Infectious nature of HEV particles in the sausages was demon-
strated by Berto et al.89 HEV presence in pig liver or meat has also been reported from countries such
as India,90 United States,91 Germany,92 Canada,93 Democratic Republic of Congo,94 Japan,95 Italy,96 and
the Netherlands.97
HEV has been detected in varied shellfish collected from European and Asian countries. Genotype 3
HEV has been detected in mussels and oysters.98–102 Human HEV infections due to the consumption of
shellfish are documented.103–105 Bivalves are known to concentrate viral particles during their filter feed-
ing process, and as they are mostly consumed raw or cooked for a very short period, they pose a serious
risk of HEV infection. Viral RNA sequences have been detected in soft fruits like strawberries and green
leafy vegetables such as lettuce, with irrigation water being the suspected contamination source.106,107
The concentration of the viable virus in an environment or food is an important factor in the outcome of
clinical hepatitis E infection. HEV is known to remain stable in frozen conditions even after 10 years.
HEV remains viable after heating to 56°C for 1 h,108 and a cooking temperature of 71°C for 20 min is
required for the complete inactivation of the virus.109 Use of contaminated water for drinking, cooking,
cleaning, and irrigation107 can spread the virus.
ALT, however, does not correlate with the degree of liver cell damage (peak levels vary from 1000 to
2000 U/L at the onset). Peak serum total bilirubin levels range from 5 to 25 mg/dL; both conjugated and
unconjugated fractions are increased. ALT progressively diminishes during the recovery phase. Patients
with anicteric or subclinical acute hepatitis E infection only exhibit elevated ALT levels, which is use-
ful in the early diagnosis of clinically suspected cases, along with the presence of anti-HEV IgM in the
serum. Liver damage results, presumably mediated by host cellular immune responses. The course of
hepatitis E in human subjects strongly suggests that HEV, like other hepatitis viruses, is noncytopathic in
most circumstances, and liver disease is probably caused by immunological and hormonal factors.113–116
The severity of hepatitis E is considered to be dose-dependent, and associated host factors such as
chronic liver disease or alcohol overuse may enhance it further.3 Patients with HEV infection superim-
posed with other viral or nonviral chronic liver disease (acute on chronic liver disease) are at the higher
risk of having a poor outcome.117,118 Case fatality rates in hepatitis E epidemics range from 0.1% to 4%,
but pregnant women, especially during the third trimester, are at a higher risk of severe disease, with
case fatality rates of 10%–25%.119–121 Hepatitis E is also associated with premature delivery, with low
birth weight and an increased risk of perinatal mortality.122 HEV transmission from mother to fetus was
reported in 33% of cases; the virus has been detected in human colostrum as well.117,123
The outcome of hepatitis E infection during pregnancy is probably influenced by nutritional status,
host immune response, and hormonal factors.113,115,116 It is not yet understood how HEV infections dur-
ing pregnancy are less severe in Egypt124 while they are more severe with higher mortality rates in
sub-Saharan Africa and South and South-East Asia,115 although genotype 1 HEV predominates in these
regions. Thus, severity during pregnancy is not the unique feature of genotype 1 or 2 viral infections, but
there seem to be additional contributing factors that influence the disease severity. Only occasional cases
of hepatitis E during pregnancy with less severity have been reported from low-endemic, developed
countries.125–127 Thus, understanding HEV pathogenesis during pregnancy still remains a big challenge.
There are certain observations to suggest that HEV strains may vary in their virulence. Genotype 3
strain (3c) circulating in Europe is known to cause mostly subclinical infections.128 A comparative study
of genotype 3 and 4 HEV-infected individuals in Japan revealed that genotype 4 HEV is associated
with significantly higher ALT and total bilirubin levels, higher viral loads, and an aggressive disease
course.129 Similar observations have been reported by Jeblaoui et al.130 in a study from France that the
clinical presentation is more severe in patients with HEV4 infections than in patients with HEV3 infec-
tions. Purcell et al.131 examined the relative virulence of human genotypes 1 and 2 and swine genotype 3
in rhesus monkeys and found that genotype 3 is significantly less virulent as compared with human
genotypes 1 and 2. There are studies indicating the association of point mutations in the HEV genome
with disease severity; however, the underlying mechanism still remains unknown. Silent substitutions of
U at the nucleotide 3148 (in the helicase domain) and C at the nucleotide 5907 (in the capsid gene) in the
genomes of HEV strains of genotypes 3 and 4 have been documented to have an association with fulmi-
nant hepatitis and disease severity in patients. C5907 mutation was also associated with high viral loads
in the infected individuals.132,133 Takahashi et al.134 have reported the association of V239A mutation (in
the helicase domain) with increased virulence of genotype 3 virus. Similarly, L1110F and V1120I muta-
tions in the helicase domain of genotype 1 sequences were found to be associated with fulminant hepa-
titis cases.135 Shukla et al.136 have recently reported the integration of human S17 rRNA sequence into
the genotype 3 HEV genome in a patient with chronic HEV infection. This patient had both neurologic
and hepatic symptoms. Nguyen et al.137 have also reported the integration of S19 in an isolate analyzed
from a chronic hepatitis E patient with organ transplant in the United States. These integrations in HEV
genomes probably altered tissue specificity and pathogenicity of the virus.
HEV normally does not lead to chronic infection in immunocompetent individuals. However, chronic
HEV infections have been observed in immunocompromised organ transplant recipients138,139 and in
patients with other conditions of immunosuppression such as HIV infection140 and hematological malig-
nancies.141,142 HEV infection results in persistent viral shedding and may rapidly progress to liver cir-
rhosis and may also cause extrahepatic manifestations such as neurological disorders and impaired renal
functions in these patients.143 Chronic HEV infections were thought to exclusively involve genotype 3
viral strains, however, recently a report from China documented the chronic course of HEV infection
with genotype 4 virus in a boy who was receiving chemotherapy for an acute lymphoblastic leukemia.144
46 Laboratory Models for Foodborne Infections
A study from India showed no evidence of chronic hepatitis E in a cohort of kidney transplant recipients,
suggesting that HEV genotype 1 may not cause chronic hepatitis E.145 However, there is a need for more
data from other endemic regions to confirm these findings.
4.1.4 Immune Response
4.1.4.1 Innate Immune Response
After entry into the host, HEV needs to overcome the host innate immune response and establish infec-
tion in the host. In the past decade, remarkable progress has been made in understanding strategies of
HEV in combating host innate immune responses. HEV interferes with type 1 interferon (IFN) induction
and IFN-activated signaling. HEV inhibits IFN-α signaling and manages to replicate in the presence of
IFN-α. There is a study demonstrating the reduced ubiquitination and degradation of IkBα in hepatoma
cells expressing the open reading frame (ORF) 2 protein, affecting the nuclear translocation of NF-κB.
Recently, the macro domain and papain-like cysteine protease (PCP) domain from HEV ORF1 were
identified as the putative IFN antagonists by Nan et al.165 In contrast, HEV ORF3 was shown to enhance
RIG-I-mediated signaling leading to enhanced IFN-α synthesis, indicating complex strategies of the
virus during its establishment in the host cells.166
A microarray-based gene expression analysis of serial liver biopsy samples from HEV and HCV in
chimpanzees has shown attenuated adaptive immune response in HEV-infected animals as compared
with HCV.167 However, there was a robust innate immune response correlating well with viremia in both
HCV- and HEV-infected chimpanzees. A major component of the host response to HEV infection was
type I interferon-induced genes (ISGs). HEV infection was completely resolved without any recurrence
in the infected chimpanzees, indicating that HEV is highly susceptible to ISGs. This indicated that HEV
infection is mostly taken care of by the robust innate immune response. Altered peripheral frequencies
and activation status of NK and NKT cells in HEV-infected individuals suggest that innate immunity
Hepatitis E Virus 47
plays a role in the pathogenesis of hepatitis E.168 It was speculated that the reduced NK cell levels during
pregnancy could contribute toward the increased severity of hepatitis E during pregnancy. Robust non-
specific IFN-γ-producing T-cell response in acute hepatitis E patients suggesting innate immune mecha-
nisms involving NK/NKT/T reg cells has been documented.169 Higher frequencies of CD4+ CD25+
Foxp3+ regulatory T cells, elevated levels of IL-10, and suppressive functionality of the regulatory T
cells in acute hepatitis E patients have been documented.170 Overall, innate immune responses seem to
play a major role during HEV infection.
4.1.5 Diagnosis
Hepatitis E diagnosis mainly depends on the clinical features and exclusion of other causes of acute
hepatitis, especially hepatitis A, in the endemic regions. The presence of anti-HEV IgM is a marker
of acute infection, while anti-HEV IgG alone is a marker of past infection. HEV was first identified by
immunoelectron microscopy.110 Serological as well as molecular assays were developed eventually after
successful cloning of the HEV genome.177,178 Anti-HEV IgM appears during the early acute phase of ill-
ness and may be detected as early as 4 days after the onset of jaundice and lasts for up to 2–5 months.179
Anti-HEV IgG has been shown to persist for long periods of time (>14 years) and provides protection
against subsequent infections. Anti-HEV IgA has also been detected in the serum of naturally infected
individuals.180 For practical purposes, HEV IgM can be detected from blood to confirm the clinical diag-
nosis. Detection of viral RNA in HEV IgM-positive patients provides additional confirmation and further
information about the virus strain, enabling molecular epidemiological studies. In immunocompromised
patients, HEV RNA testing is essential for diagnosis since patients have impaired immune response.
There are several HEV RNA detection assays developed during the past decade.181–185 However, the assay
developed by Jothikumar et al.185 seems to be very commonly used. For optimal diagnosis, a combination
of serological and nucleic acid-based assays has been recommended.186,187
HEV ORF2 protein expressed in SF9 cells has been shown to have glycosylation and performs
well in anti-HEV IgM and IgG diagnostic assays. All the four genotypes of HEV belong to a single
serotype,188 thus suggesting that diagnostic antigens from a single HEV genotype should detect
antibodies against HEV strains of different genotypes. Broad cross immunoreactivity among HEV
genotypes has been documented.189–192 Commercially available assays for hepatitis E diagnosis have
problems of sensitivity and specificity, and many studies have shown poor concordance of these
48 Laboratory Models for Foodborne Infections
tests.193,194 There is a need to have a validated assay that would have reasonable sensitivity and
specificity in detecting infections with different HEV genotypes. Such an assay would be extremely
useful in generating worldwide seroprevalence data to understand the realistic disease burden of
hepatitis E.
4.1.6 HEV Vaccine
Being refractory to cell culture, the conventional approach of developing killed or attenuated virus-
based vaccines for HEV was not possible and hence recombinant DNA technology has been employed.
Four mammalian HEV genotypes are believed to represent a single serotype. Cross-challenge studies
in rhesus monkeys using various strains of genotype 1 HEV have shown that animals are protected
from the disease up to 5 years after primary infection, indicating the possible utility of the vaccine to
prevent hepatitis E.1,12,64,195 HEV capsid (ORF2) protein is highly immunogenic and has been used as
the target antigen for vaccine development. Several approaches based on recombinant DNA technol-
ogy have been attempted to develop candidate subunit vaccines, either using a complete or partial
ORF2 protein. A 56-kDa (112–607 amino acids) recombinant protein-based candidate vaccine
phase III clinical trial, with 95% efficacy, was successfully completed in 2007 by the NIH group.196
This subunit vaccine protected against genotype 1 (homologous) as well as genotype 2 and 3 HEV
(heterologous) challenges in the monkey model.197 However, since then, there has been no progress in
the commercialization of this vaccine. The second candidate vaccine that was found to be safe and
immunogenic was based on the bacterially expressed genotype 1 HEV ORF2 protein (p239, 368–606
amino acids). Phase III clinical trial of this vaccine showed >95% efficacy and cross protection against
the heterologous genotype 4 virus.198 The vaccine (Hecolin) is currently licensed and commercially
available only in China.
TABLE 4.1
Characteristics of HEV Genotypes 1–4
Characteristics Genotype 1 Genotype 2 Genotype 3 Genotype 4
Distribution of Asia, Africa, and the Mexico, West Africa North America, China, Taiwan, and
virus in humans Middle East Europe, Latin South-East Asia
America, and Japan
Distribution of Not reported and Not reported and Widespread and China, Taiwan, and
virus in animals identified identified reported in all India, with a few
continents recent reports from
Europe and North
America
Interspecies Only human to Only human to Animal to human Animal to human
transmission human and no human and no (pigs, wild boar, and (pigs, wild boar)
interspecies interspecies deer)
transmission transmission
observed observed
Waterborne Yes, frequent (source Yes, frequent (source Not reported Not reported
transmission is from human is from human
feces) feces)
Food-borne Not reported and Not reported and Reported from Reported from
transmission recognized recognized contaminated animal contaminated
meat animal meat
Zoonotic Not reported Not reported Yes Yes
transmission
Mortality among High Not reported Not reported Not reported
pregnant women
Infection and attack Most common in Most common in Middle age and older Middle age and older
rate with respect young adults young adults above 55 years, above 55 years
to age (15–44 years) (15–44 years) males, and
immunocompromised
persons
Outbreaks Common Smaller scale Uncommon Uncommon
outbreaks
Symptoms Fever, fatigue, loss of Fever, fatigue, loss of Fever, fatigue, loss of Fever, fatigue, loss of
appetite, nausea, appetite, nausea, appetite, nausea, appetite, nausea,
vomiting, vomiting, vomiting, abdominal vomiting, abdominal
abdominal pain, abdominal pain, pain, jaundice, dark pain, jaundice, dark
jaundice, dark jaundice, dark urine, clay-colored urine, clay-colored
urine, clay-colored urine, clay-colored stool, and joint pain stool, and joint pain
stool, and joint pain stool, and joint pain
Diagnosis Anti-HEV test Anti-HEV test Anti-HEV test Anti-HEV test
Prevention Good sanitation and Good sanitation and Avoiding raw pork and Avoiding raw pork
the availability of the availability of other contaminated and other
clean drinking clean drinking animal meat can contaminated
water water reduce the risk animal meat can
reduce the risk
4.1.8 HEV Biology
HEV is a nonenveloped, spherical particle of approximately 30–34 nm in diameter.110 The HEV genome
is a positive-sense single-stranded RNA of approximately 7.2 kb with a 5ʹ-methylguanine cap and 3ʹ-poly
(A) stretch. Viral RNA contains short 5ʹ (27–35 nt) and 3ʹ (65–74 nt) untranslated regions (UTRs) and
three ORFs, ORF1, ORF2, and ORF3.178 ORF1 encodes the nonstructural polyprotein, ORF2 encodes
50 Laboratory Models for Foodborne Infections
ORF3
(nt 5131–5475)
FIGURE 4.2 Schematic diagram of the HEV genome: The three ORFs are labeled and shown as boxes with the putative
ORF1 domains indicated outside the box. JR, junction region; me7G, 5ʹ Cap; NCR, noncoding region; ORF, open reading
frame; P, Proline-rich hinge region; RdRp, RNA-dependent RNA-polymerase; Y, Y domain (nucleotide numbering done
as per prototype genotype 1 sequence SAR55).
the capsid protein, and ORF3 encodes a multifunctional protein. ORF2 and ORF3 are proposed to be
translated from a single bicistronic messenger ribonucleic acid (mRNA) and overlap with each other.210,211
Figure 4.2 shows a schematic diagram of the HEV genome.
4.1.8.1.2 Methyltransferase
It is the first functional domain of ORF1.213 Recombinant protein-encompassing amino acids 1–979 from
HEV ORF1 were shown to have guanine-7-methyltransferase as well as guanyltransferase activities. The
presence of an m7G cap at the 5ʹ end of the HEV genomic RNA confirmed the functional role of HEV
methyltransferase.214 The source of RNA triphosphatase activity, that is required for 5ʹ cap formation,
was later confirmed to be a HEV helicase protein.215
4.1.8.1.3 PCP
The most intriguing question of HEV biology that has not yet been answered is whether the ORF1 pro-
tein (pORF1) is processed into functional biochemical subunits or whether it remains as a single protein.
The postulated role of PCP is the polyprotein processing of ORF1 polyprotein. The above hypothesis
was put to test by many researchers, but none could show the evidence of ORF1 processing and the role
of PCP in polyprotein processing.216–221 A recent study from our group showed that HEV MeT-PCP pro-
tein recognizes the LXGG motif and shows in vitro deubiquitination (DUB) activity. The protein also
showed deISGylation of cellular proteins, suggesting its probable role in combating cellular antiviral
innate immunity during the HEV life cycle.222 Paliwal et al.223 recently demonstrated the role of PCP in
ORF1 polyprotein processing.
4.1.8.1.4 Helicase
The putative RNA helicase of HEV was shown to contain all the seven conserved motifs found in SF-1
helicases and was proposed to have the NTPase as well as RNA-binding domains.212 Functionality of
HEV helicase was recently demonstrated by our group. HEV Hel exhibited NTPase and RNA unwinding
activities. Enzyme hydrolyzed all rNTPs efficiently; dATP and dCTP were hydrolyzed more efficiently
as compared with dGTP and dTTP. Enzyme showed the unwinding of only RNA duplexes with 5ʹ over-
hangs showing a 5ʹ-to-3ʹ polarity.224
Hepatitis E Virus 51
4.1.8.1.5 RdRp
HEV RdRp contains eight conserved motifs (motif I–VIII) that are similar to the RdRps from other
positive-sense RNA viruses. RdRp containing polypeptide, encoded by 3546–5106 nt in ORF1, was
shown to interact with the 3ʹNCR of HEV genomic RNA and synthesizes the complementary strand
in vitro.225 Rehman et al.226 demonstrated the localization of RdRp to the endoplasmic reticulum (ER),
suggesting that HEV replicates probably in the ER in the cytosolic compartment of the cells.
4.1.8.1.7 HVR
The hypervariable region overlaps with the proline-rich sequence located between the N-terminus of the
X-domain and the C-terminal portion of the putative PCP domain. It varies both in length and sequence
among different HEV strains. HEV can tolerate small deletions in the HVR229; however, the replica-
tion efficiency of HVR deletion mutants was found to be reduced when tested in Huh7 cells.230 HVR
sequences were found to be interchangeable between different HEV genotypes, resulting in differential
replication efficiencies. From these observations, it was suggested that the HVR probably could influence
the efficiency of HEV replication by interacting with viral and/or host factors.
4.1.8.1.9 ORF3 Protein
HEV ORF3 is translated from the bicistronic subgenomic RNA.211 It overlaps with ORF2 at its 3ʹ end
and is most variable among the different HEV strains. It is a small 114-amino acid phosphoprotein that
remains associated with the cytoskeleton.236 ORF3 contains two large hydrophobic domains—D1 (amino
acids 7–23) and D2 (amino acids 28–53) at the N-terminus—and two proline-rich domains—P1 (amino
acids 66–77) and P2 (amino acids 95–111) toward the end.237,238 Of these, the cysteine-rich D1 domain
was shown to be required for the association of ORF3 to the cytoskeleton244 to bind microtubules239 and
52 Laboratory Models for Foodborne Infections
mitogen-activated protein kinase (MAPK)-phosphates,240 while the D2 domain was shown to bind to the
hemopexin and proposed to aid in viral infection by affecting cellular iron homeostasis.241 The C-terminal
region of the ORF3 protein is multifunctional and appears to be involved in virion morphogenesis and
pathogenesis. Using an in vitro replication system, it was shown that the ORF3 protein is not essential for
replication in cell lines;242,243 however, intrahepatic inoculations of these constructs in rhesus macaques
revealed that ORF3 is essential for the establishment of infection.211 ORF3 protein was detected on the
surface of HEV particles produced in cell culture, and these ORF3-coated viral particles were seen to be
lighter than uncoated particles, indicating a probable interaction with lipids.244 ORF3 coat was present on
the virus particles present in the serum, but not in the feces of HEV-infected individuals, suggesting that
the lipid-ORF3 “coat” was shed as the virus passed through the enteric system due to exposure to bile salts.
An intact PSAP motif was shown to be required for the formation of membrane-associated HEV particles
with the ORF3 protein on their surface.244 Taken together, these results suggest that the ORF3 protein is a
multifunctional protein that appears to play an important role in both HEV replication and pathogenesis.
4.1.8.2.2 3ʹNCR
The interaction of viral RdRp with 3ʹNCR and the adjacent region containing two stem-loop (SL)
structures (SL1 and SL2) and its importance in virus replication were documented by Agrawal et al.225
Using gel shift assays, they could demonstrate that RdRp bound specifically to 3ʹNCR, while 3ʹNCR
without a poly A tail failed to bind with RdRp. Similarly, a single nucleotide change in the SL2 of the
genotype 1 viral genome significantly affected virus replication, indicating the important role of both
poly A tail and SL structures in HEV replication.244,245
4.1.8.2.4 Subgenomic RNA
Graff et al.211 detected two positive-sense RNA species, approximately 7.3 and 2.2 kb in size, in HEV
replicating cells. The 2.2-kb subgenomic RNA was capped, and its 5ʹ end matched the nucleotide number
5122 in genotype 1, SAR55 HEV genome. With these results, this group proposed the ORF2 start at 5145
and 5131 as the ORF3 start, and the translation of ORF3 and ORF3 proteins from the same bicistronic
subgenomic mRNA. This model was confirmed by using the intrahepatic inoculation of wild-type and
mutant genotype 3 swine HEV replicons into pigs by Huang et al.210
entry. It is suggested that the C-terminal portion of pORF2 may bind to heat shock cognate protein 70
(HSPC70) on the cell surface to initiate cell entry.247 Heparan sulfate proteoglycans (HSPGs) appear to
be required as attachment factors.248 The intracellular trafficking following entry is also poorly under-
stood; HSP90 and tubulin appear to be involved in this process.249 A recent study has documented that
HEV enters through clathrin-mediated endocytosis.250 However, it is still not clear whether these are
specific receptors or just attachment factors. Holla et al.251 have recently reported that following inter-
nalization, the HEV-LP initially traffics into Rab5-positive compartments en route to acidic lysosomal
compartments. HEV entry requires dynamin-2, clathrin, membrane cholesterol, and actin, but is inde-
pendent of factors associated with macropinocytosis.
Once viral RNA is released in the cytosol, ORF1 translation is initiated by the cap-dependent recruitment
of ribosomes. It is not clear whether the ORF1 encoded nonstructural polyprotein functions as a single pro-
tein or whether it is processed into individual functional units. However, the regions predicted to encode viral
methyltransferase, helicase, and RdRp produce functionally active proteins when expressed in heterologous
systems. The genomic RNA is copied into a negative-sense RNA intermediate by viral RdRp. These RNA
intermediates then serve as templates for the synthesis of genomic as well as subgenomic p ositive-sense
RNA species. Genomic intermediates have been detected in replicon-transfected cells220 in the livers of
experimentally infected macaques252 and pigs.155 It was demonstrated using a replicon system that there is
an alternate cycle of positive- and negative-sense RNA synthesis.253 Single subgenomic RNA is translated
to synthesize ORF2 and ORF3 proteins.210,211,245 The ORF2 protein packages the genomic positive-sense
RNA into progeny virions. Immunocapture PCR analysis showed the association of ORF3 on the surface
of the cell-culture-generated HEV, which also showed a lower density than the ORF3 deficient virus. These
observations suggested that the ORF3 protein is present on the virion surface in association with cellular
lipids and probably plays some role during viral egress.254 Emerson et al.243 and Nagashima et al.255 have
documented the importance of the PSAP motif within the P2 domain of the ORF3 protein in the virus
egress and the probable role of SRC homology 3 signaling pathways in HEV maturation and egress. PSAP
motif is required for the formation of membrane-associated HEV particles with ORF3 on their surface,
which is mediated by cellular Tsg101 protein.256,257 These findings suggest that HEV follows the vacuolar
protein sorting pathway and uses cellular proteins such as Tsg101 for its release from infected cells.
The second report of an infectious clone from the Sar-55 strain of genotype 1 HEV (Pakistan) was
reported by Emerson et al.244 This recombinant genome could establish infection in rhesus macaques and
chimpanzees, and developed symptoms like hepatitis. They used capped transcripts for these studies. It
was controversial whether capping of RNA transcripts was essential for the infectivity since a previous
study by Panda et al. showed the successful generation of infectious particles using uncapped RNA tran-
scripts. Subsequently, Emerson et al.258 reported that HEV capped full-genome transcripts are 32–38 times
more efficiently translated than their uncapped counterparts. Several primate cell lines such as PLC/PRF/5,
Huh-7, Caco-2, HepG2/3CA, FKRHK, Vero, and AGMK were shown to support HEV replication; how-
ever, none of the nonprimate cells were able to support replication, indicating the requirement of species-
specific factors for HEV replication. Further studies from the same group using an infectious cDNA tool
demonstrated that the highly conserved regions of HEV ORF3 harbor cis-reactive regulatory elements
and that mutations in this region eliminate ORF2 and ORF3 synthesis. A single unique nucleotide change
within the stem structure at the 3ʹ end of the HEV genome similarly reduced the efficiency of replication
in both Huh-7 cells and rhesus macaques.245,257 Quantitative measurement of HEV replication became pos-
sible due to the development of fused ORF2-GFP, ORF2-luciferase subgenomic HEV replicons.246,258,259
Successful replication of a virus in a host is a complex phenomenon. With the wider range of host spe-
cies for genotype 3 and 4 HEVs, it was evident that these viruses are comparatively flexible in their host
specificity than genotype 1 and 2 viruses which have a very narrow host range. It is still not understood
how HEV determines its host specificity at the molecular level. A recent report proposes that the host
restriction for the genotype 1 virus could be due to efficiency of the virus to synthesize the ORF2 protein
in the given host.260 Lack of compatibility between cell surface receptor and receptor-binding region
(456–605 amino acids) in the capsid protein of the virus was suggested as the deciding factor. This group
used genotype 3 virus replicon (P6), developed from the Kernow C-1 virus,261 for developing chimeric
genotype 1/3 viral genomes. P6 is known to have a 171-nucleotide insertion (a sequence from the human
S17 ribosomal protein encoding gene) in the HVR region of the viral genome. It was previously reported
by this group that the P6 virus, that was isolated from a chronic HEV case, can cross the species bar-
rier.136 Ability of the S17 sequence to alter species specificity was confirmed by inserting this sequence in
SAR55 (genotype 1). This insertion enabled the replication of the chimeric virus in LLC-PK (pig) cells,
which otherwise was refractory to SAR55 1 virus infection.
Pigs being the most promising model for HEV studies, full-length infectious cDNA clones were also
developed from genotype 3 swine HEV.262 Capped transcripts from these clones were demonstrated to be
competent for forming infectious virions in Huh-7 cells, and these virions were able to establish infection
in pigs. Rescue of a genotype 4 human HEV from cloned cDNA in Huh7 cells and the infectivity of this
virus in HepG2/C3A cells were reported by Cordoba et al.263 Capped in vitro transcripts from this clone
could also establish infection in pigs. Zhu et al.264 recently reported cloning of the genotype 4 virus and
successful infection in rats.
Considering chickens as the practical animal model for HEV pathogenesis and replication studies,
cDNA clones from avian viruses were developed.265 Direct intrahepatic inoculation of RNA transcripts
from these infectious cDNA clones could establish infections in chickens and showed fecal virus shed-
ding, viremia, seroconversion, and histopathological lesions characteristic of avian HEV infection.
TABLE 4.2
Occurrence of HEV in Animal Species
Serological Evidence Experimental Applications as Animal
Animal Species HEV Genotype (Anti HEV) Susceptibility Model for HEV Studies
TABLE 4.2 (Continued)
Occurrence of HEV in Animal Species
Serological Evidence Experimental Applications as Animal
Animal Species HEV Genotype (Anti HEV) Susceptibility Model for HEV Studies
fecal shedding.150,151 The clinical presentation of hepatitis E infection in nonhuman primates was dose
dependent. Severity of infection was directly related to the infectivity titer of the challenge virus. For
demonstration of consistent hepatitis E infection in nonhuman primates, challenge virus doses at least
1000 times greater than the minimum dose needed for infection were required. Also, the oral route of
inoculation was not efficient and required a 10,000-fold higher challenge virus than that was used for
the intravenous inoculation of nonhuman primates.195 The observed high mortality (approximately 20%
particularly in the third trimester) of hepatitis E during human pregnancy was not reproducible in preg-
nant rhesus macaques.153,280
Rhesus monkeys infected with one Indian HEV isolate were found to be immune 1.5–2.75 years after
the primary infection when challenged with closely related Indian strains.1 Further, a long-term serologi-
cal follow-up of HEV immunity in experimentally infected rhesus monkeys indicated that anti-HEV IgG
titers declined to undetectable levels, but still protected the animals from homologous and heterologous
HEV challenge 5 years after the initial infection.64 It was previously documented that naturally acquired
anti-HEV in nonhuman primates renders them resistant to experimental infection.153
In general, rhesus monkeys and cynomolgus macaques were found to be most suitable model systems
for hepatitis E vaccine efficacy studies.195,281–286 Macaques immunized with an HEV candidate vaccine
developed using one genotype showed protection against homologous and heterologous HEV challenges.
The noteworthy vaccine studies that were taken further for phase 2 and phase 3 trials in populations of
hepatitis E endemic countries were from Nepal and China.
Cumbersome procurement procedures, limited animal resources, and ethical concerns have limited
the use of nonhuman primate models in HEV research today.
4.2.3.2 Pigs
HEV research and animal model development got a boost after Meng et al.159 discovered a novel strain
of the virus in pigs. As discussed earlier, swine serve as a major reservoir for zoonotic genotype 3 and 4
HEVs. Wild boars are assumed to be a natural reservoir of HEV. Although attempts to infect pigs with
genotype 1 or 2 of human HEVs were not successful,155 they were susceptible to genotype 3 and 4 human
HEV strains under experimental conditions.156,287 The pig model system has been successfully used to
study the structural and functional relationship of HEV genes and in understanding the mechanism of
HEV replication, pathogenesis, and species specificity.
Pathogenesis study with the specific-pathogen-free (SPF) pig model using a genotype 3 human HEV
strain revealed more severe and persistent hepatic lesions (multifocal lymphoplasmacytic hepatitis and
focal hepatocellular necrosis) than those infected with the swine HEV strain.156 Extra hepatic sites of
HEV replication observed were the small intestine, lymph nodes, and colon.288, 289 Pigs also shed virus in
feces, became viremic, and seroconverted to IgG anti-HEV when infected with human HEV genotype 4
(Taiwanese strain).287 The HEV pig model was used to determine the infectivity of the first infectious
clone of HEV genotype 3.262 This model also helped in the identification of the actual initiation site of the
HEV ORF3. Recently, HEV genotype 3 mutants with deletions in the HVR were shown to be infectious
in pigs, indicating dispensability of the HVR during HEV replication and infection. Thus, pigs seem to
be a promising model of HEV infection, as they are relatively inexpensive to house, feed, maintain, and
breed than nonhuman primates. However, since they do not reproduce hepatic disease with overt clinical
symptoms, their usefulness in understanding HEV pathogenesis will be limited.
4.2.3.3 Rodents
The rat strain of HEV was first identified in Hamburg, Germany, in 2009.41 Huang et al.290 have reported
the successful infection of Balb/c nude mice with a strain of genotype 4 HEV recovered from a pig; how-
ever, Li et al.291 failed to infect C57BL/6 mice with genotype 1, 3, and 4 HEV strains. Wistar rats could
be infected with the human strain of HEV,292 but subsequent studies aimed to confirm that the transmis-
sion of genotypes 1, 2, and 4 of human HEV and an avian HEV was unsuccessful. Mongolian gerbils
(desert rats) were recently shown to be susceptible to experimental infection with a genotype 4 strain
of HEV.293 Viremia and fecal virus shedding were detected in inoculated gerbils, and HEV antigen was
60 Laboratory Models for Foodborne Infections
detected in the liver, kidneys, spleen, and small intestine. Naturally infected rats do not show any HEV-
related illness. In experimental infections, laboratory rats infected with rat HEV showed seroconversion
and fecal shedding of the virus without any apparent clinical signs. Histopathological analysis of tissues
revealed mild portal inflammation, foci of parenchymal necrosis, and aggregates of lymphocytes, and
Kupffer cells within the lobules, indicating evidence of mild hepatitis consistent with human acute HEV
infection. Availability of a small animal model would give a boost to HEV research. Although labora-
tory mice and rats have been found to be susceptible to HEV, the lack of reproducibility is the major
drawback, and more work is needed in optimizing this model.
4.2.3.4 Rabbits
The potential of rabbits as an animal model was tested after the discovery of genotype 3 virus in farmed
rabbits from the Gansu province in China.26 Subsequent to these findings, HEV strains related to gen-
otype 3 were detected in farms as well as in wild rabbits in the United States and France. Rabbits
inoculated with rabbit HEV shed virus in the feces, become viremic, and have elevated levels of serum
ALT with irregular multifocal lymphohistiocytic infiltrates and local hepatocellular necrosis.294 Rabbits
experimentally inoculated with human HEV genotype 4 showed viremia and fecal virus shedding in 2/9
rabbits but in none inoculated with genotype 1 human HEV.294 This indicated that the rabbit model of
HEV infection may be useful for studying genotype 3 and 4 HEVs; however, its use for studying geno-
types 1 and 2 would be limited.
Han et al.295 utilized the rabbit model to see whether they can establish an animal model for chronic
HEV infections. On infecting SPF rabbits with a homologous rabbit HEV isolate and a heterologous
genotype 4 swine HEV, they observed the development of chronic hepatitis and an associated liver fibro-
sis in some rabbits infected with homologous HEV. Persistent fecal shedding of the virus and elevated
liver enzymes were evident for more than 6 months after infection in the chronic infections. The detec-
tion of both the positive-/negative-sense RNA and HEV antigen in the extra hepatic tissues such as
brain, stomach, duodenum, and kidney further confirmed similarity with human chronic HEV infection.
However, the underlying mechanism was not clear. In humans, HEV can establish chronic infections
only in immunocompromised individuals, this chronic disease pathology was not observed in the rabbits
infected with the heterologous genotype 4 swine HEV. Rabbit HEV has 31 amino acid insertions in the
X-domain, which probably broadened the tissue specificity of the virus as previously documented by
Shukla et al.261 in the case of chronic HEV infection in human patients.
4.2.3.5 Ferrets
HEV was first detected in ferrets (Mustela putorius) in Netherlands43 followed by detection in the United
States. Phylogenetic analysis has shown that ferret HEV strains are closer to rat strains. Nucleotide
sequence analyses indicated that the ferret HEV genome shares the highest nucleotide sequence identity
(72.3%) with rat HEV. A putative ORF4 was observed in the ferret HEV genome similar to the rat HEV
genome. Information about ferret HEV epidemiology, distribution, transmission, and pathogenesis is still
not available. Usefulness of ferret HEV as an animal model needs to be evaluated considering its small
size and extensive use as an animal model for studying the pathogenesis of respiratory viruses.
4.2.3.6 Chickens
Haqshenas et al.296 discovered avian HEV and facilitated the use of chickens as a model system for avian
HEV infection. Avian HEV has been shown to cross species barriers and infect turkeys.163 Although chick-
ens are not susceptible to human strains, upon inoculation with avian HEV, infection results in subcapsular
hemorrhages, focal lymphocytic periphlebitis, and phlebitis in the liver; extra hepatic sites of HEV repli-
cation are also evident.297 A recent study by Kwon et al.265 showed that an infectious cDNA clone of the
variant avian HEV (isolated from healthy chicken) could still induce histological liver lesions. This study
also proved that RNA transcripts may serve as an alternative for live virus in animal pathogenesis studies.
Hepatitis E Virus 61
Avian ORF2 capsid protein expressed in E. coli having a similar antigenic structure as that of
human HEV containing major neutralizing epitopes was assessed for immunogenicity in chickens.
All the tested mock-immunized control chickens developed typical avian HEV infection character-
ized by viremia, fecal virus shedding, and seroconversion to avian HEV antibodies, while none of the
tested chickens immunized with avian HEV capsid protein had detectable viremia, fecal virus shed-
ding, or observable gross hepatitis lesions. The results from this study suggested that immunization
of chickens with the avian HEV recombinant ORF2 capsid protein with aluminum as the adjuvant can
induce protective immunity against avian HEV infection.298 Overall, avian HEV seem to be useful
for HEV studies.
Animal models such as chimpanzees, rhesus/cynomolgus macaques, owl monkeys, rodents, pigs,
chickens, and rabbits have been used for studying HEV biology. Although nonhuman primates and pigs
seem to be useful animal models, ethical issues, high prices, and difficulties in handling, manipulating,
and housing these animals are major hurdles. Rats seem to be a promising laboratory model due to their
small size and easy handling. Table 4.2 shows different animal models being used for HEV studies.
4.3 Conclusions
HEV transmission occurs primarily due to contaminated water in developing countries while transmis-
sion via uncooked or partially cooked pig meat and meat products is mainly responsible for hepatitis E
infection in developed countries. For waterborne transmission, effective prevention and control depends
on ensuring a safe drinking water supply, adequate sanitation, and proper personal and environmental
hygiene. For the prevention of food-borne infections, a stringent screening of meat products is needed.
HEV infections in pigs are mostly silent and difficult to detect with traditional meat-screening proce-
dures. In such situations, a quick on-site diagnostic test is needed. PCR-based detection is comparatively
time consuming and needs well-equipped laboratories.
The proper and timely diagnosis of human HEV infections in nonendemic regions is technically chal-
lenging. Lack of consistency of serological tests and viral load quantification in terms of sensitivity and
specificity are the limiting factors. Future research should focus on developing broadly applicable sero-
logical and molecular assays for hepatitis E diagnosis.
The unsolved issues such as the relative importance of HEV transmission pathways, inactivation prop-
erties of the virus, and dose–response relationship of HEV infection need to be addressed. It is essential
to know the factors that lead to clinical hepatitis E infection in humans. For this, there is a need for a
practical animal model that would mimic the natural course of HEV infection in humans and its out-
come. With the ever expanding host range of HEV, we hope to find new animal strains that can be used
as model systems. Development of a reliable cell-culture system(s) that would support the complete life
cycle of HEV will help in knowing the virus better. Most therapeutic approaches and discovery efforts
to suppress HEV propagation will not progress unless we have these basic tools.
REFERENCES
1.
Arankalle VA, et al., Age-specific prevalence of antibodies to hepatitis A and E viruses in Pune, India,
1982 and 1992, J Infect Dis, 171, 447–450, 1995.
Chandra V, Taneja S, Kalia M, Jameel S, Molecular biology and pathogenesis of hepatitis E virus, J
2.
Biosci, 33, 451–464, 2008.
3.
Purcell RH, Emerson SU, Animal models of hepatitis A and E, ILAR J, 42, 161–177, 2001.
4.
Vishwanathan R, Infectious hepatitis in Delhi (1955–56): a critical study: epidemiology, Indian J Med
Res, 45, 49–58, 1957.
5.
Sreenivasan MA, et al., Epidemiological investigations of an outbreak of infectious hepatitis in
Ahmedabad city during 1975–76, Indian J Med Res, 67, 197–206, 1978.
6.
Khuroo MS, Study of an epidemic of non-A, non-B hepatitis possibility of another human hepatitis virus
distinct from post-transfusion non-A, non-B type, Am J Med, 68, 818–824, 1980.
62 Laboratory Models for Foodborne Infections
7. Wong DC, et al., Epidemic and endemic hepatitis in India: evidence for a non-A, non-B hepatitis etiol-
ogy, Lancet, 2, 876–879, 1980.
8. Chadha MS, Arankalle VA, Banerjee K, Follow up of cases of enterically transmitted non-A, non-B
hepatitis, J Assoc Phys India, 39, 651–652, 1991.
9. Naik SR, Aggarwal R, Salunke PN, Mehrotra NN, A large waterborne viral hepatitis E epidemic in
Kanpur, India, Bull World Health Organ, 70, 597–604, 1992.
10. Arankalle VA, Goverdhan MK, Banerjee K, Antibodies against hepatitis E virus in Old World monkeys,
J Viral Hepat, 1, 125–129, 1994.
11. Khuroo MS, Duermeyer W, Zargar SA, Ahanger MA, Shah MA, Acute sporadic non-A, non-B hepatitis
in India, Am J Epidemiol, 118, 360–364, 1983.
12. Arankalle VA, et al., Aetiology of acute sporadic non-A, non-B viral hepatitis in India, J Med Virol, 40,
121–125, 1993.
13. Chadha MS, Walimbe AM, Chobe LP, Arankalle VA, Comparison of etiology of sporadic acute and
fulminant viral hepatitis in hospitalized patients in Pune, India during 1978–81 and 1994–97, Indian
Gastroenterol, 22, 11–15, 2003.
14. Kim JH, Nelson KE, Panzner U, Kasture Y, Labrique AB, Wierzba TF, A systematic review of the epi-
demiology of hepatitis E virus in Africa, BMC Infect Dis, 14, 308, 2014.
15. Teshale EH, et al., A large outbreak of hepatitis E in northern Uganda, Emerg Infect Dis, 16, 126–129, 2010.
16. MMWR, Investigation of hepatitis E outbreak among refugees—Upper Nile, South Sudan, 2012–2013,
MMWR, 62(29), 581–586, 2013.
17. Guthmann JP, et al., A large outbreak of hepatitis E among a displaced population in Darfur, Sudan,
2004: the role of water treatment methods, Clin Infect Dis, 42, 1685–1691, 2006.
18. Purcell RH, Emerson SU, Hepatitis E: an emerging awareness of an old disease, J Hepatol, 48, 495–503,
2008.
19. Okamoto H, Genetic variability and evolution of hepatitis E virus, Virus Res, 127, 216–228, 2007.
20. Arankalle, VA, Chadha, MS, Mehendale, SM, Banerjee, K, Outbreak of enterically transmitted non-A,
non-B hepatitis among schoolchildren, Lancet, 2, 1199–1200, 1988.
21. Hyams KC, et al., Acute sporadic hepatitis E in Sudanese children: analysis based on a new western blot
assay, J Infect Dis, 165, 1001–1005, 1992.
22. Arankalle VA, Chadha MS, Dama BM, Tsarev SA, Purcell RH, Banerjee K, Role of immune serum
globulins in pregnant women during an epidemic of hepatitis E, J Viral Hepat, 5, 199–204, 1998.
23. Clayson ET, et al., Rates of hepatitis E virus infection and disease among adolescents and adults in
Kathmandu, Nepal, J Infect Dis, 176, 763–766, 1997.
24. Cossaboom C, Cordoba L, Dryman B, Meng X-J, Hepatitis E virus in rabbits, Virginia, USA, Emerg
Infect Dis, 17, 2047–2049, 2011.
25. Nakamura M, et al., Hepatitis E virus infection in wild mongooses of Okinawa, Japan: demonstration of
anti-HEV antibodies and a full genome nucleotide sequence, Hepatol Res, 34, 137–140, 2006.
26. Zhao C, et al., A novel genotype of hepatitis E virus, J Med Virol, 81, 1371–1379, 2009.
27. Wang Y, et al., A divergent genotype of hepatitis E virus in Chinese patients with acute hepatitis, J Gen
Virol, 80, 169–177, 1999.
28. Meng XJ, From barnyard to food table: the omnipresence of hepatitis E virus and risk for zoonotic infec-
tion and food safety, Virus Res, 161(1), 23–30, 2011.
29. Wang YC, et al., Prevalence, isolation, and partial sequence analysis of hepatitis E virus from domestic
animals in China, J Med Virol, 67, 516–521, 2002.
30. Balayan MS, Epidemiology of hepatitis E virus infection, J Viral Hepat, 4, 155–165, 1997.
31. Johne R, Plenge-Bönig A, Hess M, Ulrich RG, Reetz J, Schielke A, Detection of a novel hepatitis E-like
virus in faeces of wild rats using a nested broad-spectrum RT-PC, J Gen Virol, 91, 750–758, 2010.
32. Sonoda H, et al., Prevalence of hepatitis E virus (HEV) infection in wild boars and deer and genetic
identification of a genotype 3 HEV from a boar in Japan, J Clin Microbiol, 42, 5371–5374, 2004.
33. Wang H, et al., Recombination analysis reveals a double recombination event in hepatitis E virus, Virol
J, 7, 129, 2010.
34. Chandler JD, Riddell MA, Li F, Love RJ, Anderson DA, Serological evidence for swine hepatitis E virus
infection in Australian pig herds, Vet Microbiol, 68, 95–105, 1999.
35. de Deus N, et al., Hepatitis E virus infection dynamics and organic distribution in naturally infected pigs
in a farrow-to-finish farm, Vet Microbiol, 132, 19–28, 2008.
Hepatitis E Virus 63
36. Kaci S, Nockler K, Johne, R, Detection of hepatitis E virus in archived German wild boar serum sam-
ples, Vet Microbiol, 128, 380–385, 2008.
37. Martelli, F, et al., Detection of hepatitis E virus (HEV) in a demographic managed wild boar (Sus scrofa
scrofa) population in Italy, Vet Microbiol, 126, 74–81, 2008.
38. Michitaka K, et al., Prevalence of hepatitis E virus among wild boar in the Ehime area of western Japan,
Hepatol Res, 37, 214–220, 2007.
39. Rutjes SA, et al., Seroprevalence and molecular detection of hepatitis E virus in wild boar and red deer
in the Netherlands, J Virol Methods, 168, 197–206, 2010.
40. Drexler J, et al., Bats worldwide carry hepatitis E virus-related viruses that form a putative novel genus
within the family Hepeviridae, J Virol, 86, 9134–9147, 2012.
41. Takahashi M, et al., Analysis of the full-length genome of a hepatitis E virus isolate obtained from a
wild boar in Japan that is classifiable into a novel genotype, J Gen Virol, 92, 902–908, 2011.
42. Raj V, et al., Novel hepatitis E virus in ferrets, the Netherlands, Emerg Infect Dis, 18, 1369–1370, 2012.
43. Batts W, Yun S, Hedrick R, Winton J, A novel member of the family Hepeviridae from cutthroat trout
(Oncorhynchus clarkii), Virus Res, 158, 116–123, 2011.
44. Lu L, Li C, Hagedorn CH, Phylogenetic analysis of global hepatitis E virus sequences: genetic diversity,
subtypes and zoonosis, Rev Med Virol, 16, 5–36, 2006.
45. Purdy MA, Khudyakov YE, The molecular epidemiology of hepatitis E virus infection, Virus Res, 161,
31–39, 2011.
46. Johne R, et al., Novel hepatitis E virus genotype in Norway rats, Germany, Emerg Infect Dis, 16, 1452–
1455, 2011.
47. Meng X-J, Zoonotic and foodborne transmission of hepatitis E virus, Semin Liv Dis, 33, 41–49, 2013.
48. Yugo DM, Cossaboom CM, Meng XJ, Naturally occurring animal models of human hepatitis E virus
infection, ILAR J, 55, 187–199, 2014.
49. Marek A, Bilic I, Prokofieva I, Hess M, Phylogenetic analysis of avian hepatitis E virus samples
from European and Australian chicken flocks supports the existence of a different genus within the
Hepeviridae comprising at least three different genotypes, Vet Microbiol, 145, 54–61, 2010.
50. Kwon H, Sung H, Meng XJ, Serological prevalence, genetic identification, and characterization of the
first strains of avian hepatitis E virus from chickens in Korea, Virus Genes, 45, 237–245, 2012.
51. Clemente-Casares P, et al., Hepatitis E virus epidemiology in industrialized countries, Emerg Infect Dis,
9, 448–454, 2003.
52. Albinana-Gimenez N, et al., Distribution of human polyomaviruses, adenoviruses, and hepatitis E virus
in the environment and in a drinking water treatment plant, Environ Sci Technol, 40, 7416–7422, 2006.
53. Gironés R, Desire for information in the elderly: interactions with patients, family, and physicians, J
Canc Educ, doi:10.1007/s13187-014-0760-5, 2014.
54. Teo CG, Much meat, much malady: changing perceptions of the epidemiology of hepatitis E, Clin
Microbiol Infect, 16, 24–32, 2010.
55. Parashar D, Khalkar P, Arankalle VA, Survival of hepatitis A and E viruses in soil samples, Clin
Microbiol Infect, 17, e1–e4, 2011.
56. Rutjes SA, et al., Sources of hepatitis E virus genotype 3 in the Netherlands, Emerg Infect Dis, 15,
381–387, 2009.
57. McCreary C, Martelli F, Grierson S, Ostanello F, Nevel A, Banks M, Excretion of hepatitis E virus by pigs
of different ages and its presence in slurry stores in the United Kingdom, Vet Rec, 163, 261–265, 2008.
58. El-Esnawy N, Examination for hepatitis E virus in wastewater treatment plants and workers by nested
RT-PCR and ELISA, J Egypt Public Health Assoc, 75, 219–231, 2000.
59. Vaidya SR, Tilekar BN, Walimbe AM, Arankalle VA, Increased risk of hepatitis E in sewage workers
from India, J Occup Environ Med, 45, 1167–1170, 2003.
60. Arankalle VA, Chadha MS, Mehendale SM, Tungatkar SP, Epidemic hepatitis E: serological evidence
for lack of intrafamilial spread, Indian J Gastroenterol, 19, 24–28, 2000.
61. Aggarwal R, Naik SR, Hepatitis E: intrafamilial transmission versus waterborne spread, J Hepatol, 21,
718–723, 1994.
62. Robson SC, Adams S, Brink N, Woodruff B, Bradley D, Hospital outbreak of hepatitis E, Lancet, 339,
1424–1425, 1992.
63. Khuroo MS, Kamili S, Aetiology and prognostic factors in acute liver failure in India, J Viral Hepat, 10,
224–231, 2003.
64 Laboratory Models for Foodborne Infections
64. Arankalle VA, Paranjape S, Emerson SU, Purcell RH, Walimbe AM, Phylogenetic analysis of hepatitis
E virus isolates from India (1976–1993), J Gen Virol, 80, 1691–1700, 1999.
65. Boxall E, et al., Transfusion-transmitted hepatitis E in a “non-hyperendemic” country, Transfus Med,
16, 79–83, 2006.
66. Colson P, Coze C, Gallian P, Henry M, De Micco P, Tamalet C, Transfusion-associated hepatitis E,
France, Emerg Infect Dis, 13, 648–649, 2007.
67. Matsubayashi K, et al., A case of transfusion-transmitted hepatitis E caused by blood from a donor
infected with hepatitis E virus via zoonotic food-borne route, Transfusion, 48, 1368–3675, 2008.
68. Mitsui T, et al., Prevalence of hepatitis E virus infection among hemodialysis patients in Japan: evidence
for infection with a genotype 3 HEV by blood transfusion, J Med Virol, 74, 563–572, 2004.
69. Tamura A, et al., Persistent infection of hepatitis E virus transmitted by blood transfusion in a patient
with T-cell lymphoma, Hepatol Res, 37, 113–120, 2007.
70. Psichogiou M, et al., Hepatitis E virus infection in individuals at high risk of transmission of non-A,
non-B hepatitis and sexually transmitted diseases, Scand J Infect Dis, 28, 443–445, 1996.
71. Barzilai A, et al., Hepatitis E virus infection in hemophiliacs, J Med Virol, 46, 153–156, 1995.
72. Hauser L, et al., Hepatitis E transmission by transfusion of intercept blood system-treated plasma,
Blood, 123, 796–797, 2014.
73. Hewitt PE, et al., Hepatitis E virus in blood components: a prevalence and transmission study in south-
east England, Lancet, doi:10.1016/S0140-6736(14) 61034–61035, 2014.
74. Meng XJ, Hepatitis E virus: animal reservoirs and zoonotic risk, Vet Microbiol, 140, 256–265, 2010.
75. Bouquet J, et al., Close similarity between sequences of hepatitis E virus recovered from humans and
swine, France, 2008–2009, Emerg Infect Dis, 17, 2018–2025, 2011.
76. Balayan MS, Usmanov R, Zamyatina N, Brief report: experimental hepatitis E infection in domestic
pigs, J Med Virol, 32, 58–59, 1990.
77. Meng X-J, Zoonotic and xenozoonotic risks of the hepatitis E virus, Infect Dis Rev, 2, 35–41, 2000.
78. Vulcano A, et al., HEV prevalence in the general population and among workers at zoonotic risk in
Latium Region, Ann Ig, 19, 181–186, 2007.
79. Bouwknegt M, et al., Estimation of hepatitis E virus transmission among pigs due to contact-exposure,
Vet Res, 39, 40, 2008.
80. Galiana C, Fernandez-Barredo S, Garcia A, Gomez MT, Perez-Gracia MT, Occupational exposure to
hepatitis E virus (HEV) in swine workers, Am J Trop Med Hyg, 78, 1012–1015, 2008.
81. Yazaki Y, et al., Sporadic acute or fulminant hepatitis E in Hokkaido, Japan, may be food-borne, as sug-
gested by the presence of hepatitis E virus in pig liver as food, J Gen Virol, 84, 2351–2357, 2003.
82. Miyashita K, et al., Three cases of acute or fulminant hepatitis E caused by ingestion of pork meat and
entrails in Hokkaido, Japan: zoonotic food-borne transmission of hepatitis E virus and public health
concerns, Hepatol Res, 42, 870–878, 2012.
83. Li TC, et al., Hepatitis E virus transmission from wild boar meat, Emerg Infect Dis, 11, 1958–1960, 2005.
84. Matsuda H, Okada K, Takahashi K, Mishiro S, Severe hepatitis E virus infection after ingestion of
uncooked liver from a wild boar, J Infect Dis, 188, 944, 2003.
85. Tamada Y, Yano K, Yatsuhashi H, Inoue O, Mawatari F, Ishibashi H, Consumption of wild boar linked
to cases of hepatitis E, J Hepatol, 40, 869–870, 2004.
86. Hijioka S, Sato Y, Iwashita Y, Indou Y, A case of acute hepatitis E who had a history of frequent ingestion
of raw meat and viscera from wild deer and boars, Nihon Shokakibyo Gakkai Zasshi, 102, 723–728, 2005.
87. Colson P, et al., Pig liver sausage as a source of hepatitis E virus transmission to humans, J Infect Dis,
202, 825–834, 2010.
88. Renou C, et al., Association Nationale des Hépato-Gastroentérologues des Hôpitaux Généraux
(ANGH), Prospective study of hepatitis E virus infection among pregnant women in France, Virol J,
11, 68, 2014.
89. Berto A, et al., Hepatitis E virus in pork liver sausage, France, Emerg Infect Dis, 19, 264–266, 2013.
90. Kulkarni MA, Arankalle VA, The detection and characterization of hepatitis E virus in pig livers from
retail markets of India, J Med Virol, 80, 1387–1390, 2008.
91. Feagins AR, Opriessnig T, Guenette DK, Halbur PG, Meng XJ, Detection and characterization of infec-
tious hepatitis E virus from commercial pig livers sold in local grocery stores in the USA, J Gen Virol,
88, 912–917, 2007.
Hepatitis E Virus 65
92. Wenzel JJ, Preiss J, Schemmerer M, Huber B, Plentz A, Jilg W, Detection of hepatitis E virus (HEV)
from porcine livers in southeastern Germany and high sequence homology to human HEV isolate,
J Clin Virol, 52, 50–54, 2011.
93. Wilhelma B, et al., Preliminary molecular epidemiological 2 investigation of hepatitis E virus 3
sequences from Québec, Canada, Prev Vet Med, 118, 359–369, 2014.
94. Kaba M, et al., Hepatitis E virus infection in sheltered homeless persons, France, Emerg Infect Dis, 16,
1761–1763, 2010.
95. Okano H, et al., Characterization of sporadic acute hepatitis E and comparison of hepatitis E virus genomes
in acute hepatitis patients and pig liver sold as food in Mie, Japan, Hepatol Res, 44, E63–E76, 2014.
96. Di Bartolo I, Angeloni G, Ponterio E, Ostanello F, Ruggeri FM, Detection of hepatitis E virus in pork
liver sausages, Int J Food Microbiol, 193, 29–33, 2015.
97. Bouwknegt M, et al., Hepatitis E virus RNA in commercial porcine livers in the Netherlands, J Food
Prot, 70, 2889–2895, 2007.
98. Donia D, et al., Presence of hepatitis E RNA in mussels used as bio-monitors of viral marine pollution,
J Virol Methods, 186, 198–202, 2012.
99. Crossan C, Baker PJ, Craft J, Takeuchi Y, Dalton HR, Scobie L, Hepatitis E virus genotype 3 in shell-
fish, United Kingdom, Emerg Infect Dis, 18, 2085–2087, 2012.
100. Song YJ, et al., Analysis of complete genome sequences of swine hepatitis E virus and possible risk fac-
tors for transmission of HEV to humans in Korea, J Med Virol, 82, 583–591, 2010.
101. Li TC, Miyamura T, Takeda N, Detection of hepatitis E virus RNA from the bivalve Yamato-Shijimi
(Corbicula japonica) in Japan, Am J Trop Med Hyg, 76, 170–172, 2007.
102. Cacopardo B, Russo R, Preiser W, Benanti F, Brancati G, Nunnari A, Acute hepatitis E in Catania (east-
ern Sicily) 1980–1994, the role of hepatitis E virus, Infection, 25, 313–316, 1997.
103. Thorne ET, Williams E, Diseases and endangered species: the blackfooted ferret as a recent example,
Conserv Biol, 2, 6673, 1988.
104. Koizumi Y, et al., Infection of a Japanese patient by genotype 4 hepatitis E virus while traveling in
Vietnam, J Clin Microbiol, 42, 3883–3885, 2004.
105. Said B, et al., Hepatitis E Incident Investigation Team, Hepatitis E outbreak on cruise ship, Emerg Infect
Dis, 15, 1738–1744, 2009.
106. Brassard J, Gagné MJ, Généreux M, Côté C, Detection of human food-borne and zoonotic viruses on
irrigated, field-grown strawberries, Appl Environ Microbiol, 78, 3763–3766, 2012.
107. Kokkinos P, et al., Harmonised investigation of the occurrence of human enteric viruses in the leafy
green vegetable supply chain in three European countries, Food Environ Virol, 4, 179–191, 2012.
108. Emerson SU, Arankalle VA, Purcell RH, Thermal stability of hepatitis E virus, J Infect Dis, 192,
930–933, 2005.
109. Barnaud E, Rogee S, Garry P, Rose N, Pavio N, Thermal inactivation of infectious hepatitis E virus in
experimentally contaminated food, Appl Environ Microbiol, 78, 5153–5159, 2012.
110. Balayan MS, et al., Evidence for a virus in non-A, non-B hepatitis transmitted via the fecal-oral route,
Intervirology, 20, 23–31, 1983.
111. Chauhan A, Jameel S, Dilawari JB, Chawla YK, Kaur U, Ganguly NK, Hepatitis E virus transmission
to a volunteer, Lancet, 341, 149–150, 1993.
112. Aggarwal R, Jameel S, Hepatitis E, Hepatology, 54, 2218–2222, 2011.
113. Pal R, Aggarwal R, Naik SR, Das V, Das S, Naik S, Immunological alterations in pregnant women with
acute hepatitis E, J Gastroenterol Hepatol, 20, 1094–1101, 2005.
114. Bhatia V, Singhal A, Panda SK, Acharya SK, A 20-year single-center experience with acute liver failure
during pregnancy: is the prognosis really worse, Hepatology, 48, 1577–1585, 2008.
115. Navaneethan U, Al Mohajer M, Shata MT, Hepatitis E and pregnancy: understanding the pathogenesis,
Liver Int, 28, 1190–1199, 2008.
116. Kar P, et al., Does hepatitis E viral load and genotypes influence the final outcome of acute liver failure
during pregnancy? Am J Gastroenterol, 103, 2495–2501, 2008.
117. Kumar A, Beniwal M, Kar P, Sharma JB, Murthy NS, Hepatitis E in pregnancy, Int J Gynecol Obstet,
85, 240–241, 2004.
118. Kumar Acharya S, et al., Hepatitis E virus (HEV) infection in patients with cirrhosis is associated with
rapid decompensation and death, J Hepatol, 46, 387–394, 2007.
66 Laboratory Models for Foodborne Infections
119. Khuroo MS, Teli MR, Skidmore S, Sofi MA, Khuroo MI, Incidence and severity of viral hepatitis in
pregnancy, Am J Med, 70, 252–255, 1981.
120. Myint H, et al., A clinical and epidemiological study of an epidemic of non-A, non-B hepatitis in
Rangoon, Am J Trop Med Hyg, 34, 1183–1189, 1985.
121. Jilani N, et al., Hepatitis E virus infection and fulminant hepatic failure during pregnancy, J Gastroenterol
Hepatol, 22, 676–682, 2007.
122. Patra S, Kumar A, Trivedi SS, Puri M, Sarin SK, Maternal and fetal outcomes in pregnant women with
acute hepatitis E virus infection, Ann Intern Med, 147, 28–33, 2007.
123. Chibber RM, Usmani MA, Al-Sibai MH, Should HEV infected mothers breast feed? Arch Gynecol
Obstet, 270, 15–20, 2004.
124. Stoszek SK, et al., Prevalence of and risk factors for hepatitis C in rural pregnant Egyptian women,
Trans R Soc Trop Med Hyg, 100, 102–107, 2006.
125. Andersson G, Carlbring P, Grimlund A, Predicting treatment outcome in Internet versus face to face
treatment of panic disorder, Comput Human Behav, 24, 1790–1801, 2008.
126. Anty R, et al., First case report of an acute genotype 3 hepatitis E infected pregnant woman living in
South-Eastern France, J Clin Virol, 54, 76–78, 2012.
127. Mateos Lindemann ML, et al., Fulminant hepatitis E in a woman taking oral contraceptive medication,
Am J Trop Med Hyg, 82, 12–15, 2010.
128. Renou C, Pariente A, Cadranel JF, Nicand E, Pavio N, Clinically silent forms may partly explain the
rarity of acute cases of autochthonous genotype 3c hepatitis E infection in France, J Clin Virol, 51(2),
139–141, 2011.
129. Mizuo H, et al., Possible risk factors for the transmission of hepatitis E virus and for the severe form of
hepatitis E acquired locally in Hokkaido, Japan, J Med Virol, 76, 341–349, 2005.
130. Jeblaoui A, Haim-Boukobza S, Pause A, Mokhtari C, Nicand E, Roque-Afonso A, Emerging hepatitis
E genotype 4 infection in France, J Hepatol, 58, S405, 2013.
131. Purcell RH, et al., Pathobiology of hepatitis E: lessons learned from primate models, Emerg Microbes
Infect, 2, e9, doi:10.1038/emi.2013.9, 2013.
132. Inoue J, et al., Analysis of the full-length genome of genotype 4 hepatitis E virus isolates from patients
with fulminant or acute self-limited hepatitis E, J Med Virol, 78, 476–484, 2006.
133. Inoue J, et al., Nucleotide substitutions of hepatitis E virus genomes associated with fulminant hepatitis
and disease severity, Tohoku J Exp Med, 218, 279–284, 2009.
134. Takahashi K, et al., Virulent strain of hepatitis E virus genotype 3, Japan, Emerg Infect Dis, 15,
704–709, 2009.
135. Mishra N, Walimbe AM, Arankalle VA, Hepatitis E virus from India exhibits significant amino acid
mutations in fulminant hepatic failure patients, Virus Genes, 46, 47–53, 2013.
136. Shukla P, et al., Cross-species infections of cultured cells by hepatitis E virus and discovery of an infec-
tious virus-host recombinant, Proc Natl Acad Sci USA, 108, 2438–2443, 2011.
137. Nguyen HT, et al., A naturally occurring human/hepatitis E recombinant virus predominates in serum
but not in feces of a chronic hepatitis E patient and has a growth advantage in cell culture, J Gen Virol,
93, 526–530, 2012.
138. Haagsma EB, et al., Chronic hepatitis E virus infection in liver transplant recipients, Liver Transplant,
14, 547–553, 2008.
139. Kamar N, et al., Hepatitis E virus and chronic hepatitis in organ-transplant recipients, N Engl J Med,
358, 811–817, 2008.
140. Dalton HR, Bendall R, Keane F, Tedder R, Ijaz S, Persistent carriage of hepatitis E virus in patients with
HIV infection, N Engl J Med, 361, 1025–1027, 2009.
141. le Coutre P, Meisel H, Hofmann J, Reactivation of hepatitis E infection in a patient with acute lympho-
blastic leukaemia after allogeneic stem cell transplantation, Gut, 58, 699–702, 2009.
142. Ollier L, Tieulie N, Sanderson F, Chronic hepatitis after hepatitis E virus infection in a patient with non-
Hodgkin lymphoma taking Rituximab, Ann Intern Med, 150, 430–431, 2009.
143. Yugo DM, Meng XJ, Hepatitis E virus: foodborne, waterborne and zoonotic transmission, Int J Environ
Res Public Health, 10, 4507–4533, 2013.
144. Geng Y, et al., Persistent hepatitis E virus genotype 4 infection in a child with acute lymphoblastic leu-
kemia, Hepatitis Monthly, 14, e15618, 2014.
Hepatitis E Virus 67
145. Naik A, Gupta N, Goel D, Ippagunta SK, Sharma RK, Aggarwal R, Lack of evidence of hepatitis
E virus infection among renal transplant recipients in a disease-endemic area, J Viral Hepat, 20,
e138–e140, 2013.
146. Bradley DW, et al., Enterically transmitted non-A, non-B hepatitis: serial passage of disease in cynomol-
gus macaques and tamarins and recovery of disease-associated 27- to 34-nm virus like particles, Proc
Natl Acad Sci USA, 84, 6277–6281, 1987.
147. Ticehurst J, et al., Infection of owl monkeys (Aotus trivirgatus) and cynomolgus monkeys (Macaca
fascicularis) with hepatitis E virus from Mexico, J Infect Dis, 165, 835–845, 1992.
148. Tsarev SA, et al., Variation in course of hepatitis E in experimentally infected cynomolgus monkeys,
J Infect Dis, 167, 1302–1306, 1993.
149. Uchida T, et al., Serial transmission of a putative causative virus of enterically transmitted non-A, non-B
hepatitis to Macaca fascicularis and Macaca mulatto, Jpn J Exp Med, 60, 13–21, 1990.
150. Krawczynski K, Bradley DW, Enterically transmitted non-A, non-B hepatitis: identification of virus-
associated antigen in experimentally infected cynomolgus macaques, J Infect Dis, 159, 1042–1049, 1989.
151. Longer CF, et al., Experimental hepatitis E: pathogenesis in cynomolgus macaques (Macaca fascicu-
laris), J Infect Dis, 168, 602–609, 1993.
152. Arankalle VA, et al., Etiological association of a virus-like particle with enterically transmitted non-A,
non-B hepatitis, Lancet, 1, 550–554, 1988.
153. Tsarev SA, et al., Experimental hepatitis E in pregnant rhesus monkeys: failure to transmit hepatitis
E virus (HEV) to offspring and evidence of naturally acquired antibodies to HEV, J Infect Dis, 172,
31–37, 1995.
154. Li RC, et al., Seroprevalence of hepatitis E virus infection, rural southern People’s Republic of China,
Emerg Infect Dis, 12(11), 1682–1688, 2006.
155. Meng XJ, et al., Genetic and experimental evidence for cross-species infection by swine hepatitis E
virus, J Virol, 72, 9714–9721, 1998.
156. Halbur PG, et al., Comparative pathogenesis of infection of pigs with hepatitis E viruses recovered from
a pig and a human, J Clin Microbiol, 39, 918–923, 2001.
157. Arankalle VA, Chobe LP, Joshi MV, Chadha MS, Kundu B, Walimbe AM, Human and swine hepatitis
E viruses from western India belong to different genotypes, J Hepatol, 36 417–425, 2002.
158. Arankalle VA, Chobe LP, Chadha MS, Type-IV Indian swine HEV infects rhesus monkeys, J Viral
Hepat, 13, 742–745, 2006.
159. Meng J, Dubreuil P, Pillot J, A new PCR-based seroneutralization assay in cell culture for diagnosis of
hepatitis E, J Clin Microbiol, 35, 1373–1377, 1997.
160. Leblanc D, et al., Presence of hepatitis E virus in a naturally infected swine herd from nursery to slaugh-
ter, Int J Food Microbiol, 117, 160–166, 2007.
161. Arankalle VA, Chobe LP, Walimbe AM, Yergolkar PN, Jacob GP, Swine HEV infection in South India
and phylogenetic analysis (1985–1999), J Med Virol, 69, 391–396, 2003.
162. Huang FF, et al., Heterogeneity and seroprevalence of a newly identified avian hepatitis E virus from
chickens in the United States, J Clin Microbiol, 40, 4197–4202, 2002.
163. Sun ZF, et al., Generation and infectivity titration of an infectious stock of avian hepatitis E virus
(HEV) in chickens and cross-species infection of turkeys with avian HEV, J Clin Microbiol, 42, 2658–
2662, 2004.
164. Peralta B, et al., Anti-HEV antibodies in domestic animal species and rodents from Spain using a geno-
type 3-based ELISA, Vet Microbiol, 137, 66–73, 2009.
165. Nan Y, Yu Y, Ma Z, Khattar SK, Fredericksen B, Zhang YJ, Hepatitis E virus inhibits type I interferon
induction by ORF1 product, J Virol, 88, 11924–11932, 2014.
166. Nan Y, et al., Enhancement of interferon induction by ORF3 product of hepatitis E virus, J Virol, 88,
8696–8705, 2014.
167. Yu C, et al., Pathogenesis of hepatitis E virus and hepatitis C virus in chimpanzees: similarities and
differences, J Virol, 84, 11264–11278, 2010.
168. Srivastava R, Aggarwal R, Bhagat MR, Chowdhury A, Naik S, Alterations in natural killer cells and
natural killer T cells during acute viral hepatitis E, J Viral Hepat, 15, 910–916, 2008.
169. Tripathy AS, Das R, Rathod SB, Gurav YK, Arankalle VA, Peripheral T regulatory cells and cytokines
in hepatitis E infection, Eur J Clin Microbiol Infect Dis, 31, 179–184, 2012.
68 Laboratory Models for Foodborne Infections
170. Rathod SB, Thanapati S, Arankalle VA, Tripathy AS, Suppressive activity and altered conventional phe-
notype markers/mediators of regulatory T cells in patients with self-limiting hepatitis E, J Viral Hepat,
21, 141–151, 2014.
171. Prabhu SB, et al., Study of cellular immune response against hepatitis E virus (HEV), J Viral Hepat, 18,
587–594, 2011.
172. Husain MM, Aggarwal R, Kumar D, Jameel S, Naik S, Effector T cells immune reactivity among
patients with acute hepatitis E, J Viral Hepat, 18, e603–e608, 2011.
173. Srivastava R, Aggarwal R, Sachdeva S, Alam MI, Jameel S, Naik S, Adaptive immune responses during
acute uncomplicated and fulminant hepatitis E, J Gastroenterol Hepatol, 26, 306–311, 2011.
174. Saravanabalaji S, Tripathy AS, Dhoot RR, Chadha MS, Kakrani AL, Arankalle VA, Viral load, anti-
body titers and recombinant open reading frame 2 protein-induced TH1/TH2 cytokines and cellular
immune responses in self-limiting and fulminant hepatitis E, Intervirology, 52, 78–85, 2009.
175. Suneetha PV, et al., Effect of peptide pools on effector functions of antigen-specific CD8+ T cells,
J Immunol Methods, 342, 33–48, 2009.
176. Mishra N, Arankalle VA, Association of polymorphisms in the promoter regions of TNF-α (−308) with
susceptibility to hepatitis E virus and TNF-α (−1031) and IFN-γ (+874) genes with clinical outcome of
hepatitis E infection in India, J Hepatol, 55, 1227–1234, 2011.
177. Dawson GJ, Chau KH, Cabal CM, Yarbough PO, Reyes GR, Mushahwar IK, Solid-phase enzyme-
linked immunosorbent assay for hepatitis E virus IgG and IgM antibodies utilizing recombinant anti-
gens and synthetic peptides, J Virol Methods, 38, 175–186, 1992.
178. Tam AW, et al., Hepatitis E virus (HEV): molecular cloning and sequencing of the full-length viral
genome, Virology, 185, 120–131, 1991.
179. Favorov MO, et al., Serologic identification of hepatitis E virus infections in epidemic and endemic set-
tings, J Med Virol, 36, 246–250, 1992.
180. Chau KH, Dawson GJ, Bile KM, Magnius LO, Sjogren MH, Mushahwar IK, Detection of IgA class
antibody to hepatitis E virus in serum samples from patients with hepatitis E virus infection, J Med
Virol, 40, 334–338, 1993.
181. Orrù G, Masia G, Orrù G, Romanò L, Piras V, Coppola RC, Detection and quantitation of hepatitis E
virus in human faeces by real-time quantitative PCR, J Virol Methods, 118, 77–82, 2004.
182. Enouf V, et al., Validation of single real-time TaqMan PCR assay for the detection and quantitation of
four major genotypes of hepatitis E virus in clinical specimens, J Med Virol, 78, 1076–1082, 2006.
183. Gyarmati P, Mohammed N, Norder H, Blomberg J, Belák S, Widén F, Universal detection of hepatitis
E virus by two real-time PCR assays: TaqMan and primer-probe energy transfer, J Virol Methods, 146,
226–235, 2007.
184. Zhao C, et al., Comparison of real-time fluorescent RT-PCR and conventional RT-PCR for the detection
of hepatitis E virus genotypes prevalent in China, J Med Virol, 79, 1966–1973, 2007.
185. Jothikumar N, Cromeans TL, Robertson BH, Meng XJ, Hill VR, A broadly reactive one-step real-time
RT-PCR assay for rapid and sensitive detection of hepatitis E virus, J Virol Methods, 131, 65–71, 2006.
186. Huang S, et al., Profile of acute infectious markers in sporadic hepatitis E, PLoS One, 5, e13560, 2010.
187. Baylis SA, Hanschmann KM, Blumel J, Nubling CM, Standardization of hepatitis E virus (HEV)
nucleic acid amplification technique-based assays: an initial study to evaluate a panel of HEV strains
and investigate laboratory performance, J Clin Microbiol, 49, 1234–1239, 2011.
188. Emerson SU, Purcell RH, Hepatitis E virus, Rev Med Virol, 13, 145–154, 2003.
189. Arankalle VA, Lole KS, Deshmukh TM, Chobe LP, Gandhe SS, Evaluation of human (genotype 1) and
swine (genotype 4)-ORF2-based ELISAs for anti-HEV IgM and IgG detection in an endemic country
and search for type 4 human HEV infections, J Viral Hepat, 14, 435–445, 2007.
190. Herremans M, et al., Swine-like hepatitis E viruses are a cause of unexplained hepatitis in the
Netherlands, J Viral Hepat, 14, 140–146, 2007.
191. Yarbrough PO, et al., Hepatitis E virus: identification of type-common epitopes, J Virol, 65,
5790–5797, 1991.
192. Zhou YH, Purcell RH, Emerson SU, An ELISA for putative neutralizing antibodies to hepatitis E virus
detects antibodies to genotypes 1, 2, 3, and 4, Vaccine, 22, 2578–2585, 2004.
193. Drobeniuc J, et al., Serologic assays specific to immunoglobulin M antibodies against hepatitis E virus:
pangenotypic evaluation of performances, Clin Infect Dis, 1(51), e24–e27, 2010.
Hepatitis E Virus 69
194. Abravanel F, et al., Performance of anti-HEV assays for diagnosing acute hepatitis E in immunocompro-
mised patients, J Clin Virol, 58, 624–628, 2013.
195. Tsarev SA, et al., Infectivity titration of a prototype strain of hepatitis E virus in cynomolgus monkeys,
J Med Virol, 43, 135–142, 1994.
196. Shrestha MP, et al., Safety and efficacy of a recombinant hepatitis E vaccine, N Engl J Med, 356,
895–903, 2007.
197. Purcell RH, et al., Pre-clinical immunogenicity and efficacy trial of a recombinant hepatitis E vaccine,
Vaccine, 21, 2607–2615, 2003.
198. Zhu FC, et al., Efficacy and safety of a recombinant hepatitis E vaccine in healthy adults: a large-scale,
randomised, double-blind placebo-controlled, phase 3 trial, Lancet, 376, 895–902, 2010.
199. Kamar N, et al., Hepatitis E virus-induced neurological symptoms in a kidney-transplant patient with
chronic hepatitis, Am J Transplant, 10, 1321–1324, 2010.
200. Neukam K, et al., Chronic hepatitis E in HIV patients: rapid progression to cirrhosis and response to oral
ribavirin, Clin Infect Dis, 57, 465–468, 2013.
201. Mallet V, et al., Brief communication: case reports of ribavirin treatment for chronic hepatitis E, Ann
Intern Med, 153, 85–89, 2010.
202. Pischke S, Behrendt P, Bock C, Jilg W, Manns MP, Wedemeyer H, Hepatitis E in Germany—an under-
reported infectious disease, Deutsches Ärzteblatt, 111, 577–583, 2014.
203. Dalton HR, Keane FE, Bendall R, Mathew J, Ijaz S, Treatment of chronic hepatitis E in a patient with
HIV infection, Ann Intern Med, 155, 479–480, 2011.
204. Peron JM, Dalton H, Izopet J, Kamar N, Acute autochthonous hepatitis E in western patients with under-
lying chronic liver disease: a role for ribavirin? J Hepatol, 54, 1323–1324, 2011.
205. Kumar A, Saraswat VA, Hepatitis E and acute-on-chronic liver failure, J Clin Exp Hepatol, 3, 225–230, 2013.
206. Mahtab EA, et al., Podoplanin deficient mice show a rhoa-related hypoplasia of the sinus venosus myo-
cardium including the sinoatrial node, Dev Dyn, 238, 183–193, 2009.
207. Duseja A, et al., Nonalcoholic fatty liver in a developing country is responsible for significant liver dis-
ease, Hepatology, 52, 2248–2249, 2010.
208. El Sayed Zaki M, Othman W, Role of hepatitis E infection in acute on chronic liver failure in Egyptian
patients, Liver Int, 31, 1001–1005, 2011.
209. Goyal R, Kumar A, Panda SK, Paul SB, A Charaya SK, Ribavirin therapy for hepatitis E virus induced
acute on chronic liver failure: a preliminary report, Antivir Ther, 17, 1091–1096, 2012.
210. Huang YW, Opriessnig T, Halbur PG, Meng XJ, Initiation at the third in-frame AUG codon of open read-
ing frame 3 of the hepatitis E virus is essential for viral infectivity in vivo, J Virol, 81, 3018–3026, 2007.
211. Graff J, Torian U, Nguyen H, Emerson SU, A bicistronic subgenomic mRNA encodes both the ORF2
and ORF3 proteins of hepatitis E virus, J Virol, 80, 5919–5926, 2006.
212. Koonin EV, Gorbalenya AE, Purdy MA, Rozanov MN, Reyes GR, Bradley DW, Computer-assisted assign-
ment of functional domain in the nonstructural polyprotein of hepatitis E virus: delineation of an additional
group of positive-strand RNA plant and animal viruses, Proc Natl Acad Sci USA, 89, 8259–8263, 1992.
213. Magden J, et al., Virus-specific mRNA capping enzyme encoded by hepatitis E virus, J Virol, 75,
6249–6255, 2001.
214. Kabrane-Lazizi Y, et al., Evidence for widespread infection of wild rats with hepatitis E virus in the
United States, Am J Trop Med Hyg, 61, 331–335, 1999.
215. Karpe YA, Lole KS, NTPase and 5′ to 3′ RNA duplex-unwinding activities of the hepatitis E virus heli-
case domain, J Virol, 84, 3595–3602, 2010.
216. Ansari IH, et al., Cloning, sequencing, and expression of the hepatitis E virus (HEV) nonstructural open
reading frame 1 (ORF1), J Med Virol, 60, 275–283, 2000.
217. Panda SK, Ansari IH, Durgapal H, Agrawal S, Jameel S, The in vitro-synthesized RNA from a cDNA
clone of hepatitis E virus is infectious, J Virol, 74, 2430–2437, 2000.
218. Ropp SL, Tam AW, Beames B, Purdy M, Frey TK, Expression of the hepatitis E virus ORF1, Arch Virol,
145, 1321–1337, 2000.
219. Sehgal D, Thomas S, Chakraborty M, Jameel S, Expression and processing of the hepatitis E virus
ORF1 nonstructural polyprotein, Virol J, 3, 38, 2006.
220. Suppiah S, Zhou Y, Frey TK, Lack of processing of the expressed ORF1 gene product of hepatitis E
virus, Virol J, 8, 245, 2011.
70 Laboratory Models for Foodborne Infections
221. Perttila J, Spuul P, Ahola T, Early secretory pathway localization and lack of processing for hepatitis E
virus replication protein pORF1, J Gen Virol, 94, 807–816, 2013.
222. Karpe YA, Lole KS, Deubiquitination activity associated with hepatitis E virus putative papain-like
cysteine protease, J Gen Virol, 92, 2088–2092, 2011.
223. Paliwal D, Panda SK, Kapur N, Varma SP, Durgapal H, Hepatitis E virus (HEV) protease: a
chymotrypsin-like enzyme that processes both non-structural (pORF1) and capsid (pORF2) protein,
J Gen Virol, 95, 1689–1700, 2014.
224. Karpe YA, Lole KS, RNA 5′-triphosphatase activity of the hepatitis E virus helicase domain, J Virol,
84, 9637–9641, 2010.
225. Agrawal S, Gupta D, Panda SK, The 3′ end of hepatitis E virus (HEV) genome binds specifically to the
viral RNA dependent RNA polymerase (RdRp), Virology, 282, 87–101, 2001.
226. Rehman S, Kapur N, Durgapal N, Panda SK, Subcellular localization of hepatitis E virus (HEV) repli-
case, Virology, 370, 77–92, 2008.
227. Egloff M-P, et al., Structural and functional basis for ADP-ribose and poly(ADP-ribose) binding by viral
macro domains, J Virol, 80, 8493–8502, 2006.
228. Neuvonen M, Ahola T, Differential activities of cellular and viral macro domain proteins in binding of
ADP-ribose metabolites, J Mol Biol, 385, 212–225, 2009.
229. Pudupakam RS, Huang YW, Opriessnig T, Halbur PG, Pierson FW, Meng XJ, Deletions of the hyper-
variable region (HVR) in open reading frame 1 of hepatitis E virus do not abolish virus infectivity:
evidence for attenuation of HVR deletion mutants in vivo, J Virol, 83, 384–395, 2009.
230. Pudupakam RS, et al., Mutational analysis of the hyper variable region of hepatitis E virus reveals its
involvement in the efficiency of viral RNA replication, J Virol, 85, 10031–10040, 2011.
231. Zafrullah M, Ozdener MH, Kumar R, Panda SK, Jameel S, Mutational analysis of glycosylation, mem-
brane translocation, and cell surface expression of the hepatitis E virus ORF2 protein, J Virol, 73,
4074–4082, 1999.
232. Jameel S, Zafrullah M, Ozdener MH, Panda SK, Expression in animal cells and characterization of the
hepatitis E virus structural proteins, J Virol, 70, 207–216, 1996.
233. Surjit M, Jameel S, Lal SK, The ORF2 protein of hepatitis E virus binds the 5′ region of viral RNA,
J Virol, 78, 320–328, 2004.
234. Yamashita, T, et al., Biological and immunological characteristics of hepatitis E virus-like particles
based on the crystal structure, Proc Natl Acad Sci USA, 106, 12986–12991, 2009.
235. Guu TS, et al., Structure of the hepatitis E virus-like particle suggests mechanisms for virus assembly
and receptor binding, Proc Natl Acad Sci USA, 106, 12992–12997, 2009.
236. Zafrullah M, Ozdener MH, Panda SK, Jameel S, The ORF3 protein of hepatitis E virus is a phospho-
protein that associates with the cytoskeleton, J Virol, 71, 9045–9053, 1997.
237. Ahmad I, Holla RP, Jameel S, Molecular virology of hepatitis E virus, Virus Res, 161, 47–58, 2011.
238. Holla RP, Ahmad I, Ahmad Z, Jameel S, Molecular virology of hepatitis E virus, Semin Liver Dis, 33,
3–14, 2013.
239. Kannan H, Fan S, Patel D, Bossis I, Zhang YJ, The hepatitis E virus open reading frame 3 product
interacts with microtubules and interferes with their dynamics, J Virol, 83, 6375–6382, 2009.
240. Kar-Roy A, Korkaya H, Oberoi R, Lal S K, Jameel S, The hepatitis E virus open reading frame 3
protein activates ERK through binding and inhibition of the MAPK phosphatase, J Biol Chem, 279,
28345–28357, 2004.
241. Ratra R, Kar-Roy A, Lal SK, The ORF3 protein of hepatitis E virus interacts with hemopexin by means
of its 26 amino acid N-terminal hydrophobic domain II, Biochemistry, 47, 1957–1969, 2008.
242. Emerson SU, Nguyen H, Torian U, Purcell RH, ORF3 protein of hepatitis E virus is not required for
replication, virion assembly, or infection of hepatoma cells in vitro, J Virol, 80, 10457–10464, 2006.
243. Emerson SU, Nguyen HT, Torian U, Burke D, Engle R, Purcell RH, Release of genotype 1 hepatitis E
virus from cultured hepatoma and polarized intestinal cells depends on open reading frame 3 protein
and requires an intact PXXP motif, J Virol, 84, 9059–9069, 2010.
244. Emerson SU, et al., Recombinant hepatitis E virus genomes infectious for primates: importance of cap-
ping and discovery of a cis-reactive element, Proc Natl Acad Sci USA, 98, 15270–15275, 2001.
245. Graff J, et al., In vitro and in vivo mutational analysis of the 3′-terminal regions of hepatitis E virus
genomes and replicons, J Virol, 79, 1017–1026, 2005.
Hepatitis E Virus 71
246. Cao D, Huang YW, Meng XJ, The nucleotides on the stem–loop RNA structure in the junction region
of the hepatitis E virus genome are critical for virus replication, J Virol, 84, 13040–13044, 2010.
247. Zhou Y, Emerson SU, P.302 Heat shock cognate protein 70 may mediate the entry of hepatitis E virus
into host cells, J Clin Virol, 36(Suppl 2), S155, 2006.
248. Kalia M, Chandra V, Rahman SA, Sehgal D, Jameel S, Heparan sulfate proteoglycans are required
for cellular binding of the hepatitis E virus ORF2 capsid protein and for viral infection, J Virol, 83,
12714–12724, 2009.
249. Zheng ZZ, et al., Role of heat-shock protein 90 in hepatitis E virus capsid trafficking, J Gen Virol, 91,
1728–1736, 2010.
250. Kapur N, Thakral D, Durgapal H, Panda SK, Hepatitis E virus enters liver cells through receptor-
dependent clathrin-mediated endocytosis, J Viral Hepat, 19, 436–448, 2012.
251. Holla P, Ahmad I, Ahmed Z, Jameel S, Hepatitis E virus enters liver cells through a dynamin-2, clathrin
and membrane cholesterol-dependent pathway, Traffic, 16, 398–416, 2015.
252. Nanda SK, Panda SK, Durgapal H, Jameel S, Detection of the negative strand of hepatitis E virus RNA
in the livers of experimentally infected rhesus monkeys: evidence for viral replication, J Med Virol, 42,
237–240, 1994.
253. Varma SP, Kumar A, Kapur N, Durgapal H, Acharya SK, Panda SK, Hepatitis E virus replication
involves alternating negative- and positive-sense RNA synthesis, J Gen Virol, 92, 572–581, 2011.
254. Yamada K, et al., Construction of an infectious cDNA clone of hepatitis E virus strain JE03–1760F that
can propagate efficiently in cultured cells, J Gen Virol, 90, 457–462, 2009.
255. Nagashima S, et al., A PSAP motif in the ORF3 protein of hepatitis E virus is necessary for virion
release from infected cells, J Gen Virol, 92, 269–278, 2011.
256. Nagashima S, et al., Tumour susceptibility gene 101 and the vacuolar protein sorting pathway are
required for release of hepatitis E virions, J Gen Virol, 92, 2838–2848, 2011.
257. Graff J, et al., The open reading frame 3 gene of hepatitis E virus contains a cis-reactive element and
encodes a protein required for infection of macaques, J Virol, 79, 6680–6689, 2005.
258. Emerson SU, et al. (Eds.), Virus Taxonomy, VIIIth Report of the ICTV, Elsevier/Academic Press,
London, 851–855, 2004.
259. Thakral D, Nayak B, Rehman S, Durgapal H, Panda SK, Replication of a recombinant hepatitis E virus
genome tagged with reporter genes and generation of a short-term cell line producing viral RNA and
proteins, J Gen Virol, 86, 1189–1200, 2005.
260. Nguyen, HT, Shukla P, Torian U, Faulk K, Emerson SU, Hepatitis E virus genotype 1 infection of swine
kidney cells in vitro is inhibited at multiple levels, J Virol, 88, 868–877, 2014.
261. Shukla P, et al., Adaptation of a genotype 3 hepatitis E virus to efficient growth in cell culture depends
on an inserted human gene segment acquired by recombination, J Virol, 86, 5697–5707, 2012.
262. Huang YW, Haqshenas G, Kasorndorkbua C, Halbur PG, Emerson SU, Meng XJ, Capped RNA tran-
scripts of full-length cDNA clones of swine hepatitis E virus are replication competent when transfected
into Huh7 cells and infectious when intrahepatically inoculated into pigs, J Virol, 79, 1552–1558, 2005.
263. Cordoba L, et al., Rescue of a genotype 4 human hepatitis E virus from cloned cDNA and character-
ization of intergenotypic chimeric viruses in cultured human liver cells and in pigs, J Gen Virol, 93,
2183–2194, 2012.
264. Zhu Y, et al., Infectivity of a genotype 4 hepatitis E virus cDNA clone by intrahepatic inoculation of
laboratory rats, Vet Microbiol, 166, 405–411, 2013.
265. Kwon HM, et al., Construction of an infectious cDNA clone of avian hepatitis E virus (avian HEV)
recovered from a clinically healthy chicken in the United States and characterization of its pathogenicity
in specific-pathogen-free chickens, Vet Microbiol, 147, 310–319, 2011.
266. Tam AW, et al., In vitro propagation and production of hepatitis E virus from in vivo-infected primary
macaque hepatocytes, Virology, 215, 1–9, 1996.
267. Tam AW, et al., In vitro infection and replication of hepatitis E virus in primary cynomolgus macaque
hepatocytes, Virology, 238, 94–102, 1997.
268. Huang R, et al., Cell culture of sporadic hepatitis E virus in China, Clin Diagn Lab Immunol, 6, 729–733, 1999.
269. Huang RT, Li DR, Wei J, Huang XR, Yuan XT, Tian X, Isolation and identification of hepatitis E virus
in Xinjiang, China, J Gen Virol, 73, 1143–1148, 1992.
270. Kazachkov Yu, et al., Hepatitis E virus in cultivated cells, Arch Virol, 127, 399–402, 1992.
72 Laboratory Models for Foodborne Infections
271. Li DR, Huang RT, Pang JJ, Yuan XT, Li XY, Biological feature and genome analysis of HEV isolated
by cell-culture. In: Buisson Y, Coursaget P, Kane M (Eds.), Enterically-Transmitted Hepatitis Viruses.
La Simarre, Joue-les-Tours, 349–361, 1996.
272. Meng J, Guinet R, Pillot J, Infection of PLC/PRF-5 cells with the hepatitis E virus. In: Buisson
Y, Coursaget P, Kane M. (Eds.), Enterically-Transmitted Hepatitis Viruses, La Simarre, Joue-les-Tours,
336–345, 1996.
273. Wei S, Walsh P, Huang R, To SS, 93G, a novel sporadic strain of hepatitis E virus in South China iso-
lated by cell culture, J Med Virol, 61, 311–318, 2000.
274. Tanaka T, Takahashi M, Kusano E, Okamoto H, Development and evaluation of an efficient cell-culture
system for hepatitis E virus, J Gen Virol, 88, 903, 2007.
275. Rogee S, et al., New models of hepatitis E virus replication in human and porcine hepatocyte cell lines,
J Gen Virol, 94, 549–558, 2013.
276. Tanaka T, et al., Development and characterization of a genotype 4 hepatitis E virus cell culture
system using a HE-Jf5/15f strain recovered from a fulminant hepatitis patient, J Clin Microbiol, 47,
1906–1910, 2009.
277. Zhang HY, Chen DS, Wu YQ, He QG, Chen HC, Liu ZF, Both swine and human cells are capable to
support the replication of swine hepatitis E virus type 4 in vitro, Virus Res, 158, 289–293, 2011.
278. Takahashi M, et al., Hepatitis E Virus (HEV) strains in serum samples can replicate efficiently in cul-
tured cells despite the coexistence of HEV antibodies: characterization of HEV virions in blood circula-
tion, J Clin Microbiol, 48, 1112–1125, 2010.
279. McCaustland KA, et al., Hepatitis E virus infection in chimpanzees: a retrospective analysis, Arch
Virol, 145, 1909–1918, 2000.
280. Arankalle VA, Favorov MO, Chadha MS, Phule DM, Banerjee K, Rhesus monkeys infected with hepa-
titis E virus (HEV) from the former USSR are immune to subsequent challenge with an Indian strain of
HEV, Acta Virol, 37, 515–518, 1993.
281. Tsarev SA, et al., Recombinant vaccine against hepatitis E: dose response and protection against heter-
ologous challenge, Vaccine, 15, 1834–1838, 1997.
282. Zhang M, et al., Recombinant vaccine against hepatitis E: duration of protective immunity in rhesus
macaques, Vaccine, 20, 3285–3291, 2002.
283. Zhou YH, Purcell RH, Emerson SU, Truncated ORF2 protein contains the most immunogenic site
on ORF2: antibody responses to non-vaccine sequences following challenge of vaccinated and non-
vaccinated macaques with hepatitis E virus, Vaccine, 23, 3157–3165, 2005.
284. Li TC, Suzuki Y, Ami Y, Dhole TN, Miyamura T, Takeda N, Protection of cynomolgus monkeys
against HEV infection by oral administration of recombinant hepatitis E virus-like particles, Vaccine,
22, 370–377, 2004.
285. Im SW, et al., A bacterially expressed peptide prevents experimental infection of primates by the hepa-
titis E virus, Vaccine, 19, 3726–3732, 2001.
286. Ma H, et al., Immunogenicity and efficacy of a bacterially expressed HEV ORF3 peptide, assessed by
experimental infection of primates, Arch Virol, 154, 1641–1648, 2009.
287. Feagins AR, Opriessnig T, Huang YW, Halbur PG, Meng XJ, Cross-species infection of specific-
pathogen-free pigs by a genotype 4 strain of human hepatitis E virus, J Med Virol, 80, 1379–1386, 2008.
288. Williams TP, et al., Evidence of extra hepatic sites of replication of the hepatitis E virus in a swine
model, J Clin Microbiol, 39 3040–3046, 2001.
289. Huang FF, Pierson FW, Toth TE, Meng XJ, Construction and characterization of infectious cDNA
clones of a chicken strain of hepatitis E virus (HEV), avian HEV, J Gen Virol, 86, 2585–2593, 2005.
290. Huang F, et al., Experimental infection of Balb/c nude mice with hepatitis E virus, BMC Infect Dis, 9,
93, doi: 10.1186/1471-2334-9-93, 2009.
291. Li TC, Suzaki Y, Ami Y, Tsunemitsu H, Miyamura T, Takeda N, Mice are not susceptible to hepatitis E
virus infection, J Vet Med Sci, 70, 1359–1362, 2008.
292. Maneerat Y, Clayson ET, Myint KS, Young GD, Innis BL, Experimental infection of the laboratory rat
with the hepatitis E virus, J Med Virol, 48, 121–128, 1996.
293. Li WG, et al., Experimental infection of Mongolian gerbils by a genotype 4 strain of swine hepatitis E
virus, J Med Virol, 81, 1591–1596, 2009.
294. Ma H, et al., Experimental infection of rabbits with rabbit and genotypes 1 and 4 hepatitis E viruses,
PLoS One, 5, e9160, 2010.
Hepatitis E Virus 73
295. Han J, et al., SPF rabbits infected with rabbit hepatitis E virus isolate experimentally showing the chro-
nicity of hepatitis, PLoS One, 9, e99861, 2014.
296. Haqshenas G, Shivaprasad HL, Woolcock PR, Read DH, Meng XJ, Genetic identification and charac-
terization of a novel virus related to human hepatitis E virus from chickens with hepatitis-splenomegaly
syndrome in the United States, J Gen Virol, 82, 2449–2462, 2001.
297. Billam P, Pierson FW, Li W, LeRoith T, Duncan RB, Meng XJ, Development and validation of a
negative-strand-specific reverse transcription-PCR assay for detection of a chicken, J Clin Microbiol,
46, 2630–2634, 2008.
298. Guo H, Zhou EM, Sun ZF, Meng XJ, Protection of chickens against avian hepatitis E virus (avian HEV)
infection by immunization with recombinant avian HEV capsid protein, Vaccine, 25, 2892–2899, 2007.
5
Noroviruses: Laboratory Surrogates for
Determining Survival and Inactivation
CONTENTS
5.1 Introduction..................................................................................................................................... 75
5.2 HNoVs and the Use of Surrogates................................................................................................... 76
5.3 Human Feeding Studies and Associated Drawbacks..................................................................... 77
5.4 Animal Viruses as HNoV Surrogates and Model Systems to Study HNoVs................................. 77
5.4.1 Feline Calicivirus............................................................................................................... 77
5.4.2 Murine Norovirus............................................................................................................... 80
5.4.3 Tulane Virus....................................................................................................................... 84
5.4.4 Porcine Sapovirus............................................................................................................... 85
5.4.5 Bovine Noroviruses............................................................................................................ 86
5.4.6 Rabbit Caliciviruses........................................................................................................... 86
5.4.7 Norovirus-Like Particles ................................................................................................... 86
5.5 Bacteriophage MS2......................................................................................................................... 87
5.6 Conclusions..................................................................................................................................... 88
Acknowledgments..................................................................................................................................... 88
References................................................................................................................................................. 88
5.1 Introduction
Noroviruses (NoVs) belong to the Caliciviridae family that currently comprises of five genera: Norovirus,
Sapovirus, Lagovirus, Vesivirus, and Nebovirus, with potentially additional genera as new sequence
information becomes available on unclassified viruses [1,2]. The name “Norovirus” was shortened from
the initial “Norwalk-like” virus term that was associated with an epidemic outbreak that occurred in
Norwalk, Ohio, in 1968 in an elementary school [3]. Infected school children upon returning home trans-
mitted the secondary infection to family members resulting in 50% of students and teachers developing
nonbacterial gastroenteritis.
NoVs are small viruses about 27–32 nm in size and round in structure with an icosahedral sym-
metry. The human norovirus (HNoV) genome contains a single-stranded positive-sense RNA about
7.6 kb in length that is enclosed in a capsid without an envelope [3]. The capsid is made of 90 cap-
somers protruding from the shell that has 90 dimers of capsid protein. The genome has three open
reading frames (ORFs). ORF1 (nucleotides 146–5359) is about 5 kb in size and encodes a ∼200 kDa
nonstructural polyprotein. This nonstructural protein is cleaved to produce the N-terminal protein, the
enzyme nucleoside triphosphatase, a 3A-like protein, a genome-linked viral protein (VpG), a 3C-like
protease, and RNA-dependent RNA-polymerase (RdRp) [4]. ORF2 (nucleotides 5346–6935) is ∼1.8 kb
in size and encodes the 57 kDa major structural capsid viral protein VP1; ORF3 (nucleotides 6938–
7573) is ∼0.6 kb in size and encodes a small 22 kDa minor viral structural protein, VP2, reported to
package the genome into virions [5,6]. The NoV genus at the time of this submission, is composed of
75
76 Laboratory Models for Foodborne Infections
five genogroups based on sequence analysis: genogroup I (GI) (prototype Norwalk virus); GII (pro-
totype Snow Mountain virus); GIII (prototype bovine enteric calicivirus); GIV (prototype Alphatron
and Ft. Lauderdale viruses); and GV (prototype Murine NoV) [7,8].
From the five genogroups of NoV, genogroups GI, GII, and GIV specifically infect humans (and are
commonly referred to as human noroviruses or HNoV) and as indicated above, GIII includes bovine
enteric calicivirus that infects cattle, and GV is associated with the infection of mice. There have been
at least 32 genetic clusters identified based on the amino acid-sequence similarity between these geno-
groups [1,9]. Eight genotypes are recognized in GI and 19 genotypes are recognized in GII, and of these,
HNoV genotype GII.4 has been the most prevalent during the past few decades. Indeed, the majority of
HNoV outbreaks are reported to be caused by the GII.4 genocluster and its variants, where in 2002, a
GII.4 variant, called the Farmington Hill strain, was responsible for 80% of acute HNoV outbreaks in the
United States [10]. In addition, the Hunter strain GII.4 variant was found to be circulating in Australia,
Europe, and Asia in 2004, and then the Sakai strain (reported in Southeast Asia) and Minerva strain
(found in the United States and the Netherlands) were found to be circulating in 2006, replacing the
Hunter GII.4 variant [11]. Another HNoV GII.6 strain (previously not reported in California) classified
as Seacroft was reported to be responsible for causing illness in 30 individuals at a local university in
Los Angeles, California, in October 2008 [12]. It is also noteworthy to report that outbreaks of the HNoV
genogroup GII.4 in health-care settings in the United States, Europe, and Oceania were responsible for
70%–80% of all HNoV outbreaks [13]. The HNoV strains continue to remain a worldwide concern and
are continually evolving, with these emerging HNoV GII.4 variants also becoming virulent and known
to cause death in the elderly and immunocompromised [14].
HNoVs have a very short incubation period of 18–48 h, with typically self-limiting infection that lasts
for up to 72 h, characterized by mild gastroenteritis symptoms that include nausea, vomiting, abdomi-
nal cramps, fever, and malaise. “Winter vomiting disease” is the common seasonal syndrome associ-
ated with HNoV infection, with peak outbreaks in winter, even though HNoV transmission reportedly
occurs all year round [15]. It is also important to keep in mind that from a public health aspect, infected
individuals can shed viruses for 72 h (and even longer, greater than a week) after signs of the first symp-
toms appear [16]. Outbreaks have been reported in restaurants, cruise ships, health-care settings, closed
environments, and nursing homes, either due to the consumption of at-risk ready-to-eat or undercooked
contaminated foods, such as seafood, shellfish, produce, deli items, and bakery products, and also due to
environmental transmission and person-to-person transmission [17]. Therefore, adequate control strate-
gies, proper sanitation, food processing, storage, and hygienic practices of workers are important to
prevent further transmission especially in food environments and health-care settings.
pH of 2.0–4.0 encountered in the gastrointestinal tract [6], unlike HNoVs that are known to be resistant
to low pH conditions, alternate surrogates and model systems are being researched.
FCV-F9 has been utilized as a surrogate in determining improved inactivation strategies against
HNoVs in foods and on contact surfaces, such as chemical, nonthermal, and thermal treatments (refer
to Tables 5.1 through 5.3). Broad-spectrum contact surface disinfectants have been evaluated using
TABLE 5.1
Effect of Chemicals on the Feline Calicivirus (FCV-F9) Used as a Cultivable HNoV Surrogate to Develop
Disinfection and Control Strategies
Virus Treatment Reduction (log PFU/mL) References
Feline calicivirus Hydrogen peroxide vapors; 25 mL volume: [70,109]
70% ethanol, 5 min 2.6
Formaldehyde; 0.7%, 60 min at 20°C 4 [110]
Glutaraldehyde; 0.5%, 1 min at room temperature 5 [26]
Bacoban WB (quaternary ammonium compound); 4 [110]
2%; 240 min at 20°C
Benzalkonium chloride; 0.25 mg/mL, 2 h at room 3.1 [28]
temperature
β-Propiolactone; 0.1%, 60 min at 22°C 5.2 [111]
Free chlorine; 500 ppm, 10 min at room temperature 1.9 [112]
TABLE 5.2
Effects of Surface Inactivation Methods Using Feline Calicivirus (FCV-F9) as a Cultivable HNoV Surrogate
Virus Medium Treatment Reduction References
Feline calicivirus Plastic surfaces Steam (130°C)–ultrasound 30–50 kHz for 3 s 4.8 log [36]
Low protein virus stock Gamma radiation 5.9 log PFU [113]
1 µM methylene blue; human plasma, 5 min 3.9 log [114]
illumination
Virus + cell-culture media 200 MPa—0°C, 4 min 4.4 log [115]
250 MPa—0°C, 4 min 4.8 log
300 MPa—10°C, 3 min 5 log
Glass dishes ClO2 gas (1.7 mg/h at 25°C for 5 h) 3 log [116]
TABLE 5.3
Evaluation of Heat Treatments on Feline Calicivirus (FCV-F9) as a HNoV Surrogate
Virus Medium Inactivation Parameters References
Feline calicivirus Buffer 56°C; D-value 6.36 [73]
65°C; D-value 0.32
72°C; D-value 0.11
Seafood 56°C; D-value 3.33 [75]
65°C; D-value 0.33
72°C; D-value 0.07
Spinach 56°C; D-value 5.83 [40]
65°C; D-value 0.27
72°C; D-value 0.15
Virus stock 56°C; D-value 3.473 [6]
63°C; D-value 0.435
72°C; D-value 0.166
Noroviruses: Laboratory Surrogates for Determining Survival and Inactivation 79
FCV-F9 for their antiviral properties. Potassium peroxymonosulfate (KPMS) at concentrations of 5, 10,
and 20 mg/mL was reported to reduce FCV-F9 titers to undetectable levels from an initial 5 and 7 log
PFU/mL within 2 h at room temperature [28]. Similarly, benzalkonium chloride (BAC) at 0.1, 0.25, and
0.5 mg/mL was reported to reduce FCV-F9 by 2.87, 3.08, and 3.25 log PFU/mL from an initial 7 log PFU/mL
after 2 h at room temperature [28]. D’Souza and Su reported that FCV-F9 could be inactivated to non-
detectable levels (6.84 log PFU/mL reduction) after a contact time of 30 s with 10% bleach (0.6% sodium
hypochlorite, 5000 ppm available chlorine) and 2% trisodium phosphate (TSP) [29]. A neutral solution
of electrochemically activated anolyte Ecasol was reported to reduce FCV-F9 titers by more than 5 log
within 1 min of treatment at room temperature [30].
FCV-F9 was also used as a model virus in a study to determine the effect of chlorine dioxide (ClO2) gas
against the virus inoculated on glass surfaces in a test room (39 m3), where a constant low-concentration
ClO2 gas was produced [31]. These researchers found that a low-concentration ClO2 gas (mean 0.05 ppmv,
0.14 mg/m3) inactivated FCV-F9 (by >2 logs) in the wet state on glass dishes within 5 h. Other research-
ers used FCV-F9 as a cultivable HNoV surrogate and model to determine the effect of aqueous ClO2
at concentrations ranging from 0.2 to 0.8 mg/L at pH 7 and 20°C, and found that at a concentration of
0.8 mg/L ClO2, a complete elimination of FCV-F9 was obtained in 2 min, while lower concentrations
of 0.2 mg/L required 30 min for inactivation [32]. FCV-F9 was also used as a model to determine the
inactivation of enteric viruses by ozone treatment, where for a 4-log (99.99%) inactivation of FCV at 5°C
and pH 7 about <0.01–0.03 mg/L min of ozone was required [the Ct value which is described as the con-
centration of ozone multiplied by the virus contact time, using application of the efficiency factor Hom
(EFH) model] [33]. In another study using FCV-F9 as an HNoV surrogate, ozone treatment at 6.25 ppm
for 4 min showed >6 log TCID50/mL reduction of FCV-F9 in water and ∼2-log TCID50/mL on lettuce and
green onions [34].
High-pressure processing and ultraviolet (UV) light treatments are commonly used processing methods
in the food industry. A study reported a decrease of 3.48, 3.82, and 4.62 log TCID50 in FCV-F9 titers on let-
tuce after exposure to UV light at doses of 16, 40, and 75 mW s/cm2, respectively [35]. High HPP treatments
of 500 and 600 MPa were reported to reduce FCV-F9 titers to undetectable levels after 1, 3, 5, and 7 min,
while lower pressures of 300 and 400 MPa caused 1.13 and 0.55 log decreases after 1 min, respectively [32].
A combination of pressurized steam (130°C) and high-power ultrasound (30–50 kHz) was reported to
reduce FCV-F9 titers on plastic surfaces by >99.99% corresponding to >4.8-log after 3 s of treatment [36].
FCV-F9 in milk showed reductions of ≥4 log PFU/mL after high-pressure homogenization at 300 MPa,
∼1.3 log PFU/mL at 250 MPa, and insignificant reduction at ≤200 MPa [37]. These researchers showed that
in orange juice, FCV-F9 was reduced by ≥4 and ∼1 log PFU/mL only after high-pressure homogenization at
300 and 250 MPa, respectively [37]. FCV-F9 in PBS at titers of ∼4 log PFU/mL after a treatment at 20 kHz
of high-intensity ultrasound (HIU) for 5 min could be reduced to undetectable levels [38].
FCV-F9 has also been used as a model/surrogate for small, round-structured viruses on lettuce, where
the lettuce was inoculated with FCV-F9 by immersion to simulate contamination from irrigation or wash
water. Then the lettuce was subjected to electron beam irradiation at various dose levels [39]. These
researchers reported the D10-value of FCV-F9 on lettuce to be 2.95 kGy. When FCV-F9 was used as a
model to determine heat inactivation of HNoVs in spinach, the D-values calculated from the first-order
model (50°C–72°C) ranged from 0.15 to 17.39 min [40].
Significant research has been conducted on natural oils and extracts for their antiviral properties,
including 0.25% natural mulberry (Morus alba) juice that caused a 50% reduction in FCV-F9 titers [41].
Essential oil thymol at 0.5% and 1% was reported to reduce FCV-F9 titers to undetectable levels from an
initial 6 log TCID50 after 2 h at 37°C [42]. Black raspberry juice showed antiviral activity when Crandell
Reese feline kidney cells were pretreated with the juice or directly or FCV-F9 itself [43]. After treatment
with cranberry juice and cranberry proanthocyanidins at 0.30, 0.60, and 1.20 mg/mL for 1 h at room
temperature, FCV-F9 from initial 5 log PFU/mL was reported to be reduced to undetectable levels [44].
When FCV-F9 was used as a surrogate to determine the effect of grape-seed extract (GSE) as a wash to
disinfect lettuce and jalapeno peppers, FCV-F9 was reported to be reduced by 2.33, 2.58, and 2.71 log
PFU on lettuce, and 2.20, 2.74, and 3.05 log PFU on peppers after 1 min using 0.25, 0.50, and 1 mg/mL
GSE, respectively [45]. Thus, FCV-F9 has also been extensively used as a model to understand antiviral
options.
80 Laboratory Models for Foodborne Infections
The survivability/persistence of FCV is surprisingly high on surfaces and was detectable for up to
28 days at room temperature [26]. FCV-F9 was detected on three surfaces (formica, stainless steel,
and ceramic) for up to 7 days postinoculation at room temperature [46]. A persistence/survival study of
FCV-F9 at storage (7°C) and room temperatures over 70 days showed that the infectivity of FCV-F9 at
room temperature decreased after 6 h, 1 day, and 7 days by 1 log, 3 log, and to below the detection limit
(∼4.5 log), respectively [47]. However, FCV-F9 was shown to survive longer at 7°C, with only a reduction
of 2 log after 7 days and a titer decrease of 3 log after 70 days being reported [47]. Similarly, FCV-F9
at 6 log PFU/mL was reported to be reduced to undetectable levels after 14 days in orange as well as
pomegranate juice and by ∼3 logs after 21 days in milk at 4°C [48]. However, FCV-F9 was reported to
be completely reduced to undetectable levels after 1 day in the blend of orange and pomegranate juice
at 4°C [48]. In another study, FCV-F9 (at initial titers of ∼5 log PFU/mL) was reported to be reduced to
undetectable levels after 1 day in blueberry juice at 4°C [49].
Thus, FCV-F9 has been widely used as a surrogate for HNoVs due to its ease of cultivation in vitro,
its relatively straightforward infectivity assays to develop and understand antiviral agents and for envi-
ronmental persistence and stability studies, and because it is readily manipulated genetically for use
as a model for replication and translation [20]. The development of the FCV reverse genetics system
made FCV a desirable model to study the molecular mechanism of calicivirus translation and genome
replication [50]. Infection with FCV was reported to lead to the inhibition of cellular protein synthesis
associated with the cleavage of host translation initiation factors [51] and also to the first report and
identification of a functional protein receptor molecule for a calicivirus [52]. Subsequent studies with
FCV showed conformational changes in the viral capsid protein upon receptor binding and FCV was
used to identify the first functional host cell factor–viral genome interaction required for FCV replica-
tion. Similar interactions are reported to occur for HNoVs [20]. However, when compared to the other
surrogates including MNV-1 and the newer cultivable TV, FCV does not appear to be the most robust
surrogate model for the determination of HNoV survival and inactivation, specifically because it causes
a respiratory or systemic disease in some cases, unlike the gastrointestinal illness caused by HNoVs.
5.4.2 Murine Norovirus
MNV-1 is a member of the Caliciviridae family and since it falls under the Norovirus genus, it shares
closer biological and molecular properties with HNoVs as compared to FCV-F9 [53]. Besides, MNV-1
can replicate to high titers in tissue culture having a dendritic cell and macrophage tropism and the infec-
tious assays are relatively straightforward as well. MNV-1 is clearly an intestinal pathogen spread via
the fecal–oral route, and MNV infection exacerbates the onset of inflammatory bowel disease similar to
HNoVs [20]. These properties make MNV-1 a suitable model or surrogate to study HNoVs. MNV-1 was
isolated from mice brain tissue with clinical symptoms including diarrhea, fever, nausea, and abdom-
inal pain [53,54]. Similar to HNoVs, with transmission via the fecal–oral route, MNV-1 is shed for
up to 8 weeks after infection and/or inoculation [2]. It is the only NoV that currently replicates in cell
culture in a reproducible manner, which has made HNoV surrogate studies adaptable and somewhat eco-
nomical [54]. On an average the size of this nonenveloped virus lies between 28 and 35 nm in diameter,
with an icosahedral symmetry. MNV-1 contains a single-stranded positive-sense RNA with three ORFs,
where ORF1 encodes a predicted 187.5-kDa polyprotein that comprises of the 2C helicase, 3C protease,
and 3D polymerase motifs typically observed in other caliciviruses and picornaviruses, while ORF2
encodes a 58.9-kDa capsid protein which self-assembles into virus-like particles (VLPs) in baculovirus
expression systems, and ORF3 encodes a putative 22.1-kDa basic protein [54].
As a laboratory model to understand HNoV survival, MNV-1 has been shown to be more stable and
survive longer under various environmental conditions than FCV-F9. Upon exposure to a low pH of 2
at 37°C for 30 min, MNV-1 titers were reported to be reduced by <1 log [6]. MNV-1 titers were found
to decrease by a mere 0.09 log PFU/mL/day in environmental waters at 25°C over 3–5 weeks [55]. In a
study conducted for its survival in the hospital environment, MNV-1 infectivity was reduced by <2 log
over 40 days at both −20°C and 4°C on the surface of gauze and diaper materials; however, at a higher
temperature of 30°C, a higher reduction of 5 log was reported after 24 days [56]. On stainless steel, under
wet and dry conditions, MNV-1 was shown to survive for 7 days with <2 log PFU/mL reduction at 4°C,
Noroviruses: Laboratory Surrogates for Determining Survival and Inactivation 81
with similar reductions at room temperature [6]. MNV-1 is also known to survive freezing temperatures
of −21°C, and no change in infectivity on frozen onions and frozen spinach was reported after 6 months
[57]. MNV-1 was found to survive in commercial blueberry juice (BJ) even after 21 days at 4°C [37]. In
orange juice and milk, MNV-1 titers were not reported to be reduced after 21 days; however, in pome-
granate juice a reduction of 1.4 log PFU/mL was reported. MNV-1 was reduced to undetectable levels
after 7 days in the orange and pomegranate juice blend at 4°C [48]. Escudero et al. also showed that infec-
tious MNV-1 could be detected until 21 days on inoculated stainless steel, formica, or ceramic surfaces,
and did not persist on lettuce stored at room temperature or 4°C [58]. Taken together, these survival stud-
ies provide an indication of the ease of transmission of HNoVs in the environment.
MNV-1 has also been studied as a laboratory model to understand the effect of various inactivation
methods against HNoV (Tables 5.4 through 5.6). With free chlorine concentrations of 0.193 mg/L, MNV
infective titers were found to be reduced to undetectable levels within 2 min of contact time at 5°C and
1 min of contact time at 20°C [59]. Upon exposure to 0.255 mg/L ClO2, MNV-1 was reduced to unde-
tectable levels (<3.5 log) within 1 min of contact time at 5°C and 30 s of contact time at 20°C [59]. It
TABLE 5.4
Inactivation of Murine Norovirus (MNV-1) as an HNoV Surrogate by Physical and Chemical Approaches
Virus Treatment Reduction References
Murine norovirus 15 ppm active chlorine 1.4 log [60]
0.193 mg/L free chlorine; 2 min at 5°C 5 log PFU/mL [59]
0.255 mg/L ClO2; 1 min at 5°C 3.5-log PFU/mL [59]
100 ppm peroxyacetic-biocide; 2 min, room temperature 2.3 log RT-PCR units [60]
2.5% liquid H2O2; 5 min, room temperature 1 log PFU/mL [61]
6.25 ppm ozone; 10 min, room temperature 2.5 log PFU/plant [86]
2% trisodium phosphate; 30 s, room temperature 3 log PFU/mL [86]
0.2 mg/mL benzalkonium chloride; 2 h, room temperature 5 log PFU/mL [28]
5 mg/mL potassium peroxymonosulfate; 2 h, room temperature 5 log PFU/mL [28]
5% levulinic acid and 2% sodium dodecyl sulfate; 1 min 1.50 log PFU/mL [65]
Tween 20 (50 ppm) and chlorine (200 ppm); 1 h 3 log PFU/mL [66]
450-MPa; high hydrostatic pressure in cell-culture media 6.85 log PFU [117]
400-MPa; high hydrostatic pressure; oysters 4.05 log PFU [69]
300 MPa; high-pressure homogenization; blueberry juice 0.71 log PFU/mL [37]
UV at 25 mJ/cm2; 200 s 3.6 log [56]
Sonication; 20 kHz 4 log PFU/mL [38]
TABLE 5.5
Thermal Inactivation of Murine Norovirus (MNV-1) as a Cultivable Surrogate for HNoVs
Murine norovirus Buffered cell-culture media 56°C; D-value 3.74 [73]
65°C; D-value 0.77
72°C; D-value 0.25
Seafood 56°C; D-value 6.12 [75]
65°C; D-value 0.14
72°C; D-value 0.18
Spinach 56°C; D-value 3.29 [40]
65°C; D-value 0.40
72°C; D-value 0.16
Virus stock in cell-culture media, 72°C for 25 s 1 log PFU/mL [6]
Soft shelled clams; 90 s at 90°C 3.33 log cycles [76]
60°C for 5 min; buffer 3.03 log TCID50/mL [77]
60°C for 5 min 0.78 log TCID50/mL
82 Laboratory Models for Foodborne Infections
TABLE 5.6
Inactivation of Murine Norovirus (MNV-1) as a Cultivable HNoV Surrogate Using Natural Plant Extracts
Murine norovirus Grape seed extract (0.35 mg/mL); 37°C for 2 h 0.82 log PFU/mL [78]
Cranberry proanthocyanidins (0.6 mg/mL); 2 h, room temperature 2.9 log PFU/mL [44]
Pomegranate juice; 1 h, room temperature 1.32 log PFU/mL [79]
Pomegranate polyphenols (4 mg/mL); 1 h, room temperature 1.30 log PFU/mL [79]
Black raspberry juice (6%); 1 h at 37°C 99% reduction in plaque titers [43]
Aqueous hibiscus extracts (40 mg/mL); 6 h, 37°C 1.78 log PFU/mL [80]
Oregano oil (4%); 24 h, room temperature 1.07 log TCID50 [82]
was reported that 15 ppm of active chlorine could reduce MNV-1 by 1.4 log units (detected by PCR)
and the application of 100 ppm peroxyacetic-based biocide on lettuce as a produce wash could decrease
MNV-1 titers by 2.3 log RT-PCR units in 2 min [60]. Liquid hydrogen peroxide (H2O2) at 2.5% when used
as an antiviral produce wash was reported to reduce MNV-1 by ∼1 log after 5 min [61]. Ozone is known to
decompose in the water phase of foods, though ozone might be able to diffuse and act on viruses as shown
in a study where the internalized MNV-1 in green onions was reduced by 2.5 log PFU/plant after an ozone
treatment of 6.25 ppm for 10 min [62]. When the green onion plants that were inoculated with MNV-1
were sprayed with calcium hypochlorite (150 ppm, 4°C) or ozone (6.25 ppm for 10 min), a reduction of 2
and 2.5 log PFU/plant, respectively, from an initial titer of 4.92 log PFU/plant was reported [63]. TSP at
2% and 5% after 30 s as a produce wash for lettuce and peppers was reported to reduce MNV-1 by 3 log
PFU/mL and to undetectable levels, respectively, from an initial titer of 5 log PFU/mL [64]. BAC, KPMS,
tannic acid (TA), and gallic acid (GA) were also evaluated for their antiviral effect using MNV-1 as a labo-
ratory model, and MNV-1 (5 log PFU/mL) was reported to be reduced to undetectable levels with BAC
at 0.2, 0.5, and 1 mg/mL after 2 h at room temperature [28]. KPMS at 2.5 and 5 mg/mL was reported to
reduce low-titer MNV-1 (5 log PFU/mL) to undetectable levels and high-titer MNV-1 (7 log PFU/mL) by
0.92 and 3.44 log PFU/mL, respectively, after 2 h at room temperature. TA at 0.1 mg/mL and GA at 0.2 and
0.4 mg/mL were reported to have caused no reduction in MNV-1 titers after 2 h at room temperature [28].
Another novel sanitizer consisting of 0.5% levulinic acid in combination with an anionic detergent
sodium dodecyl sulfate (SDS) at 0.5% was shown to decrease MNV-1 titers by 3 log PFU/mL after
1 min, while a solution of 5% levulinic acid and 2% SDS was reported to reduce MNV-1 titer by 1.50 log
PFU/mL after 1 min and by 3.3 log PFU/mL after 5 min on stainless steel surfaces [65]. Another study
explored the effects of combinations of surfactants (SDS) and polysorbates (Tween 20, Tween 65, and
Tween 80) along with the traditional chlorine (200 ppm) washes against MNV-1 from fresh produce [66].
The study reported that the combination of Tween 20 (50 ppm) and chlorine (200 ppm) after 1 h at room
temperature was most effective, with MNV-1 being reduced by 3 log PFU/mL from an initial titer of
∼6.5 log PFU/mL.
MNV-1 was also used as a laboratory model and surrogate for HNoV to determine inactivation by
physical and chemical inactivation methods that include high HPP, high-pressure homogenization, UV
light, and ozone. The high hydrostatic pressure inactivation of viruses is shown to be due to changes in the
capsid proteins or by disruption of capsid proteins and thereby their inability to bind to host cells [67,68].
MNV-1 titers after a high hydrostatic pressure treatment of 450-MPa were shown to decrease by 6.85 log
PFU in cell-culture media [69]. A reduction of 4.05 log was reported in MNV-1-contaminated oysters
after 5 min at 5°C at 400-MPa high hydrostatic pressure [69]. The high hydrostatic pressure treatment
of 300 MPa was shown to inactivate MNV-1 completely from initial titers of ∼7–8 log PFU/mL after
1 min [70], while MNV-1 in inoculated clams was reported to be reduced by a mere 1 log after high
hydrostatic pressure treatment of 400 MPa for 5 min [71]. When the effect of UV treatment on MNV-1
infectivity was evaluated, 3.6 log reductions after 200 s with UV at 25 mJ/cm2 were obtained [56]. When
green onions were inoculated with MNV-1 and treated with UV (240 mJ s/cm2), 1.2 log PFU reduction
per plant was obtained from an initial titer of 4.92 log PFU/plant [62]. These researchers also showed
that high hydrostatic pressures (500 MPa for 5 min at 20°C) could decrease MNV-1 to undetectable lev-
els. When MNV-1 was used as a surrogate to understand the effect of electron beam irradiation against
Noroviruses: Laboratory Surrogates for Determining Survival and Inactivation 83
enteric viruses, the electron-beam dose required to reduce MNV-1 titers by 90% (D10-value) in whole
oysters was reported to be 4.05 ± 0.63 kGy [72].
When sonication was studied as another physical inactivation method for HNoV, MNV-1 titers were
found to be decreased by ∼4 log PFU/mL after a 30-min treatment at 20 kHz [44]. When exposed to high-
pressure homogenization, MNV-1 in blueberry juice was reported to be reduced by 0.33 log PFU/mL at
250 MPa and by 0.71 log PFU/mL at 300 MPa [37]. Studies carried out by Bozkurt et al. aimed to charac-
terize the thermal inactivation kinetics of MNV-1 as a laboratory surrogate to understand the inactivation
of HNoV at 50°C, 56°C, 60°C, 65°C, and 72°C in buffer as well as different food samples including spin-
ach and blue mussels [40,73,74]. The D-values for MNV-1 using the capillary tube method at 50°C, 56°C,
60°C, 65°C, and 72°C were reported to be 34.49, 3.65, 0.57, 0.30, and 0.15 min, respectively [73]. In buff-
ered cell-culture media, D-values of 36.28, 3.74, 1.09, 0.77, and 0.25 at 50°C, 56°C, 60°C, 65°C, and 72°C,
respectively, were reported [74]. In blue mussels, the reported D-values were 20.19, 6.12, 2.64, 0.41, and
0.18 at temperatures of 50°C, 56°C, 60°C, 65°C, and 72°C, respectively [75]. In spinach, the D-values were
comparatively lower than in blue mussels, where values of 14.57, 3.29, 0.98, 0.40, and 0.16 were obtained
at 50°C, 56°C, 60°C, 65°C, and 72°C, respectively [40]. The effect of heat treatment on MNV-1-infected
soft shell clams was studied using RT-PCR as a method of detection that showed a reduction of 3.33 log
cycles after 90 s at 90°C and 5.47 log cycles after 180 s at 90°C [76]. MNV-1 was reported to be reduced by
3.03, 3.69, 4.35, 5.05, and 5.88 log TCID50/mL in suspensions at temperature–time combinations of 60°C
for 5 min, 60°C for 15 min, 60°C for 30 min, 85°C for 3 min, and 85°C for 6 min, respectively, and in dried
mussels, titer reductions of 0.78, 2.00, 3.35, 1.95, and 3.20 log, respectively, were reported [77].
A variety of natural plant and fruit extracts have been examined for their antiviral properties against
enteric viruses using MNV-1 as a cultivable laboratory surrogate for HNoVs. GSE at 0.25, 0.5, and 1.0 mg/
mL was shown to reduce MNV-1 by 0.82, 1.35, and 1.73 log PFU/mL, respectively, at 37°C for 2 h [78].
Cranberry proanthocyanidins were also reported to decrease MNV-1 titers by 1.6, 2.4, and 2.9 log PFU/
mL after 0, 10, and 60 min, respectively [44]. Su et al. showed that commercial pomegranate juice could
reduce MNV-1 by 1.32 log PFU/mL, while pomegranate polyphenols at 4 mg/mL reduced MNV-1 by
1.30 log PFU/mL after 1 h at room temperature [79]. Black raspberry juice (6%) was also shown to have
antiviral effects on MNV-1 causing reduction in plaque titers by 99% after 1 h [43]. Cotreatment (simul-
taneous addition of treatment and virus to the host cells) was demonstrated to exert maximal antiviral
activity on MNV-1, suggesting that the juice exerted its antiviral effect by inhibiting viral attachment to
host cells, though gallic acid and quercetin were not shown to have any significant effect [43]. Aqueous
hibiscus extracts were shown to reduce MNV-1 by 1.89 and 1.78 log PFU/mL after 6 h with 100 and
40 mg/mL of the extract, respectively, and to undetectable levels after 24 h with both 100 and 40 mg/mL
of the extract at 37°C, respectively [80]. When host cells RAW 264.7 were pretreated with Korean Red
Ginseng at 5, 6.7, and 10 μg/mL, a reduction in the infectivity of MNV-1 by 0.38 ± 0.41, 0.73 ± 0.19,
and 1.48 ± 0.27 log TCID50/mL from an initial titer of 6.7 ± 0.2 log TCID50/mL after 24 h at 37°C was
reported [81]. Oregano oil at 4% was reported to cause a reduction of 1.07 log TCID50 in MNV-1 titers after
24 h, while carvacrol caused a 3.87 log TCID50 decrease after 1 h at room temperature [82].
As immune knockout mice are readily available, MNV-1 is often used as a model to study the immune
responses to HNoV infection and to examine the host responses required for norovirus pathogenesis,
clearance and establishment of immunity, and insights for the future development of vaccines [21].
MNV-1 has also gained popularity as the surrogate or model system of choice to study HNoVs due to the
development of two reverse genetics systems, one based on the polymerase II-driven expression of viral
RNA and the second based on the T7 RNA polymerase-driven expression of viral RNA with the recov-
ery of fully infectious virions by both systems that are used to determine the fundamentals of norovirus
genome translation and replication [21]. The cleavage of the MNV protease–polymerase precursor form
of RdRp into separate units and RNA secondary structures was shown to be essential for replication,
while the correct sequence of the last nucleotide of the MNV genome, immediately upstream of the poly
A tail was found to be essential for the recovery of infectious virus. Terminal sialic acid moieties on
gangliosides function as an attachment receptor for MNV on murine macrophages [20,83].
However, the limitations of using MNV-1 as a model and surrogate need to be addressed. MNV-1
causes a persistent disease in its host that is different from HNoV as mice do not routinely develop diar-
rhea and cannot vomit, while HNoV causes acute rapid infection and subsequent shedding and clearance
84 Laboratory Models for Foodborne Infections
with sometimes longer shedding periods that can last for more than a week. MNV-1 is less genetically
variable than the variable HNoV genome that is associated with repeated infection and short-term immu-
nity [20]. The relevance of the apparent immune-cell-tropism of MNV-1 is currently unknown as HNoV
tropism in vivo remains unclear to date [20]. Therefore, research on suitable cultivable surrogates and
model systems for understanding HNoV infections and inactivation is ongoing.
5.4.3 Tulane Virus
Tulane virus (TV) is a cultivable enteric calicivirus and is also known to recognize the same receptors
(human histoblood group antigens) as HNoVs [84]. TV is 36 nm in size with a genome size of about
6.7 kb single-stranded RNA with a poly(A) tail comprising of one of the shortest known genomes in
the Caliciviridae family and belongs to the Recovirus genus. The viral genome that is enclosed in a
nonenveloped capsid is divided into three ORFs, where ORF1 encodes for one nonstructural polyprot-
ein, ORF2 encodes for the capsid protein, and ORF3 encodes for a minor structural protein [85]. TV is
closely genetically related to HNoVs and pairwise homology has revealed the highest amino acid identity
with HNoVs among other members of the Caliciviridae family [85]. TV was isolated from stool samples
of monkeys without typical symptoms of gastrointestinal disease, questioning the ability of monkeys to
be robust animal models. TV is known to bind to all B antigens (types 1–4) and type 3 of the A antigen.
Since TV can be easily cultured and grown in vitro, it remains an important cultivable surrogate for
HNoVs and a strong candidate as a laboratory model to study HNoVs.
TV has been used as a cultivable HNoV surrogate to determine inactivation and survival under
different environmental stresses and conditions that include different pH, temperature, and chlorine
levels [86]. Hirneisen and Kniel reported that TV could be reduced by 1.59, 0.96, and 0.82 log PFU/mL
at pH values 2.0, 3.0, and 4.0, after 30 min at room temperature, respectively [62]. At temperatures of
50°C, 55°C, 60°C, and 65°C for 2 min, TV was reported to be reduced by 1.79, 1.83, 2.90, and 3.07 log
PFU/mL, respectively [62]. Upon chlorine treatments of 0.2, 2, 20, and 200 ppm, TV was reported to be
reduced by 1.33, 2.11, 1.53, and 2.93 log PFU/mL after 5 min. TV was also examined for its survival on
spinach plants with and without exposure to sunlight [62]. It was found to persist on whole spinach plants
and on both semisavoy and smooth spinach plants maintained in a greenhouse chamber for 7 days [62].
To assess the role of sunlight, these researchers exposed the spinach plants to UVA/UVB using a lamp to
mimic sunlight for 10 h/day for 7 days; however, they did not find any significant difference in TV titers
from the initial titer values.
The survival and dissemination of TV was also studied by inoculating the roots of romaine lettuce
with TV and growing them for 2 weeks in hydroponic feed water [87]. These researchers detected about
3 log PFU/g of TV infectious particles in leaves and shoots after 2 days postinoculation; however, the
levels of detected infectious particles in leaves and shoots reached up to 6 log PFU/g after 7 days postin-
oculation. Thus, TV was found to internalize in the roots of plants of lettuce and can be disseminated to
the leaves and root shoots. It was also found to persist on alfalfa seeds for over 50 days at 22°C, where
TV was reported to be reduced from an initial titer of 3.87–0.85 log PFU/g [88].
TV was also used to determine the effects of high hydrostatic pressure on its inactivation in cell-
culture media, blueberries, and oysters [89]. These researchers showed that in cell-culture media, a
treatment of 350 MPa caused a reduction of 3.8 and 2.4 log PFU/mL of TV at pH 7 and 4, respectively,
after 2 min at 21°C, while 600 MPa at temperatures of 4°C, 21°C, and 35°C was shown to not cause any
reduction of TV on dry blueberries after 2 min. However, it was shown that complete inactivation of TV
occurred when blueberries were immersed in phosphate-buffered saline after 300 MPa treatment for
2 min at 4°C and also at 400 MPa for 2 min at 4°C. In oysters, TV reductions of 2.9 and 2 log PFU/mL
were reported after a treatment of 250 MPa at 4°C and 21°C, respectively.
TV was also used as a surrogate to understand the effects of ionizing radiation by an electron beam,
as a nonthermal processing approach on the viral infectivity of spiked produce, and to determine the
mechanism of inactivation [90]. TV was reported to be reduced from 7 log PFU/mL to undetectable
levels with 16 kGy or higher doses in strawberries and lettuce, while with lower target doses of 4 kGy,
reductions of 1.5 and 1.8 log PFU/mL were reported in PBS and cell-culture media (Opti-MEM), respec-
tively (Table 5.7). Transmission electron microscopy (TEM) did not reveal any significant damage to
Noroviruses: Laboratory Surrogates for Determining Survival and Inactivation 85
TABLE 5.7
Thermal and Nonthermal Approaches for the Inactivation of Tulane Virus (TV) as a Cultivable HNoV Surrogate
Virus Treatment Reduction References
Tulane virus 50°C, 2 min 1.79 log PFU/mL [86]
55°C, 2 min 1.83
60°C, 2 min 2.90
65°C, 2 min 3.07
High hydrostatic pressure; 350 MPa; cell-culture media 3.8 log PFU/mL [89]
High hydrostatic pressure; 600 MPa; dry blueberries No reduction [89]
High hydrostatic pressure; 600 MPa; oysters 2.9 log PFU/mL [89]
16 kGy electron beam; strawberries 7 log PFU/mL [90]
70% ethanol; 20 s 5 log TCID50 [91]
UVC; 60 mJ/cm2; 4 min 4 log TCID50 [91]
300 ppm free chlorine; 10 min 4 log TCID50 [91]
the treated viral particles, though the numbers of detectable virions were reported to be reduced in the
treated samples. Sodium dodecyl sulfate–polyacrylamide gel electrophoresis (SDS–PAGE) of the treated
viral particles showed degraded viral proteins compared to the controls posttreatment [90]. TV was also
used as an HNoV surrogate to determine inactivation by 50%–70% ethanol [91], where TV was reduced
by 5 log TCID50 after 20 s treatment, and heating at 63°C for 5 min and 56°C for 30 min, UVC exposure
of 60 mJ/cm2 for 4 min, and 300 ppm of free chlorine for 10 min were also reported to cause a reduction
of 4 log TCID50 [91].
TV infectivity was also determined in the presence of bacterial cell-free supernatants (CFS), show-
ing reduction by 0.44, 0.37, 0.38, and 0.81 logs with CFS of Bacillus coagulans, Escherichia coli,
Enterococcus faecalis, Staphylococcus epidermidis grown in tryptic soy broth (TSB), when host cells
were overlaid with CFS at 37°C for 44 h [92]. These results should help toward understanding the factors
affecting viral infections and how colonization of bacteria in the gut or the native microbiome can inhibit
HNoV or other viral infections through competitive attachment [92].
Replication and biological studies have demonstrated that an authentic 5′ end of TV RNA or the
N-terminal protein appears to be essential for infectivity, indicating that while nonstructural proteins
appear to be sufficient for TV genomic RNA replication and translation, structural proteins are still
needed to generate progeny viruses [20].
However, TV causes an uncharacterized disease in its rhesus macaque host and represents a separate
genus than HNoV. Nonhuman primates such as common marmosets, cotton-top tamarins, and rhesus
macaques serve as a good alternate model to study HNoV biology, as they seem to be susceptible to
the HNoV GI Norwalk virus and shed virus after infection, though without exhibiting any disease symp-
toms [20]. Thus, based on the information provided above, TV may be another appropriate alternate
model to study HNoV pathogenesis, survival, and transmission.
5.4.4 Porcine Sapovirus
Porcine sapovirus belongs to the genus Sapovirus within the Caliciviridae family, and the porcine sapovi-
rus Cowden strain has been successfully propagated in the continuous porcine kidney cell line (LLC-PK)
[93]. The presence of bile acid or intestinal content fluid filtrate obtained from uninfected gnotobiotic
pigs was found to be necessary during preinfection for viral growth in cell-culture media [94]. Human
sapoviruses are known to cause gastroenteritis in humans with similar symptoms to HNoVs. However,
similar to HNoVs, they currently remain unculturable in the laboratory. From an epidemiological per-
spective, the HNoVs are considered to be more significant due to their prevalence/number of outbreaks as
compared to Sapoviruses. The properties of porcine sapovirus that make it a suitable surrogate are that
it is an enteric virus, is genetically related to HNoVs, is cultivable in the lab, replicates in intestinal cells
of pigs, and causes gastroenteritis in piglets [95–97].
86 Laboratory Models for Foodborne Infections
TABLE 5.8
Evaluation of Various Inactivation Methods on Porcine Enteric Virus (PEV) as a Cultivable Surrogate
Virus Treatment Reduction References
Porcine enteric virus 56°C, 20 min 2 log TCID50 [70]
70% and 90% ethanol, 1 min 0.38 × 106 TCID50 [70]
200 ppm chlorine, 5 min 0.4 log TCID50 [70]
1000 ppm Chlorine, 5 min 1.3 log TCID50
High hydrostatic pressure; 700 MPa 7 log TCID50 [70]
High hydrostatic pressure; 400 MPa 4 log TCID50
Porcine enteric caliciviruses (PEC) are reported to infect pigs of all ages, but cause diarrhea only
in young piglets. Gnotobiotic pigs can become infected with HNoVs and therefore have been reported
to be used to study and model antibody and cytokine responses [20]. PEC is subclinical in pigs, yet
may play an important role in evolutionary modeling if they are a natural reservoir or are involved in
zoonotic disease transfer and have been used as a model for tissue culture–derived enteric calicivirus
attenuation [20]. To gain insight into PEC replication, a reverse genetics system that was developed for
PEC showed that capped RNA transcripts derived from a cDNA clone were fully infectious [20].
Porcine sapovirus appears to be suitable as a cultivable HNoV for determining inactivation by various
methods (Table 5.8) as PEC infectious (using TCID50 assays) titers were reported to have no significant
changes after exposure to pH 4–8 at room temperature for 1 h and showed <l-log reduction at pH 3 [88].
Porcine sapovirus was also reported to attach to lettuce leaves at its capsid isoelectric point (pH 5.0), and
even remained infectious on lettuce after 1 week of storage at 4°C [88]. These researchers also showed
that porcine sapovirus and HNoV shared similar resistance to heat and chlorine treatment. However, the
stability of porcine sapovirus to food-processing technologies is yet to be determined. Hence, further
studies using PEC as a model or surrogate for HNoVs and human sapoviruses are ongoing.
5.4.5 Bovine Noroviruses
Bovine NoVs are reported to be less closely related to HNoVs than porcine viruses, and the prototype
bovine NoV strain Newbury agent 2 was identified in calves suffering from diarrhea. The overall simi-
larity of bovine NoVs and their potential zoonotic transfer make them an alternative model for in vivo
studies, although recently NoVs have also been identified in sheep [20].
5.4.6 Rabbit Caliciviruses
Rabbit hemorrhagic disease virus (RHDV), rabbit calicivirus (RCV) and, European brown hare syn-
drome virus (EBHSV) are caliciviruses of the Lagovirus genus, mainly of concern in America and
China, that most often cause fatal disease with symptoms such as hepatitis and hemorrhages, and are
virulent viruses of rabbits [20]. Their suitability as model systems for HNoVs is unclear as they cause
different disease symptoms than HNoVs, while RCV is nonpathogenic and has not been completely
characterized.
However, for RHDV, an animal model, a tissue culture system, and a reverse genetics system are
reportedly available, and it has been demonstrated that the minor capsid protein VP2 of RHDV does not
play a role in virus infectivity and also that the 3′ poly A tail has no effect on virus replication in vitro
[20]. Thus, RHDV may be considered for use in an alternate animal model system to study HNoV
biology.
5.4.7 Norovirus-Like Particles
Norovirus-like particles have also been used as a model and alternate surrogate in the laboratory for
inactivation studies on HNoVs in order to investigate the structural and biochemical characteristics of
Noroviruses: Laboratory Surrogates for Determining Survival and Inactivation 87
HNoVs after inactivation treatments. The recombinant expression of the VP1 major capsid protein of
HNoV has been shown to result in self-assembly to form empty, noninfectious VLPs that have similar
morphology and antigenic properties to the native infectious virus particle [114]. The protruding (P)
domain of the major structural capsid protein that forms the outermost surface of the capsid contains the
elements required for viral capsid binding to host carbohydrate receptors [89,95,98]. A variety of expres-
sion systems have been reported to be used for the expression of VLPs, including baculovirus insect cell
and transgenic plant expression systems [99–102]. When the P protein is expressed in E. coli, subviral
particles, namely P particles, are formed that may be used as a surrogate to study the binding ability of
HNoVs, and it is the VLP receptor binding ability that has been proposed as a unique indicator for virus
survival and infectivity [95], as well as for the determination of the damage of VLPs by using electron
microscopy and SDS–PAGE techniques. These VLPs have been used in research to demonstrate the
ability of gamma irradiation to disrupt the VLP structure causing degradation of the VP1 protein [61].
VLPs have also been used as a surrogate for determining the interaction/binding of HNoV with fresh
produce, seafood, and other at-risk foods [95]. HNoV P particles, using saliva binding enzyme-linked
immunosorbent assays, were used as an HNoV surrogate, and the presence of cranberry and pomegran-
ate juices, showed reduced HNoV binding [103]. However, some fresh produce extracts that included
strawberry, blackberry, blueberry, cherry tomato, spinach, romaine lettuce, or raspberry did not demon-
strate any effect on the binding ability of these particles [103].
5.5 Bacteriophage MS2
Bacteriophage MS2 is a bacterial virus, commonly found in sewage and wastewaters, that infects the
bacteria E. coli [in particular, American Type Culture Collection (ATCC) 15597B]. Bacteriophage MS2
is a single-stranded icosahedral RNA virus, between 27 and 34 nm in diameter, that belongs to the
Leviviridae family in group 1 of the RNA coliphages and is adapted to the intestinal tract [104].
Bacteriophage MS2 has been used by various researchers as a gastrointestinal virus surrogate to deter-
mine disinfection and inactivation efficacy as well as survival and transmission in the environment,
including the use of electrochemical oxidants (ECOs) for surface disinfection, due to its ease of propaga-
tion and relatively simplistic assays [105]. ECOs were generated using battery power to electrolyze brine
(NaCl) solutions, which contain both chlorine [HOCl, OCl(−)] and reactive oxygen species (e.g., ·OH,
O3, H2O2, and O2−), and at free available chlorine concentrations of 2500 ppm for a 30-s contact time,
infective MS2 bacteriophage was reported to be inactivated by >7 log compared to the MNV-1 inactiva-
tion of ∼2 log [105]. When these researchers compared genomic RNA inactivation, MS2 RNA inactiva-
tion was reported to be around 5 log compared to the MNV-1 RNA inactivation of around 1.5 log, with
comparable inactivation efficacy to household bleach at similar free available chlorine concentrations.
MS2 was reported to be reduced by ≥6 log PFU/mL at 5% TSP, and a 4.5 log PFU/mL reduction was
reported to be achieved with 1% TSP after a contact time of 30 s [29]. Lee and Ko reported that MS2 was
highly resistant to disinfection by UVA, but the addition of TiO2 enhanced the efficacy of UVA, where a
UVA dose of 1379 mJ/cm2 resulted in a 4 log reduction, while UVB alone inactivated both MS2 by 4 log
with a dose of 367 mJ/cm2 [59]. A study of the virucidal inactivation efficacy of an in-house-designed
atmospheric pressure, nonthermal plasma jet operated at varying helium–oxygen feed gas concentrations
used MS2 bacteriophage as a surrogate and model for HNoV, showed that the inactivation rate constant
increased with increasing oxygen concentrations up to 0.75% [106]. These researchers reported that
3 log (99.9%) reductions in infectious MS2 were obtained after 3 min of exposure in a helium–oxygen
(99.25%:0.75%) gas mixture, with increased reduction after 9 min exposure by >7 log. Black et al. found
that MS2 was inactivated by <1 log PFU/mL at hydrostatic pressures of 500 MPa for 5 min at 20°C,
showing its resistance as compared to other surrogates and making it a suitable surrogate and model
virus for hydrostatic pressure studies [107]. Dawson et al. studied the survivability/persistence of MS2
bacteriophage on fresh iceberg lettuce, baton carrot, cabbage, spring onion, curly leaf parsley, capsicum
pepper, tomato, cucumber, raspberries, and strawberries and showed that MS2 survived for a very long
time and even extended the shelf life of the produce, where <1 log PFU/mL reduction was observed after
50 days at 4°C and 8°C [104]. In a study determining the survival of HNoV in juices, bacteriophage MS2
88 Laboratory Models for Foodborne Infections
(∼6 log PFU/mL) was used as a surrogate and model that showed significant reduction (1.93 log PFU/mL)
after 2 days and was undetectable after 7 days in blueberry juice at 4°C [48].
5.6 Conclusions
HNoVs are a worldwide public health and food safety concern. However, in the absence of reproducible
cell-culture-based infectivity assays and due to the lack of available vaccines to date, cultivable surro-
gates and model systems are used in order to determine survival, transmission, and inactivation methods
to prevent outbreaks and minimize the risk of HNoV contamination. Typically, cultivable surrogates that
mimic the characteristics of HNoVs are genetically related and are used in laboratory settings. These
surrogates include cultivable animal viruses from the Caliciviridae family as well as bacteriophages and
noroviral-like particles. The application of these surrogates in environmental studies and to enhance
food safety remains ongoing. However, the ideal situation of having reproducible infectivity assays for
HNoVs appears to be a challenge and research efforts continue to be made toward attaining this goal.
It is evident that a reproducible and validated cell-culture system for the propagation of HNoVs is
needed to understand HNoV biology, replication, and molecular aspects. Toward this end, researchers
have still to validate a B-cell replication system for HNoVs [19]. Virus entry and genome uncoating are
considered the “vital missing link” for HNoV propagation [20]. Despite the availability of a variety of
animal models and surrogates for understanding HNoVs, there does not appear to be one single overall
“ideal” model system, whose suitability varies and depends on the topic being researched. As indicated
and described in detail by Vashist et al., for water-related and environmental studies, bacteriophages
such as MS2 and phiX-174 have been used as indicators and surrogates in the past to determine survival
and transmission routes. For studying pathogenesis, animal caliciviruses appear to be most suitable, yet
none of them reproduce all the HNoV disease aspects. Most animal caliciviruses appear to be suitable
models for genome translation and replication studies, and MNV-1 appears to be most closely related
or suitable for HNoVs [20]. With regards to vaccines and antivirals, progress seems to be limited and
slow, and small molecule inhibitors, natural plant polyphenols, and antisense RNA technologies show
potential for future application (further suggested reading on NoVs in reference [20,108]). Thus, in
conclusion, research emphasis should continue to be placed on the propagation of HNoV in cell-culture
systems in vitro to obtain reproducible HNoV infectivity assays.
Acknowledgments
The authors gratefully acknowledge the support provided by the University of Tennessee-Institute of
Agriculture. The authors also wish to thank Carrie Yard for her assistance with the formatting some of
the references during the final stage of this chapter submission.
REFERENCES
1.
Scipioni, A., et al., Animal noroviruses. Vet J, 2008. 178(1): 32–45.
Green, K., et al., Taxonomy of the caliciviruses. J Infect Dis, 2000. 181(Suppl 2): S322–S330.
2.
Kapikian, A.Z., The discovery of the 27-nm Norwalk virus: an historic perspective. J Infect Dis, 2000.
3.
181(Suppl 2): S295–S302.
4.
Seah, E.L., J.A. Marshall, and P.J. Wright, Open reading frame 1 of the Norwalk-like virus Camberwell:
completion of sequence and expression in mammalian cells. J Virol, 1999. 73(12): 10531–10535.
D’Souza, D.H. and S.S. Joshi, Foodborne viruses of human health concern, in The Encyclopedia of
5.
Food and Health, vol. 3, Caballero, B., Finglas, P., and Toldrá, F., (Eds). 2016, Academic Press, Oxford,
pp. 87–93.
Cannon, J.L., et al., Surrogates for the study of norovirus stability and inactivation in the environment:
6.
a comparison of murine norovirus and feline calicivirus. J Food Prot, 2006. 69(11): 2761–2765.
7.
Zheng, D.P., et al., Norovirus classification and proposed strain nomenclature. Virology, 2006. 346(2):
312–323.
Noroviruses: Laboratory Surrogates for Determining Survival and Inactivation 89
8. Xerry, J., et al., Genetic characterization of genogroup I norovirus in outbreaks of gastroenteritis. J Clin
Microbiol, 2010. 48(7): 2560–2562.
9. Zheng, D.P., et al., Molecular epidemiology of genogroup II-genotype 4 noroviruses in the United States
between 1994 and 2006. J Clin Microbiol, 2010. 48(1): 168–177.
10. Fankhauser, R.L., et al., Epidemiologic and molecular trends of “Norwalk-like viruses” associated with
outbreaks of gastroenteritis in the United States. J Infect Dis, 2002. 186(1): 1–7.
11. Donaldson, E.F., et al., Norovirus pathogenesis: mechanisms of persistence and immune evasion in
human populations. Immunol Rev, 2008. 225(1): 190–211.
12. CDC, Norovirus outbreaks on three college campuses—California, Michigan, and Wisconsin, 2008.
MMWR, 2009. 58(39): 1095–1100.
13. Pang, X.L., et al., Influence of novel norovirus GII.4 variants on gastroenteritis outbreak dynamics in
Alberta and the Northern Territories, Canada between 2000 and 2008. PLoS One, 2010. 5(7): e11599.
14. Siebenga, J.J., et al., Norovirus illness is a global problem: emergence and spread of norovirus GII.4
variants, 2001–2007. J Infect Dis, 2009. 200(5): 802.
15. Lopman, B.A., et al., A summertime peak of “winter vomiting disease”: surveillance of noroviruses in
England and Wales, 1995 to 2002. BMC Public Health, 2003. 3: 1–4.
16. Malek, M., et al., Outbreak of norovirus infection among river rafters associated with packaged delica-
tessen meat, Grand Canyon, 2005. Clin Infect Dis, 2009. 48(1): 31–37.
17. Escudero-Abarca, B., et al., Molecular methods used to estimate thermal inactivation of a prototype
human norovirus: more heat resistant than previously believed? Food Microbiol, 2014. 41: 91–95.
18. Papafragkou, E., et al., Challenges of culturing human norovirus in three-dimensional organoid intesti-
nal cell culture models. PLoS One, 2013. 8(6): e63485.
19. Jones, M.K., et al., Enteric bacteria promote human and mouse norovirus infection of B cells. Science,
2014. 346(6210): 755–759.
20. Vashist, S., et al., Model systems for the study of human norovirus biology. Future Virol, 2009. 4(4):
353–367.
21. Leon, J., et al., Randomized, double-blinded clinical trial for human norovirus inactivation in oysters by
high hydrostatic pressure processing. Appl Environ Microbiol, 2011. 77(15): 5476–5482.
22. Grohmann, G., et al., Norwalk virus gastroenteritis in volunteers consuming depurated oysters. Aust J
Exp Biol Med Sci, 1981. 59(2): 219–228.
23. Richards, G.P., Critical review of norovirus surrogates in food safety research: rationale for considering
volunteer studies. Food Environ Virol, 2012. 4(1): 6–13.
24. Thiel, H.-J. and M. König, Caliciviruses: an overview. Vet Microbiol, 1999. 69(1): 55–62.
25. Radford, A.D., et al., Feline calicivirus. Vet Res, 2007. 38(2): 319–335.
26. Doultree, J.C., et al., Inactivation of feline calicivirus, a Norwalk virus surrogate. J Hosp Infect, 1999.
41(1): 51–57.
27. Greening, G.E., et al., Molecular epidemiology of norovirus gastroenteritis outbreaks in New Zealand
from 2002–2009. J Med Virol, 2012. 84(9): 1449–1458.
28. Su, X. and D.H. D’Souza, Inactivation of human norovirus surrogates by benzalkonium chloride,
potassium peroxymonosulfate, tannic acid, and gallic acid. Foodborne Pathog Dis, 2012. 9(9):
829–834.
29. D’Souza, D.H. and X. Su, Efficacy of chemical treatments against murine norovirus, feline calicivirus,
and MS2 bacteriophage. Foodborne Pathog Dis, 2010. 7(3): 319–326.
30. Chander, Y., et al., Antiviral activity of Ecasol against feline calicivirus, a surrogate of human norovi-
rus. J Infect Public Health, 2012. 5(6): 420–424.
31. Morino, H., et al., Effect of low-concentration chlorine dioxide gas against bacteria and viruses on a
glass surface in wet environments. Lett Appl Microbiol, 2011. 53(6): 628–634.
32. Tibollo, S., et al., High hydrostatic pressure activity on the disinfection of clams artificially contami-
nated with feline calicivirus. Ann Ig, 2013. 25(3): 201–208.
33. Thurston-Enriquez, J.A., et al., Inactivation of enteric adenovirus and feline calicivirus by ozone. Water
Res, 2005. 39(15): 3650–3656.
34. Hirneisen, K.A. and K.E. Kniel, Inactivation of internalized and surface contaminated enteric viruses
in green onions. Int J Food Microbiol, 2013. 166(2): 201–206.
35. Fino, V.R. and K.E. Kniel, UV light inactivation of hepatitis A virus, Aichi virus, and feline calicivirus
on strawberries, green onions, and lettuce. J Food Prot, 2008. 71(5): 908–913.
90 Laboratory Models for Foodborne Infections
36. Schultz, A., et al., Inactivation of norovirus surrogates on surfaces and raspberries by steam-ultrasound
treatment. J Food Prot, 2012. 75(2): 376–381.
37. Horm, K.M., et al., Survival and inactivation of human norovirus surrogates in blueberry juice by high-
pressure homogenization. Foodborne Pathog Dis, 2012. 9(11): 974–979.
38. Su, X., S. Zivanovic, and D. D’Souza, Inactivation of human enteric virus surrogates by high-intensity
ultrasound. Foodborne Pathog Dis, 2010. 7(9): 1055–1061.
39. Zhou, F., et al., Inactivation of feline calicivirus as a surrogate for norovirus on lettuce by electron beam
irradiation. J Food Prot, 2011. 74(9): 1500–1503.
40. Bozkurt, H., D.H. D’Souza, and P.M. Davidson, Thermal inactivation of human norovirus surrogates in
spinach and measurement of its uncertainty. J Food Prot, 2014. 77(2): 276–283.
41. Lee, J.H., et al., Antiviral effects of mulberry (Morus alba) juice and its fractions on foodborne viral
surrogates. Foodborne Pathog Dis, 2014. 11(3): 224–229.
42. Sanchez, G. and R. Aznar, Evaluation of natural compounds of plant origin for inactivation of enteric
viruses. Food Environ Virol, 2015: 183–187.
43. Oh, M., et al., Antiviral effects of black raspberry (Rubus coreanus) juice on foodborne viral surrogates.
Foodborne Pathog Dis, 2012. 9(10): 915–921.
44. Su, X., A.B. Howell, and D.H. D’Souza, Antiviral effects of cranberry juice and cranberry proantho-
cyanidins on foodborne viral surrogates—a time dependence study in vitro. Food Microbiol, 2010.
27(8): 985.
45. Su, X. and D.H. D’Souza, Grape seed extract for foodborne virus reduction on produce. Food Microbiol,
2013. 34(1): 1–6.
46. D’Souza, D.H., et al., Persistence of caliciviruses on environmental surfaces and their transfer to food.
Int J Food Microbiol, 2006. 108(1): 84–91.
47. Mormann, S., et al., Tenacity of human norovirus and the surrogates feline calicivirus and murine
norovirus during long-term storage on common nonporous food contact surfaces. J Food Prot, 2015.
78(1): 224–229.
48. Horm, K.M. and D.H. D’Souza, Survival of human norovirus surrogates in milk, orange, and pome-
granate juice, and juice blends at refrigeration (4°C). Food Microbiol, 2011. 28(5): 1054–1061.
49. Horm, K., F. Harte, and D. D’Souza, Human norovirus surrogate reduction in milk and juice blends by
high pressure homogenization. J Food Prot, 2012. 75(11): 1984–1990.
50. Sosnovtsev, S. and K. Green, RNA transcripts derived from a cloned full-length copy of the feline
calicivirus genome do not require VPG for infectivity. Virology, 1995. 210(2): 383–390.
51. Willcocks, M.M., M.J. Carter, and L.O. Roberts, Cleavage of eukaryotic initiation factor elF4G and
inhibition of host-cell protein synthesis during feline calicivirus infection. J Gen Virol, 2004. 85(5):
1125–1130.
52. Makino, A., et al., Junctional adhesion molecule 1 is a functional receptor for feline calicivirus. J Virol,
2006. 80(9): 4482–4490.
53. Karst, S.M., et al., STAT1-dependent innate immunity to a Norwalk-like virus. Science, 2003. 299(5612):
1575–1578.
54. Wobus, C.E., L.B. Thackray, and H.W. Virgin, 4th, Murine norovirus: a model system to study norovirus
biology and pathogenesis. J Virol, 2006. 80(11): 5104–5112.
55. Bae, J. and K.J. Schwab, Evaluation of murine norovirus, feline calicivirus, poliovirus, and MS2 as
surrogates for human norovirus in a model of viral persistence in surface water and groundwater. Appl
Environ Microbiol, 2008. 74(2): 477–484.
56. Lee, J., K. Zoh, and G. Ko, Inactivation and UV disinfection of murine norovirus with TiO2 under
various environmental conditions. Appl Environ Microbiol, 2008. 74(7): 2111–2117.
57. Baert, L., et al., Survival and transfer of murine norovirus 1, a surrogate for human noroviruses, during
the production process of deep-frozen onions and spinach. J Food Prot, 2008. 71(8): 1590–1597.
58. Escudero, B., et al., Persistence and transferability of noroviruses on and between common surfaces and
foods. J Food Prot, 2012. 75(5): 927–935.
59. Lim, M.Y., J.M. Kim, and G. Ko, Disinfection kinetics of murine norovirus using chlorine and chlorine
dioxide. Water Res, 2010. 44(10): 3243–3251.
60. Fraisse, A., et al., Comparison of chlorine and peroxyacetic-based disinfectant to inactivate feline cali-
civirus, murine norovirus and hepatitis A virus on lettuce. Int J Food Microbiol, 2011. 151(1): 98–104.
Noroviruses: Laboratory Surrogates for Determining Survival and Inactivation 91
61. Herdt, J. and H. Feng, Aqueous antimicrobial treatments to improve fresh and fresh‐cut produce safety,
in Microbial Safety of Fresh Produce, Fan, X., et al., (Eds). 2009, IFT Press, Wiley-Blackwell, Ames,
IA, pp. 167–190.
62. Hirneisen, K. and K. Kniel, Norovirus surrogate survival on spinach during preharvest growth.
Phytopathology, 2013. 103(4): 389–394.
63. Hirneisen, K.A., S.M. Markland, and K.E. Kniel, Ozone inactivation of norovirus surrogates on fresh
produce. J Food Prot, 2011. 74(5): 836–839.
64. Su, X. and D.H. D’Souza, Trisodium phosphate for foodborne virus reduction on produce. Foodborne
Pathog Dis, 2011. 8(6): 713–717.
65. Cannon, J.L., et al., Efficacy of a levulinic acid plus sodium dodecyl sulfate-based sanitizer on inactiva-
tion of human norovirus surrogates. J Food Prot, 2012. 75(8): 1532–1535.
66. Predmore, A. and J. Li, Enhanced removal of a human norovirus surrogate from fresh vegetables and
fruits by a combination of surfactants and sanitizers. Appl Environ Microbiol, 2011. 77(14): 4829–4838.
67. Tang, Q., et al., Mechanism of inactivation of murine norovirus-1 by high pressure processing. Int
J Food Microbiol, 2010. 137(2–3): 186–189.
68. Kingsley, D.H., et al., Inactivation of hepatitis A virus and a calicivirus by high hydrostatic pressure.
J Food Prot, 2002. 65(10): 1605–1609.
69. Kingsley, D.H., et al., Inactivation of a norovirus by high-pressure processing. Appl Environ Microbiol,
2007. 73(2): 581–585.
70. Cromeans, T., et al., Comprehensive comparison of cultivable norovirus surrogates in response to dif-
ferent inactivation and disinfection treatments. Appl Environ Microbiol, 2014. 80(18): 5743–5751.
71. Arcangeli, G., et al., Effect of high hydrostatic pressure on murine norovirus in Manila clams. Lett Appl
Microbiol, 2012. 54(4): 325–329.
72. Praveen, C., et al., Susceptibility of murine norovirus and hepatitis A virus to electron beam irradia-
tion in oysters and quantifying the reduction in potential infection risks. Appl Environ Microbiol, 2013.
79(12): 3796–3801.
73. Bozkurt, H., D.H. D’Souza, and P.M. Davidson, Determination of the thermal inactivation kinetics
of the human norovirus surrogates, murine norovirus and feline calicivirus. J Food Prot, 2013. 76(1):
79–84.
74. Bozkurt, H., D.H. D’Souza, and P.M. Davidson, A comparison of the thermal inactivation kinetics of
human norovirus surrogates and hepatitis A virus in buffered cell culture medium. Food Microbiol,
2014. 42: 212–217.
75. Bozkurt, H., et al., Thermal inactivation kinetic modeling of human norovirus surrogates in blue mussel
(Mytilus edulis) homogenate. Int J Food Microbiol, 2014. 172: 130–136.
76. Sow, H., et al., Heat inactivation of hepatitis A virus and a norovirus surrogate in soft-shell clams
(Mya arenaria). Foodborne Pathog Dis, 2011. 8(3): 387–393.
77. Park, S.Y., et al., Thermal inactivation of murine norovirus-1 in suspension and in dried mussels (Mytilus
edulis). J Food Safety, 2014. 34(3): 193–198.
78. Su, X. and D.H. D’Souza, Grape seed extract for control of human enteric viruses. Appl Environ
Microbiol, 2011. 77(12): 3982–3987.
79. Su, X., M.Y. Sangster, and D.H. D’Souza, In vitro effects of pomegranate juice and pomegranate
polyphenols on foodborne viral surrogates. Foodborne Pathog Dis, 2010. 7(12): 1473–1479.
80. Joshi, S.S., L. Dice, and D.H. D’Souza, Aqueous extracts of Hibiscus sabdariffa calyces decrease hepa-
titis A virus and human norovirus surrogate titers. Food Environ Virol, 2015 7(4): 366–373.
81. Lee, M.H., et al., Antiviral effect of Korean red ginseng extract and ginsenosides on murine norovirus
and feline calicivirus as surrogates for human norovirus. J Ginseng Res, 2011. 35(4): 429–435.
82. Gilling, D.H., et al., Antiviral efficacy and mechanisms of action of oregano essential oil and its primary
component carvacrol against murine norovirus. J Appl Microbiol, 2014. 116(5): 1149–1163.
83. Taube, S., et al., Ganglioside-linked terminal sialic acid moieties on murine macrophages function as
attachment receptors for murine noroviruses. J Virol, 2009. 83(9): 4092–4101.
84. Zhang, D., et al., Tulane virus recognizes the A type 3 and B histo-blood group antigens. J Virol, 2015.
89(2): 1419–1427.
85. Farkas, T., et al., Characterization of a rhesus monkey calicivirus representing a new genus of
Caliciviridae. J Virol, 2008. 82(11): 5408–5416.
92 Laboratory Models for Foodborne Infections
86. Hirneisen, K.A. and K.E. Kniel, Comparing human norovirus surrogates: murine norovirus and Tulane
virus. J Food Prot, 2013. 76(1): 139–143.
87. Dicaprio, E., et al., Internalization and dissemination of human norovirus and animal caliciviruses in
hydroponically grown romaine lettuce. Appl Environ Microbiol, 2012. 78(17): 6143–6152.
88. Wang, Q., et al., Survival of murine norovirus, Tulane virus, and hepatitis A virus on alfalfa seeds and
sprouts during storage and germination. Appl Environ Microbiol, 2013. 79(22): 7021–7027.
89. Li, X., et al., Pressure inactivation of Tulane virus, a candidate surrogate for human norovirus and its
potential application in food industry. Int J Food Microbiol, 2013. 162(1): 37.
90. Predmore, A., et al., Electron beam inactivation of Tulane virus on fresh produce, and mechanism of
inactivation of human norovirus surrogates by electron beam irradiation. Int J Food Microbiol, 2015.
198: 28–36.
91. Tian, P., et al., Inactivation of the Tulane virus, a novel surrogate for the human norovirus. J Food Prot,
2013. 76(4): 712–718.
92. Shearer, A., D.G. Hoover, and K. Kniel, Effect of bacterial cell-free supernatants on infectivity of
norovirus surrogates. J Food Prot, 2014. 77(1): 145–149.
93. Saif, L.J., et al., Rotavirus-like, calicivirus-like, and 23-nm virus-like particles associated with diarrhea
in young pigs. J Clin Microbiol, 1980. 12(1): 105–111.
94. Chang, K.-O., et al., Bile acids are essential for porcine enteric calicivirus replication in association with
down-regulation of signal transducer and activator of transcription 1. Proc Natl Acad Sci USA, 2004.
101(23): 8733–8738.
95. Li, J., et al., New interventions against human norovirus: progress, opportunities, and challenges. Annu
Rev Food Sci Technol, 2012. 3: 331–352.
96. Flynn, W.T. and L.J. Saif, Serial propagation of porcine enteric calicivirus-like virus in primary porcine
kidney cell cultures. J Clin Microbiol, 1988. 26(2): 206–212.
97. Guo, M., et al., Comparative pathogenesis of tissue culture-adapted and wild-type Cowden porcine
enteric calicivirus (PEC) in gnotobiotic pigs and induction of diarrhea by intravenous inoculation of
wild-type PEC. J Virol, 2001. 75(19): 9239–9251.
98. Tan, M., R.S. Hegde, and X. Jiang, The P domain of norovirus capsid protein forms dimer and binds to
histo-blood group antigen receptors. J Virol, 2004. 78(12): 6233–6242.
99. Mason, H.S., et al., Expression of Norwalk virus capsid protein in transgenic tobacco and potato and its
oral immunogenicity in mice. Proc Natl Acad Sci USA, 1996. 93(11): 5335–5340.
100. Santi, L., et al., An efficient plant viral expression system generating orally immunogenic Norwalk
virus-like particles. Vaccine, 2008. 26(15): 1846–1854.
101. Jiang, X., et al., Expression, self-assembly, and antigenicity of the Norwalk virus capsid protein. J Virol,
1992. 66(11): 6527–6532.
102. Koho, T., et al., Production and characterization of virus-like particles and the P domain protein of GII.
4 norovirus. J Virol Methods, 2012. 179(1): 1–7.
103. Li, D., et al., Effects of a variety of food extracts and juices on the specific binding ability of norovirus
GII.4 P particles. J Food Prot, 2012. 7(75): 1350–1354.
104. Dawson, D., et al., Survival of viruses on fresh produce, using MS2 as a surrogate for norovirus. J Appl
Microbiol, 2005. 98(1): 203–209.
105. Julian, T.R., J.M. Trumble, and K.J. Schwab, Evaluating efficacy of field-generated electrochemical oxi-
dants on disinfection of fomites using bacteriophage MS2 and mouse norovirus MNV-1 as pathogenic
virus surrogates. Food Environ Virol, 2014. 6(2): 145–155.
106. Alshraiedeh, N., et al., Atmospheric pressure, nonthermal plasma inactivation of MS2 bacteriophage:
effect of oxygen concentration on virucidal activity. J Appl Microbiol, 2013. 115(6): 1420–1426.
107. Black, E., et al., Coliphage as pressure surrogates for enteric viruses in foods. Innov Food Sci Emerg
Technol, 2010. 11(2): 239–244.
108. Jaykus, L., D.H. D’Souza, and C.L. Moe, Foodborne viral pathogens, in Food Microbiology, Fundamentals
and Frontiers, Doyle, M.P. and Buchanan, R.L., (Eds). 2014, ASM Press, Washington, DC.
109. Goyal, S.M., et al., Evaluating the virucidal efficacy of hydrogen peroxide vapour. J Hosp Infect, 2014.
86(4): 255–259.
110. Steinmann, J., Evaluation of effectiveness of Bacoban WB against feline calicivirus as surro-
gate for human norovirus. 2005; Available from: http://www.bacoban.de/downloads/zertifikate/
Bacoban%20WB/Norovirus_EN14476_EN.pdf.
Noroviruses: Laboratory Surrogates for Determining Survival and Inactivation 93
111. Nims, R. and M. Plavsic, Inactivation of caliciviruses. Pharmaceuticals (Basel), 2013. 6(3): 358–392.
112. Park, G.W. and M.D. Sobsey, Simultaneous comparison of murine norovirus, feline calicivirus,
coliphage MS2, and GII.4 norovirus to evaluate the efficacy of sodium hypochlorite against human
norovirus on a fecally soiled stainless steel surface. Foodborne Pathog Dis, 2011. 8(9): 1005–1010.
113. De Roda Husman, A.M., et al., Calicivirus inactivation by nonionizing (253.7-nanometer-wavelength
[UV]) and ionizing (gamma) radiation. Appl Environ Microbiol, 2004. 70(9): 5089–5093.
114. Mohr, H., B. Lambrecht, and A. Selz, Photodynamic virus inactivation of blood components. Immunol
Invest, 1995. 24(1–2): 73–85.
115. Chen, H., D.G. Hoover, and D.H. Kingsley, Temperature and treatment time influence high hydrostatic
pressure inactivation of feline calicivirus, a norovirus surrogate. J Food Prot, 2005. 68(11): 2389–2394.
116. Morino, H., et al., Inactivation of feline calicivirus by chlorine dioxide gas-generating gel. Yakugaku
Zasshi, 2013. 133(9): 1017–1022.
117. Kingsley, D.H., H. Chen, and D.G. Hoover, Inactivation of selected picornaviruses by high hydrostatic
pressure. Virus Res, 2004. 102(2): 221–224.
6
Rotavirus
CONTENTS
6.1 Introduction..................................................................................................................................... 95
6.2 Overview of Animal Models for Human Rotavirus (HRV) Research........................................... 97
6.2.1 History of Mouse Models for Rotavirus Research............................................................. 98
6.2.2 History of the Gnotobiotic (Gn) Pig Model for HRV Research......................................... 99
6.2.3 History of Nonhuman Primate Models for HRV Research............................................... 99
6.3 Rotavirus Replication and Pathogenesis........................................................................................101
6.3.1 Recent Major Advancements Made Using Mouse Models...............................................101
6.3.2 Major Advancements Made Using the Gn Pig Model..................................................... 102
6.4 Rotavirus Immunity and Vaccine Development........................................................................... 102
6.4.1 Mouse Models in the Study of Rotavirus Protective Immunity and Vaccines................ 102
6.4.2 Gnotobiotic Pig Model in the Study of Rotavirus Immunity and Vaccines.................... 103
6.4.2.1 Unique Features of the Gn Pig Model.............................................................. 103
6.4.2.2 HRV Vaccines and Therapeutics Evaluated in the Gn Pig Model of Wa
HRV Infection and Diarrhea���������������������������������������������������������������������������� 104
6.4.2.3 Gnotobiotic Pig Model Colonized with Probiotic Bacteria or Human Gut
Microbiota�������������������������������������������������������������������������������������������������������� 104
6.5 Cell Lines and Organoid/Enteroid Models for Rotavirus Infection............................................. 106
6.6 Concluding Remarks..................................................................................................................... 107
References............................................................................................................................................... 107
6.1 Introduction
Rotaviruses are recognized as the leading cause of severe gastroenteritis in infants and young chil-
dren worldwide since the discovery of human rotavirus (HRV) in the early 1970s.1–3 Rotaviruses are
transmitted via the fecal oral route, are highly contagious and stable in the environment,4 and post a
high risk of foodborne infections. Rotavirus is a nonenveloped, 70-nm icosahedral virus in the family
of Reoviridae. It has a genome consisting of 11 double-stranded RNA segments surrounded by a dis-
tinctive three-layered protein capsid.5 These 11 segments encode six structural proteins, VP1–4, VP6,
and VP7, and six nonstructural proteins, NSP1–6.6 Among the 11 viral genes, the properties of the
proteins encoded by genes 3, 4, 5, 9, and 10 are known to be related to rotavirus virulence in the host.7
Gene 3 encodes the capping enzyme that affects the level of viral RNA replication.8,9 Genes 4 and 9
produce the outer capsid proteins VP4 and VP7 required to initiate infection, and they induce virus
neutralizing antibodies independently from each other.10,11 Gene 5 codes NSP1 that functions as an
interferon antagonist.12–14 Gene 10 codes for the nonstructural protein NSP4, which regulates calcium
homeostasis and virus replication and acts as an enterotoxin.7 NSP2 (encoded by gene segment 8) is
also involved in virulence, especially in mice.15
Seven rotavirus groups (A to G) are known based on the antigenicity of VP6 and sequences of
genomic RNA.16 Among them, Groups A, B, and C are associated with illness in humans and animals,
95
96 Laboratory Models for Foodborne Infections
and Groups D through G with illness in animals only. Group A rotaviruses are the most common
cause of illness in infants and young children worldwide. The antigenicity of Group A rotaviruses
is highly diverse, and about 19 G (based on VP7) and 28 [P] (based on VP4) sero/genotypes have
been identified.17 Serotypes G1, G3, G4, and G9 are responsible for 90% of all HRV infections in
North America and Europe but for less than 70% infections in Africa.18 Genotypes P[8] and P[4]
account for over 90% of circulating P types worldwide, whereas in Africa, the relative frequency of
these two P types is lower and P[6] accounts for around a third of all detected P types.18 There are
potentially “211” different combinations of G and P proteins that can be generated; however, the actual
number of G and P combinations is less than the possible number because most combinations do not
survive subsequent rounds of replication in the host due to lack of fitness. The different rotaviruses
circulating in humans are categorized as (1) common human genotypes (G1P[8], G2P[4], G3P[8],
G4P[8]); (2) reassortants among human genotypes (G1P[4], G2P[8], G4P[4]); (3) reassortants between
animal and human genotypes (G1P[9], G4P[6], G9P[8], G12P[8]); and (4) likely zoonotic introductions
(G9P[6], G9P[11], G10P[11], G12P[6]).17,19 Among these, G1, G2, G3, and G4 in combination with P[4]
and P[8] represented over 88% of strains worldwide based on studies published between 1998 and
2004.18 The data from the surveillance networks of the WHO also indicate that G1P[8], G9P[8], and
G2P[4] account for 75% of samples genotyped in North America, Europe, southeast Asia, and western
Pacific regions.
Rotavirus replicates in the small intestine and causes gastroenteritis. The symptoms of rotavirus
infection are severe watery diarrhea, fever, and vomiting, leading to fluid and electrolyte disequi-
librium. Severe diarrhea without fluid and electrolyte replacement may result in other secondary
complications (e.g., renal failure and death).20 Before the implementation of rotavirus vaccines in
many developed and some developing countries starting from 2006, it was estimated that rotavirus
infections annually accounted for more than 125 million cases of diarrhea (20%–50% of hospitaliza-
tions for gastroenteritis) and 600,000–870,000 deaths (around 20% of all diarrhea-associated deaths)
in children under 5 years of age worldwide.21 In the United States, rotavirus infections caused an
estimated 410,000 physician visits and 50,000–70,000 hospitalizations each year before rotavirus
vaccines were implemented in the vaccination program.22 Following implementation of rotavirus vac-
cination in 2006, all-cause acute gastroenteritis hospitalization rates among US children younger than
5 years declined by 31%–55% in each of the postvaccine years from 2008 to 201223 and the laboratory
detection rate of rotavirus declined by 57.8%–89.9% in each of the seven postvaccine years from 2007
to 2014.24
Because rotaviruses are highly contagious and stable in the environment, exposure in closed envi-
ronments usually results in rapid, widespread infections among susceptible individuals. Outbreaks of
rotavirus infection are common in childcare centers, and rotavirus is one of the most common causes of
nosocomial infection, especially in pediatric hospitals.25,26 In spite of improved sanitation and water sup-
ply, rotavirus infections remain an important cause of pediatric hospitalization in many countries where
rotavirus vaccines have not been implemented.
Given that rotaviruses are the single most important cause of severe acute gastroenteritis in infants and
young children, the worldwide impact of rotaviruses on public health has led to major research efforts in
the past 40 years in understanding rotavirus replication, host–pathogen interactions, pathogenesis, and
immunity, and in vaccine development to control rotavirus diarrhea. Animal models including mice,
rats, rabbits, pigs, lambs, calves, and nonhuman primates have been used extensively in these stud-
ies. Among these animal models, gnotobiotic (Gn) piglets were among the first animal models used to
study HRV right after the virus was associated with infantile gastroenteritis. Since then, the model has
continuously contributed to our understanding of rotavirus pathogenesis and immunity. Various mouse
models with different genetic backgrounds and immune competence are used extensively for studies
of rotavirus–host interactions, mechanisms of host range restriction, and protective immunity. In this
chapter, animal models pertaining to the establishment and applications in studies of rotavirus–host
interactions, pathogenesis, immunity, and vaccine development are reviewed with emphasis on mouse
and Gn pig models. The advantages, disadvantages, and limitations of the animal models are discussed.
Cell lines and tissue culture models that support rotavirus replication and have been used in rotavirus
research are summarized.
Rotavirus 97
TABLE 6.1
Advantages and Disadvantages of the Major Animal Models Used in Rotavirus Research
Animal Species and Age Advantages Disadvantages
Neonatal mice (between • Small size • Resistant to infection by HRV
1 and 14 days of age) • Inbred; homogeneous response; allow immune • Different from humans in
cell transfer experiments gastrointestinal physiology and
• Availability of many genetically modified immune system
strains (gene knockout mice) • Different histopathology from
• Availability of molecular biology and humans after rotavirus infection
immunology reagents • Cannot be used to study infection-
• Produce clinical signs of disease (diarrhea) for or vaccine-induced active immunity
a limited time after infection by murine against rotavirus diarrhea due to
rotavirus and high-dose heterologous rotavirus age-dependent disease
strains • Maternal antibodies can interfere
• Can be used to study passive protective with rotavirus infectivity and
immunity against rotavirus diarrhea by development of immune responses
immunizing dams
Adult mice (older than • Small size • Resistant to infection by HRV
15 days of age) • Inbred; homogeneous response; allow immune • Different from humans in
cell transfer experiments gastrointestinal physiology and
• Availability of many genetically modified immune system
strains (gene knockout mice) • Mouse strains with different genetic
• Availability of molecular biology and backgrounds display different
immunology reagents susceptibility to infection for the
• Can be used to study rotavirus infection or same rotavirus strain, and shed
vaccine-induced protective immunity using different titers of rotavirus
virus shedding as the readout • Cannot be used to study infection- or
vaccine-induced active immunity
against rotavirus diarrhea
Gnotobiotic pigs • Susceptible to infection by HRV (Wa and M • Outbred; variability in host response
strains) without prior adaptation and develop • Although increasing, the availability
diarrhea up to 8 week of age of reagents is still less than that for
• Display clinical signs of disease (diarrhea) and mice and humans
histopathological changes similar to human • Expensive
infants • Special facilities and trainings are
• Similarity to human infants in physiology, required
immune system, body size, milk diet, and
metabolism
• Large amount of cells can be isolated from
lymphoid tissues for comprehensive
immunological analysis
• Devoid of maternal antibodies and other
maternal and environmental confounding
factors
• Can be colonized with human gut microbiota
to more closely mimic humans
Nonhuman primates • High similarity with humans in genomics, • Fail to induce clinical signs of
gastrointestinal physiology, and immune disease by second passage HRV
system (after the HRV isolate is passaged in
• Neonates are susceptible to HRV infection and nonhuman primates)
diarrhea • Outbred; high variability in host
• Broad availability of immunology and response
molecular biology reagents • Varying levels of maternal
antibodies
• Ethical concerns
• Very expensive
• Special facilities and trainings are
required
Rotavirus 101
(human 116E and bovine B223 strains). In vitro studies have suggested that the P[11] rotavirus specifi-
cally recognizes LacNAc that are likely expressed on the intestinal epithelial cells of only neonates and
infants, not in older children or adults, as potential receptors for P[11] rotaviruses.157 Rotavirus can also
infect adults, but severe symptomatic disease is relatively uncommon and can result from immunosup-
pression,159 infection with an unusual virus strain, or extremely high doses of rotavirus.7 Further studies
are needed to better understand the age-dependent susceptibility and pathogenesis of rotaviruses, and
to fully identify the host genetic factors in susceptibility to rotavirus illness and the mechanisms behind
asymptomatic rotavirus shedding in adults.
immunity. In sucking mice orally inoculated with a heterologous simian or bovine rotavirus (RRV or
NCDV) strain or a homologous murine rotavirus (wild-type or tissue culture-adapted EHP) strain and
challenged after 6 weeks with a virulent murine rotavirus (wild-type strain ECW), a fecal IgA antibody
induced by homologous infection was identified as the correlate of protection.74 A murine model with
“backpack tumor” transplantation was used to determine the protective effects of antibodies against VP4
(the outer capsid viral protein) and VP6 (the major inner capsid viral protein). The study indicated that
in vivo intracellular viral inactivation by anti-VP6 secretory IgA during transcytosis is a mechanism of
host defense against rotavirus infection.170
Various gene knockout mice have been used to identify the determinants of rotavirus protective immu-
nity,78–85 including Rag-2 (devoid of both T and B cells), β2m (lack cytotoxic T cell responses), JHD (lack
B cell responses), and IgA knockout mice (no detectable IgA in the serum or in any secretions). These
studies have examined the roles of various components of humoral and cellular immunity in the resolu-
tion of primary infection or protection against rotavirus reinfection. The findings indicate that the IgA
antibody, CD4, and CD8 T cells are all important in mediating protective immunity; however, none of
the cellular effectors is absolutely indispensable in mediating protective immunity against rotavirus infec-
tion, suggesting the redundant nature of the host immune defense system of mice. The B-cell-dependent
humoral immunity appears to be the main mechanism of protection from rotavirus infection.171
The effectiveness of various novel rotavirus vaccines including DNA plasmids,172 VLP,173 and sub-
unit174 vaccines was first evaluated in mouse models of rotavirus infection for protection against virus
shedding before it was further evaluated in the Gn pig model of rotavirus diarrhea.104,127,136
Although there are many similarities between porcine and human physiology and immune systems,
there are also differences that need to be considered. One significant difference between humans and
pigs is that humans acquire maternal antibodies transplacentally, whereas the placenta of pigs acts
as a barrier to the transfer of macromolecules, including maternal antibodies and cytokines. Unlike
mice and humans, no maternal antibodies are transferred from the mother to the porcine fetus during
intrauterine development due to the special structure of the placenta wall in swine; hence, Gn pigs
are agammaglobulinemic but are immunocompetent at birth.184 Piglets acquire immunoglobulins (Ig)
solely by intestinal absorption of colostral Ig for about 36 h after birth before gut closure occurs.185
This feature permits basic studies of the ontogeny of neonatal immune responses and a true primary
antibody response to rotavirus to be evaluated. This is also an advantage for using this model to study
the effects of maternal antibodies and other maternal immune regulators on rotavirus vaccine efficacy
because the levels and titers of those introduced into the circulation or intestines of piglets can be
manipulated experimentally.125,144,145,150
The mechanisms of probiotic LGG on reducing HRV pathogenesis were examined in Gn pigs fed with
high-dose LGG and challenged with virulent Wa HRV.108,110 LGG treatment before and during HRV
gastroenteritis partially prevented virus-induced intestinal tissue damage, reduced autophagy marker
expression to normal levels, induced apoptosis,108 and partially prevented HRV-induced compensatory
increases of the adherent junction proteins, α-catenin, and β-catenin; tight junction proteins, occludin,
claudin-3, and claudin-4; and leak protein claudin-2 in the ileal epithelium.110
The immune-modulating effects of probiotic Lactobacillus strains L. acidophilus NCFM, L. reuteri
and LGG on intestinal and systemic B cells, CD4 and CD8 T cells, γ/δ T cell, macrophage/dendritic
cell, toll-like receptor, and cytokine responses to virulent HRV infection or the live attenuated Wa HRV
vaccine were comprehensively examined in a series of studies on Gn pigs fed with different doses of
L. acidophilus NCFM or LGG101,106,113,191 or the mixture of L. acidophilus NCFM and L. reuteri.116,117,120–123
L. acidophilus NCFM and LGG are shown to have therapeutic as well as immune-stimulating effects,
and have dual functions in partial protection against HRV diarrhea and as adjuvants for the HRV vac-
cine when used in the proper dosage.101,106 These studies also demonstrated that both the dose (CFU/day)
and dosing regimen (total number of days’ intake) of probiotics have significant effects on the immune
modulatory functions of probiotics. Different LGG dosages differentially modulated immune responses
to favor either the mucosal IgA response (five doses) or the T-cell response (nine doses).101 High-dosage
L. acidophilus NCFM intake induced regulatory immune responses, similar to the effect of microbiota
colonization in infants, which had a negative impact on the immunostimulatory effect of probiotics and
abolished its effectiveness as a vaccine adjuvant.106
The lack of gut microbiota is a unique feature of the Gn pig model that provides an indispensable tool
for the study of the consequences of bacterial colonization, but it can also be a drawback when results
from preclinical vaccine studies need to be extrapolated to humans. In normal humans and conven-
tional animals, commensal microbiota helps the host to defend against pathogenic microorganisms.190
These bacteria provide the colonization resistance against attachment, multiplication, and invasion of
pathogenic microorganisms into epithelial cells and their potential circulation in the host. Furthermore,
they exert a major influence on the development of the mucosal immune system and regulation of host
immune responses. In the very early stage of the postnatal period, the immune responses are biased
toward Th2, and the Th1-mediated immune response is lacking.192,193 Thereafter, intestinal commensal
microbiota stimulates the development of both local and systemic immune systems, preferentially Th1
immune responses, but later induces regulatory mechanisms to maintain homeostasis.194 The intesti-
nal colonization of germfree animals with commensal microbes not only significantly stimulates the
development of mucosal and systemic immune systems, but may also promote integrity of the epithe-
lial barrier by regulating tight junctions and protecting it from injury with enhanced proliferation and
cytoprotective protein production.195–199 In addition, commensal bacteria regulate cell migration to rein-
force the epithelial barrier.200 Intestinal colonization also stimulates mucin secretion from goblet cells
to limit intestinal infections through binding between mucin and pathogens.201,202 Intestinal microbiota
also drive the development of Th17 cells203 and Treg cells.204 Therefore, the lack of gut microbiota in Gn
pigs can cause deviations in the pathogenesis of HRV infection and in the patterns of immune responses
induced by HRV infection or on vaccination, as compared to those observed in human infants.
In order to identify the influence of microbiota on the Gn pig’s response to HRV and to more closely
mimic human infants, Gn pigs transplanted with newborn HGM and infected with HRV have been estab-
lished. This model has been used to test the effects of probiotics on the gut microbiome structure during
an HRV infection.102,105 The development of HRV vaccine-induced immune responses has been compared
between the HGM and non-HGM-transplanted Gn pigs. HGM successfully colonized the Gn pig intestine
after three oral inoculations. Sequencing of the V4 region of 16S rRNA genes demonstrated that the pigs
carried a microbiome similar to that of a C-section delivered human infant.102 The attenuated Wa HRV
vaccine conferred similar overall protection against rotavirus diarrhea and virus shedding in Gn pigs and
HGM-transplanted Gn pigs. HGM promoted the development of the neonatal immune system, significantly
enhanced IFN-γ-producing T-cell responses, and reduced Treg cell responses in the AttHRV-vaccinated
pigs.105 Given the many advantages of HGM pigs (reviewed by Wang et al.151), the HGM pig model will
have wide applications for the studies of viral pathogenesis; interactions between host-microbiota, host-
pathogens, and microbiota-pathogens; and for the evaluation of vaccines and therapeutics.143
106 Laboratory Models for Foodborne Infections
TABLE 6.2
Cell Lines Used in Rotavirus Research
Cell Line Rotavirus Strain References
MA104 cells Bovine rotavirus RF and UK 141,210–218
Simian rotavirus RRV and SA-11
HRV Wa, RV4, M4, Wi, M69 strains, and from clinical
stool samples
Porcine rotavirus OSU, CRW-8, and YM
Murine rotavirus EB
Caco-2 RRV and SA-11 217,219,220
HRV Wa, Wi, and M69
Porcine small intestinal epithelial cell Porcine rotavirus OSU 221,222
line (IPEC-J2) HRV Wa
Vero cells HRV Wa and CDC-9 223,224
Primary AGMK cells HRV from clinical stool samples, murine rotavirus EB 100,207,218
Cos-7 cells Bovine rotavirus RF 216,225
HRV M4
HT-29 SA11–4F, Wa, Bovine rotavirus A5–13 226
Stem-cell-derived human intestinal SA11 and rotavirus from clinical stool samples 208
organoids
Human enteroids Laboratory strains and rotavirus from clinical stool 209
samples
Rotavirus 107
6.6 Concluding Remarks
Animal models have been critical in our understanding of rotavirus pathogenesis and immunity and in
the preclinical assessment of the safety and efficacy of rotavirus vaccines and therapeutics. The most
widely used animal models, namely mice and Gn pigs, present different sets of advantages and limita-
tions, and will continue to be indispensable tools for rotavirus research. Further optimizations of the
models, including humanization of the immune system through stem cell transfer, transplantation with
HGM from donors representing different health and immune statuses, and genetic modification using
CRISPR/Cas9 technology, will further improve the usefulness and reliability of the models for mimick-
ing rotavirus infection in humans.
REFERENCES
1. Davidson, G.P., Bishop, R.F., Townley, R.R. & Holmes, I.H. Importance of a new virus in acute sporadic
enteritis in children. Lancet 1, 242–6 (1975).
2. Bishop, R.F. et al. An epidemic of diarrhoea in human neonates involving a reovirus-like agent and
“enteropathogenic” serotypes of Escherichia coli. J Clin Pathol 29, 46–9 (1976).
3. Kapikian, A.Z. et al. Human reovirus-like agent as the major pathogen associated with “winter” gastro-
enteritis in hospitalized infants and young children. N Engl J Med 294, 965–72 (1976).
4. Hurst, C.J. & Gerba, C.P. Stability of simian rotavirus in fresh and estuarine water. Appl Environ
Microbiol 39, 1–5 (1980).
5. McDonald, S.M. & Patton, J.T. Assortment and packaging of the segmented rotavirus genome. Trends
Microbiol 19, 136–44 (2011).
6. Patton, J.T., Vasquez-Del Carpio, R. & Spencer, E. Replication and transcription of the rotavirus
genome. Curr Pharm Des 10, 3769–77 (2004).
7. Greenberg, H.B. & Estes, M.K. Rotaviruses: from pathogenesis to vaccination. Gastroenterology 136,
1939–51 (2009).
8. Chen, D., Luongo, C.L., Nibert, M.L. & Patton, J.T. Rotavirus open cores catalyze 5ʹ-capping and meth-
ylation of exogenous RNA: evidence that VP3 is a methyltransferase. Virology 265, 120–30 (1999).
9. Liu, M., Mattion, N.M. & Estes, M.K. Rotavirus VP3 expressed in insect cells possesses guanylyltrans-
ferase activity. Virology 188, 77–84 (1992).
10. Li, Y.J. et al. Oral vaccination with the porcine rotavirus VP4 outer capsid protein expressed by
Lactococcus lactis induces specific antibody production. J Biomed Biotechnol 2010, 708460 (2010).
11. Ward, R. Mechanisms of protection against rotavirus infection and disease. Pediatr Infect Dis J 28,
S57–9 (2009).
12. Barro, M. & Patton, J.T. Rotavirus nonstructural protein 1 subverts innate immune response by inducing
degradation of IFN regulatory factor 3. Proc Natl Acad Sci USA 102, 4114–9 (2005).
13. Pina-Vazquez, C., De Nova-Ocampo, M., Guzman-Leon, S. & Padilla-Noriega, L. Post-translational
regulation of rotavirus protein NSP1 expression in mammalian cells. Arch Virol 152, 345–68 (2007).
14. Graff, J.W., Mitzel, D.N., Weisend, C.M., Flenniken, M.L. & Hardy, M.E. Interferon regulatory factor 3
is a cellular partner of rotavirus NSP1. J Virol 76, 9545–50 (2002).
15. Lundgren, O. & Svensson, L. Pathogenesis of rotavirus diarrhea. Microbes Infect 3, 1145–56 (2001).
16. Saif, L.J. & Jiang, B. Nongroup A rotaviruses of humans and animals. Curr Top Microbiol Immunol
185, 339–71 (1994).
17. Hyser, J.M. & Estes, M.K. Rotavirus vaccines and pathogenesis: 2008. Curr Opin Gastroenterol 25,
36–43 (2009).
18. Santos, N. & Hoshino, Y. Global distribution of rotavirus serotypes/genotypes and its implication for the
development and implementation of an effective rotavirus vaccine. Rev Med Virol 15, 29–56 (2005).
19. Gray, J. et al. Rotavirus. J Pediatr Gastroenterol Nutr 46 Suppl 2, S24–31 (2008).
20. Offit, P.A. & Rubin, D.H. Viral diseases: infections of the gastrointestinal tract. Compr Ther 8, 21–6
(1982).
21. Bresee, J.S., Glass, R.I., Ivanoff, B. & Gentsch, J.R. Current status and future priorities for rotavi-
rus vaccine development, evaluation and implementation in developing countries. Vaccine 17, 2207–22
(1999).
108 Laboratory Models for Foodborne Infections
22. Glass, R.I., Gentsch, J.R. & Ivanoff, B. New lessons for rotavirus vaccines. Science 272, 46–8 (1996).
23. Leshem, E. et al. Acute gastroenteritis hospitalizations among US children following implementation of
the rotavirus vaccine. JAMA 313, 2282–4 (2015).
24. Aliabadi, N., Tate, J.E., Haynes, A.K. & Parashar, U.D. Sustained decrease in laboratory detection of
rotavirus after implementation of routine vaccination-United States, 2000–2014. MMWR Morb Mortal
Wkly Rep 64, 337–42 (2015).
25. O’Ryan, M.L., Matson, D.O., Estes, M.K., Bartlett, A.V. & Pickering, L.K. Molecular epidemiology of
rotavirus in children attending day care centers in Houston. J Infect Dis 162, 810–6 (1990).
26. Matson, D.O. & Estes, M.K. Impact of rotavirus infection at a large pediatric hospital. J Infect Dis 162,
598–604 (1990).
27. Ward, R.L., McNeal, M.M., Farone, M.B. & Farone, A.L. Reoviridae. In The Mouse in Biomedical
Research. Vol. 2, Diseases (eds. Fox, J.G. et al.), pp. 235–268. Academic Press, New York/London
(2007).
28. Matthijnssens, J. et al. Full genome-based classification of rotaviruses reveals a common origin between
human Wa-Like and porcine rotavirus strains and human DS-1-like and bovine rotavirus strains. J Virol
82, 3204–19 (2008).
29. Papp, H. et al. Zoonotic transmission of reassortant porcine G4P[6] rotaviruses in Hungarian pediatric
patients identified sporadically over a 15 year period. Infect Genet Evol 19, 71–80 (2013).
30. Zeller, M., Heylen, E., De Coster, S., Van Ranst, M. & Matthijnssens, J. Full genome characterization of
a porcine-like human G9P[6] rotavirus strain isolated from an infant in Belgium. Infect Genet Evol 12,
1492–500 (2012).
31. My, P.V. et al. Novel porcine-like human G26P[19] rotavirus identified in hospitalized paediatric diar-
rhoea patients in Ho Chi Minh City, Vietnam. J Gen Virol 95, 2727–33 (2014).
32. Steyer, A., Poljsak-Prijatelj, M., Barlic-Maganja, D. & Marin, J. Human, porcine and bovine rotaviruses
in Slovenia: evidence of interspecies transmission and genome reassortment. J Gen Virol 89, 1690–8
(2008).
33. Zhou, X. et al. Genomic characterization of G3P[6], G4P[6] and G4P[8] human rotaviruses from
Wuhan, China: evidence for interspecies transmission and reassortment events. Infect Genet Evol 33,
55–71 (2015).
34. Bohl, E.H. et al. Rotavirus as a cause of diarrhea in pigs. J Am Vet Med Assoc 172, 458–63 (1978).
35. Cordle, C.T. et al. Passive immune protection from diarrhea caused by rotavirus or E. coli: an animal
model to demonstrate and quantitate efficacy. Adv Exp Med Biol 310, 317–27 (1991).
36. Torres, A. & Ji-Huang, L. Diarrheal response of gnotobiotic pigs after fetal infection and neonatal chal-
lenge with homologous and heterologous human rotavirus strains. J Virol 60, 1107–12 (1986).
37. Ward, L.A., Rosen, B.I., Yuan, L. & Saif, L.J. Pathogenesis of an attenuated and a virulent strain of
group A human rotavirus in neonatal gnotobiotic pigs. J Gen Virol 77(Pt 7), 1431–41 (1996).
38. Snodgrass, D.R., Madeley, C.R., Wells, P.W. & Angus, K.W. Human rotavirus in lambs: infection and
passive protection. Infect Immun 16, 268–70 (1977).
39. Torres-Medina, A., Wyatt, R.G., Mebus, C.A., Underdahl, N.R. & Kapikian, A.Z. Diarrhea caused in
gnotobiotic piglets by the reovirus-like agent of human infantile gastroenteritis. J Infect Dis 133, 22–7
(1976).
40. Mebus, C.A. et al. Diarrhea in gnotobiotic calves caused by the reovirus-like agent of human infantile
gastroenteritis. Infect Immun 14, 471–4 (1976).
41. Varshney, K.C. et al. The lesions of rotavirus infection in 1- and 10-day-old gnotobiotic calves. Vet
Pathol 32, 619–27 (1995).
42. Wyatt, R.G. et al. Rotaviral immunity in gnotobiotic calves: heterologous resistance to human virus
induced by bovine virus. Science 203, 548–50 (1979).
43. Vesikari, T. et al. Immunogenicity and safety of live oral attenuated bovine rotavirus vaccine strain RIT
4237 in adults and young children. Lancet 2, 807–11 (1983).
44. Zissis, G. et al. Protection studies in colostrum-deprived piglets of a bovine rotavirus vaccine candidate
using human rotavirus strains for challenge. J Infect Dis 148, 1061–8 (1983).
45. Van Pinxteren, L.A., Campbell, I., Clarke, C.J., Snodgrass, D.R. & Bruce, M.G. A single oral dose of
inactivated rotavirus and ISCOM matrices induces partial protection in lambs. Biochem Soc Trans 25,
340S (1997).
Rotavirus 109
46. van Pinxteren, L.A., Bruce, M.G., Campbell, I., Clarke, C.J. & Snodgrass, D.R. Characterisation of
the primary local and systemic immune response in gnotobiotic lambs against rotavirus infection. Vet
Immunol Immunopathol 64, 349–65 (1998).
47. van Pinxteren, L.A. et al. Effect of oral rotavirus/iscom vaccines on immune responses in gnotobiotic
lambs. Vet Immunol Immunopathol 71, 53–67 (1999).
48. McNeal, M.M. et al. Development of a rotavirus-shedding model in rhesus macaques, using a homolo-
gous wild-type rotavirus of a new P genotype. J Virol 79, 944–54 (2005).
49. Westerman, L.E. et al. Isolation and characterization of a new simian rotavirus, YK-1. Virol J 3, 40
(2006).
50. Soike, K.F., Gary, G.W. & Gibson, S. Susceptibility of nonhuman primate species to infection by simian
rotavirus SA-11. Am J Vet Res 41, 1098–1103 (1980).
51. Hoshino, Y. et al. A rotavirus strain isolated from pig-tailed macaque (Macaca nemestrina) with diar-
rhea bears a P6[1]:G8 specificity. Virology 345, 1–12 (2006).
52. Westerman, L.E., Xu, J., Jiang, B., McClure, H.M. & Glass, R.I. Experimental infection of pigtailed
macaques with a simian rotavirus, YK-1. J Med Virol 75, 616–25 (2005).
53. Zhao, W. et al. Evaluation of rotavirus dsRNA load in specimens and body fluids from experimentally
infected juvenile macaques by real-time PCR. Virology 341, 248–56 (2005).
54. Wyatt, R.G. et al. Induction of diarrhea in colostrum-deprived newborn rhesus monkeys with the human
reovirus-like agent of infantile gastroenteritis. Arch Virol 50, 17–27 (1976).
55. Majer, M. et al. Diarrhea in newborn cynomologus monkeys infected with human rotavirus. Infection 6,
71–2 (1978).
56. Chege, G.K. et al. Experimental infection of non-human primates with a human rotavirus isolate.
Vaccine 23, 1522–8 (2005).
57. Sestak, K. et al. Defining T-cell-mediated immune responses in rotavirus-infected juvenile rhesus
macaques. J Virol 78, 10258–64 (2004).
58. Conner, M.E., Estes, M.K. & Graham, D.Y. Rabbit model of rotavirus infection. J Virol 62, 1625–33
(1988).
59. Ciarlet, M., Conner, M.E., Finegold, M.J. & Estes, M.K. Group A rotavirus infection and age-dependent
diarrheal disease in rats: a new animal model to study the pathophysiology of rotavirus infection. J Virol
76, 41–57 (2002).
60. Ciarlet, M. et al. Rotavirus disease, but not infection and development of intestinal histopathological
lesions, is age restricted in rabbits. Virology 251, 343–60 (1998).
61. Ciarlet, M. et al. Subunit rotavirus vaccine administered parenterally to rabbits induces active protective
immunity. J Virol 72, 9233–46 (1998).
62. Ciarlet, M., Estes, M.K., Barone, C., Ramig, R.F. & Conner, M.E. Analysis of host range restriction
determinants in the rabbit model: comparison of homologous and heterologous rotavirus infections. J
Virol 72, 2341–51 (1998).
63. Crawford, S.E. et al. Heterotypic protection and induction of a broad heterotypic neutralization response
by rotavirus-like particles. J Virol 73, 4813–22 (1999).
64. Bertolotti-Ciarlet, A., Ciarlet, M., Crawford, S.E., Conner, M.E. & Estes, M.K. Immunogenicity and
protective efficacy of rotavirus 2/6-virus-like particles produced by a dual baculovirus expression vector
and administered intramuscularly, intranasally, or orally to mice. Vaccine 21, 3885–900 (2003).
65. Crawford, S.E. et al. Rotavirus viremia and extraintestinal viral infection in the neonatal rat model.
J Virol 80, 4820–32 (2006).
66. Eydelloth, R.S., Vonderfecht, S.L., Sheridan, J.F., Enders, L.D. & Yolken, R.H. Kinetics of viral replication
and local and systemic immune responses in experimental rotavirus infection. J Virol 50, 947–50 (1984).
67. Kraft, L.M. Studies on the etiology and transmission of epidemic diarrhea of infant mice. J Exp Med
106, 743–55 (1957).
68. Kraft, L.M. Observations on the control and natural history of epidemic diarrhea of infant mice (EDIM).
Yale J Biol Med 31, 121–37 (1958).
69. Ward, R.L., McNeal, M.M. & Sheridan, J.F. Development of an adult mouse model for studies on pro-
tection against rotavirus. J Virol 64, 5070–5 (1990).
70. Ward, R.L. Mechanisms of protection against rotavirus in humans and mice. J Infect Dis 174 Suppl 1,
S51–8 (1996).
110 Laboratory Models for Foodborne Infections
71. McNeal, M.M., Belli, J., Basu, M., Choi, A.H. & Ward, R.L. Discovery of a new strain of murine rota-
virus that is consistently shed in large quantities after oral inoculation of adult mice. Virology 320, 1–11
(2004).
72. Choi, A.H., McNeal, M.M., Basu, M. & Ward, R.L. Immunity to homologous rotavirus infection in
adult mice: response. Trends Microbiol 8, 52 (2000).
73. Franco, M.A. & Greenberg, H.B. Immunity to homologous rotavirus infection in adult mice. Trends
Microbiol 8, 50–2 (2000).
74. Feng, N., Burns, J.W., Bracy, L. & Greenberg, H.B. Comparison of mucosal and systemic humoral
immune responses and subsequent protection in mice orally inoculated with a homologous or a heter-
ologous rotavirus. J Virol 68, 7766–73 (1994).
75. Reimerink, J.H. et al. Systemic immune response after rotavirus inoculation of neonatal mice depends
on source and level of purification of the virus: implications for the use of heterologous vaccine candi-
dates. J Gen Virol 88, 604–12 (2007).
76. Ball, J.M., Tian, P., Zeng, C.Q., Morris, A.P. & Estes, M.K. Age-dependent diarrhea induced by a rota-
viral nonstructural glycoprotein. Science 272, 101–4 (1996).
77. Horie, Y. et al. Diarrhea induction by rotavirus NSP4 in the homologous mouse model system. Virology
262, 398–407 (1999).
78. McNeal, M.M. et al. CD4 T cells are the only lymphocytes needed to protect mice against rotavirus
shedding after intranasal immunization with a chimeric VP6 protein and the adjuvant LT(R192G). J
Virol 76, 560–8 (2002).
79. Franco, M.A., Tin, C. & Greenberg, H.B. CD8+ T cells can mediate almost complete short-term and
partial long-term immunity to rotavirus in mice. J Virol 71, 4165–70 (1997).
80. Franco, M.A. & Greenberg, H.B. Immunity to rotavirus infection in mice. J Infect Dis 179 Suppl 3,
S466–9 (1999).
81. Dharakul, T. et al. Immunization with baculovirus-expressed recombinant rotavirus proteins VP1, VP4,
VP6, and VP7 induces CD8+ T lymphocytes that mediate clearance of chronic rotavirus infection in
SCID mice. J Virol 65, 5928–32 (1991).
82. Dharakul, T., Rott, L. & Greenberg, H.B. Recovery from chronic rotavirus infection in mice with severe
combined immunodeficiency: virus clearance mediated by adoptive transfer of immune CD8+ T lym-
phocytes. J Virol 64, 4375–82 (1990).
83. Blutt, S.E., Miller, A.D., Salmon, S.L., Metzger, D.W. & Conner, M.E. IgA is important for clearance
and critical for protection from rotavirus infection. Mucosal Immunol 5, 712–9 (2012).
84. Vancott, J.L., McNeal, M.M., Choi, A.H. & Ward, R.L. The role of interferons in rotavirus infections
and protection. J Interferon Cytokine Res 23, 163–70 (2003).
85. VanCott, J.L. et al. Role for T cell-independent B cell activity in the resolution of primary rotavirus
infection in mice. Eur J Immunol 31, 3380–7 (2001).
86. Holloway, G. & Coulson, B.S. Innate cellular responses to rotavirus infection. J Gen Virol 94, 1151–60
(2013).
87. Pott, J. et al. IFN-λ determines the intestinal epithelial antiviral host defense. Proc Natl Acad Sci USA
108, 7944–9 (2011).
88. Broquet, A.H., Hirata, Y., McAllister, C.S. & Kagnoff, M.F. RIG-I/MDA5/MAVS are required to
signal a protective IFN response in rotavirus-infected intestinal epithelium. J Immunol 186, 1618–26
(2011).
89. Pott, J. et al. Age-dependent TLR3 expression of the intestinal epithelium contributes to rotavirus sus-
ceptibility. PLoS Pathog 8, e1002670 (2012).
90. Feng, N., Franco, M.A. & Greenberg, H.B. Murine model of rotavirus infection. Adv Exp Med Biol 412,
233–40 (1997).
91. Franco, M.A., Feng, N. & Greenberg, H.B. Rotavirus immunity in the mouse. Arch Virol Suppl 12,
141–52 (1996).
92. Franco, M.A., Angel, J. & Greenberg, H.B. Immunity and correlates of protection for rotavirus vac-
cines. Vaccine 24, 2718–31 (2006).
93. Ward, R.L. Possible mechanisms of protection elicited by candidate rotavirus vaccines as determined
with the adult mouse model. Viral Immunol 16, 17–24 (2003).
94. Boshuizen, J.A. et al. Changes in small intestinal homeostasis, morphology, and gene expression during
rotavirus infection of infant mice. J Virol 77, 13005–16 (2003).
Rotavirus 111
95. Torres-Medina, A., Wyatt, R.G., Mebus, C.A., Underdahl, N.R. & Kapikian, A.Z. Patterns of shedding
of human reovirus-like agent in gnotobiotic newborn piglets with experimentally-induced diarrhea.
Intervirology 7, 250–55 (1976).
96. Wentzel, J.F., Yuan, L., Rao, S., van Dijk, A.A. & O’Neill, H.G. Consensus sequence determination and
elucidation of the evolutionary history of a rotavirus Wa variant reveal a close relationship to various Wa
variants derived from the original Wa strain. Infect Genet Evol 20, 276–83 (2013).
97. Tzipori, S.R., Makin, T.J. & Smith, M.L. The clinical response of gnotobiotic calves, pigs and lambs
to inoculation with human, calf, pig and foal rotavirus isolates. Aust J Exp Biol Med Sci 58, 309–18
(1980).
98. Hoshino, Y. et al. Identification of group A rotavirus genes associated with virulence of a porcine rotavi-
rus and host range restriction of a human rotavirus in the gnotobiotic piglet model. Virology 209, 274–80
(1995).
99. Saif, L.J., Ward, L.A., Yuan, L., Rosen, B.I. & To, T.L. The gnotobiotic piglet as a model for studies of
disease pathogenesis and immunity to human rotaviruses. Arch Virol Suppl 12, 153–61 (1996).
100. Wyatt, R.G. et al. Human rotavirus type 2: cultivation in vitro. Science 207, 189–91 (1980).
101. Wen, K. et al. Lactobacillus rhamnosus GG dosage affects the adjuvanticity and protection against
rotavirus diarrhea in gnotobiotic pigs. J Pediatr Gastroenterol Nutr 60, 834–843 (2015).
102. Zhang, H. et al. Probiotics and virulent human rotavirus modulate the transplanted human gut micro-
biota in gnotobiotic pigs. Gut Pathog 6, 39 (2014).
103. Yang, X. et al. Dietary rice bran protects against rotavirus diarrhea and promotes Th1-type immune
responses to human rotavirus vaccine in gnotobiotic pigs. Clin Vaccine Immunol 21, 1396–403
(2014).
104. Wen, X. et al. Inclusion of a universal tetanus toxoid CD4 + T cell epitope P2 significantly enhanced
the immunogenicity of recombinant rotavirus ΔVP8* subunit parenteral vaccines. Vaccine 32, 4420–7
(2014).
105. Wen, K. et al. Probiotic Lactobacillus rhamnosus GG enhanced Th1 cellular immunity but did not
affect antibody responses in a human gut microbiota transplanted neonatal gnotobiotic pig model. PLoS
One 9, e94504 (2014).
106. Liu, F. et al. Dual functions of Lactobacillus acidophilus NCFM as protection against rotavirus diar-
rhea. J Pediatr Gastroenterol Nutr 58, 169–76 (2014).
107. Kandasamy, S., Chattha, K.S., Vlasova, A.N., Rajashekara, G. & Saif, L.J. Lactobacilli and bifidobacte-
ria enhance mucosal B cell responses and differentially modulate systemic antibody responses to an oral
human rotavirus vaccine in a neonatal gnotobiotic pig disease model. Gut Microbes 5, 639–51 (2014).
108. Wu, S. et al. Probiotic Lactobacillus rhamnosus GG mono-association suppresses human rotavirus-
induced autophagy in the gnotobiotic piglet intestine. Gut Pathog 5, 22 (2013).
109. Vega, C.G. et al. Recombinant monovalent llama-derived antibody fragments (VHH) to rotavirus
VP6 protect neonatal gnotobiotic piglets against human rotavirus-induced diarrhea. PLoS Pathog 9,
e1003334 (2013).
110. Liu, F. et al. Lactobacillus rhamnosus GG on rotavirus-induced injury of ileal epithelium in gnotobiotic
pigs. J Pediatr Gastroenterol Nutr 57, 750–8 (2013).
111. Chattha, K.S. et al. Probiotics and colostrum/milk differentially affect neonatal humoral immune
responses to oral rotavirus vaccine. Vaccine 31, 1916–23 (2013).
112. Wen, K. et al. CD4 + CD25− FoxP3+ regulatory cells are the predominant responding regulatory T cells
after human rotavirus infection or vaccination in gnotobiotic pigs. Immunology 137, 160–71 (2012).
113. Wen, K. et al. High dose and low dose Lactobacillus acidophilus exerted differential immune modulat-
ing effects on T cell immune responses induced by an oral human rotavirus vaccine in gnotobiotic pigs.
Vaccine 30, 1198–207 (2012).
114. Wen, K. et al. Characterization of immune modulating functions of γδ T cell subsets in a gnotobiotic pig
model of human rotavirus infection. Comp Immunol Microbiol Infect Dis 35, 289–301 (2012).
115. Vega, C.G. et al. IgY antibodies protect against human rotavirus induced diarrhea in the neonatal gno-
tobiotic piglet disease model. PLoS One 7, e42788 (2012).
116. Azevedo, M.S. et al. Lactobacillus acidophilus and Lactobacillus reuteri modulate cytokine responses
in gnotobiotic pigs infected with human rotavirus. Benef Microbes 3, 33–42 (2012).
117. Wen, K. et al. Development of γδ T cell subset responses in gnotobiotic pigs infected with human rota-
viruses and colonized with probiotic lactobacilli. Vet Immunol Immunopathol 141, 267–75 (2011).
112 Laboratory Models for Foodborne Infections
118. Wang, Y. et al. Inactivated rotavirus vaccine induces protective immunity in gnotobiotic piglets. Vaccine
28, 5432–6 (2010).
119. Azevedo, M.S. et al. An oral versus intranasal prime/boost regimen using attenuated human rotavi-
rus or VP2 and VP6 virus-like particles with immunostimulating complexes influences protection
and antibody-secreting cell responses to rotavirus in a neonatal gnotobiotic pig model. Clin Vaccine
Immunol 17, 420–8 (2010).
120. Wen, K. et al. Toll-like receptor and innate cytokine responses induced by lactobacilli colonization and
human rotavirus infection in gnotobiotic pigs. Vet Immunol Immunopathol 127, 304–15 (2009).
121. Zhang, W. et al. Lactic acid bacterial colonization and human rotavirus infection influence distribu-
tion and frequencies of monocytes/macrophages and dendritic cells in neonatal gnotobiotic pigs. Vet
Immunol Immunopathol 121, 222–31 (2008).
122. Zhang, W. et al. Probiotic Lactobacillus acidophilus enhances the immunogenicity of an oral rotavirus
vaccine in gnotobiotic pigs. Vaccine 26, 3655–61 (2008).
123. Zhang, W. et al. Influence of probiotic Lactobacilli colonization on neonatal B cell responses in a gnotobi-
otic pig model of human rotavirus infection and disease. Vet Immunol Immunopathol 122, 175–81 (2008).
124. Yuan, L. et al. Virus-specific intestinal IFN-γ producing T cell responses induced by human rotavirus
infection and vaccines are correlated with protection against rotavirus diarrhea in gnotobiotic pigs.
Vaccine 26, 3322–31 (2008).
125. Nguyen, T.V. et al. High titers of circulating maternal antibodies suppress effector and memory B-cell
responses induced by an attenuated rotavirus priming and rotavirus-like particle-immunostimulating
complex boosting vaccine regimen. Clin Vaccine Immunol 13, 475–85 (2006).
126. Azevedo, M.S. et al. Cytokine responses in gnotobiotic pigs after infection with virulent or attenuated
human rotavirus. J Virol 80, 372–82 (2006).
127. Yuan, L. et al. Mucosal and systemic antibody responses and protection induced by a prime/boost
rotavirus-DNA vaccine in a gnotobiotic pig model. Vaccine 23, 3925–36 (2005).
128. Azevedo, M.S. et al. Viremia and nasal and rectal shedding of rotavirus in gnotobiotic pigs inoculated
with Wa human rotavirus. J Virol 79, 5428–36 (2005).
129. Nguyen, T.V. et al. Protection and antibody responses to oral priming by attenuated human rotavirus
followed by oral boosting with 2/6-rotavirus-like particles with immunostimulating complexes in gno-
tobiotic pigs. Vaccine 21, 4059–70 (2003).
130. Yuan, L. & Saif, L.J. Induction of mucosal immune responses and protection against enteric viruses:
rotavirus infection of gnotobiotic pigs as a model. Vet Immunol Immunopathol 87, 147–60 (2002).
131. Iosef, C. et al. Systemic and intestinal antibody secreting cell responses and protection in gnotobiotic
pigs immunized orally with attenuated Wa human rotavirus and Wa 2/6-rotavirus-like-particles associ-
ated with immunostimulating complexes. Vaccine 20, 1741–53 (2002).
132. Iosef, C., Chang, K.O., Azevedo, M.S. & Saif, L.J. Systemic and intestinal antibody responses to NSP4
enterotoxin of Wa human rotavirus in a gnotobiotic pig model of human rotavirus disease. J Med Virol
68, 119–28 (2002).
133. Yuan, L. et al. Protective immunity and antibody-secreting cell responses elicited by combined oral
attenuated Wa human rotavirus and intranasal Wa 2/6-VLPs with mutant Escherichia coli heat-labile
toxin in gnotobiotic pigs. J Virol 75, 9229–38 (2001).
134. Yuan, L., Geyer, A. & Saif, L.J. Short-term immunoglobulin A B-cell memory resides in intestinal
lymphoid tissues but not in bone marrow of gnotobiotic pigs inoculated with Wa human rotavirus.
Immunology 103, 188–98 (2001).
135. Chang, K.O., Vandal, O.H., Yuan, L., Hodgins, D.C. & Saif, L.J. Antibody-secreting cell responses to
rotavirus proteins in gnotobiotic pigs inoculated with attenuated or virulent human rotavirus. J Clin
Microbiol 39, 2807–13 (2001).
136. Yuan, L. et al. Intranasal administration of 2/6-rotavirus-like particles with mutant Escherichia coli
heat-labile toxin (LT-R192G) induces antibody-secreting cell responses but not protective immunity in
gnotobiotic pigs. J Virol 74, 8843–53 (2000).
137. Parreno, V. et al. Serum and intestinal isotype antibody responses to Wa human rotavirus in gnotobiotic
pigs are modulated by maternal antibodies. J Gen Virol 80(Pt 6), 1417–28 (1999).
138. Yuan, L., Kang, S.Y., Ward, L.A., To, T.L. & Saif, L.J. Antibody-secreting cell responses and protec-
tive immunity assessed in gnotobiotic pigs inoculated orally or intramuscularly with inactivated human
rotavirus. J Virol 72, 330–8 (1998).
Rotavirus 113
139. To, T.L., Ward, L.A., Yuan, L. & Saif, L.J. Serum and intestinal isotype antibody responses and cor-
relates of protective immunity to human rotavirus in a gnotobiotic pig model of disease. J Gen Virol 79,
2661–72 (1998).
140. Saif, L., Yuan, L., Ward, L. & To, T. Comparative studies of the pathogenesis, antibody immune
responses, and homologous protection to porcine and human rotaviruses in gnotobiotic piglets. Adv Exp
Med Biol 412, 397–403 (1997).
141. Yuan, L., Ward, L.A., Rosen, B.I., To, T.L. & Saif, L.J. Systematic and intestinal antibody-secreting
cell responses and correlates of protective immunity to human rotavirus in a gnotobiotic pig model of
disease. J Virol 70, 3075–83 (1996).
142. Ward, L.A., Yuan, L., Rosen, B.I., To, T.L. & Saif, L.J. Development of mucosal and systemic lym-
phoproliferative responses and protective immunity to human group A rotaviruses in a gnotobiotic pig
model. Clin Diagn Lab Immunol 3, 342–50 (1996).
143. Yang, X. & Yuan, L. Neonatal gnotobiotic pig models for studying viral pathogenesis, immune
responses, and for vaccine evaluation. Br J Virol 1, 87–91 (2014).
144. Hodgins, D.C. et al. Effects of maternal antibodies on protection and development of antibody responses
to human rotavirus in gnotobiotic pigs. J Virol 73, 186–97 (1999).
145. Nguyen, T.V. et al. Low titer maternal antibodies can both enhance and suppress B cell responses to a
combined live attenuated human rotavirus and VLP-ISCOM vaccine. Vaccine 24, 2302–16 (2006).
146. Azevedo, M.S. et al. Magnitude of serum and intestinal antibody responses induced by sequential rep-
licating and nonreplicating rotavirus vaccines in gnotobiotic pigs and correlation with protection. Clin
Diagn Lab Immunol 11, 12–20 (2004).
147. Azevedo, M.P., Vlasova, A.N. & Saif, L.J. Human rotavirus virus-like particle vaccines evaluated in a
neonatal gnotobiotic pig model of human rotavirus disease. Expert Rev Vaccines 12, 169–81 (2013).
148. Gonzalez, A.M. et al. Innate immune responses to human rotavirus in the neonatal gnotobiotic piglet
disease model. Immunology 131, 242–56 (2010).
149. Gonzalez, A.M. et al. Antibody responses to human rotavirus (HRV) in gnotobiotic pigs following a
new prime/boost vaccine strategy using oral attenuated HRV priming and intranasal VP2/6 rotavirus-
like particle (VLP) boosting with ISCOM. Clin Exp Immunol 135, 361–72 (2004).
150. Nguyen, T.V. et al. Transfer of maternal cytokines to suckling piglets: in vivo and in vitro models with
implications for immunomodulation of neonatal immunity. Vet Immunol Immunopathol 117, 236–48
(2007).
151. Wang, M. & Donovan, S.M. Human microbiota-associated swine: current progress and future opportu-
nities. ILAR J 56, 63–73 (2015).
152. Angel, J., Franco, M.A., Greenberg, H.B. & Bass, D. Lack of a role for type I and type II interferons in
the resolution of rotavirus-induced diarrhea and infection in mice. J Interferon Cytokine Res 19, 655–9
(1999).
153. Fu, C. et al. Effectiveness of the Lanzhou lamb rotavirus vaccine against gastroenteritis among children.
Vaccine 31, 154–8 (2012).
154. Ciarlet, M. & Schodel, F. Development of a rotavirus vaccine: clinical safety, immunogenicity, and
efficacy of the pentavalent rotavirus vaccine, RotaTeq®. Vaccine 27 Suppl 6, G72–81 (2009).
155. Feng, N., Yasukawa, L.L., Sen, A. & Greenberg, H.B. Permissive replication of homologous murine
rotavirus in the mouse intestine is primarily regulated by VP4 and NSP1. J Virol 87, 8307–16 (2013).
156. Ray, P.G., Kelkar, S.D., Walimbe, A.M., Biniwale, V. & Mehendale, S. Rotavirus immunoglobulin levels
among Indian mothers of two socio-economic groups and occurrence of rotavirus infections among
their infants up to six months. J Med Virol 79, 341–9 (2007).
157. Liu, Y. et al. Poly-LacNAc as an age-specific ligand for rotavirus P[11] in neonates and infants. PLoS
One 8, e78113 (2013).
158. Huang, P. et al. Spike protein VP8* of human rotavirus recognizes histo-blood group antigens in a type-
specific manner. J Virol 86, 4833–43 (2012).
159. Liakopoulou, E. et al. Rotavirus as a significant cause of prolonged diarrhoeal illness and morbidity
following allogeneic bone marrow transplantation. Bone Marrow Transplant 36, 691–4 (2005).
160. Barnes, G.L. & Townley, R.R. Duodenal mucosal damage in 31 infants with gastroenteritis. Arch Dis
Child 48, 343–9 (1973).
161. Davidson, G.P. & Barnes, G.L. Structural and functional abnormalities of the small intestine in infants
and young children with rotavirus enteritis. Acta Paediatr Scand 68, 181–6 (1979).
114 Laboratory Models for Foodborne Infections
162. Yuan, L., Stevenson, G. & Saif, L. Rotavirus and reovirus. In Diseases of Swine (eds. Straw, B.E.,
Zimmerman, J.J., D’Allaire, S. & Taylor, D.J.), pp. 435–454. Blackwell Publishing, Ames, IA (2006).
163. Conner, M.E. & Ramig, R.F. Viral enteric diseases. In Viral Pathogenesis (ed. Nathanson, N.),
pp. 713–742. Lippincott-Raven, Philadelphia, PA (1997).
164. Davidson, G.P., Gall, D.G., Petric, M., Butler, D.G. & Hamilton, J.R. Human rotavirus enteritis induced
in conventional piglets. Intestinal structure and transport. J Clin Invest 60, 1402–9 (1977).
165. Blutt, S.E. et al. Rotavirus antigenemia in children is associated with viremia. PLoS Med 4, e121 (2007).
166. Blutt, S.E. & Conner, M.E. Rotavirus: to the gut and beyond! Curr Opin Gastroenterol 23, 39–43
(2007).
167. Blutt, S.E., Fenaux, M., Warfield, K.L., Greenberg, H.B. & Conner, M.E. Active viremia in rotavirus-
infected mice. J Virol 80, 6702–5 (2006).
168. Mendicino, M. et al. Generation of antibody- and B cell-deficient pigs by targeted disruption of the
J-region gene segment of the heavy chain locus. Transgenic Res 20, 625–41 (2011).
169. Lee, K. et al. Engraftment of human iPS cells and allogeneic porcine cells into pigs with inactivated
RAG2 and accompanying severe combined immunodeficiency. Proc Natl Acad Sci USA 111, 7260–5
(2014).
170. Burns, J.W., Siadat-Pajouh, M., Krishnaney, A.A. & Greenberg, H.B. Protective effect of rotavirus VP6-
specific IgA monoclonal antibodies that lack neutralizing activity. Science 272, 104–7 (1996).
171. Franco, M.A. & Greenberg, H.B. Immunity to rotavirus in T cell deficient mice. Virology 238, 169–79
(1997).
172. Chen, S.C. et al. Protective immunity induced by rotavirus DNA vaccines. Vaccine 15, 899–902 (1997).
173. O’Neal, C.M., Crawford, S.E., Estes, M.K. & Conner, M.E. Rotavirus virus-like particles administered
mucosally induce protective immunity. J Virol 71, 8707–17 (1997).
174. Choi, A.H., Basu, M., McNeal, M.M., Clements, J.D. & Ward, R.L. Antibody-independent protection
against rotavirus infection of mice stimulated by intranasal immunization with chimeric VP4 or VP6
protein. J Virol 73, 7574–81 (1999).
175. Meyer, R.C., Bohl, E.H. & Kohler, E.M. Procurement and maintenance of germ-free swine for micro-
biological investigations. Appl Microbiol 12, 295–300 (1964).
176. Graham, D.Y., Dufour, G.R. & Estes, M.K. Minimal infective dose of rotavirus. Arch Virol 92, 261–71
(1987).
177. Butler, J.E. Isolator and other neonatal piglet models in developmental immunology and identification
of virulence factors. Anim Health Res Rev 10, 35–52 (2009).
178. Butler, J.E. et al. The piglet as a model for B cell and immune system development. Vet Immunol
Immunopathol 128, 147–70 (2009).
179. Sinkora, M. & Butler, J.E. The ontogeny of the porcine immune system. Dev Comp Immunol 33, 273–83
(2009).
180. Butler, J.E., Wertz, N., Deschacht, N. & Kacskovics, I. Porcine IgG: structure, genetics, and evolution.
Immunogenetics 61, 209–30 (2009).
181. Butler, J.E., Zhao, Y., Sinkora, M., Wertz, N. & Kacskovics, I. Immunoglobulins, antibody repertoire
and B cell development. Dev Comp Immunol 33, 321–33 (2009).
182. Bianchi, A.T., Zwart, R.J., Jeurissen, S.H. & Moonen-Leusen, H.W. Development of the B- and T-cell
compartments in porcine lymphoid organs from birth to adult life: an immunohistological approach. Vet
Immunol Immunopathol 33, 201–21 (1992).
183. Meurens, F., Summerfield, A., Nauwynck, H., Saif, L. & Gerdts, V. The pig: a model for human infec-
tious diseases. Trends Microbiol 20, 50–7 (2012).
184. Butler, J.E. & Sinkora, M. The isolator piglet: a model for studying the development of adaptive immu-
nity. Immunol Res 39, 33–51 (2007).
185. Mehrazar, K., Gilman-Sachs, A. & Kim, Y.B. Intestinal absorption of immunologically intact mac-
romolecules in germfree colostrum-deprived piglets maintained on total parenteral nutrition. JPEN J
Parenter Enteral Nutr 17, 8–15 (1993).
186. Yuan, L. & Saif, L.J. Rotavirus-like particle vaccines evaluated in a pig model of human rotavirus
diarrhea and in cattle. In Viral Gastroenteritis (eds. Desselberger, U. & Gray, J.), pp. 357–368. Elsevier
Sciences, Netherlands (2003).
Rotavirus 115
187. Kandasamy, S., Chattha, K.S., Vlasova, A.N. & Saif, L.J. Prenatal vitamin A deficiency impairs adap-
tive immune responses to pentavalent rotavirus vaccine (RotaTeq®) in a neonatal gnotobiotic pig model.
Vaccine 32, 816–24 (2014).
188. Yuan, L. et al. Rotavirus-specific IFN-γ producing and proliferating T cell responses to human rotavirus
infection and vaccination in gnotobiotic pigs. The 4th International Conference on Vaccines for Enteric
Diseases, Apr. 25–27, Lisbon, Portugal (2007).
189. Yang, X. et al. High protective efficacy of rice bran against human rotavirus diarrhea via enhancing
probiotic growth, gut barrier function, and innate immunity. Sci Rep 5, 15004 (2015).
190. Tlaskalova-Hogenova, H. et al. The role of gut microbiota (commensal bacteria) and the mucosal barrier
in the pathogenesis of inflammatory and autoimmune diseases and cancer: contribution of germ-free
and gnotobiotic animal models of human diseases. Cell Mol Immunol 8, 110–20 (2011).
191. Yuan, L., Wen, K., Liu, F. & G., L. Dose effects of LAB on modulation of rotavirus vaccine induced
immune responses. In Lactic Acid Bacteria—R & D for Food, Health and Livestock Purposes (ed.
Kongo, J.M.). InTech, Rijeka, http://www.intechopen.com/books/lactic-acid-bacteria-r-d-for-food-
health-and-livestock-purposes/dose-effects-of-lab-on-modulation-of-rotavirus-vaccine-induced-
immune-responses (2013).
192. Smith, P.M. & Garrett, W.S. The gut microbiota and mucosal T cells. Front Microbiol 2, 111 (2011).
193. McLoughlin, R.M. & Mills, K.H. Influence of gastrointestinal commensal bacteria on the immune
responses that mediate allergy and asthma. J Allergy Clin Immunol 127, 1097–107; quiz 1108–9 (2011).
194. Walker, W.A. Initial intestinal colonization in the human infant and immune homeostasis. Ann Nutr
Metab 63 Suppl 2, 8–15 (2013).
195. Hooper, L.V. et al. Molecular analysis of commensal host-microbial relationships in the intestine.
Science 291, 881–4 (2001).
196. O’Hara, A.M. & Shanahan, F. The gut flora as a forgotten organ. EMBO Rep 7, 688–93 (2006).
197. Ukena, S.N. et al. Probiotic Escherichia coli Nissle 1917 inhibits leaky gut by enhancing mucosal integ-
rity. PLoS One 2, e1308 (2007).
198. Ewaschuk, J.B. et al. Secreted bioactive factors from Bifidobacterium infantis enhance epithelial cell
barrier function. Am J Physiol Gastrointest Liver Physiol 295, G1025–34 (2008).
199. Rakoff-Nahoum, S., Paglino, J., Eslami-Varzaneh, F., Edberg, S. & Medzhitov, R. Recognition of com-
mensal microflora by toll-like receptors is required for intestinal homeostasis. Cell 118, 229–41 (2004).
200. Swanson, P.A. et al. Enteric commensal bacteria potentiate epithelial restitution via reactive oxygen
species-mediated inactivation of focal adhesion kinase phosphatases. Proc Natl Acad Sci USA 108,
8803–8 (2011).
201. Linden, S.K., Florin, T.H. & McGuckin, M.A. Mucin dynamics in intestinal bacterial infection. PLoS
One 3, e3952 (2008).
202. Linden, S.K. et al. MUC1 limits Helicobacter pylori infection both by steric hindrance and by acting as
a releasable decoy. PLoS Pathog 5, e1000617 (2009).
203. Atarashi, K., Tanoue, T. & Honda, K. Induction of lamina propria Th17 cells by intestinal commensal
bacteria. Vaccine 28, 8036–8 (2010).
204. Kunisawa, J. & Kiyono, H. Peaceful mutualism in the gut: revealing key commensal bacteria for the
creation and maintenance of immunological homeostasis. Cell Host Microbe 9, 83–4 (2011).
205. Vashist, S., Bailey, D., Putics, A. & Goodfellow, I. Model systems for the study of human norovirus
biology. Future Virol 4, 353–367 (2009).
206. Ward, R.L., Knowlton, D.R. & Pierce, M.J. Efficiency of human rotavirus propagation in cell culture.
J Clin Microbiol 19, 748–53 (1984).
207. Ward, R.L. et al. Culture adaptation and characterization of group A rotaviruses causing diarrheal ill-
nesses in Bangladesh from 1985 to 1986. J Clin Microbiol 29, 1915–23 (1991).
208. Finkbeiner, S.R. et al. Stem cell-derived human intestinal organoids as an infection model for rotavi-
ruses. mBio 3, e00159-12 (2012).
209. Foulke-Abel, J. et al. Human enteroids as an ex-vivo model of host–pathogen interactions in the gastro-
intestinal tract. Exp Biol Med (Maywood) 239, 1124–34 (2014).
210. Razavinikoo, H. et al. Activation of calcium/calmodulin-dependent kinase II following bovine rotavirus
enterotoxin NSP4 expression. Iran J Basic Med Sci 18, 393–7 (2015).
116 Laboratory Models for Foodborne Infections
211. Donato, C.M. et al. Characterization of a G1P[8] rotavirus causing an outbreak of gastroenteritis in the
Northern Territory, Australia, in the vaccine era. Emerg Microbes Infect 3, e47 (2014).
212. Lipson, S.M., Ozen, F.S., Louis, S. & Karthikeyan, L. Comparison of α-glucosyl hesperidin of citrus
fruits and epigallocatechin gallate of green tea on the loss of rotavirus infectivity in cell culture. Front
Microbiol 6, 359 (2015).
213. Torres-Flores, J.M., Silva-Ayala, D., Espinoza, M.A., Lopez, S. & Arias, C.F. The tight junction protein
JAM-A functions as coreceptor for rotavirus entry into MA104 cells. Virology 475, 172–8 (2015).
214. Teimoori, A., Soleimanjahi, H. & Makvandi, M. Characterization and transferring of human rotavirus
double-layered particles in MA104 cells. Jundishapur J Microbiol 7, e10375 (2014).
215. Pane, J.A., Dang, V.T., Holloway, G., Webster, N.L. & Coulson, B.S. VP7 of rhesus monkey rotavi-
rus RRV contributes to diabetes acceleration in association with an elevated anti-rotavirus antibody
response. Virology 468–470, 504–9 (2014).
216. Duponchel, S. et al. Transfection of exogenous rotavirus rearranged RNA segments in cells infected
with a WT rotavirus results in subsequent gene rearrangements. J Gen Virol 95, 2089–98 (2014).
217. Guerrero, C.A., Murillo, A. & Acosta, O. Inhibition of rotavirus infection in cultured cells by N-acetyl-
cysteine, PPARγ agonists and NSAIDs. Antiviral Res 96, 1–12 (2012).
218. Tsugawa, T., Tatsumi, M. & Tsutsumi, H. Virulence-associated genome mutations of murine rotavirus
identified by alternating serial passages in mice and cell cultures. J Virol 88, 5543–58 (2014).
219. Bautista, D., Rodriguez, L.S., Franco, M.A., Angel, J. & Barreto, A. Caco-2 cells infected with rota-
virus release extracellular vesicles that express markers of apoptotic bodies and exosomes. Cell Stress
Chaperones 20, 697–708 (2015).
220. Huang, H., Liao, D., Liang, L., Song, L. & Zhao, W. Genistein inhibits rotavirus replication and upregu-
lates AQP4 expression in rotavirus-infected Caco-2 cells. Arch Virol 160, 1421–33 (2015).
221. Zhao, Y. et al. Effect of 25-hydroxyvitamin D3 on rotavirus replication and gene expressions of RIG-I
signalling molecule in porcine rotavirus-infected IPEC-J2 cells. Arch Anim Nutr 69, 227–35 (2015).
222. Liu, F. et al. Porcine small intestinal epithelial cell line (IPEC-J2) of rotavirus infection as a new model
for the study of innate immune responses to rotaviruses and probiotics. Viral Immunol 23, 135–49
(2010).
223. Lee do, K. et al. Probiotic bacteria, B. longum and L. acidophilus inhibit infection by rotavirus in vitro
and decrease the duration of diarrhea in pediatric patients. Clin Res Hepatol Gastroenterol 39, 237–44
(2015).
224. Esona, M.D. et al. Molecular characterization of human rotavirus vaccine strain CDC-9 during sequen-
tial passages in vero cells. Hum Vaccines 6, pii: 10409 (2010).
225. Diaz, Y. et al. Dissecting the Ca2+ entry pathways induced by rotavirus infection and NSP4-EGFP
expression in Cos-7 cells. Virus Res 167, 285–96 (2012).
226. Bagchi, P. et al. Identification of common human host genes involved in pathogenesis of different rota-
virus strains: an attempt to recognize probable antiviral targets. Virus Res 169, 144–53 (2012).
7
Prions
CONTENTS
7.1 Introduction....................................................................................................................................117
7.2 In Vitro Models..............................................................................................................................119
7.3 Cell-Culture Models..................................................................................................................... 120
7.4 Animal Models............................................................................................................................. 122
7.5 Conclusions................................................................................................................................... 123
Acknowledgments................................................................................................................................... 123
References............................................................................................................................................... 124
7.1 Introduction
Transmissible spongiform encephalopathies (TSEs), also known as prion diseases, are fatal neurological
disorders that include Creutzfeldt–Jakob disease (CJD) in humans, scrapie in sheep and goats, bovine
spongiform encephalopathy (BSE) in cattle, chronic wasting disease (CWD) in cervids, transmissible
mink encephalopathy (TME) in mink, feline spongiform encephalopathy (FSE) in cats, and exotic ungu-
late encephalopathy (EUE) in zoo animals such as kudu, nyala, gemsbok, eland, and oryx [1]. The prion
agent associated with each disease is named after the disease itself (e.g., CJD agent in CJD, scrapie
agent in scrapie, etc.). A key event in the development of prion diseases is the conversion of the cellular,
host-encoded prion protein (PrPC) to its abnormal isoform (PrPSc) predominantly in the central nervous
system (CNS) of the infected host [1]. Numerous compelling observations support the notion that PrPSc
is the main component of prion agents [2,3]. PrPSc, in contrast to PrPC, is resistant to complete digestion
with proteinase K (PK). Consequently, the detection of PK-resistant prion protein (PrPres) is generally
used for identifying the presence of a prion agent, because PrPres contains PrPSc. Therefore, most diag-
nostic methods for prion diseases are based on the index of PrPSc.
Several diagnostic methods have been used for prion diseases. These include enzyme-linked immu-
nosorbent assay (ELISA), western blotting, and immunohistochemistry (IHC) [4]. As an index of prion
infection, representative changes in the brain are generally assayed. Prion protein (PrP) accumulates in
the brain to form deposits. PrPres is biochemically detected after treatment with PK using western blot-
ting (Figure 7.1) and ELISA (Figure 7.2). In the case of ELISA for BSE, a homogenate is usually prepared
from the obex region of the brain, which is subsequently treated with PK. The PK-treated sample is then
applied to a microtiter plate for absorption and reacted with an anti-PrP antibody. This method is com-
mercially exploited in the Bio-Rad TeSE BSE kit (Bio-Rad, France), Enfer-TSE kit (Abbott Laboratories,
USA), and FRELISA BSE Kit (Fujirebio Inc., Japan). Although the extensively employed ELISA is a
sensitive and high-throughput method, the large number of false positives remains a significant problem.
Therefore, if a result is positive, ELISA must be repeated to validate the finding. If a positive result is
obtained again, western blotting and IHC are performed.
Western blotting initially involves separation of the PK-treated proteins by sodium dodecyl sulfate
(SDS)–polyacrylamide gel electrophoresis (PAGE) followed by electroblotting onto a suitable membrane
support. After blotting, PrPres present as a membrane-bound protein can be detected using an anti-PrP
117
118 Laboratory Models for Foodborne Infections
PrPC PrPSc
PrPC
PK PK
Band
pattern
FIGURE 7.1 Western blotting for detection of PrPSc. Most methods for diagnosing prion diseases are based on the char-
acteristics of the PrPSc, which is resistant to PK. PK completely degrades the PrPC but only partially digests PrPSc because
the latter forms protease-resistant aggregates. After protease treatment, PrPres is detected by western blotting with an
anti-PrP antibody. Western blotting reveals that PK-resistant PrP shifts to a lower molecular weight compared to untreated
PrP (−) because the N-terminal region of PrPSc is susceptible to PK (+). A schematic representation of a Western blot for
PrP derived from representative tissues of prion-infected and uninfected mice before (−) and after (+) PK treatment is
shown. The tissue of choice for PrPSc analysis is the brain.
PK PK
ELISA
Well
color
FIGURE 7.2 ELISA for detection of PrPSc. ELISA diagnosis of prion diseases is based on the characteristics of PrPSc,
which is resistant to PK. PK completely degrades PrP C but only partially digests PrPSc, because PrPSc forms protease-
resistant aggregates. After PK treatment, ELISA is performed using an anti-PrP antibody to detect PK-resistant PrP
(when PrPSc is present in the sample). Because the N-terminal portion of PrPSc is digested with PK, an antibody should
be chosen that recognizes an alternative region of PrP. In the case of a sandwich ELISA, which is becoming increas-
ingly popular, two discrete anti-PrP antibodies recognizing different epitopes of PrP are used. Specifically, one acts as
a capture antibody, which is coated onto the well of the microplate, while the other is a detection antibody conjugated
with an enzyme such as horseradish peroxidase (HRP) or alkaline phosphatase (AP). After reaction with the detection
antibody, the corresponding substrate for the conjugated enzyme is added. The resulting color change caused by the
enzyme reaction is then measured using a microplate reader on the basis of absorbance.
Prions 119
antibody. Importantly, western blotting provides information on both the prion infection and the mobil-
ity of peptides, which is influenced by the host genotype and strain of prion [5]. In the case of IHC, the
indexes for prion infection are neuronal cell loss, astrocytosis, and vacuolation in addition to PrP accu-
mulation (amyloid plaques) [6]. In IHC-based analyses of brain sections, these changes are examined by
light microscopy.
A number of novel, alternative diagnostic methods for prion diseases have also been developed, and
recently reviewed [4,7,8]. Readers may refer to these articles for further technical details.
In this chapter, we describe laboratory models used for analyzing prion infections. The laboratory
models for prion infections are divided into in vitro models, cellular models, and animal models. In all
three cases, prion infection is diagnosed by detection of PrPSc as described earlier.
PrPSc
PrPC Incubation
Conformational
change
Sonication/
incubation
FIGURE 7.3 PMCA for amplification of PrPSc. Recently, a powerful diagnostic method for prion diseases, known as
PMCA, has been developed for the amplification of PrPSc prions from body fluids such as blood, cerebral spinal fluid,
semen, milk, urine, and saliva during the preclinical and clinical phases of prion diseases [61]. In this method, a PrPSc seed
is incubated with the PrPC source. PrPSc in the presence of excess PrPC induces the formation of large aggregates that are
subsequently broken up by sonication. Repeated cycles of this procedure efficiently amplify PrPSc.
120 Laboratory Models for Foodborne Infections
prions in blood [12]. PMCA has been successfully used to diagnose both terminally diseased hamsters
and prion-infected presymptomatic hamsters [13] and deer [14]. In summary, this method can be used to
amplify PK-resistant PrP derived from various species including cervid, ferret, hamster, mouse, sheep,
bovine, and human [12–17].
Although conventional PMCA utilizes normal brain homogenate as a source of PrP C, a novel
adaptation of the PMCA procedure is to amplify PrP res and generate highly infectious prions using
recombinant PrP derived from Escherichia coli [18] or baculovirus [19] in the absence of brain
homogenate. This novel procedure requires RNA and lipids, such as 1-palmitoyl-2-oleolyl-sn-
glycero-3-phospho(1′-rac-glycerol) (POPG) or phosphatidylethanolamine (PE), to act as conversion
cofactors [18,20,21].
Recently, the real-time quaking-induced conversion (RT-QuIC) test [22], which is a modified version
of PMCA, has been developed. This method is based on the prion-seeded fibrillization of recombi-
nant PrP. In RT-QuIC reactions, prion-associated seeds induce the amyloid fibril formation of bacte-
rially expressed recombinant PrP in multiwell plates. The resulting amyloid fibrils are then detected
by the enhanced fluorescence of an amyloid sensitive dye, thioflavin T, present in the reaction mix.
RT-QuIC is known to be highly specific and sensitive for the detection of multiple human and animal
prion diseases [23]. Epidemiological surveillance of CJD, which currently relies heavily on autopsy-
based diagnosis, could be more efficient, cost-effective, and broadly applicable with RT-QuIC testing for
cerebrospinal fluid (CSF) samples that can be obtained without autopsies [24]. The second-generation
RT-QuIC assay markedly improves the speed and sensitivity of detecting prion seeds in CSF specimens
from CJD patients [24].
In addition, the RT-QuIC assay has been used successfully to detect multiple human, bovine, cer-
vid, ovine hamster, and mouse strains in a variety of biological tissues such as saliva, blood, and nasal
fluids [25]. Indeed, numerous CJD diagnostic laboratories around the world are currently implement-
ing and validating RT-QuIC testing for human sCJD CSF using standard conditions [26]. RT-QuIC
enables an estimation of the quantity of CWD prion infectivity in tissues, such as the obex, left ventricle,
pancreas, jejunum, and spleen, as well as body fluids, such as saliva and urine [27]. Such an assay will
contribute to the risk assessment of deer tissues, biological fluids, excreta, or environmental samples
obtained from CWD-endemic areas.
To date, numerous cell lines have been established for prion infection (Table 7.1). The representa-
tive neuronal cell, rat pheochromocytoma PC12, can be differentiated by nerve growth factor (NGF)
and infected with prions [32], probably due to increased levels of PrP through NGF stimulation. Rabbit
kidney–derived RK13 cells overexpressed with ovine PrP are susceptible to sheep scrapie infection [33].
Furthermore, mouse fibroblast cells NIH/3T3 and L929 are also susceptible to mouse-adapted scrapie
infection [34]. In other cell types, microglial cells as well as epithelial cells and myoblasts can be suc-
cessfully infected with prions. Thus, under certain conditions, nonneuronal cells can also be infected
with prions. Indeed, this is consistent with the observation that PrPSc accumulates not only in the CNS
but also in other tissues such as the placenta, lymphoreticular system, and muscle tissue [35,36].
The use of the prion-susceptible cell line N2a is attractive because it is easily cultured. The N2a cell
line has been extensively used since 1970 to study prion infection [37], although the observed low postin-
fection titers and rapid attenuation in titers remain problematic [38,39]. The hypothalamic cell line GT1
displays higher PrPC expression levels and is more susceptible to prion infection than other cell lines
[40,41]. Although the human neuroblastoma cell line SH-SYS5 was reported to be infected with CJD pri-
ons, reproducibility of the result has yet to be confirmed by other laboratories [42]. Therefore, cell-culture
systems for CJD prion-infection have not been established. For BSE prion, only two cell lines, the mouse
microglial cell line MG20 overexpressing mouse PrP and the rabbit epithelial cell line RK13 overexpress-
ing bank vole PrP, can be infected with mouse-adapted BSE and bank vole-proliferated BSE, respectively.
TABLE 7.1
Representative Cell Lines Used for Prion Infection
Cell Lines Origins Prion Strains References
PC12 Rat pheochromocytoma 139A, ME7 [32,62,63]
C2C12 Mouse skeletal myoblast cell 22L [64]
CAD Mouse catecholaminergic cell RML, 22L, 22F, 79A, 139A, ME7 [44,65–67]
CF10 Mouse brain–derived PrP- 22L [68]
deficient cell
GT1 Mouse hippocampal neuron Chandler, RML, 139A, 22L, kCJD, FU [40,41,69–72]
CJD, M1000
HaB Hamster brain–derived cell Sc237 [73]
HpL3–4 Mouse hippocampal neuronal cell 22L, Chandler [74,75]
L. fibroblasts Mouse fibroblast ME7, Chandler [76]
L929 Mouse fibroblast 22L, ME7, RML [34]
MDB Deer immortalized brain cell CWD [77]
MG20 Mouse microglial cell Chandler, ME7, Obihiro, mouse-adapted [78]
BSE
MNB Mouse neuroblastoma Chandler [39]
MovS Mouse Schwann cell-like dorsal PG127, SSBP/1, scrapie field isolates [79,80]
root ganglia
MSC-80 Mouse Schwann cell Chandler [81]
N2a Mouse neuroblastoma Chandler, RML, 139A, 22L, C506, [38–40,71,82–86]
Fukuoka-1, FU CJD
NIH/3T3 Mouse fibroblast 22L [34]
RK13 Rabbit kidney epithelial cell Fukuoka-1, 22L, Chandler, M1000, [33,69,80,87–91]
mo sCJD, voBSE, PG127, LA404,
SSBP/1, scrapie field isolates, CWD
ScHB Hamster brain cell Scrapie (Chandler) [31]
SH-SY5Y Human neuroblastoma CJD [42]
SMB Mouse brain cell Chandler, 139A, 22F, 79A [31,92,93]
SN56 Mouse cholinergic septal cell Chandler, ME7, 22L [94]
BSE, bovine spongiform encephalopathy; CJD, Creutzfeldt–Jakob disease; kCJD, Kuru CJD; sCJD, Sporadic CJD; CWD,
chronic wasting disease; RML, Rocky Mountain Laboratory isolate.
122 Laboratory Models for Foodborne Infections
It should be noted that cell lines susceptible to some prion strains demonstrate a remarkable resistance
to other strains [43,44]. For example, mouse fibroblast 3T3 cells are only susceptible to prion strain 22L,
while mouse fibroblast L929 cells can be infected with prion strain 22L, RML, and ME7 [34,44]. In addi-
tion, although most of the cell lines and primary cells have been shown to express PrPC [45,46], only a lim-
ited number of these cells are susceptible to prion infection. Moreover, it is intriguing that both neuronal
and nonneuronal cell lines are susceptible to prion infection [47]. However, the reason for this difference
in susceptibility to prion infection among cell lines and prion strains remains unclear. Such differences
may be related to the cellular properties of the receptor expression or possibly cofactors contributing to
PrPSc internalization, such as laminin receptor precursor (LRP/LR) [48], low-density lipoprotein receptor-
related protein 1 (LPR1) [49], proteoglycans, and glycosaminoglycans (GAGS) [50]. Indeed, some cells
infected with prions secrete exosomes associated with the prion agent, suggesting that exosome release
from cells into the cell-culture medium might be related to susceptibility to infection [51,52].
Intracerebral inoculation
Intraperitoneal inoculation
FIGURE 7.4 Animal infection models of prion diseases. Animal infection models of prion diseases generally involve
intracerebral or intraperitoneal inoculation. In both cases, the survival time and incubation period are measured as an
index of prion infection and infectious titer. Both the survival time and incubation period after intracerebral infection are
shorter than after intraperitoneal infection. The survival curve of the mice is then examined by statistical analysis (e.g.,
the log-rank test).
Prions 123
prion strain combination. Nonetheless, markedly different incubation times are observed when different
prion strains are tested against a single inbred mouse strain [56]. Likewise, strikingly different incubation
times are observed when a single prion strain is tested against different mouse strains, especially those
having different genotypes of the PrP gene (Prnp). There are two alleles of mouse Prnp, namely Prnpa and
Prnpb, encoding PrP that differ by two amino acid residues at codons 108 and 189 [57]. Mice with differ-
ent PrP genotypes may have an incubation period that differs by hundreds of days after infection with the
same prion strain. Subtle changes in the behavior of infected mice are often seen for a few weeks before
the onset of definite neurological signs during the last 2–3 weeks of the incubation period.
Behavioral changes are closely related to neuropathology. Images of histological sections of the brains
of infected animals are dramatically different among prion strains [58]. The areas of vacuolation and
PrP deposits depend on the prion strain, but also to some extent on PrP and other genetic factors. The
so-called “lesion profile” is an objective analysis that involves scoring the area of vacuolation and/or PrP
deposits in nine gray matter and three white matter brain areas. Interestingly, each combination of prion
strain and mouse strain gives a characteristic lesion profile [56,59,60].
Due to the extended incubation period, animal bioassays take a very long time to complete (e.g., up to
2.5 years for primary transmission in mice, then at least 6 months for each serial mouse passage). In addi-
tion, a special biosafety level facility is required, which makes the whole procedure very costly. Therefore,
an appropriate sample size (number of animals, type of animals) for each assay should be carefully con-
sidered. Given the difficulties and expense of performing such experiments, researchers should consider
whether an alternative approach, such as in vitro testing or cell-culture assays, can be used.
7.5 Conclusions
Studies of prion biology have until recently used conventional laboratory models for prion infection,
cell-culture assays, and bioassays. The development of novel tube-based methods involving PrPres
amplification, such as PMCA and RT-QuIC, has revolutionized the analysis of prion diseases [61].
In p articular, PMCA and RT-QuIC have dramatically improved the diagnosis of prion diseases by
facilitating the sensitive detection of prions. Furthermore, RT-QuIC provides quantitative data on PrP res,
while PMCA enables amplification of PrPSc in vitro. Novel future applications of these methods in prion
biology are anticipated.
Studies using cell lines suggest that PrPC is not the sole determinant for prion susceptibility. Indeed,
other cellular factors may be required for efficient infection. Under certain conditions, loss of host factors
for susceptibility to prion infection may occur. Furthermore, most cell lines have restricted prion-strain
specificity, that is, matching pairs between the host cell and prion strain. Similarly, constituents of the
buffer used for PMCA vary depending on the particular prion strain under investigation. Research into
anti-prion drugs using cell lines and PMCA systems must take these factors into consideration, because
some of the anti-prion activity might be attributed to a special property of the prion strain or cell line. For
example, an anti-prion activity may be caused by an indirect effect on cellular metabolism. Thus, studies
using different strains and cell lines are necessary in order to identify an anti-prion compound effec-
tive against a broad range of prion strains. Unfortunately, animal bioassays are often time consuming,
laborious, and expensive to perform. Nonetheless, animal bioassays are still valuable in determining the
biological activity of prions and remain the gold standard laboratory model for prion infections.
Acknowledgments
This work was supported by Grant-in-Aids for Scientific Research from the Ministry of Education,
Science, Culture and Technology of Japan (25450447, 24110717, 23780299, 22110514, 20780219,
17780228), Grants-in-Aid from the Research Committee of Prion Disease and Slow Virus Infection,
the Ministry of Health, Labour and Welfare of Japan, and Japan Agency for Medical Research and
Development (AMED).
124 Laboratory Models for Foodborne Infections
REFERENCES
1. Prusiner, S.B. Prions. Proc Natl Acad Sci USA 95, 13363–83 (1998).
2. Aguzzi, A. & Polymenidou, M. Mammalian prion biology: one century of evolving concepts. Cell 116,
313–27 (2004).
3. Prusiner, S.B. et al. Scrapie agent contains a hydrophobic protein. Proc Natl Acad Sci USA 78, 6675–9
(1981).
4. Sakudo, A., Nakamura, I., Ikuta, K. & Onodera, T. Recent developments in prion disease research:
diagnostic tools and in vitro cell culture models. J Vet Med Sci 69, 329–37 (2007).
5. Thuring, C.M. et al. Discrimination between scrapie and bovine spongiform encephalopathy in sheep
by molecular size, immunoreactivity, and glycoprofile of prion protein. J Clin Microbiol 42, 972–80
(2004).
6. Imran, M. & Mahmood, S. An overview of animal prion diseases. Virol J 8, 493 (2011).
7. Sakudo, A. & Onodera, T. Chapter 98: Bovine spongiform encephalopathy (BSE), In Molecular
Detection of Animal Viral Pathogens (Liu, D., ed.). Taylor & Francis, CRC Press, Boca Raton, FL,
pp. 901–912 (2016).
8. Sakudo, A. & Onodera, T. Chapter 99: Chronic wasting disease (CWD), In Molecular Detection of
Animal Viral Pathogens (Liu, D., ed.). Taylor & Francis, CRC Press, Boca Raton, FL, pp. 913–921 (2016).
9. Saborio, G.P., Permanne, B. & Soto, C. Sensitive detection of pathological prion protein by cyclic ampli-
fication of protein misfolding. Nature 411, 810–3 (2001).
10. Castilla, J., Saá, P. & Soto, C. Cyclic amplification of prion protein misfolding, In Techniques in Prion
Research (Methods and Tools in Biosciences and Medicine) (Lehmann, S. & Grassi, J., eds.). Birkhäuser
Verlag, Basel, Switzerland, pp. 198–213, 2005.
11. Castilla, J., Saa, P., Hetz, C. & Soto, C. In vitro generation of infectious scrapie prions. Cell 121,
195–206 (2005).
12. Thorne, L. & Terry, L.A. In vitro amplification of PrPSc derived from the brain and blood of sheep
infected with scrapie. J Gen Virol 89, 3177–84 (2008).
13. Saa, P., Castilla, J. & Soto, C. Presymptomatic detection of prions in blood. Science 313, 92–4 (2006).
14. Rubenstein, R. et al. Prion disease detection, PMCA kinetics, and IgG in urine from sheep naturally/
experimentally infected with scrapie and deer with preclinical/clinical chronic wasting disease. J Virol
85, 9031–8 (2011).
15. Kurt, T.D., Telling, G.C., Zabel, M.D. & Hoover, E.A. Trans-species amplification of PrPCWD and cor-
relation with rigid loop 170N. Virology 387, 235–43 (2009).
16. Murayama, Y. et al. Ultrasensitive detection of PrPSc in the cerebrospinal fluid and blood of macaques
infected with bovine spongiform encephalopathy prion. J Gen Virol 95, 2576–88 (2014).
17. Murayama, Y. et al. Efficient in vitro amplification of a mouse-adapted scrapie prion protein. Neurosci
Lett 413, 270–3 (2007).
18. Wang, F., Wang, X., Yuan, C.G. & Ma, J. Generating a prion with bacterially expressed recombinant
prion protein. Science 327, 1132–5 (2010).
19. Faburay, B., Tark, D., Kanthasamy, A.G. & Richt, J.A. In vitro amplification of scrapie and chronic wast-
ing disease PrPres using baculovirus-expressed recombinant PrP as substrate. Prion 8, 393–403 (2014).
20. Deleault, N.R. et al. Isolation of phosphatidylethanolamine as a solitary cofactor for prion formation in
the absence of nucleic acids. Proc Natl Acad Sci USA 109, 8546–51 (2012).
21. Deleault, N.R. et al. Cofactor molecules maintain infectious conformation and restrict strain properties
in purified prions. Proc Natl Acad Sci USA 109, E1938–46 (2012).
22. Atarashi, R., Sano, K., Satoh, K. & Nishida, N. Real-time quaking-induced conversion: a highly sensi-
tive assay for prion detection. Prion 5, 150–3 (2011).
23. Orru, C.D. et al. Detection and discrimination of classical and atypical L-type bovine spongiform
encephalopathy by real-time quaking-induced conversion. J Clin Microbiol 53, 1115–20 (2015).
24. Orru, C.D. et al. Rapid and sensitive RT-QuIC detection of human Creutzfeldt–Jakob disease using
cerebrospinal fluid. mBio 6, e02451-14 (2015).
25. Orru, C.D., Wilham, J.M., Vascellari, S., Hughson, A.G. & Caughey, B. New generation QuIC assays for
prion seeding activity. Prion 6, 147–52 (2012).
26. McGuire, L.I. et al. Real time quaking-induced conversion analysis of cerebrospinal fluid in sporadic
Creutzfeldt–Jakob disease. Ann Neurol 72, 278–85 (2012).
Prions 125
27. Henderson, D.M. et al. Quantitative assessment of prion infectivity in tissues and body fluids by real-
time quaking-induced conversion. J Gen Virol 96, 210–9 (2015).
28. Grassmann, A., Wolf, H., Hofmann, J., Graham, J. & Vorberg, I. Cellular aspects of prion replication in
vitro. Viruses 5, 374–405 (2013).
29. Beranger, F., Mange, A., Solassol, J. & Lehmann, S. Cell culture models of transmissible spongiform
encephalopathies. Biochem Biophys Res Commun 289, 311–6 (2001).
30. Lehmann, S., Solassol, J. & Perrier, V. Cell culture models of TSEs, In Techniques in Prion Research
(Methods and Tools in Biosciences and Medicine) (Lehmann, S. & Grassi, J., eds.). Birkhäuser Verlag,
Basel, Switzerland, pp. 72–81, 2005.
31. Clarke, M.C. & Haig, D.A. Evidence for the multiplication of scrapie agent in cell culture. Nature 225,
100–1 (1970).
32. Rubenstein, R., Carp, R.I. & Callahan, S.M. In vitro replication of scrapie agent in a neuronal model:
infection of PC12 cells. J Gen Virol 65 (Pt 12), 2191–8 (1984).
33. Vilette, D. et al. Ex vivo propagation of infectious sheep scrapie agent in heterologous epithelial cells
expressing ovine prion protein. Proc Natl Acad Sci USA 98, 4055–9 (2001).
34. Vorberg, I., Raines, A., Story, B. & Priola, S.A. Susceptibility of common fibroblast cell lines to trans-
missible spongiform encephalopathy agents. J Infect Dis 189, 431–9 (2004).
35. Thomzig, A., Kratzel, C., Lenz, G., Kruger, D. & Beekes, M. Widespread PrPSc accumulation in muscles
of hamsters orally infected with scrapie. EMBO Rep 4, 530–3 (2003).
36. Bosque, P.J. et al. Prions in skeletal muscle. Proc Natl Acad Sci USA 99, 3812–7 (2002).
37. Clarke, M.C. & Haig, D.A. Multiplication of scrapie agent in cell culture. Res Vet Sci 11, 500–1 (1970).
38. Butler, D.A. et al. Scrapie-infected murine neuroblastoma cells produce protease-resistant prion pro-
teins. J Virol 62, 1558–64 (1988).
39. Race, R.E., Fadness, L.H. & Chesebro, B. Characterization of scrapie infection in mouse neuroblastoma
cells. J Gen Virol 68 (Pt 5), 1391–9 (1987).
40. Nishida, N. et al. Successful transmission of three mouse-adapted scrapie strains to murine neuroblas-
toma cell lines overexpressing wild-type mouse prion protein. J Virol 74, 320–5 (2000).
41. Schatzl, H.M. et al. A hypothalamic neuronal cell line persistently infected with scrapie prions exhibits
apoptosis. J Virol 71, 8821–31 (1997).
42. Ladogana, A., Liu, Q., Xi, Y.G. & Pocchiari, M. Proteinase-resistant protein in human neuroblastoma
cells infected with brain material from Creutzfeldt–Jakob patient. Lancet 345, 594–5 (1995).
43. Kleer, C.G. et al. EZH2 is a marker of aggressive breast cancer and promotes neoplastic transformation
of breast epithelial cells. Proc Natl Acad Sci USA 100, 11606–11 (2003).
44. Mahal, S.P. et al. Prion strain discrimination in cell culture: the cell panel assay. Proc Natl Acad Sci
USA 104, 20908–13 (2007).
45. Sakudo, A. & Onodera, T. Prion protein (PrP) gene-knockout cell lines: insight into functions of the PrP.
Front Cell Dev Biol 2, 75 (2015).
46. Sakudo, A. et al. Recent advances in clarifying prion protein functions using knockout mice and derived
cell lines. Mini Rev Med Chem 6, 589–601 (2006).
47. Solassol, J., Crozet, C. & Lehmann, S. Prion propagation in cultured cells. Br Med Bull 66, 87–97 (2003).
48. Gauczynski, S. et al. The 37-kDa/67-kDa laminin receptor acts as the cell-surface receptor for the cel-
lular prion protein. EMBO J 20, 5863–75 (2001).
49. Taylor, D.R. & Hooper, N.M. The low-density lipoprotein receptor-related protein 1 (LRP1) mediates
the endocytosis of the cellular prion protein. Biochem J 402, 17–23 (2007).
50. Holmes, B.B. et al. Heparan sulfate proteoglycans mediate internalization and propagation of specific
proteopathic seeds. Proc Natl Acad Sci USA 110, E3138–47 (2013).
51. Alais, S. et al. Mouse neuroblastoma cells release prion infectivity associated with exosomal vesicles.
Biol Cell 100, 603–15 (2008).
52. Fevrier, B. et al. Cells release prions in association with exosomes. Proc Natl Acad Sci USA 101, 9683–8
(2004).
53. Bruce, M.E., Boyle, A. & McConnell, I. TSE strain typing in mice, In Techniques in Prion Research
(Methods and Tools in Biosciences and Medicine) (Lehmann, S. & Grassi, J., eds.). Birkhäuser Verlag,
Basel, Switzerland, pp. 132–146, 2005.
54. Inoue, Y. et al. Infection route-independent accumulation of splenic abnormal prion protein. Jpn J Infect
Dis 58, 78–82 (2005).
126 Laboratory Models for Foodborne Infections
55. Fichet, G. et al. Novel methods for disinfection of prion-contaminated medical devices. Lancet 364,
521–6 (2004).
56. Bruce, M.E., McConnell, I., Fraser, H. & Dickinson, A.G. The disease characteristics of different strains
of scrapie in Sinc congenic mouse lines: implications for the nature of the agent and host control of
pathogenesis. J Gen Virol 72 (Pt 3), 595–603 (1991).
57. Westaway, D. et al. Distinct prion proteins in short and long scrapie incubation period mice. Cell 51,
651–62 (1987).
58. Fraser, H. Diversity in the neuropathology of scrapie-like diseases in animals. Br Med Bull 49, 792–809
(1993).
59. Fraser, H. & Dickinson, A.G. The sequential development of the brain lesion of scrapie in three strains
of mice. J Comp Pathol 78, 301–11 (1968).
60. Bruce, M.E., McBride, P.A. & Farquhar, C.F. Precise targeting of the pathology of the sialoglycoprotein,
PrP, and vacuolar degeneration in mouse scrapie. Neurosci Lett 102, 1–6 (1989).
61. Saa, P. & Cervenakova, L. Protein misfolding cyclic amplification (PMCA): current status and future
directions. Virus Res 207, 47–61 (2015).
62. Rubenstein, R. et al. Demonstration of scrapie strain diversity in infected PC12 cells. J Gen Virol
73 (11), 3027–31 (1992).
63. Rubenstein, R., Deng, H., Scalici, C.L. & Papini, M.C. Alterations in neurotransmitter-related enzyme
activity in scrapie-infected PC12 cells. J Gen Virol 72 (Pt 6), 1279–85 (1991).
64. Dlakic, W.M., Grigg, E. & Bessen, R.A. Prion infection of muscle cells in vitro. J Virol 81, 4615–24
(2007).
65. Dron, M., Dandoy-Dron, F., Farooq Salamat, M.K. & Laude, H. Proteasome inhibitors promote the
sequestration of PrPSc into aggresomes within the cytosol of prion-infected CAD neuronal cells. J Gen
Virol 90, 2050–60 (2009).
66. Browning, S. et al. Abrogation of complex glycosylation by swainsonine results in strain- and cell-
specific inhibition of prion replication. J Biol Chem 286, 40962–73 (2011).
67. Julius, C. et al. Transcriptional stability of cultured cells upon prion infection. J Mol Biol 375, 1222–33
(2008).
68. McNally, K.L., Ward, A.E. & Priola, S.A. Cells expressing anchorless prion protein are resistant to
scrapie infection. J Virol 83, 4469–75 (2009).
69. Vella, L.J. et al. Packaging of prions into exosomes is associated with a novel pathway of PrP processing.
J Pathol 211, 582–90 (2007).
70. Lewis, V. et al. Increased proportions of C1 truncated prion protein protect against cellular M1000 prion
infection. J Neuropathol Exp Neurol 68, 1125–35 (2009).
71. Arjona, A., Simarro, L., Islinger, F., Nishida, N. & Manuelidis, L. Two Creutzfeldt–Jakob disease agents
reproduce prion protein-independent identities in cell cultures. Proc Natl Acad Sci USA 101, 8768–73
(2004).
72. Miyazawa, K., Emmerling, K. & Manuelidis, L. High CJD infectivity remains after prion protein is
destroyed. J Cell Biochem 112, 3630–7 (2011).
73. Taraboulos, A., Serban, D. & Prusiner, S.B. Scrapie prion proteins accumulate in the cytoplasm of per-
sistently infected cultured cells. J Cell Biol 110, 2117–32 (1990).
74. Maas, E. et al. Scrapie infection of prion protein-deficient cell line upon ectopic expression of mutant
prion proteins. J Biol Chem 282, 18702–10 (2007).
75. Sakudo, A., Wu, G., Onodera, T. & Ikuta, K. Octapeptide repeat region of prion protein (PrP) is required
at an early stage for production of abnormal prion protein in PrP-deficient neuronal cell line. Biochem
Biophys Res Commun 365, 164–9 (2008).
76. Clarke, M.C. & Millson, G.C. Infection of a cell line of mouse L fibroblasts with scrapie agent. Nature
261, 144–5 (1976).
77. Raymond, G.J. et al. Inhibition of protease-resistant prion protein formation in a transformed deer cell
line infected with chronic wasting disease. J Virol 80, 596–604 (2006).
78. Iwamaru, Y. et al. Microglial cell line established from prion protein-overexpressing mice is susceptible
to various murine prion strains. J Virol 81, 1524–7 (2007).
79. Archer, F. et al. Cultured peripheral neuroglial cells are highly permissive to sheep prion infection.
J Virol 78, 482–90 (2004).
Prions 127
80. Neale, M.H. et al. Infection of cell lines with experimental and natural ovine scrapie agents. J Virol 84,
2444–52 (2010).
81. Follet, J. et al. PrP expression and replication by Schwann cells: implications in prion spreading. J Virol
76, 2434–9 (2002).
82. Enari, M., Flechsig, E. & Weissmann, C. Scrapie prion protein accumulation by scrapie-infected neuro-
blastoma cells abrogated by exposure to a prion protein antibody. Proc Natl Acad Sci USA 98, 9295–9
(2001).
83. Markovits, P., Dautheville, C., Dormont, D., Dianoux, L. & Latarjet, R. In vitro propagation of the
scrapie agent. I. Transformation of mouse glia and neuroblastoma cells after infection with the mouse-
adapted scrapie strain c-506. Acta Neuropathol 60, 75–80 (1983).
84. Ostlund, P., Lindegren, H., Pettersson, C. & Bedecs, K. Up-regulation of functionally impaired insulin-
like growth factor-1 receptor in scrapie-infected neuroblastoma cells. J Biol Chem 276, 36110–5 (2001).
85. Race, R. The scrapie agent in vitro. Curr Top Microbiol Immunol 172, 181–93 (1991).
86. Scott, M.R., Kohler, R., Foster, D. & Prusiner, S.B. Chimeric prion protein expression in cultured cells
and transgenic mice. Protein Sci 1, 986–97 (1992).
87. Courageot, M.P. et al. A cell line infectible by prion strains from different species. J Gen Virol 89,
341–7 (2008).
88. Lawson, V.A. et al. Mouse-adapted sporadic human Creutzfeldt–Jakob disease prions propagate in cell
culture. Int J Biochem Cell Biol 40, 2793–801 (2008).
89. Sabuncu, E. et al. PrP polymorphisms tightly control sheep prion replication in cultured cells. J Virol 77,
2696–700 (2003).
90. Bian, J. et al. Cell-based quantification of chronic wasting disease prions. J Virol 84, 8322–6 (2010).
91. Kim, H.J. et al. Establishment of a cell line persistently infected with chronic wasting disease prions.
J Vet Med Sci 74, 1377–80 (2012).
92. Birkett, C.R. et al. Scrapie strains maintain biological phenotypes on propagation in a cell line in cul-
ture. EMBO J 20, 3351–8 (2001).
93. Kanu, N. et al. Transfer of scrapie prion infectivity by cell contact in culture. Curr Biol 12, 523–30 (2002).
94. Baron, T.G., Biacabe, A.G., Bencsik, A. & Langeveld, J.P. Transmission of new bovine prion to mice.
Emerg Infect Dis 12, 1125–8 (2006).
Section II
CONTENTS
8.1 Introduction....................................................................................................................................131
8.2 Virulence Factors: An Overview...................................................................................................133
8.3 In Vitro Models............................................................................................................................. 134
8.3.1 Cultured Eukaryotic Cells................................................................................................ 134
8.3.2 Regulation of Toxin Production....................................................................................... 138
8.3.3 Direct Interactions with Eukaryotic Cells: Other Virulence Factors............................... 139
8.3.4 Virulence Factors in Other Bacillus spp.......................................................................... 140
8.4 Ex Vivo and In Vivo Models.......................................................................................................... 140
8.4.1 Ex Vivo Models..................................................................................................................141
8.4.2 In Vivo Models...................................................................................................................141
8.5 Invertebrates...................................................................................................................................141
8.5.1 Nematodes.........................................................................................................................141
8.5.2 Insects................................................................................................................................142
8.5.2.1 Galleria mellonella.............................................................................................142
8.5.2.2 Bombyx mori.....................................................................................................143
8.6 Vertebrates.....................................................................................................................................143
8.6.1 Rabbit.................................................................................................................................143
8.6.2 Zebrafish........................................................................................................................... 144
8.6.3 Monkeys........................................................................................................................... 144
8.6.4 Asian House Shrew.......................................................................................................... 144
8.6.5 Mice.................................................................................................................................. 144
8.6.6 Rats................................................................................................................................... 146
8.7 Concluding Remarks..................................................................................................................... 146
Acknowledgments....................................................................................................................................147
References................................................................................................................................................147
8.1 Introduction
The ability of prokaryotes to sporulate is one of the evolutionary strategies to cope with stress. Spore-
forming microorganisms invest considerable amounts of energy in a refined differentiation process
(sporogenesis) that involves the expression of hundreds of genes in order to give a morphologically
distinct cell that presents several barriers related to the resistance to physical, chemical, and biological
challenges.1
Among spore-forming microorganisms, those belonging to the genus Bacillus play an important role
in several aspects of human activities. Indeed, the ability of fast growth and the diversity of metabolic
capabilities along with the capacity to survive in adverse conditions lead to a versatile genus that actively
131
132 Laboratory Models for Foodborne Infections
participates in shaping the environment. However, some members of the genus Bacillus have evolved
pathogenic potential and are thus associated with distinct pathological processes.
The genus Bacillus includes spore-forming Gram-positive, low G + C content, rod-shaped bacteria.
Members of the genus are ubiquitous and capable of surviving and growing in very different, even
extreme, conditions such as high temperatures, high saline concentrations, and low pH.2
The early works of Pollender, Brauell, Delafond, and Davaine on anthrax or carbuncle prompted
Robert Koch to enunciate the paramount postulates of the germ theory of disease and Louis Pasteur
to establish effective immunization for anthrax prevention in the seminal experiment of massive live-
stock vaccination at Pouilly-le-Fort.3–5 It is known that some Bacillus species constitute a very definite
cluster sharing many characteristics. This so-called “cereus group” includes microorganisms that can
be identified as Bacillus cereus (B. cereus sensu lato) on the basis of conventional microbiological pro-
cedures, but the use of more refined techniques lead to a more precise identification. The B. cereus
group encompasses seven species, that is, Bacillus anthracis—the etiological agent of anthrax, B. cereus
(sensu stricto), Bacillus thuringiensis, Bacillus mycoides, Bacillus pseudomycoides, Bacillus weihen-
stephanensis, and Bacillus cytotoxicus.6–8 The division between species of the B. cereus group is based
on phenotypic characteristics related to differences in ecology and pathogenesis. However, adoption of
molecular approaches strongly suggests that members of the B. cereus group belong to one single spe-
cies.9–12 In this chapter, we will include bacterial species stated in the original publications irrespective
of the exact taxonomic nomenclature.
It has been demonstrated that the B. cereus group diversified along evolution. This evolutionary path-
way includes thermotolerant lineages as the older microorganisms evolved into mesophilic members,
with a more recent shift to psychrotrophic groups.13 Interestingly, the food-poisoning risk is not neces-
sarily related to species but to the phylogenetic group.7,14
Although the exact cause of many historical epidemics may never be known, there are relevant refer-
ences compatible with epidemic anthrax. Even though no diagnostic methods were available, descriptions
from historians and physicians revealed an illness; their symptoms probably implicated anthrax, that is,
the fifth and sixth plagues of Egypt described in the Book of Exodus, the Plague of Athens (430–427
BCE), and the epizootics described in Virgil’s Aeneid (70–19 BCE).15 However, there are other reports
that postulated that anthrax is unlikely the basis of the Plague of Athens16–18 or other biblical plagues.19
Beyond controversial historical and epidemiological aspects, it is evident that B. anthracis is an impor-
tant zoonotic pathogen and that several outbreaks are well documented. In the 18th century, there was
an outbreak of intestinal anthrax due to consumption of smoked or salted meat from ill animals after
an earthquake occurred in Saint Domingue (modern Haiti). In this episode, 15,000 people were killed.3
Human gastrointestinal anthrax outbreaks are described in different studies, and they are often associ-
ated with consumption of foods originating from diseased animals.20–24 However, unusual sources of
contamination such as animal-hide drums have been reported.24 Intestinal anthrax constitutes the main
presentation for livestock that normally ingests spores in foods.25,26
The B. cereus group represents an example of how natural plasmid transfer within a genus leads to
different microorganisms that are allocated to different species.
Indeed, in the same chromosomal genetic background, the presence of PXO1 and PXO2 character-
izes B. anthracis, a microorganism with high pathogenic potential, and the presence of large transfer-
able plasmids in B. thuringiensis enables its production of insecticidal proteins.27 It is worth noting that
B. cereus harboring B. anthracis toxin genes leads to life-threatening pathology.28–30 Because of these
particularities, members of the group other than B. anthracis were not clearly differentiated until the
late 1960s. Indeed, aerobic endospore-forming rods were not identified at all or assigned to the Bacillus
subtilis species.31 Earlier reports that univocally identified B. cereus as the causative agent of foodborne
diseases were published between 1950 and 1955.31
Foods are suitable culture media for Bacillus spp., and thus, growth in high numbers can occur in
many steps of food preparation and preservation. In addition, some strains are psychrotrophic.32 To
a lesser extent, other Bacillus species have been associated with gastrointestinal illness, for example,
B. subtilis, Bacillus pumilus, Bacillus licheniformis, Bacillus brevis, and Bacillus mojavensis, although
these microorganisms are more often involved in food spoilage.31,33–35
Bacillus 133
Other virulence factors have been described in different species of the Bacillus genus. Interestingly,
B. subtilis and B. mojavensis involved in food poisoning have been demonstrated to produce amylosin,
a heat-stable ion channel forming toxin, which also reported for B. amyloliquefaciens.54,55 Some strains
are able to produce this toxin during growth at refrigeration temperatures.33 In addition, different factors
with biological activity on mammalian cells have been associated to B. licheniformis (lichenysin)56 and
B. pumilus (pumilacidin).57
In the next sections, models that contributed to the understanding of the pathogenesis of Bacillus spp.
will be discussed in the context of the biological activities they are able to detect.
TABLE 8.1
In Vitro, In Vivo, and Ex Vivo Models Used for the Study of the Biological Activity of Bacillus spp.
Model
In vitro Epithelial Caco-2 (human colorectal HT1080 (human Vero (kidney epithelial cells from
cells adenocarcinoma) fibrosarcoma cells) an African green monkey)
CHO (Chinese hamster Hep-2 (human laryngeal Hep-G2 (human hepatocellular
ovary) carcinoma) carcinoma)
A549 (human lung HeLa (cervical cancer from Calu-3 (human lung
carcinoma) human) adenocarcinoma)
Paju (human neural Ptk6 null colonic epithelial A204 (human
crest-derived cell line) cells rhabdomyosarcoma)
McCoy (murine Huvet (human umbilical
fibroblasts) vein endothelial cells)
Immune Raw264.7 (monocytes J774 (monocyte/ Neutrophils (hPMNs)
cells from murine origin) macrophage from murine
origin)
Jurkat (human T Ped-2E9 (lymphocyte from U937 (monocytes from human
lymphoblastoid cell line) murine origin) origin)
Ex vivo Spermatozoon test
In vivo Invertebrates Caenorhabditis elegans
Galleria mellonella
Bombyx mori
Vertebrates Ligated rabbit or mouse ileal loop
Vascular permeability reaction
Zebrafish
Suncus murinus
Mice
Rats
Monkeys
This simple experimental approach allowed for the differentiation of the biological activity of B. cereus
strains, even though this activity is related to several extracellular factors present in spent culture super-
natants. In addition, a multivariate analysis demonstrated that high detaching strains were positive for
the sequences of the virulence genes entS, nheC, and sph.51
Cytotoxicity is strain-dependent51,58,68 with species-to-species variations.63 Since nutritional factors
influence the expression of virulence factors, the culture medium modifies the values of cytotoxicity, cell
adhesion, and invasion.63
Fractioning and purification of factors present in the culture supernatants allowed for the identifica-
tion and further study of several toxins of B. cereus. Hemolysin BL (HBL) and nonhemolytic enterotoxin
(NHE) are consistently described as key virulence factors. They are tripartite protein toxins expressed
in different proportions by members of the B. cereus group.61 HBL components are HBLB (37.5 kDa),
HBLL1 (38.2 kDa), and HBLL2 (45 kDa),81 and NHE components are NHA (41 kDa), NHB (39.8 kDa),
and NHC (36.5 kDa).67,82 HBL is expressed in 42%–73% of food-poisoning-associated strains, whereas
NHE is expressed in 97%–99% of strains.61,83,84 Both toxins are responsible for the cytotoxic effect on
several cell lines such as Vero, Hep-2, Hep-G2, Caco-2, Jurkat, RPMI 8226, A204, A549, HUVET, and
U937 cells.62 For NHE, the specificity of the observed biological effects was demonstrated in neutraliza-
tion experiments with monoclonal antibodies.85
The presence of the three components for maximum cytotoxic effect was necessary for the activity
of both HBL and NHE.60,66,81,82 Furthermore, the relative concentrations of the components are relevant
136 Laboratory Models for Foodborne Infections
FIGURE 8.1 Disorganization of the F-actin network following incubation of cultured human enterocytes (Caco-2 cells)
with spent culture supernatants of B. cereus. References: control cells (top), strain 2 (middle), and strain M2 (bottom).
Bacillus 137
for the observed effects (e.g., inhibition of the protein synthesis of Vero cells by NHE44,82). NHEB and
HBLL2 are crucial for the cytotoxic effect since when these components were not present significantly,
lower cytotoxic effects were observed in some cell lines.62
Studies on the structures involved in the binding of toxins to target cells by using CHO and red blood
cells demonstrated that for HBL, the binding component is B (Hbl-B) and L1 and L2 bind to Hbl-B.61,86
On the other hand, the NHE component responsible for the binding to eukaryotic cells is Nhe-C.61,87
Then, there is a sequential binding of Nhe-B and Nhe-A.87 It is worth noting that despite its name (non-
hemolytic enterotoxin), the NHE toxin also has hemolytic activity.88
Even though several extracellular factors have been described in the B. cereus group, NHE was pro-
posed as the main virulence factor84,89 showing the highest cytotoxic effect on Vero (fibroblasts) and
HUVEC (endothelial) cells.62 Albeit to a lesser extent, NHE is also active on U937 cells62 and is respon-
sible for the impairment of the plasma membrane of Caco-2 cells.72 Vero cells treated with spent culture
supernatants showed blebs, which subsequently burst, and these effects were not observed when super-
natants from nheBC mutants were used.72 By using a nheBC mutant strain and Ptk6 cells, it has been
demonstrated that NHE and sphingomyelinase have a concerted effect.71
In relation to the hemolytic action of toxins, B. cereus isolates with the ability to produce trimeric
toxin HBL lead to a characteristic ring-shaped zone of hemolysis in blood agar plates.86,90,91 Hemolysis
depends on the source of erythrocytes: Sheep or calf erythrocytes are the most susceptible, and human
or horse erythrocytes are the least susceptible.86
There is a high similarity in the structure and sequence between HBL and NHE toxins,72 but it is not
possible to complement or substitute components to give an active toxin.61 So far, little is known about
the mechanisms involved in the biological effects of these toxins. Studies performed with osmotic pro-
tectants such as carbohydrates or polyethylene glycol of increasing size allowed researchers to determine
that the mechanism involved in HBL and NHE lysis cells is colloid-osmotic72,92 leading to pore forma-
tion in planar lipid bilayers.72 Spent culture supernatants lead to the release of intracellular ATP and
LDH from Caco-2 and Vero cells; these effects were not detected when supernatants from cultures of
nheBC mutants were employed.72 Because of structural similarities between NHE and HBL components
with hemolysin E from E. coli, it is proposed that the biological effects of both toxins share common
mechanisms.61,93,94
Cytotoxin K (CytK) is another relevant extracellular factor of B. cereus associated with necrotic
enteritis. It is a single-component protein toxin of 34 kDa that is hemolytic to rabbit red blood cells (it
is a β-barrel pore-forming toxin) and cytotoxic to several cells lines.8,62,70 However, CytK has lower
biological activity as compared with other virulence factors such as NHE and HBL.62 Caco-2 and
Vero cells have been seen to be particularly susceptible to CytK.62 Toxicity to Caco-2 cells is related
to the ability to form nonselective pores in the plasma membrane.8,70 This effect is similar to that of C.
perfringens C toxin, although this toxin is cytotoxic and noncytolytic due to the formation of cation
selective pores.70
CytK, also known as hemolysin IV,95 has two variants: CytK1 (firstly reported by Lund et al.8) and
CytK2 which shares 89% identity with CytK1 and has a similar size.96 CytK2 is hemolytic to bovine
erythrocytes and cytotoxic to Caco-2 although to a lesser extent than the original variant, and it did not
show toxic effects on Vero-2 cells.96 Both variants lead to different distributions of pore sizes on lipid
bilayers.96
HlyII is another single-component protein toxin (42.3 kDa), which is a member of the family of
β-barrel pore-forming toxins.8,97–99 HlyII leads to a decrease of the mitochondrial membrane potential,
and it is lytic to Paju and Caco-2 cells.99 Besides, it has hemolytic activity on erythrocytes of different
species, with the highest effects on human and rabbit red blood cells.99 However, participation of HlyII
in diarrhea caused by B. cereus has not been demonstrated.95
The apoptosis of macrophages (J774 cell line) was induced by purified HlyII, and the involvement
of this factor in the observed effect was supported by studies with mutants with deletions in the hlyII
gene.47 This effect is related to the ability of HlyII to form pores because macrophages incubated with
purified HlyII in the presence of polyethylene glycol abolished apoptosis and preserved the perme-
ability of the cell membrane. There is also a contribution of caspase activation on the observed pro-
apoptotic effect.47
138 Laboratory Models for Foodborne Infections
Members of the B. cereus group are able to produce other pore-forming toxins such as cereolysin O,
hemolysin III, and three phospholipases C.95,100–102 Cereolysin O (or Hemolysin I), a heat-labile protein
toxin, is a member of the cholesterol-dependent cytolysin family; its best known members are listerio-
lysin O and perfringolysin O. They are able to form large pores in the cell membrane.103 Concerning
hemolysin III, it was first reported by Baida and Kuzmin in 1995.100 This is a heat-labile factor that is
not inhibited by cholesterol, but no further reports were found. Sphingomyelinase, phosphatidylcholine–
phospholipase C, and phosphatidylinositol–phospholipase C (PI-PLC) have cytolytic activity.42,46,104–107
Interestingly, PI-PLC from B. anthracis inhibits the activation of murine dendritic cells.108
The B. cereus emetic toxin is cereulide, a heat-stable dodecadepsipeptide preformed in food.109,110 It
is a toxin that causes mitochondria dysfunction due to its activity on K+ -channels.111,112 Since cereulide
is structurally closely related to valinomycin, several studies used this antibiotic as a surrogate mol-
ecule.110,113,114 However, the emetic toxin showed higher affinity for potassium than valinomycin thus
leading to a higher biological effect.113,114
Cereulide is produced by a non ribosomal peptide synthetase (NRPS) encoded in an operon situated
in a virulence plasmid related to the B. anthracis toxin plasmid pXO1.115,116 The gene sequence (ces) that
encodes the enzymatic machinery required for the synthesis of cereulide also includes genes encoding
a putative hydrolase (cesH), a phosphopantetheinyl transferase (cesP), a type II thioesterase (cesT), and
a putative ABC transporter (cesC7D).115,116 Because of its small size and low antigenicity, cereulide is
difficult to detect using immunological methods. Instead, assays with cultured cells (Hep-2) allow for the
determination of its biological activity leading to vacuole formation and impairment of mitochondrial
dehydrogenase activity.109,117–119
It has been demonstrated that there are different cereulide variants (isocereulides) that differ in toxic-
ity to Hep-2 and lead to different effects on the conductance of lipid bilayers.75,120 These in vitro effects
could correlate with the severity of the gastrointestinal disease.75
Although it has been established that cereulide doses of around 8 μg/kg body weight can be associ-
ated to severe emetic outbreaks,102 in vitro assays in Caco-2 cells showed that there is an accumulative
effect with successive subemetic doses of cereulide ranging from 0.125 to 4 ng/mL. Indeed, the effects on
viability, proliferation, and release of lactate dehydrogenase were detected at cereulide concentrations of
0.5–5 μg/mL.74 Interestingly, cereulide treatments also change the expression of proteins such as PCSK9,
cathepsin D, apolipoprotein A-I, proapolipoprotein, and apolipoprotein preprotein.74
Although Hep-2 cells are most widely used to evaluate the toxicity of cereulide, other cell lines such
as HepG2 (inhibition of RNA-synthesis),76 Hepa-1 (cytotoxicity),76 fetal porcine Langerhans islets (necro-
sis),121 natural killer (NK) cells (apoptosis and swelling of mitochondria)122 and HeLa, Calu-3, and Paju
(dissipation of the mitochondrial membrane potential)77 have also been used. These assays of cereulide
activity correlate with the gold standard assay for this toxin, that is, the boar sperm motility assay (see
ex vivo assays).
Also, environmental factors control the production of extracellular factors in B. cereus.102 Indeed,
growth in anaerobic conditions and low oxidation–reduction potential favor HBL and NHE production
through the participation of the two-component system, ResDE, and the transcriptional redox regulator,
Fnr.102 B. cereus has the ability to adapt to low pH conditions and survive within the gastrointestinal
tract,135,136 but the adaptation to sublethal low pH values inhibits HBL production.136
FIGURE 8.2 Adhesion of vegetative cells of B. cereus onto cultured human enterocytes (Caco-2 cells).
140 Laboratory Models for Foodborne Infections
Adhesion onto the epithelial cells of bacteria of the B. cereus group is related to the presence of flagella
components (i.e., FLhA) whose expression is not controlled by the PlcR regulator.138
Concerning models of professional phagocytic cells, studies with J774 macrophages demonstrate
that the metalloprotease InhA1 (main component of the exosporium) is involved in the escape of
Bacillus from macrophages.140 These findings are related to the ability of InhA1 to increase membrane
permeability.140
Spores of B. anthracis adhered to a diversity of epithelial cells like A549, CHO, and Caco-2.141
Interestingly, a model of M-like cells was developed by using cocultures of Caco-2 (epithelial) and Raji
B cells (human Burkitt’s lymphoma). In this model, there was an efficient translocation of B. anthracis
as compared with single cultures of Caco-2 cells.142 These findings encourage further studies on the
mechanisms involved in the pathogenesis of intestinal anthrax.
8.4 E
x Vivo and In Vivo Models
Different in vivo and ex vivo models of infection have been used to study the pathogenesis of Bacillus
spp. These models can determine both the importance of the route of infection and the virulence factors
involved. Table 8.1 summarizes different models reported in the scientific literature.
Bacillus 141
8.5 Invertebrates
Despite evolutionary distances between mammals and invertebrates, they share common mechanisms
concerning detection, signaling, and response to pathogens. These characteristics have encouraged mod-
eling mammals’ host–microbe interactions by using different invertebrates.158
8.5.1 Nematodes
Caenorhabditis elegans is a bacteriovorous soil nematode that has gained attention as a model for the
study of host–microbe interactions.159 It is a natural host for Bacillus spp. and can be involved in the
spreading of these microorganisms.160 In addition, it is considered as a potential vector for human patho-
gens related to food borne diseases.161 It is worth noting that the defenses of C. elegans against pathogens
rely on epithelial cells as key players of its innate immune system.159 Interestingly, there are similarities
between structures of the intestinal epithelia of C. elegans and those of mammals.159
Since C. elegans feeds on a variety of microbes, it is usually maintained on agar plates seeded with
E. coli. Worms are motile and curvilinear, displaying typical internal structures when in healthy condi-
tions.162 The effects of pathogens on C. elegans are evidenced by assessing the survival ratio, ratio of
142 Laboratory Models for Foodborne Infections
infected worms, fecundity, development, microbial load, and more refined readouts including transcrip-
tional responses.159,162
C. elegans and other nematode species (Pristionchus pacificus) have been used to study virulence fac-
tors of Bacillus spp. isolated from different sources (soil, horse dung, and beetles).160 There was a strain-
dependent effect on the survival ratio of infected nematodes, and there were different responses for the
fecundity of the two nematodes studied. Interestingly, virulence patterns depended on both the genetic
background of worms and bacteria.160
By using this model, Franks et al.162 determined the correlation between the presence of genes related
to tellurite resistance and the virulence of B. anthracis. In this study, they demonstrated a correlation
between results obtained in this model and resistance to reactive oxygen species and antimicrobial pep-
tides as well as survival in the blood of humans and mice.162
8.5.2 Insects
8.5.2.1 Galleria mellonella
Caterpillars of the greater wax moth, G. mellonella, have been widely used to mimic gastrointestinal
infection caused by bacteria of the genus Bacillus.
G. mellonella eggs are incubated at 30°C, and the larvae are reared on beeswax and pollen at 25°C.
There are three infection routes: free ingestion, force feeding, or intrahemocoelic injection.163
The use of this model allowed for great strides in the knowledge of the factors involved in the viru-
lence of Bacillus spp. In this context, studies conducted with G. mellonella permitted the identification
of virulence factors that both B. cereus and B. thuringiensis have in common. These factors include the
pleiotropic regulator PlcR, the internaline/siderophore-like protein IlsA, the PlcR-regulated metallopro-
tease InhA2, the flagellar protein FlhA, and other adaptation and pathogenic genes.101,124,164,166
As described previously in this chapter, production of secreted virulence factors by B. thuringiensis
and B. cereus is regulated by the PlcR regulon, which is the major virulence regulator in these species.
This regulation has been demonstrated in G. mellonella where disruption of the plcR gene mitigates
the virulence of B. thuringiensis and B. cereus.124 These results were also seen in mice, suggesting that
extracellular factors that depend on the plcR regulon are required for the pathogenesis in both hosts.124
In a series of experiments, spores of strains from B. cereus of a clinical origin were administered by two
methods: free ingestion and force feeding. The survival ratio was evaluated and compared with those of
other microorganisms including some human pathogens such as Listeria monocytogenes. The lowest sur-
vival ratios were obtained with the entomopathogen Pseudomonas entomophila. Some B. cereus strains lead
to low ratios of mortality that were significantly increased in the presence of Cry toxin (entomotoxin from
B. thuringiensis). In contrast, one attenuated strain of B. anthracis and L. monocytogenes did not lead to lar-
vae mortality even in the presence of Cry toxin. These results demonstrate that the suitability of this model to
assess the biological activity of human pathogens by the oral route is both strain- and species-dependent.165
B. weihenstephanensis virulence was assayed for the first time in an in vivo model using G. mellonella.
B. weihenstephanensis is a psychrotolerant species of the B. cereus group that is potentially patho-
genic for humans. In this model, it has been demonstrated that the virulence is temperature-dependent.
A comparison between the virulence of B. cereus and B. weihenstephanensis showed that the virulence
of the latter is low at 37°C and maximal at 15°C. Interestingly, these findings correlated with those found
in vitro with cultured Vero cells.167
B. cereus sphingomyelinase (SMase) acquired relevance using this infection model. Doll et al. sug-
gested that SMase is a relevant virulence factor involved in the mortality rate of insect larvae infected with
B. cereus. In addition, their results have shown that Nhe and SMase complement each other for full virulence.71
Furthermore, this infection model, along with assays with cultured eukaryotic cells, has been used to
test the virulence of B. cereus strains isolated in a severe intestinal infection in neonates.168 Also, the rel-
evance of InhA1, NprA, and HlyII to differentiate pathogenic from nonpathogenic B. cereus strains was
assessed in G. mellonella.169 This model was also appropriate to gain insight into the expression of viru-
lent genes in vivo by using in vivo expression technology (IVET). Through this experimental approach,
the relevance of the expression of internalin under iron deprivation was demonstrated.164
Bacillus 143
8.6 Vertebrates
8.6.1 Rabbit
Early studies used this model to analyze crude cell-free spent culture supernatants of B. cereus cul-
tures.170 Biological activity was detected through two main experimental approaches: accumulation
of fluid in ligated rabbit ileal loops (RILs) and vascular permeability reaction (VPR). In the RIL test,
samples are injected into ileal loops and fluid accumulation is evaluated after euthanasia. Results
obtained with this technique correlated with those obtained in the VPR. This latter assay involves the
intradermal injection of samples in the shaved back of rabbits. Next, a Blue Evans solution is admin-
istered intravenously (ear vein), and the diameter of blue zones in the inoculation zone is evaluated. In
some cases, edema and hemorrhage are also present.170 It has been suggested that capillaries of blood
were affected by B. cereus enterotoxin, giving a local and also a systemic response since histologi-
cal changes were observed in the liver.171 Comparing with ligated ileal loop assays, the VPR is faster,
cheaper, and easier to perform.172
Both vegetative cells and cell-free culture filtrates from B. cereus cultures induce fluid accumulation.
However, ileal loop response did not differentiate strains isolated from food-poisoning outbreaks from
those isolated from other sources.173 Fluid accumulation was also positive for other Bacillus species, for
example, some B. thuringiensis strains. It is important to highlight that several strains that were positive
for the ileal loop assays failed to elicit diarrhea neither when injected intraluminally into normal ileum
rabbits nor when administered by a stomach catheter.173 This experimental approach (conducted in rab-
bits or mice) demonstrated histological changes and, in some instances, lethality in mice.64,171,173,174
Ligated RIL has been used to analyze purified toxins or their components. For example, Beecher
et al. studied hemolysin BL activity, a three-component toxin associated with diarrheal food poisoning.
The authors showed that all three components were necessary for maximal activity.42 In addition, Agata
et al. cloned the bceT gene, encoding one of the enterotoxic proteins of B. cereus. Supernatants of E. coli
cultures bearing this gene were positive in the VPR, resulting in fluid accumulation in murine ileal loops
(mouse), and had mouse lethal activity.64
Several enteropathogenic bacteria that induced fluid accumulation in ligated intestinal loop can also
produce a characteristic reaction when injected into the rabbit skin.172 However, other authors have found
no relationship between permeability reaction size and fluid accumulation in ligated RIL.175
Rabbit vascular permeability reaction has been used for the study of enterotoxins, either for the analy-
sis of toxins that could be related to outbreaks176 or for the study of isolated toxins.
Sometimes toxins caused vascular permeability, fluid accumulation, and lethal activity in mice— all
requested features for a diarrheal enterotoxin.64
144 Laboratory Models for Foodborne Infections
8.6.2 Zebrafish
Zebrafish (Danio rerio) is a vertebrate organism that shares some level of the functional conservation of genes
and signaling pathways with mammals. In addition, developmental particularities of the immune system of
zebrafish allow for the study of different levels of complexity of the immune response, that is, innate immune
response in embryos and larvae or fully developed adaptive response in adult fishes. It is noteworthy that
there are many similarities between components of zebrafish and human immune systems. Other advantages
are that each single experiment could include hundreds of embryos for examination of several conditions
and that the transparency of some stages of the fish cycle allows for the localization of infecting bacteria. In
the framework of intestinal infections, bacteria can be administered by the oral route or static immersion.177
There are few reports on the use of this model to study the virulence factors of Bacillus spp., and to
our knowledge, there is only one study conducted by using the oral route of infection with these micro-
organisms.178 In contrast, Bolcome et al. developed a zebrafish embryonic model to assess the effect
of B. anthracis toxins. They demonstrated an increased endothelial permeability by extravasation of
fluorescent microspheres in toxin-injected embryos.179 Further studies demonstrated the involvement of
the MAPK pathway in the biological effects of the lethal toxin. Interestingly, in this study, genetically
modified zebrafishes were developed.180
8.6.3 Monkeys
The concept of diarrheal and emetic syndromes in foodborne B. cereus pathologies started in the 1970s,
when most of the in vivo studies included the analysis of fluid accumulation in ligated rabbit or mice ileal
loop and alteration of vascular permeability.
Nausea and vomiting are pathognomonic signs of gastrointestinal infections and intoxications in
humans. However, emetic reflex is restricted to some mammals (e.g., pig, goat, dog, cat, seal, sperm
whale, and nonhuman primates) and nonmammals (e.g., some fish, reptiles, amphibians, and birds).
Although, overall, some of the above-mentioned animals could be suitable to study emesis following
food poisoning, there are intrinsic differences between humans and other animals.181 Consequently, stud-
ies in nonhuman primates fulfil most of the expected clinic outcomes although their implementation is
restricted due to ethical considerations.
Early studies in rhesus monkeys were used to analyze the effect (diarrhea and vomiting) of B. cereus
ingested with foods.182,183 Along with studies in rabbits (ligated ileal loop), the monkey feeding test
permitted the demonstration of factors involved in vomiting, and diarrheal syndromes were distinct enti-
ties.184 Some examples of cats and dogs suffering from diarrhea were described, but these models were
not practical for the study of B. cereus infection.173
8.6.5 Mice
Mice (Mus musculus) have long been used for modeling host–pathogen interactions. They constitute an
easy-to-handle mammalian animal model that presents a fully developed immune system and shares
Bacillus 145
with humans many mechanisms of detection, signaling, and response to microorganisms. Furthermore,
a wide range of tools have been developed for this model and genetic engineering resulted in obtaining
strains of mice deficient in key steps of the host response against microorganisms.
Since Koch’s early reports on B. anthracis, mice were used in studies on this microorganism.5 Models
became more and more refined and mouse strains with specific deficiencies in key steps of the immune
response allowed for gaining further insight into the interactions between B. anthracis and the host.
As indicated in Section 8.1, the capsule of B. anthracis endows bacteria with the ability to evade
phagocytosis. It has been shown that A/J mice are susceptible to the noncapsulated Sterne strain of
B. anthracis. This infection model resembles the situation of conventional mice infected with wild-type
strains but, concomitantly, has reduced safety concerns. The susceptibility of A/J mice to noncapsulated
B. anthracis is due to deficiency in C5, a key component of the complement system. With this strain,
Duong et al. developed a murine model of systemic anthrax. Using both spores and vegetative cells, and
a subcutaneous infection, all infected animals became moribund or died.188 Mice showed severe dam-
age at the thoracic nodes, thymus, and spleen. However, there was a remarkable lack of hemorrhage or
inflammation in Peyer’s patches, mesenteric lymph nodes, and bronchial-associated lymphoid tissue.
Interestingly, in this model of infection, pathologies in the gastrointestinal tract were absent in contrast
with human systemic anthrax.188
A/J mice were also used for the development of a murine model of gastrointestinal anthrax.189 In this
study, infection was done by gavage with suspensions of Sterne strain (vegetative form). Results showed
animal death in a dose-dependent manner. It is worth mentioning that bacilli penetrated and grew within the
intestinal villi. As a consequence, bacteremia and systemic dissemination were observed with pathological
changes similar to those of systemic anthrax. The animals showed a decrease in weight, body temperature,
and activity along with evidence of intestinal hemorrhage. However, no immune activation was detected.189
A remarkable result obtained in this model was the early compromise of the intestinal epithelium.
Disruption of the normal villus structures, villus blunting, and ulceration at the ileum and jejunum showed
that gastrointestinal anthrax mainly affects the small intestine instead of the colon. Gastrointestinal tract
epithelium was the first site of infection. Xie et al. hypothesized that extracellular factors produced by
vegetative bacteria (e.g., LF) could be involved in the early steps of pathogenesis as well as in the immu-
nosuppressive effect of anthrax.189 An antibiotic treatment (amoxicillin + gentamicin) assayed 24 h after
infection resulted in complete recuperation with no deaths of the infected animals. These findings encour-
age the use of this in vivo model of gastrointestinal anthrax for the evaluation of potential therapeutic
approaches. Interestingly, A/J mice orally infected with spores of the Sterne strain showed a significant
breakdown of the intestinal barrier function. This leads to the systemic dissemination of B. anthracis and
commensal microorganisms.190 This model of gastrointestinal anthrax allowed for the demonstration of
the role of the MAPK pathway in the impairment of the immune response triggered by B. anthracis.
In Balb/cJ mice, a bioluminescent B. anthracis strain, unable to produce PA, was used for the real-time
analysis of gastrointestinal infection.26 In this study, the importance of the selection of the inoculation
technique was demonstrated. Rigid feeding needles cause abrasions in the laryngopharynx, which in
turn leads to systemic dissemination, whereas by using a flexible plastic tube, the infection initiated in
Peyer’s patches resembles gastrointestinal infection.26
The DBA/2 strain is a complement-deficient mouse susceptible to the Sterne strain. Tonry et al. ana-
lyzed gastrointestinal infection using this model. To maximize spore survival, neutralization of the stom-
ach acidity was performed before infection. In addition, a thiabendazole paste containing spores was
formulated to protect spores from gastric acidity. Intragastric inoculation of animals was performed by a
catheter syringe. The infected animals showed signs similar to those of human gastrointestinal anthrax,
that is, edema and hemorrhage.142
Also, the effects of B. anthracis toxins were studied in murine models. The effect of purified EF was
assayed by intravenous or intraperitoneal injection in Balb/cJ mice. The toxin caused rapid death of mice,
even at low doses, and these findings are related to the expression of toxin receptors in various tissues,
which in turn correlates with histological damage. The animals showed a marked intestinal dilatation due
to the extensive intestinal fluid accumulation in the gastrointestinal tract. Necrotic lesions were observed
in the mucosa. This experimental model demonstrated the toxic effects induced by purified EF, suggesting
that the death of animals could be related to multiorgan failure.191 On the other hand, anthrax LF caused
146 Laboratory Models for Foodborne Infections
intestinal damage in both C57BL/6J and Balb/c mice after being injected intravenously. Likewise, when A/J
mice were orally infected with spores of the Sterne strain,190 a systemic infection with commensal enteric
organisms was seen. An early breakdown of the gastrointestinal barrier, villus blunting, hemorrhage, and
ulceration, along with the known immunosuppressive effect of LF, provided a potential mechanism for
invasion via the enteric route.192 This toxin causes a dose-dependent disruption of the intestinal epithelial
integrity. Interestingly, the effects were partially attenuated by the co administration of antibiotics.
To investigate the toxic activity of B. cereus-secreted proteins, DBA/2J mice were inoculated intra-
peritoneally with filtered spent culture supernatants.61 Shortly after inoculation (10 min), a high ratio of
cells was observed in the peritoneal cavity. When viable bacteria were inoculated, the recruitment of
high numbers of neutrophils masked the effect on other cells. The authors concluded that Hbl and Nhe
cannot complement each other in spite of the coexpression of these factors.61
A murine model of B. cereus gastrointestinal infection was developed by Rolny et al.193 The vegeta-
tive B. cereus strain B10502 isolated from a food-borne outbreak was used to infect C57BL/6J mice by
gavage. To our knowledge, this was the first published data of an in vivo model of B. cereus gastrointes-
tinal infection. In contrast to murine gastrointestinal anthrax, which produced intestinal hemorrhage,
edema, systemic dissemination, and death, mouse infection with B. cereus led to a transient passage of
the bacteria through the digestive tract. Although self-limiting, B. cereus infection modifies the balance
of relevant immune cells in different regions of the intestinal mucosa and immune-associated tissues.
Ratios and activation statuses of different cell populations were studied in the spleen, Peyer’s patches,
and mesenteric lymph nodes. These results correlated with an increase in the size of Peyer’s patches in
infected mice. Additionally, the infected mice showed a higher ratio of intestinal goblet cells and the
presence of mononuclear cell infiltrates in the spleen. Cytokine mRNA expression showed a significant
increase in IFN-γ in mesenteric lymph nodes. At the same time, a slight increase in IL-12 mRNA and
TNF-α mRNA was observed. This study established relevant immune readouts to assess gastrointestinal
infection with B. cereus.193
8.6.6 Rats
The rat model is seldom used for the study of Bacillus spp. infections. Human-flora-associated rats
(Germfree Sprague–Dawley rats) were used to analyze the effect of B. cereus in the gastrointestinal
tract. Evaluation of the composition of the indigenous gut flora and examination of whether spores and
vegetative cells were able to persist in the gut were carried out. Experiments included administration
of irradiated spores, untreated spores, heat-activated spores, and vegetative cells of a k nown-to-cause
food-poisoning B. cereus strain.194 By DGGE analysis, it was demonstrated that when animals were
administered with spores, significant changes in the intestinal microbiota were evident. However,
culture-dependent approaches did not reveal changes during the passage of B. cereus. Using in vitro
assays and commercial kits for detection of toxins, no enterotoxins were detected in the intestinal tract.
In addition, in those infectious conditions, only spores were able to resist the gastrointestinal passage.194
8.7 Concluding Remarks
The interaction of the genus Bacillus with different hosts leads to various outcomes: from a beneficial
relationship195 to life-threatening pathologies such as anthrax. The use of in vitro and in vivo mod-
els allowed for establishing key steps and mechanisms involved in infection and foodborne outbreaks.
Experimental models with cultured eukaryotic cells such as enterocyte-like Caco-2 cells helped unravel
extracellular factors involved in the gastrointestinal pathologies as well as to demonstrate the partici-
pation of direct bacteria–enterocyte interactions in the virulence. Further insights were obtained with
in vivo invertebrate and vertebrate models that elucidated many aspects of the pathogenesis in a more
complex cellular context. The recent advances in the field of in vivo expression technologies herald fur-
ther gains in our understanding of the control and treatment of foodborne pathologies associated with
Bacillus spp.
Bacillus 147
Acknowledgments
Jessica Minnaard is a Research Scientist at the Consejo Nacional de Investigaciones Científicas y Técnicas
(CONICET, Argentina), Ivanna S. Rolny is a Research Scientist at the Biological Sciences Department
(Facultad de Ciencias Exactas, Universidad Nacional de La Plata, Argentina), and Pablo F. Pérez is
a Research Scientist at the Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET,
Argentina) and a Microbiology Professor at the Biological Sciences Department (Facultad de Ciencias
Exactas, Universidad Nacional de La Plata, Argentina).
REFERENCES
1. Higgins, D. & Dworkin, J. Recent progress in Bacillus subtilis sporulation. FEMS Microbiol Rev 36,
131–48 (2012).
2. Maughan, H. & Van der Auwera, G. Bacillus taxonomy in the genomic era finds phenotypes to be essen-
tial though often misleading. Infect Genet Evol 11, 789–97 (2011).
3. Morens, D.M. Characterizing a “new” disease: epizootic and epidemic anthrax, 1769–1780. Am J Public
Health 93, 886–93 (2003).
4. Brock, T.D. Milestones in Microbiology: 1546 to 1940. American Society for Microbiology, Washington,
DC (1999).
5. Brock, T.D. Robert Koch: A Life in Medicine and Bacteriology, p. 363. American Society for
Microbiology, Washington, DC (1999).
6. Guinebretiere, M.H. et al. Bacillus cytotoxicus sp. nov. is a novel thermotolerant species of the Bacillus
cereus group occasionally associated with food poisoning. Int J Syst Evol Microbiol 63, 31–40 (2013).
7. Guinebretiere, M.H. et al. Ability of Bacillus cereus group strains to cause food poisoning varies
according to phylogenetic affiliation (groups I to VII) rather than species affiliation. J Clin Microbiol
48, 3388–91 (2010).
8. Lund, T., De Buyser, M.L. & Granum, P.E. A new cytotoxin from Bacillus cereus that may cause
necrotic enteritis. Mol Microbiol 38, 254–61 (2000).
9. Helgason, E. et al. Bacillus anthracis, Bacillus cereus, and Bacillus thuringiensis—one species on the
basis of genetic evidence. Appl Environ Microbiol 66, 2627–30 (2000).
10. Priest, F.G., Barker, M., Baillie, L.W., Holmes, E.C. & Maiden, M.C. Population structure and evolution
of the Bacillus cereus group. J Bacteriol 186, 7959–70 (2004).
11. Rasko, D.A., Altherr, M.R., Han, C.S. & Ravel, J. Genomics of the Bacillus cereus group of organisms.
FEMS Microbiol Rev 29, 303–29 (2005).
12. Tourasse, N.J., Helgason, E., Okstad, O.A., Hegna, I.K. & Kolsto, A.B. The Bacillus cereus group: novel
aspects of population structure and genome dynamics. J Appl Microbiol 101, 579–93 (2006).
13. Guinebretiere, M.H. et al. Ecological diversification in the Bacillus cereus group. Environ Microbiol 10,
851–65 (2008).
14. Kamar, R. et al. Pathogenic potential of Bacillus cereus strains as revealed by phenotypic analysis. J
Clin Microbiol 51, 320–3 (2013).
15. Sternbach, G. The history of anthrax. J Emerg Med 24, 463–7 (2003).
16. Littman, R.J. The plague of Athens: epidemiology and paleopathology. Mt Sinai J Med 76, 456–67
(2009).
17. Dagnino, J. What was the plague of Athens? Rev Chilena Infectol 28, 374–80 (2011).
18. Papagrigorakis, M.J., Yapijakis, C., Synodinos, P.N. & Baziotopoulou-Valavani, E. DNA examination
of ancient dental pulp incriminates typhoid fever as a probable cause of the plague of Athens. Int J Infect
Dis 10, 206–14 (2006).
19. Ehrenkranz, N.J. & Sampson, D.A. Origin of the old testament plagues: explications and implications.
Yale J Biol Med 81, 31–42 (2008).
20. Hatami, H., Ramazankhani, A. & Mansoori, F. Two cases of gastrointestinal anthrax with an unusual
presentation from Kermanshah (western Iran). Arch Iran Med 13, 156–9 (2010).
21. Meric, M., Willke, A., Muezzinoglu, B., Karadenizli, A. & Hosten, T. A case of pneumonia caused by
Bacillus anthracis secondary to gastrointestinal anthrax. Int J Infect Dis 13, e456–8 (2009).
148 Laboratory Models for Foodborne Infections
22. Babamahmoodi, F., Aghabarari, F., Arjmand, A. & Ashrafi, G.H. Three rare cases of anthrax arising
from the same source. J Infect 53, e175–9 (2006).
23. Bravata, D.M. et al. Inhalational, gastrointestinal, and cutaneous anthrax in children: a systematic
review of cases: 1900 to 2005. Arch Pediatr Adolesc Med 161, 896–905 (2007).
24. CDC. Gastrointestinal anthrax after an animal-hide drumming event—New Hampshire and
Massachusetts, 2009. Morb Mortal Wkly Rep 59, 872–7 (2010).
25. Owen, J.L., Yang, T. & Mohamadzadeh, M. New insights into gastrointestinal anthrax infection. Trends
Mol Med 21(3), 154–63 (2015).
26. Glomski, I.J., Piris-Gimenez, A., Huerre, M., Mock, M. & Goossens, P.L. Primary involvement of phar-
ynx and peyer’s patch in inhalational and intestinal anthrax. PLoS Pathog 3, e76 (2007).
27. Jensen, G.B., Hansen, B.M., Eilenberg, J. & Mahillon, J. The hidden lifestyles of Bacillus cereus and
relatives. Environ Microbiol 5, 631–40 (2003).
28. Avashia, S.B. et al. Fatal pneumonia among metalworkers due to inhalation exposure to Bacillus cereus
containing Bacillus anthracis toxin genes. Clin Infect Dis 44, 414–6 (2007).
29. Hoffmaster, A.R. et al. Characterization of Bacillus cereus isolates associated with fatal pneumo-
nias: strains are closely related to Bacillus anthracis and harbor B. anthracis virulence genes. J Clin
Microbiol 44, 3352–60 (2006).
30. Johnson, S.L. et al. Finished genome sequence of Bacillus cereus strain 03BB87, a clinical isolate with
B. anthracis virulence genes. Genome Announc 3, e01446-14 (2015).
31. Logan, N.A. Bacillus and relatives in foodborne illness. J Appl Microbiol 112, 417–29 (2012).
32. Jan, S. et al. Biodiversity of psychrotrophic bacteria of the Bacillus cereus group collected on farm and
in egg product industry. Food Microbiol 28, 261–5 (2011).
33. Apetroaie-Constantin, C. et al. Bacillus subtilis and B. mojavensis strains connected to food poisoning
produce the heat stable toxin amylosin. J Appl Microbiol 106, 1976–85 (2009).
34. Ostensvik, O., From, C., Heidenreich, B., O’Sullivan, K. & Granum, P.E. Cytotoxic Bacillus spp. belonging
to the B. cereus and B. subtilis groups in Norwegian surface waters. J Appl Microbiol 96, 987–93 (2004).
35. From, C., Pukall, R., Schumann, P., Hormazabal, V. & Granum, P.E. Toxin-producing ability among
Bacillus spp. outside the Bacillus cereus group. Appl Environ Microbiol 71, 1178–83 (2005).
36. Liu, S., Moayeri, M. & Leppla, S.H. Anthrax lethal and edema toxins in anthrax pathogenesis. Trends
Microbiol 22, 317–25 (2014).
37. Tonello, F. & Zornetta, I. Bacillus anthracis factors for phagosomal escape. Toxins (Basel) 4, 536–53 (2012).
38. Koehler, T.M. Bacillus anthracis physiology and genetics. Mol Aspects Med 30, 386–96 (2009).
39. Moayeri, M. et al. Inflammasome sensor Nlrp1b-dependent resistance to anthrax is mediated by
caspase-1, IL-1 signaling and neutrophil recruitment. PLoS Pathog 6, e1001222 (2010).
40. Terra, J.K. et al. Cutting edge: resistance to Bacillus anthracis infection mediated by a lethal toxin sensi-
tive allele of Nalp1b/Nlrp1b. J Immunol 184, 17–20 (2010).
41. Drysdale, M. et al. Capsule synthesis by Bacillus anthracis is required for dissemination in murine
inhalation anthrax. EMBO J 24, 221–7 (2005).
42. Beecher, D.J., Schoeni, J.L. & Wong, A.C. Enterotoxic activity of hemolysin BL from Bacillus cereus.
Infect Immun 63, 4423–8 (1995).
43. Beecher, D.J. & Wong, A.C. Tripartite haemolysin BL: isolation and characterization of two distinct
homologous sets of components from a single Bacillus cereus isolate. Microbiology 146 (Pt 6), 1371–80
(2000).
44. Lund, T. & Granum, P.E. Comparison of biological effect of the two different enterotoxin complexes
isolated from three different strains of Bacillus cereus. Microbiology 143 (Pt 10), 3329–36 (1997).
45. Beecher, D.J., Olsen, T.W., Somers, E.B. & Wong, A.C. Evidence for contribution of tripartite hemolysin
BL, phosphatidylcholine-preferring phospholipase C, and collagenase to virulence of Bacillus cereus
endophthalmitis. Infect Immun 68, 5269–76 (2000).
46. Granum, P.E. & Nissen, H. Sphingomyelinase is part of the “enterotoxin complex” produced by Bacillus
cereus. FEMS Microbiol Lett 110, 97–100 (1993).
47. Tran, S.L. et al. Haemolysin II is a Bacillus cereus virulence factor that induces apoptosis of macro-
phages. Cell Microbiol 13, 92–108 (2011).
48. Fricke, B. et al. The cell envelope-bound metalloprotease (camelysin) from Bacillus cereus is a possible
pathogenic factor. Biochim Biophys Acta 1537, 132–46 (2001).
Bacillus 149
49. Ehling-Schulz, M., Fricker, M. & Scherer, S. Identification of emetic toxin producing Bacillus cereus
strains by a novel molecular assay. FEMS Microbiol Lett 232, 189–95 (2004).
50. Horwood, P.F., Burgess, G.W. & Oakey, H.J. Evidence for non-ribosomal peptide synthetase production
of cereulide (the emetic toxin) in Bacillus cereus. FEMS Microbiol Lett 236, 319–24 (2004).
51. Minnaard, J. et al. Virulence of Bacillus cereus: a multivariate analysis. Int J Food Microbiol 116,
197–206 (2007).
52. Minnaard, J., Lievin-Le Moal, V., Coconnier, M.H., Servin, A.L. & Perez, P.F. Disassembly of F-actin
cytoskeleton after interaction of Bacillus cereus with fully differentiated human intestinal Caco-2 cells.
Infect Immun 72, 3106–12 (2004).
53. Minnaard, J., Rolny, I.S. & Perez, P.F. Interaction between Bacillus cereus and cultured human entero-
cytes: effect of calcium, cell differentiation, and bacterial extracellular factors. J Food Prot 76, 820–6
(2013).
54. Mikkola, R. et al. Amylosin from Bacillus amyloliquefaciens, a K+ and Na+ channel-forming toxic pep-
tide containing a polyene structure. Toxicon 49, 1158–71 (2007).
55. Rasimus-Sahari, S. et al. The peptide toxin amylosin of Bacillus amyloliquefaciens from moisture-
damaged buildings is immunotoxic, induces potassium efflux from mammalian cells, and has
antimicrobial activity. Appl Environ Microbiol 81, 2939–49 (2015).
56. Salkinoja-Salonen, M.S. et al. Toxigenic strains of Bacillus licheniformis related to food poisoning.
Appl Environ Microbiol 65, 4637–45 (1999).
57. From, C., Hormazabal, V. & Granum, P.E. Food poisoning associated with pumilacidin-producing
Bacillus pumilus in rice. Int J Food Microbiol 115, 319–24 (2007).
58. Minnaard, J., Humen, M. & Perez, P.F. Effect of Bacillus cereus exocellular factors on human intestinal
epithelial cells. J Food Prot 64, 1535–41 (2001).
59. Gray, K.M., Banada, P.P., O’Neal, E. & Bhunia, A.K. Rapid Ped-2E9 cell-based cytotoxicity analysis
and genotyping of Bacillus species. J Clin Microbiol 43, 5865–72 (2005).
60. Lund, T. & Granum, P.E. Characterisation of a non-haemolytic enterotoxin complex from Bacillus
cereus isolated after a foodborne outbreak. FEMS Microbiol Lett 141, 151–6 (1996).
61. Sastalla, I. et al. The Bacillus cereus Hbl and Nhe tripartite enterotoxin components assemble sequen-
tially on the surface of target cells and are not interchangeable. PLoS One 8, e76955 (2013).
62. Jeßberger, N., Dietrich, R., Bock, S., Didier, A. & Märtlbauer, E. Bacillus cereus enterotoxins act as major
virulence factors and exhibit distinct cytotoxicity to different human cell lines. Toxicon 77, 49–57 (2014).
63. Rowan, N.J. et al. Putative virulence factor expression by clinical and food isolates of Bacillus spp. after
growth in reconstituted infant milk formulae. Appl Environ Microbiol 67, 3873–81 (2001).
64. Agata, N., Ohta, M., Arakawa, Y. & Mori, M. The bceT gene of Bacillus cereus encodes an enterotoxic
protein. Microbiology 141, 983–8 (1995).
65. Phung, D., Granum, P.E., Dietrich, R., Martlbauer, E. & Hardy, S.P. Inhibition of cytotoxicity by the Nhe
cytotoxin of Bacillus cereus through the interaction of dodecyl maltoside with the NheB component.
FEMS Microbiol Lett 330, 98–104 (2012).
66. Dietrich, R., Fella, C., Strich, S. & Martlbauer, E. Production and characterization of monoclonal
antibodies against the hemolysin BL enterotoxin complex produced by Bacillus cereus. Appl Environ
Microbiol 65, 4470–4 (1999).
67. Granum, P.E. & Lund, T. Bacillus cereus and its food poisoning toxins. FEMS Microbiol Lett 157,
223–8 (1997).
68. Buchanan, R.L. & Frankie, J.S. Evaluation of the Oxoid BCET-RPLA kit for the detection of Bacillus
cereus diarrheal enterotoxin as compared to cell culture cytotonicity. J Food Prot 55, 440–3 (1992).
69. Fletcher, P. & Logan, N.A. Improved cytotoxicity assay for Bacillus cereus diarrhoeal enterotoxin. Lett
Appl Microbiol 28, 394–400 (1999).
70. Hardy, S.P., Lund, T. & Granum, P.E. CytK toxin of Bacillus cereus forms pores in planar lipid bilayers
and is cytotoxic to intestinal epithelia. FEMS Microbiol Lett 197, 47–51 (2001).
71. Doll, V.M., Ehling-Schulz, M. & Vogelmann, R. Concerted action of sphingomyelinase and non-
hemolytic enterotoxin in pathogenic Bacillus cereus. PLoS One 8, e61404 (2013).
72. Fagerlund, A., Lindback, T., Storset, A.K., Granum, P.E. & Hardy, S.P. Bacillus cereus Nhe is a pore-
forming toxin with structural and functional properties similar to the ClyA (HlyE, SheA) family of
haemolysins, able to induce osmotic lysis in epithelia. Microbiology 154, 693–704 (2008).
150 Laboratory Models for Foodborne Infections
73. Andreeva, Z.I. et al. Purification and cytotoxic properties of Bacillus cereus hemolysin II. Protein Expr
Purif 47, 186–93 (2006).
74. Rajkovic, A. et al. Sub-emetic toxicity of Bacillus cereus toxin cereulide on cultured human enterocyte-
like Caco-2 cells. Toxins (Basel) 6, 2270–90 (2014).
75. Marxen, S. et al. Chemodiversity of cereulide, the emetic toxin of Bacillus cereus. Anal Bioanal Chem
407, 2439–53 (2015).
76. Andersson, M.A. et al. Toxicological profile of cereulide, the Bacillus cereus emetic toxin, in functional
assays with human, animal and bacterial cells. Toxicon 49, 351–67 (2007).
77. Jääskeläinen, E.L. et al. In vitro assay for human toxicity of cereulide, the emetic mitochondrial toxin
produced by food poisoning Bacillus cereus. Toxicol In Vitro 17, 737–44 (2003).
78. Mikami, T. et al. An improved method for detecting cytostatic toxin (emetic toxin) of Bacillus cereus
and its application to food samples. FEMS Microbiol Lett 119, 53–7 (1994).
79. Bonventre, P.F. Differential cytotoxicity of Bacillus anthracis and Bacillus cereus culture filtrates. J
Bacteriol 90, 284–5 (1965).
80. Lievin-Le Moal, V. & Servin, A.L. Pathogenesis of human enterovirulent bacteria: lessons from cul-
tured, fully differentiated human colon cancer cell lines. Microbiol Mol Biol Rev 77, 380–439 (2013).
81. Beecher, D.J. & Macmillan, J.D. Characterization of the components of hemolysin BL from Bacillus
cereus. Infect Immun 59, 1778–84 (1991).
82. Lindback, T., Fagerlund, A., Rodland, M.S. & Granum, P.E. Characterization of the Bacillus cereus Nhe
enterotoxin. Microbiology 150, 3959–67 (2004).
83. Guinebretiere, M.H., Broussolle, V. & Nguyen-The, C. Enterotoxigenic profiles of food-poisoning and
food-borne Bacillus cereus strains. J Clin Microbiol 40, 3053–6 (2002).
84. Moravek, M. et al. Determination of the toxic potential of Bacillus cereus isolates by quantitative
enterotoxin analyses. FEMS Microbiol Lett 257, 293–8 (2006).
85. Didier, A. et al. Monoclonal antibodies neutralize Bacillus cereus Nhe enterotoxin by inhibiting ordered
binding of its three exoprotein components. Infect Immun 80, 832–8 (2012).
86. Beecher, D.J. & MacMillan, J.D. A novel bicomponent hemolysin from Bacillus cereus. Infect Immun 58,
2220–7 (1990).
87. Lindback, T. et al. Cytotoxicity of the Bacillus cereus Nhe enterotoxin requires specific binding order
of its three exoprotein components. Infect Immun 78, 3813–21 (2010).
88. Haug, T.M. et al. Formation of very large conductance channels by Bacillus cereus Nhe in Vero and
GH(4) cells identifies NheA + B as the inherent pore-forming structure. J Membr Biol 237, 1–11 (2010).
89. Dietrich, R., Moravek, M., Burk, C., Granum, P.E. & Martlbauer, E. Production and characterization of
antibodies against each of the three subunits of the Bacillus cereus nonhemolytic enterotoxin complex.
Appl Environ Microbiol 71, 8214–20 (2005).
90. Beecher, D.J. & Wong, A.C. Identification and analysis of the antigens detected by two commercial
Bacillus cereus diarrheal enterotoxin immunoassay kits. Appl Environ Microbiol 60, 4614–6 (1994).
91. Ghelardi, E. et al. Identification and characterization of toxigenic Bacillus cereus isolates responsible
for two food-poisoning outbreaks. FEMS Microbiol Lett 208, 129–34 (2002).
92. Beecher, D.J. & Wong, A.C.L. Tripartite hemolysin BL from Bacillus cereus. Hemolytic analysis
of component interactions and a model for its characteristic paradoxical zone phenomenon. J Biol
Chem 272, 233–9 (1997).
93. Madegowda, M., Eswaramoorthy, S., Burley, S.K. & Swaminathan, S. X-ray crystal structure of the B
component of Hemolysin BL from Bacillus cereus. Proteins 71, 534–40 (2008).
94. Phung, D. et al. Crystallization and preliminary crystallographic analysis of the NheA component of the
Nhe toxin from Bacillus cereus. Acta Crystallogr Sect F Struct Biol Cryst Commun 68, 1073–6 (2012).
95. Ramarao, N. & Sanchis, V. The pore-forming haemolysins of Bacillus cereus: a review. Toxins (Basel)
5, 1119–39 (2013).
96. Fagerlund, A., Ween, O., Lund, T., Hardy, S.P. & Granum, P.E. Genetic and functional analysis of the
cytK family of genes in Bacillus cereus. Microbiology 150, 2689–97 (2004).
97. Baida, G., Budarina, Z.I., Kuzmin, N.P. & Solonin, A.S. Complete nucleotide sequence and molecular
characterization of hemolysin II gene from Bacillus cereus. FEMS Microbiol Lett 180, 7–14 (1999).
98. Menestrina, G., Serra, M.D. & Prevost, G. Mode of action of β-barrel pore-forming toxins of the staphy-
lococcal α-hemolysin family. Toxicon 39, 1661–72 (2001).
Bacillus 151
99. Andreeva, Z.I. et al. The properties of Bacillus cereus hemolysin II pores depend on environmental
conditions. Biochim Biophys Acta 1768, 253–63 (2007).
100. Baida, G.E. & Kuzmin, N.P. Cloning and primary structure of a new hemolysin gene from Bacillus
cereus. Biochim Biophys Acta 1264, 151–4 (1995).
101. Fedhila, S., Nel, P. & Lereclus, D. The InhA2 metalloprotease of Bacillus thuringiensis strain 407 is
required for pathogenicity in insects infected via the oral route. J Bacteriol 184, 3296–304 (2002).
102. Stenfors Arnesen, L.P., Fagerlund, A. & Granum, P.E. From soil to gut: Bacillus cereus and its food
poisoning toxins. FEMS Microbiol Rev 32, 579–606 (2008).
103. Alouf, J.E. Cholesterol-binding cytolytic protein toxins. Int J Med Microbiol 290, 351–6 (2000).
104. Gilmore, M.S., Cruz-Rodz, A.L., Leimeister-Wachter, M., Kreft, J. & Goebel, W. A Bacillus cereus
cytolytic determinant, cereolysin AB, which comprises the phospholipase C and sphingomyelinase
genes: nucleotide sequence and genetic linkage. J Bacteriol 171, 744–53 (1989).
105. Pomerantsev, A.P., Kalnin, K.V., Osorio, M. & Leppla, S.H. Phosphatidylcholine-specific phospholipase C
and sphingomyelinase activities in bacteria of the Bacillus cereus group. Infect Immun 71, 6591–606 (2003).
106. Beecher, D.J. & Wong, A.C. Cooperative, synergistic and antagonistic haemolytic interactions between
haemolysin BL, phosphatidylcholine phospholipase C and sphingomyelinase from Bacillus cereus.
Microbiology 146 (Pt 12), 3033–9 (2000).
107. Bottone, E.J. Bacillus cereus, a volatile human pathogen. Clin Microbiol Rev 23, 382–98 (2010).
108. Zenewicz, L.A., Wei, Z., Goldfine, H. & Shen, H. Phosphatidylinositol-specific phospholipase C of
Bacillus anthracis down-modulates the immune response. J Immunol 174, 8011–6 (2005).
109. Agata, N., Ohta, M., Mori, M. & Isobe, M. A novel dodecadepsipeptide, cereulide, is an emetic toxin of
Bacillus cereus. FEMS Microbiol Lett 129, 17–9 (1995).
110. Agata, N. et al. A novel dodecadepsipeptide, cereulide, isolated from Bacillus cereus causes vacuole
formation in HEp-2 cells. FEMS Microbiol Lett 121, 31–4 (1994).
111. Mikkola, R., Saris, N.E., Grigoriev, P., Andersson, M.A. & Salkinoja-Salonen, M. Ionophoretic proper-
ties and mitochondrial effects of cereulide: the emetic toxin of B. cereus. Eur J Biochem 263, 112–7
(1999).
112. Haggblom, M.M., Apetroaie, C., Andersson, M.A. & Salkinoja-Salonen, M.S. Quantitative analysis
of cereulide, the emetic toxin of Bacillus cereus, produced under various conditions. Appl Environ
Microbiol 68, 2479–83 (2002).
113. Teplova, V.V., Mikkola, R., Tonshin, A.A., Saris, N.E. & Salkinoja-Salonen, M.S. The higher toxicity
of cereulide relative to valinomycin is due to its higher affinity for potassium at physiological plasma
concentration. Toxicol Appl Pharmacol 210, 39–46 (2006).
114. Biesta-Peters, E.G. et al. Quantification of the emetic toxin cereulide in food products by liquid
chromatography-mass spectrometry using synthetic cereulide as a standard. Appl Environ Microbiol
76, 7466–72 (2010).
115. Ehling-Schulz, M. et al. Toxin gene profiling of enterotoxic and emetic Bacillus cereus. FEMS Microbiol
Lett 260, 232–40 (2006).
116. Ehling-Schulz, M. et al. Identification and partial characterization of the nonribosomal peptide synthe-
tase gene responsible for cereulide production in emetic Bacillus cereus. Appl Environ Microbiol 71,
105–13 (2005).
117. Agata, N., Ohta, M. & Yokoyama, K. Production of Bacillus cereus emetic toxin (cereulide) in various
foods. Int J Food Microbiol 73, 23–7 (2002).
118. Shinagawa, K., Ueno, S., Matsusaka, N. & Sugii, S. In vitro stability in biological activity and antigenic-
ity of the vascular permeability factor produced by Bacillus cereus. J Vet Med Sci 53, 317–9 (1991).
119. Finlay, W.J., Logan, N.A. & Sutherland, A.D. Semiautomated metabolic staining assay for Bacillus
cereus emetic toxin. Appl Environ Microbiol 65, 1811–2 (1999).
120. Stark, T. et al. Mass spectrometric profiling of Bacillus cereus strains and quantitation of the emetic
toxin cereulide by means of stable isotope dilution analysis and HEp-2 bioassay. Anal Bioanal Chem
405, 191–201 (2013).
121. Virtanen, S.M. et al. In vitro toxicity of cereulide on porcine pancreatic Langerhans islets. Toxicon 51,
1029–37 (2008).
122. Paananen, A. et al. Inhibition of human natural killer cell activity by cereulide, an emetic toxin from
Bacillus cereus. Clin Exp Immunol 129, 420–8 (2002).
152 Laboratory Models for Foodborne Infections
123. Okstad, O.A. et al. Sequence analysis of three Bacillus cereus loci carrying PlcR-regulated genes encod-
ing degradative enzymes and enterotoxin. Microbiology 145 (Pt 11), 3129–38 (1999).
124. Salamitou, S. et al. The plcR regulon is involved in the opportunistic properties of Bacillus thuringiensis
and Bacillus cereus in mice and insects. Microbiology 146, 2825–32 (2000).
125. Gohar, M. et al. Two-dimensional electrophoresis analysis of the extracellular proteome of Bacillus
cereus reveals the importance of the PlcR regulon. Proteomics 2, 784–91 (2002).
126. Slamti, L. et al. Distinct mutations in PlcR explain why some strains of the Bacillus cereus group are
nonhemolytic. J Bacteriol 186, 3531–8 (2004).
127. Slamti, L. & Lereclus, D. Specificity and polymorphism of the PlcR-PapR quorum-sensing system in the
Bacillus cereus group. J Bacteriol 187, 1182–7 (2005).
128. Brillard, J. & Lereclus, D. Characterization of a small PlcR-regulated gene co-expressed with cereolysin
O. BMC Microbiol 7, 52 (2007).
129. Slamti, L. & Lereclus, D. A cell–cell signaling peptide activates the PlcR virulence regulon in bacteria
of the Bacillus cereus group. EMBO J 21, 4550–9 (2002).
130. Bouillaut, L. et al. Molecular basis for group-specific activation of the virulence regulator PlcR by PapR
heptapeptides. Nucleic Acids Res 36, 3791–801 (2008).
131. Budarina, Z.I. et al. A new Bacillus cereus DNA-binding protein, HlyIIR, negatively regulates expres-
sion of B. cereus haemolysin II. Microbiology 150, 3691–701 (2004).
132. Sineva, E. et al. Iron regulates expression of Bacillus cereus hemolysin II via global regulator Fur.
J Bacteriol 194, 3327–35 (2012).
133. Lucking, G., Dommel, M.K., Scherer, S., Fouet, A. & Ehling-Schulz, M. Cereulide synthesis in emetic
Bacillus cereus is controlled by the transition state regulator AbrB, but not by the virulence regulator
PlcR. Microbiology 155, 922–31 (2009).
134. Frenzel, E. et al. CodY orchestrates the expression of virulence determinants in emetic Bacillus cereus
by impacting key regulatory circuits. Mol Microbiol 85, 67–88 (2012).
135. Wijnands, L.M., Dufrenne, J.B., van Leusden, F.M. & Abee, T. Germination of Bacillus cereus spores
is induced by germinants from differentiated Caco-2 cells, a human cell line mimicking the epithelial
cells of the small intestine. Appl Environ Microbiol 73, 5052–4 (2007).
136. Berthold-Pluta, A., Pluta, A. & Garbowska, M. The effect of selected factors on the survival of Bacillus
cereus in the human gastrointestinal tract. Microb Pathog 82, 7–14 (2015).
137. Andersson, A., Granum, P.E. & Rönner, U. The adhesion of Bacillus cereus spores to epithelial cells
might be an additional virulence mechanism. Int J Food Microbiol 39, 93–9 (1998).
138. Ramarao, N. & Lereclus, D. Adhesion and cytotoxicity of Bacillus cereus and Bacillus thuringiensis to
epithelial cells are FlhA and PlcR dependent, respectively. Microbes Infect 8, 1483–91 (2006).
139. Ronner, U., Husmark, U. & Henriksson, A. Adhesion of bacillus spores in relation to hydrophobicity.
J Appl Bacteriol 69, 550–6 (1990).
140. Ramarao, N. & Lereclus, D. The InhA1 metalloprotease allows spores of the B. cereus group to escape
macrophages. Cell Microbiol 7, 1357–64 (2005).
141. Bozue, J. et al. Bacillus anthracis spores of the bclA mutant exhibit increased adherence to epithelial
cells, fibroblasts, and endothelial cells but not to macrophages. Infect Immun 75, 4498–505 (2007).
142. Tonry, J.H. et al. In vivo murine and in vitro M-like cell models of gastrointestinal anthrax. Microbes
Infect 15, 37–44 (2013).
143. Lucking, G., Stoeckel, M., Atamer, Z., Hinrichs, J. & Ehling-Schulz, M. Characterization of aerobic
spore-forming bacteria associated with industrial dairy processing environments and product spoilage.
Int J Food Microbiol 166, 270–9 (2013).
144. Fernandez-No, I.C. et al. Detection and quantification of spoilage and pathogenic Bacillus cereus,
Bacillus subtilis and Bacillus licheniformis by real-time PCR. Food Microbiol 28, 605–10 (2011).
145. De Bellis, P. et al. Toxigenic potential and heat survival of spore-forming bacteria isolated from bread
and ingredients. Int J Food Microbiol 197, 30–9 (2015).
146. Lopez, A.C., Minnaard, J., Perez, P.F. & Alippi, A.M. In vitro interaction between Bacillus megaterium
strains and Caco-2 cells. Int Microbiol 16, 27–33 (2013).
147. Taylor, J.M., Sutherland, A.D., Aidoo, K.E. & Logan, N.A. Heat-stable toxin production by strains of
Bacillus cereus, Bacillus firmus, Bacillus megaterium, Bacillus simplex and Bacillus licheniformis.
FEMS Microbiol Lett 242, 313–7 (2005).
Bacillus 153
148. Krause, N. et al. Performance characteristics of the Duopath(R) cereus enterotoxins assay for rapid
detection of enterotoxinogenic Bacillus cereus strains. Int J Food Microbiol 144, 322–6 (2010).
149. Buchanan, R.L. & Schultz, F.J. Comparison of the Tecra VIA kit, Oxoid BCET-RPLA kit and CHO cell cul-
ture assay for the detection of Bacillus cereus diarrhoeal enterotoxin. Lett Appl Microbiol 19, 353–6 (1994).
150. Day, T.L., Tatani, S.R., Notermans, S. & Bennett, R.W. A comparison of ELISA and RPLA for detection
of Bacillus cereus diarrhoeal enterotoxin. J Appl Bacteriol 77, 9–13 (1994).
151. Beattie, S.H. & Williams, A.G. Detection of toxigenic strains of Bacillus cereus and other Bacillus spp.
with an improved cytotoxicity assay. Lett Appl Microbiol 28, 221–5 (1999).
152. McKillip, J.L. Prevalence and expression of enterotoxins in Bacillus cereus and other Bacillus spp., a
literature review. Antonie Van Leeuwenhoek 77, 393–9 (2000).
153. Tallent, S.M., Hait, J.M. & Bennett, R.W. Analysis of Bacillus cereus toxicity using PCR, ELISA and a
lateral flow device. J Appl Microbiol 118, 1068–75 (2015).
154. Andersson, M.A., Mikkola, R., Helin, J., Andersson, M.C. & Salkinoja-Salonen, M. A novel sensi-
tive bioassay for detection of Bacillus cereus emetic toxin and related depsipeptide ionophores. Appl
Environ Microbiol 64, 1338–43 (1998).
155. Madslien, E.H. et al. Lichenysin is produced by most Bacillus licheniformis strains. J Appl Microbiol
115, 1068–80 (2013).
156. Suominen, I. et al. Toxic Bacillus pumilus from indoor air, recycled paper pulp, Norway spruce, food
poisoning outbreaks and clinical samples. Syst Appl Microbiol 24, 267–76 (2001).
157. Rajkovic, A. et al. Dynamics of boar semen motility inhibition as a semi-quantitative measurement of
Bacillus cereus emetic toxin (cereulide). J Microbiol Methods 65, 525–34 (2006).
158. Garcia-Lara, J., Needham, A.J. & Foster, S.J. Invertebrates as animal models for Staphylococcus aureus
pathogenesis: a window into host-pathogen interaction. FEMS Immunol Med Microbiol 43, 311–23
(2005).
159. Balla, K.M. & Troemel, E.R. Caenorhabditis elegans as a model for intracellular pathogen infection.
Cell Microbiol 15, 1313–22 (2013).
160. Rae, R., Iatsenko, I., Witte, H. & Sommer, R.J. A subset of naturally isolated Bacillus strains show
extreme virulence to the free-living nematodes Caenorhabditis elegans and Pristionchus pacificus.
Environ Microbiol 12, 3007–21 (2010).
161. Anderson, G.L., Caldwell, K.N., Beuchat, L.R. & Williams, P.L. Interaction of a free-living soil nem-
atode, Caenorhabditis elegans, with surrogates of foodborne pathogenic bacteria. J Food Prot 66,
1543–9 (2003).
162. Franks, S.E. et al. Novel role for the yceGH tellurite resistance genes in the pathogenesis of Bacillus
anthracis. Infect Immun 82, 1132–40 (2014).
163. Ramarao, N., Nielsen-Leroux, C. & Lereclus, D. The insect Galleria mellonella as a powerful infection
model to investigate bacterial pathogenesis. J Vis Exp 70, e4392 (2012).
164. Fedhila, S., Daou, N., Lereclus, D. & Nielsen-LeRoux, C. Identification of Bacillus cereus internalin
and other candidate virulence genes specifically induced during oral infection in insects. Mol Microbiol
62, 339–55 (2006).
165. Fedhila, S. et al. Comparative analysis of the virulence of invertebrate and mammalian pathogenic
bacteria in the oral insect infection model Galleria mellonella. J Invertebr Pathol 103, 24–9 (2010).
166. Bouillaut, L. et al. FlhA influences Bacillus thuringiensis PlcR-regulated gene transcription, protein
production, and virulence. Appl Environ Microbiol 71, 8903–10 (2005).
167. Stenfors Arnesen, L., Granum, P.E., Buisson, C., Bohlin, J. & Nielsen-LeRoux, C. Using an insect
model to assess correlation between temperature and virulence in Bacillus weihenstephanensis and
Bacillus cereus. FEMS Microbiol Lett 317, 196–202 (2011).
168. Decousser, J.W. et al. Bacillus cereus and severe intestinal infections in preterm neonates: putative role
of pooled breast milk. Am J Infect Control 41, 918–21 (2013).
169. Cadot, C. et al. InhA1, NprA, and HlyII as candidates for markers to differentiate pathogenic from non-
pathogenic Bacillus cereus strains. J Clin Microbiol 48, 1358–65 (2010).
170. Shinagawa, K. Analytical methods for Bacillus cereus and other Bacillus species. Int J Food Microbiol
10, 125–41 (1990).
171. Singh, D.K., Narayan, K.G. & Gupta, M.K. Mechanisms of Bacillus cereus enteropathy. Indian J Exp
Biol 30, 324–6 (1992).
154 Laboratory Models for Foodborne Infections
172. Glatz, B.A., Spira, W.M. & Goefert, J.M. Alteration of vascular permeability in rabbits by culture fil-
trates of Bacillus cereus and related species. Infect Immun 10(2), 299–303 (1974).
173. Spira, W.M. & Goepfert, J.M. Bacillus cereus-induced fluid accumulation in rabbit ileal loops. Appl
Microbiol 24, 341–8 (1972).
174. Shinagawa, K., Sato, K., Konuma, H., Matsusaka, N. & Sugii, S. Fluid accumulation in mouse ligated
intestine inoculated with the vascular permeability factor produced by Bacillus cereus. J Vet Med Sci
53, 167–71 (1991).
175. Hostacká, A., Kosiarová, A., Majtán, V. & Kohútová, S. Toxic properties of Bacillus cereus strains iso-
lated from different foodstuffs. Zentralbl Bakteriol 276, 303–12 (1992).
176. DeBuono, B.A., Brondum, J., Kramer, J.M., Gilbert, R.J. & Opal, S.M. Plasmid, serotypic, and entero-
toxin analysis of Bacillus cereus in an outbreak setting. J Clin Microbiol 26, 1571–4 (1988).
177. Meijer, A.H. & Spaink, H.P. Host-pathogen interactions made transparent with the zebrafish model.
Curr Drug Targets 12, 1000–17 (2011).
178. Grisolia, C.K. et al. Acute toxicity and cytotoxicity of Bacillus thuringiensis and Bacillus sphaericus
strains on fish and mouse bone marrow. Ecotoxicology 18, 22–6 (2009).
179. Bolcome, R.E., 3rd et al. Anthrax lethal toxin induces cell death-independent permeability in zebrafish
vasculature. Proc Natl Acad Sci USA 105, 2439–44 (2008).
180. Bolcome, R.E., 3rd & Chan, J. Constitutive MEK1 activation rescues anthrax lethal toxin-induced vas-
cular effects in vivo. Infect Immun 78, 5043–53 (2010).
181. Holmes, A.M., Rudd, J.A., Tattersall, F.D., Aziz, Q. & Andrews, P.L. Opportunities for the replacement
of animals in the study of nausea and vomiting. Br J Pharmacol 157, 865–80 (2009).
182. Turnbull, P.C.B. Studies on the production of enterotoxins by Bacillus cereus. J Clin Pathol 29, 941–8
(1976).
183. Melling, J., Capel, B.J., Turnbull, P.C.B. & Gilbert, R.J. Identification of a novel enterotoxigenic activity
associated with Bacillus cereus. J Clin Pathol 29, 938–40 (1976).
184. Turnbull, P.C.B., Kramer, J.M., Jorgensen, K., Gibbert, R.J. & Melling, J. Properties and production
characteristics of vomiting, diarrheal, and necrotizing toxins of Bacillus cereus. Am J Clin Nutr 32,
219–28 (1979).
185. Ueno, S., Matsuki, N. & Saito, H. Suncus murinus: a new experimental model in emesis research. Life
Sci 41, 513–8 (1987).
186. Isobe, M., Ishikawa, T., Suwan, S., Agata, N. & Ohta, M. Synthesis and activity of cereulide, a cyclic
dodecadepsipeptide lonophore as emetic toxin from Bacillus cereus. Bioorg Med Chem Lett 5, 2855–8
(1995).
187. Ueda, S., Nakajima, H., Iwase, M., Shinagawa, K. & Kuwabara, Y. LC-MS analysis of the emetic toxin,
cereulide, produced by Bacillus cereus. Biocontrol Sci 17, 191–5 (2012).
188. Duong, S., Chiaraviglio, L. & Kirby, J.E. Histopathology in a murine model of anthrax. Int J Exp Pathol
87, 131–7 (2006).
189. Xie, T. et al. A new murine model for gastrointestinal anthrax infection. PLoS One 8, e66943 (2013).
190. Lightfoot, Y.L. et al. Colonic immune suppression, barrier dysfunction, and dysbiosis by gastrointesti-
nal Bacillus anthracis infection. PLoS One 9, e100532 (2014).
191. Firoved, A.M. et al. Bacillus anthracis edema toxin causes extensive tissue lesions and rapid lethality in
mice. Am J Pathol 167, 1309–20 (2005).
192. Sun, C. et al. Anthrax lethal toxin disrupts intestinal barrier function and causes systemic infections
with enteric bacteria. PLoS One 7, e33583 (2012).
193. Rolny, I.S., Minnaard, J., Racedo, S.M. & Perez, P.F. Murine model of Bacillus cereus gastrointestinal
infection. J Med Microbiol 63, 1741–9 (2014).
194. Wilcks, A., Hansen, B.M., Hendriksen, N.B. & Licht, T.R. Fate and effect of ingested Bacillus cereus
spores and vegetative cells in the intestinal tract of human-flora-associated rats. FEMS Immunol Med
Microbiol 46, 70–7 (2006).
195. Cutting, S.M. Bacillus probiotics. Food Microbiol 28, 214–20 (2011).
9
Clostridium
Emilio Aranda, María G. Córdoba, María J. Benito, and Juan José Córdoba
CONTENTS
9.1 Introduction: Clostridium Neurotoxins and Enterotoxins.............................................................155
9.2 Characteristics and Incidence of Foodborne Outbreaks due to Clostridium Neurotoxins
and Enterotoxins........................................................................................................................... 156
9.3 Investigation of Foodborne Intoxication by Clostridium Neurotoxins......................................... 157
9.3.1 Investigation of C. botulinum and BoNTs in Food Samples............................................ 157
9.3.2 Investigation of C. perfringens in Food Samples............................................................. 159
9.4 Laboratory Models for Foodborne Clostridium Intoxication....................................................... 160
9.4.1 Animal Models for Foodborne BoNT.............................................................................. 160
9.4.2 Animal Models for Foodborne C. perfringens Enterotoxins............................................162
9.4.3 Animal Models for Foodborne C. difficile....................................................................... 164
9.5 Cell Culture Models.......................................................................................................................165
9.6 Conclusions................................................................................................................................... 166
Acknowledgments....................................................................................................................................167
References................................................................................................................................................167
155
156 Laboratory Models for Foodborne Infections
Other members of the genus Clostridium (Clostridium baratii and Clostridium butyricum) have occa-
sionally been reported to be involved in foodborne botulism and thus should also be considered as
potentially foodborne pathogens.
Furthermore, C. perfringens, which produces enterotoxins and is responsible for foodborne intoxica-
tion,7 and Clostridium difficile, which is involved in nosocomial, or institution-acquired, diarrhea,8 must
be considered as potentially pathogenic clostridia associated to foodborne outbreaks.
C. perfringens produces clostridial toxins activated in the gastrointestinal tract. Among 17 toxins
produced by C. perfringens, alpha (CPA), beta (CPB), epsilon (ETX), and iota (ITX) toxins are the four
major toxins present in five different toxinotypes (A, B, C, D, and E) of the bacterium.9–11 In addition,
C. perfringens may produce other toxins such as C. perfringens enterotoxin (CPE), C. perfringens beta2
toxin (CPB2), C. perfringens NetB toxin, and TpeL.9,12–14 CPE causes C. perfringens type A food poison-
ing as well as antibiotic-associated diarrhea (AAD) and sporadic diarrheas in humans. This toxin is also
responsible for enteric diseases and enterotoxemias in animals.15,16
Accurate methods for isolating and detecting these pathogenic clostridia in raw materials and pro-
cessed foods are necessary to take corrective measures throughout processing, avoiding clostridial food-
borne cases. In addition, laboratory (animal or cell culture) should be available to investigate these types
of foodborne cases.
each year), Argentina, Australia, Canada, Italy, and Japan have reported most of the cases.23 IB is a rare
disorder in Europe. From 1976, the year in which IB was first recognized,24 through 2006, 65 cases were
identified in 13 European countries, most of them in Italy and Spain, followed by the UK.25 In Europe,
IB is less common than foodborne botulism; however, IB is the most common form of human botulism
recognized in the United States.25
Foodborne illness caused by CPEs commonly occurs in industrialized countries.7 This food
poisoning is the second most common bacterial illness in the USA, where about 1 million cases/
year are reported.26 Outbreaks typically involve a large number of victims and are associated with
temperature-abused meat or poultry dishes. The high incidence of C. perfringens type A food poison-
ing is well documented. From 1998 to 2010, 289 confirmed outbreaks of C. perfringens illness were
reported in the United States with 15,208 illnesses, 83 hospitalizations, and eight deaths.27 Sudden
infant death syndrome (SIDS) accounts for unexpected deaths in infants under the age of 1 year.
Strains of C. perfringens type A have been discovered to be commonly present in the intestines of
babies dying with SIDS.28
C. difficile belongs to the normal microbiota of the mammalian gastrointestinal tract. C. difficile prolif-
eration and infection in the human colon often occur after use of broad-spectrum antibiotics. C. difficile
destroys the intestinal lining and causes AAD, colitis, and pseudomembranous colitis. The pathogenicity
depends on the production of enterotoxin A and cytotoxin B. C. difficile toxin A is an enterotoxin that
induces fluid accumulation in the bowel, and toxin B is a cytopathic toxin that is extremely lethal. Both
toxins are highly unstable and tend to degrade at room temperature.10
To identify C. botulinum, unselective plating media such as blood agar and egg yolk agar (EYA)36 are
commonly used, since it could enable typical lipase production by this microorganism. However, other
clostridia species have lipase and may therefore confuse the identification.37 EYA medium alone does
not contain any inhibitory compounds, but it has been reported that medium supplemented with cyclo-
serine, sulfamethoxazole, and trimethoprim can be used for identifying select group I C. botulinum.38,39
Selection of the correct incubation temperature is essential to differentiate strains from the different
physiologies of groups I and II of Clostridium. Group I strains grow optimally at 35°C–37°C, whereas
temperatures of 25°C–30°C or even lower favor growth of group II strains.40
Quantification of C. botulinum by use of plate count procedures in samples containing other bac-
teria is difficult, since prevalence of C. botulinum in naturally contaminated samples is generally low
(10–1000 spores/kg) and proper selective media are not available.29
Polymerase chain reaction (PCR) represents an alternative to the traditional microbiological culture
technique to detect C. botulinum in foods. Fakruddin et al.41 described improvements and alternatives to
PCR such as loop-mediated isothermal amplification (LAMP), nucleic acid sequence based amplification
(NASBA), self-sustained sequence replication (3SR), and rolling circle amplification (RCA).
PCR detection of C. botulinum often targets the BoNT genes although other types of toxin genes are
also of diagnostic value.29 Use of multiplex PCR assays enables the simultaneous detection of two or more
types of BoNT genes.42–44 One such multiplex method was able to discriminate among BoNT serotypes
A, B, E, and F, corroborating mouse bioassay results.45 Furthermore, Peck and colleagues1 developed cul-
ture enrichment methods that when coupled with multiplex PCR can identify strains of C. botulinum that
are nonproteolytic (BoNT serotypes B, E, and F).1 However, PCR detection of neurotoxin genes does not
provide details on the physiological group and epidemiology of C. botulinum isolates. For differentiation
between group I (proteolytic) and group II (nonproteolytic) strains of C. botulinum, several molecular
typing methods are useful.46–48 Furthermore, Janda and Whitehouse49 have developed a multilocus PCR
followed by electrospray ionization mass spectrometry to identify C. botulinum, and Fach et al.50 have
described an innovative molecular detection tool based on the GeneDisc cycler for tracking all types
A, B, E, and F. Detection of C. botulinum in foods by the above mentioned traditional culture techniques
are highly time consuming and require good microbiological expertise. PCR offers the possibility to
detect any of the toxic strains regardless of the type of C. botulinum and BoNT produced. This strategy
is possible due to the use of universal primers that recognize all BoNT genes, but the nucleotide diversity
among the BoNT genes may present a potential problem.51 Nonetheless, BoNTs are generated as part of a
progenitor toxin complex, and a conserved component among serotypes is the nontoxic nonhemaglutinin
(NTNH). East and Collins52 demonstrated that the gene encoding NTNH shows a high level of similarity
and is present in all strains that produce BoNTs and absent from strains that are nontoxic. Aranda et al.53
developed a PCR protocol to detect all BoNTs-producing strains by using a single set of degenerate
primers. This protocol yields a single PCR product of 1.1 kb in agarose gel, providing a more specific
detection than hybridization with a BoNT probe.
Real-time PCR, biosensors, and DNA microarray offer the possibility of continuous and real-time
monitoring of the environment for the presence of infectious agents. Therefore, several real-time PCR
protocols54 along with biosensor technology55 and DNA microarrays56 have been reported to detect
Clostridium. More recently, Fenicia et al.57 proposed a real-time PCR method for detecting and typing
BoNT-producing C. botulinum types A, B, E, and F in clinical, food, and environmental samples and
thus support use of it as an international standard method. Microarray technology for toxin identifi-
cation of contaminated food has not been widely used. This may be due to the challenge in isolating
high-quality RNA samples from clostridia in food matrices. A recent oligonucleotide microarray with
62 different sequences based on known strain variable regions in the genome of C. botulinum strain
ATCC 3502 was constructed and used to differentiate different C. botulinum type A strains.58 Regions
corresponding to BoNT genes of various serotypes, and other markers were observed. Further develop-
ment of microarray-based assay approaches may provide a means to rapidly identify toxin-producing
strains.
In most of the cases of foodborne botulism investigation, detection of C. botulinum should be comple-
mented by detection of BoNT toxins. For BoNT detection, several procedures such as enzyme-linked
immunosorbent assay (ELISA)59,60 or mass spectrometric-based endopeptidase methods61 have been
Clostridium 159
reported. This approach has been successful in identifying BoNT serotypes A, B, E, and F in a variety of
food and clinical sample matrices with submouse bioassay sensitivities. To advance this technique even
further, a single, high-affinity mAb (4E17.1) that can simultaneously identify BoNT serotypes A, B, E,
and F has been developed.62 Recently, surface plasmon resonance (SPR) sensors have been also applied
for detecting BoNTs serotypes A, B, and F.63,64 This method uses a label-free biosensor assay for BoNT
B detection in food and human serum based on protein chip assay. Lastly, Ching et al.65 have reported
the use of an immunochromatographic test strip for the detection of BoNT serotypes A and B and Liu
et al.66 have developed an immunobiochemical assay to detect BoNT. In addition, ELISA and Endopep
MS analyses using fluorogenic reporters to detect BoNT in drinking water have been reported.67,68
Several protocols such as ELISA and immuno-PCR and ELISA based on protein antibody microarray
have been recently reported to detect BoNT neurotoxins A, B, C, D, E, and F in complex clinical, food,
and environmental samples at higher sensitivity than mouse bioassay.69,70 A number of rapid affinity
immunochromatography column (AICC) assays for the detection of BoNT serotypes A, B, E, and F in
food matrices have been developed. Brunt et al.71 reported a detection limit for BoNT/A of 0.5 ng, two-
fold more sensitive than earlier reported lateral flow methods. For serotypes B, E, and F, the minimum
detection limit ranged from 5 to 50 ng. Although not as sensitive as ELISA or mouse bioassays, immuno-
chromatographic methods generally are rapid assays, requiring only 15–30 min to complete, and do not
require enrichment steps, making them highly amenable to use in the field.
reported for the detection of six toxic genes of C. perfringens.89,90 Recently, Chon et al.91 developed an
accurate real-time PCR for detection and quantification of C. perfringens in foods.
Given that not all C. perfringens produce toxins and consequently not all strains are related to dis-
eases or cause food poisonings, the confirmation of toxin productions is the most accepted criterion in
conventional diagnosis.92 Among the techniques for detection of major toxins of C. perfringens are the
mouse neutralization test (MNT),92 ELISAs,93–95 counter-immunoelectrophoresis,96 and latex agglutina-
tion test.97,98 Lastly, Seyer et al.99 reported a method to detect C. perfringens toxins in complex foods and
biological matrixes by immunopurification and ultra performance liquid chromatography-tandem mass
spectrometry.
The above methods give information about the presence of pathogenic Clostridium species and/
or the corresponding toxins in foods. However, no information is supplied about the effect of these
microorganisms in live cell that it is required in most of the cases during clostridial foodborne investi-
gation. Bioassays using laboratory animal or cells culture methods report data on effects of neurotoxin
or enterotoxins in live organisms. They are based on the ability of suspected food extracts to induce
symptoms in laboratory animal models and/or superantigenic action in cell culture models. These
methods offer a very valuable alternative for foodborne clostridia investigation and will be explored in
a separate section.
TABLE 9.1
Animal Models Used for the Detection of Clostridium Neurotoxins and Enterotoxins
Animal Studies Toxins Tested References
Mouse Mouse neutralization bioassay— BoNT A, B, C, D, E, and F 25,60
binding, translocation, and enzymatic
activity of neurotoxins
Inactivation/neutralization effect of BoNT A, B, C, D, E, and F 102–109,111,115–117
botulinum toxin
Neuroimmunological changes BoNT A 114
Changes of ECG pattern of CPE 121
hyperpotassemia, fall of blood
pressure, and transient hyperpnea and
respiratory depression of
C. perfringens enterotoxins and death
Mucosal necrosis of small intestinal CPE 122
loops, without accumulation of fluid
in the lumen
Absorption of CPE from the intestine CPE 122,123
and formation of CH-1-like
complexes in the liver and kidney
Hemorrhagic cecitis C. difficile C. difficile Enterotoxin A and B 148–152
enterotoxins
Rat Developing, maintaining, and BoNT A 110
recovering from neuropathic pain
Neuroimmunological changes BoNT A 108,112,113
Changes of ECG pattern of CPE C. perfringens Enterotoxin 121
hyperpotassemia, fall of blood Type A
pressure, and transient hyperpnea and
respiratory depression caused by
C. perfringens enterotoxins and death
121 Rabbit Fluid and electrolyte secretion and CPE C. perfringens Enterotoxin 119,125,128
changes to the mucosa consist mainly Type A
of mucosal necrosis and hemorrhage
in small intestinal and colonic mucosa
Toxin binds to liver and kidney CPE C. perfringens Enterotoxin 122,123
Type A
Hemorrhagic lesions and luminal fluid C. perfringens Enterotoxin CPB 130
accumulation in these intestinal loops and CPE
Hamster Hemorrhagic cecitis C. difficile C. difficile Enterotoxin A and B 146,147
enterotoxins
Piglets, goats Endothelial damage of intestinal loops CPB in C. perfringens type C 134,135
Primates Developed vomiting and diarrhea CPE C. perfringens Enterotoxin 140
C. perfringens enterotoxins type A
Wohlfarth et al.103 reported that oral poisoning with BoNT serotype B was capable of activating the
immune system. These authors studied two cases of foodborne botulism following a meal consisting of
homemade canned beans. Four months later, one person developed a moderate and the other a borderline
titer of serum antibodies to BoNT serotype B, detected using an ex vivo assay (mouse phrenic-nerve
hemidiaphragm assay).
Moreover, bioassays in mice are used not only to confirm the type of C. botulinum, but also to investigate of
substances that neutralize the effect of Botulinum toxin. Examples in recent years are studies that use animal
models to find a small molecular weight (SMW) inhibitor of BoNTs that will neutralize the toxin. There have
been numerous publications reporting finding SMW inhibitors to BoNTA,104–106 but few reports on SMW
162 Laboratory Models for Foodborne Infections
inhibitors to BoNTB, one report on BoNTE,107 and none on BoNTF. Recently, Pirazzini et al.108 reported on
several inhibitors that were effective against BoNTA, BoNTC, and BoNTE, but Montgomery et al.109 found
an SMW inhibitor that might be useful against multiple serotypes of BoNT (Table 9.1). They tested two
promising SMW inhibitors to BoNTA against BoNTs B, C, E, and F in mice phrenic nerve-hemidiaphragm.
Although most studies are performed in mice, to have a confirmation in different species, additional
experiments must be carried out in other animal models, where it is also easier to verify the toxin effects.
Thus, Marinelli et al.110 studied in rats the effects of BoNT/A in developing, maintaining, and recover-
ing from neuropathic pain induced by the ligature of the sciatic nerve. They saw this effect was evident
starting 24 h after the administration of BoNT/A, and it was long-lasting—present 81 or 25 days after
intraplantar injection of the higher dose in mice (15 pg/paw) and 35 days after injection in rats (75 pg/rat).
Moreover, BoNT/A-injected mice showed a quicker recovery of the walking pattern and weight bearing
compared to control groups.
Botulinum toxin type A used in the treatment of strabismus and other human diseases characterized
by hyperactivity of peripheral cholinergic nerve terminals, it could get some patients are or become
resistant to it. This can be overcome by using other botulinum toxins. Eleopra et al.111 investigated the
action of BoNT type D in mouse and human muscles and showed that botulinum toxin type D is poorly
effective in inducing human skeletal muscle paralysis but is very potent in mice. Mika et al.112 studied the
effect of BoNT A on sciatic-nerve-injury-induced neuroimmunological changes in rats and found evi-
dence that BoNT/A impedes injury-activated neuronal function in structures distant from the injection
site using doses based on an approximate equivalence of 100 U of botox (4.8 ng tox) as reported by Cui
et al.,113 the dose injected in this experiment was 75 pg per paw, which corresponded to about 4–5 U/kg
in rats weighing 300–350 g. Also, Kato et al.,114 using 8 weeks old male animals, showed that BoNT A2
reduces incidence of seizures in mouse models of temporal lobe epilepsy.
Pirazzini et al.108 studied a Thioredoxin Reductase-thioredoxin Redox System cleaves the interchain
disulfide bond of BoNTs on the cytosolic surface of synaptic rat vesicles which blocked the differ-
ent BoNTs tested within a very similar concentration range, suggesting that the interchain disulfide
reduction is closely similar for different BoNTs serotypes. These in vitro results were validated in vivo
using digit abduction score (DAS) assay, a well-established model to compare potency and duration
of BoNTs.115 All tested molecules were very effective in reducing the degree and duration of paralysis
induced by the local injection of BoNT/A and BoNT/C. For the first time, it was shown that small mol-
ecules effectively prevent the paralytic activity of BoNTs. As a proof of concept, they also tested one
of these inhibitors, Ebselen, a compound reported to target both TrxR and Trx,116 in the lethality assay.
Ebselen, preventively administered via intraperitoneal injection, was very effective in protecting animals
from a lethal amount of BoNT/A, both by prolongation of the time to death and by reduction of the num-
ber of deaths108 (Table 9.1).
In another report, Pellet et al.117 analyzed in vivo the onset and duration of action of BoNT/A1–5 in female
ICR mice (Harlan) that were injected with sublethal amounts of BoNT/A1,/A2,/A3,/A4, or/A5 in 10 μL of
GelPhos buffer [30 mM sodium phosphate (pH 6.3) and 0.2% gelatin] into the right gastrocnemius muscle.
The amount of toxin injected IM per dilution was confirmed by mouse bioassay. Analyses of several of these
studied subtypes revealed distinct characteristics, ranging from differences in cell entry and enzyme kinetics
to differences in potency in mice and cell-model-specific potency. In a long-term activity study in cultured
primary neurons, it was indicated that BoNT/A1, 2, 4, and 5 have a similar duration of action, whereas
BoNT/A3 has a significantly shorter duration of action. This report describes an in vivo mouse study, show-
ing that local injection of BoNT/A2 resulted in faster onset of local paralysis than BoNT/A1, 3, 4, and 5,
whereas BoNT/A3 resulted in significantly faster recovery of motor-neuron deficiency.
transient hyperpnea followed by respiratory depression were observed. Analysis of plasma levels of
rations revealed hyperpotassemia in both animal species. On the other hand, enterotoxin (up to 100 µg)
showed little direct cardiotoxicity on the isolated heart.121 The in vivo effects of CPE have also been stud-
ied in rats,121 although much less extensively than in mice and rabbits. In rats, as in mice, this toxin pro-
duces lethality when injected i.v., and death in rats is preceded by respiratory difficulty, ECG alterations,
and hyperkalemia, because these effects were accompanied by an increase in liver enzymes (GPT, GOT,
and LDH). Sugimoto et al.121 suggested that CPE-induced hyperkalemia was a consequence of cytotoxic
action of CPE on hepatocytes. Mice have also been used to study the intestinal and systemic effects of
CPE.122 In these animals, lethality was associated with a rapid fall in blood pressure, respiratory dif-
ficulty, and changes in ECG. In the small intestinal loops of mice, as in rabbits, CPE causes dose- and
time-dependent mucosal necrosis. However, toxin administered in this manner does not cause fluid accu-
mulation in mice.122 Results of experiments inoculating CPE into intestinal loops of mice suggest that
death observed in constipated human patients with CPE-positive C. perfringens type A infection123 could
have been a consequence of absorption of CPE from the intestine. Mice that were inoculated with CPE in
the intestinal loops showed that CPE bound to and formed CH-1-like complexes in the liver and kidney.
A mouse intravenous lethality model was used to demonstrate that CPB is the main factor responsible for
systemic lethality in type C culture supernatants.124 In that study, lethality was abolished when culture
supernatants were preincubated with a CPB monoclonal antibody, but not when the cultures were incu-
bated with a CPA monoclonal antibody, which confirmed the role of CPB in mouse lethality.124
Furthermore, for many years, rabbit intestinal loops have been used, and are still used today, to study
the effects of CPE in vivo.125 Most of these rabbit models have been followed to evaluate the effect of CPE
in the small intestine, where it causes fluid and electrolyte secretion and produces significant damage to
the mucosa of the jejunum and ileum, but less damage in the duodenum.125–127
García et al.119 have also demonstrated that rabbit colon is sensitive to the action of purified CPE, with
both fluid secretion and mucosal damage observed. The histological changes caused by CPE in both
small intestinal and colonic loops of rabbits consist mainly of mucosal necrosis and hemorrhage. These
changes are both time- and dose-dependent.119,125
Rabbits have also been used to study the binding of CPE to extraintestinal tissues, which led to the
demonstration that this toxin binds to liver and kidney.122,123 This finding suggests that CPE absorbed
from the intestine can be responsible for systemic alterations, which may help explain the lethality
observed in some cases of experimental animals and human patients with the disease.122,123
A rabbit intestinal loop model was also used to determine the spatial distribution of the effects of CPB
along the alimentary canal.128 In that study, fluid accumulation and necrotizing enteritis were observed
only in the small intestine, with the jejunum and ileum being most severely affected. This result is in
agreement with natural type C disease in animals and humans, in which the jejunum and ileum are pri-
marily affected.73,129
Synergism between CPB and CPE for the virulence of CPE-positive type C strains of C. perfringens
was demonstrated by using a rabbit-ligated intestinal loop model,130 inducing significant hemorrhagic
lesions and luminal fluid accumulation in these intestinal loops. However, when lysate supernatants of
the cpb or cpe knockout mutants of these strains were inoculated into rabbit-ligated intestinal loops, no
significant damage or fluid accumulation were observed. Complementing the cpe mutant, or reversing
the cpb mutation, restored the virulent effects of culture lysates. Purified CPB and CPE, inoculated
together at concentrations similar to those found in wild-type CN3758 culture lysates, also produced
lesions and fluid accumulation in rabbit intestines. However, when either of these toxins was inoculated
independently, only higher doses caused damage to the intestine, suggesting that at low concentration,
both toxins act synergistically in the intestine.130 These experiments provided the first evidence of syner-
gistic toxin activity during intestinal C. perfringens infections.
Large animal models have also been used to study the pathogenesis of C. perfringens type C dis-
ease. Initially, piglets were experimentally used for this purpose.131–133 However, those experiments
were performed using whole cultures or crude culture supernatants, and although the results confirmed
that C. perfringens type C is a pathogen for piglets, they did not identify the main virulence factor(s)
involved in the pathogenesis of those infections. The mechanism of action of CPB in type C infection
was recently studied in intestinal loops of piglets.134 This study indicated that there is a tropism of CPB
164 Laboratory Models for Foodborne Infections
toward endothelial cells, suggesting that endothelial damage induced by CPB plays a role in the early
stages of C. perfringens type C enteritis in pigs. Koch’s postulates for type C disease were fulfilled in
goats,135 using the same set of C. perfringens type C mutants previously used to fulfill those postulates
in rabbits and mice.118,136 The results of the experiments in goats confirmed, this time in a natural host of
the disease, the key role of CPB in the pathogenesis of natural C. perfringens type C disease.
The effects of i.v. administration of extracts of sporulating cultures of CPE-positive C. perfringens
type A, and of CPE into ligated loops, also have been studied in calves and lambs, respectively.137–139 The
results of these experiments were variable and included diarrhea in calves and mild mucosal changes in
the intestinal loops of lambs.
Nonhuman primates have rarely been used to study the pathogenesis of CPE intoxication and/or
enterotoxigenic C. perfringens type A-associated disease.140 In the only published study, cynomolgus
monkeys fed purified CPE developed vomiting and diarrhea, while monkeys given CPE-positive C. per-
fringens type A orally developed only diarrhea. These effects were only observed when either CPE or
CPE-positive C. perfringens type A were given together with sodium bicarbonate to neutralize the low
gastric pH. Lethality was not observed in these nonhuman primates.140
TABLE 9.2
Cell Culture Models Used for the Studies of Clostridium Neurotoxins and Enterotoxins
Cell Culture Studies Toxins Tested References
Rat spinal cord cells 153,163
Chick embryo neuronal cell Receptor binding, BoNT A, B, C, D, E, and F 154
translocation, and enzymatic
activity of neurotoxins
Neuroblastoma cells N2A 155
BE(2)-M17 cells 156
Murine embryonic stem cells BoNT A, B, and E 157
Rat spinal cord cells 153
Neuro-2a, SK-N-SH, M17, Sensitivity to BoNT BoNT A 158,159
SHSY5Y, PC12 NT2
Neuroblastoma cells NG108–15 161
Rodent ganglia cell 162
Cortical neurons
Human neuronal cell
Human intestinal epithelial Cytotoxicity of C. perfringens C. perfringens Enterotoxin 165–167
Caco-2 cells enterotoxins type A
Human colonic epithelial cell Cytotoxicity of C. difficile C. difficile Enterotoxin A and B 169,170
lines Caco-2, T84, HT-29 enterotoxins
Human monocytic THP-1 cells Inflammatory cytokine release C. difficile Enterotoxin A and B 168,171–173
stimulation of C. difficile
enterotoxins
166 Laboratory Models for Foodborne Infections
BoNT/A1 sensitivity comparable with rat SCN, but another human cell model, the SiMa cells, had the
lowest BoNT/A1 sensitivity of all the models tested.
Delaflotte et al.163 developed a procedure to isolate, enrich, and culture spinal motor neurons from rat
embryonic spinal cords and to assess sensitivity to BoNT/A1 (Table 9.2). This model obtained a highly
enriched culture derived from neonatal rat ventral spinal cord and provides a physiologically relevant
model system to understand the biology and characteristics of BoNTs acting at cholinergic terminals.
These cultures’ exhibited high sensitivity an EC50 (median effective concentration) below 0.5 pM to
BoNT/A1, which is more than five times the sensitivity measured in the standard rat spinal cord neuron
cultures and could be an ideal model to assess differences between BoNT serotypes and subtypes.
Regarding C. perfringens foodborne investigation, cell culture assays are possible alternatives to
replace in vivo neutralization tests. Animal experiments still play a central role in this kind of quality
control test; however, potency tests such as clostridial toxoids require a very high number of animals for
quality control testing.164 In this sense, Allaart et al.165 demonstrated that CPE expression is necessary
and sufficient for C. perfringens strains SM101 and F4969 to cause fluid accumulation and GI damage
in a rabbit ileal loop model; however, it remains unclear whether CPE is indispensable for bacterial cyto-
toxicity in vitro (Table 9.2).
The significance of toxins in the induction of in vitro cytotoxicity has been investigated using human
intestinal epithelial Caco-2 cells infected with toxin-gene-harboring C. perfringens strains and their
mutants or anti-toxin antibody.165,166 These authors revealed that β2 toxin is not involved in Caco-2 cell
cytotoxicity during infection with a cpb2-harboring C. perfringens strain. Yasugi et al.167 examined the
cytotoxicity of cpe-harboring C. perfringens isolates cocultured with human intestinal epithelial Caco-2
cells. The food poisoning strains showed severe cytotoxicity during sporulation and CPE production,
but not during vegetative cell growth. While Caco-2 cells were intact during coculturing with cpe-null
mutant derivative of strain SM101 (a food poisoning strain carrying a chromosomal cpe gene), the wild-
type level cytotoxicity was observed with cpe-complemented strain. In contrast, both wild-type and
cpe-null mutant derivative of the non-foodborne strain F4969 induced Caco-2 cell death during both
vegetative and sporulation growth. The Caco-2 cell cytotoxicity caused by C. perfringens strain SM101
is considered to be exclusively dependent on CPE production.
Regarding C. difficile investigation, the human colonic epithelial cell lines Caco-2, T84, HT-29, and
human monocytic THP-1 cells have been used to study direct effects on intestinal cells168–170 (Table 9.2).
In human intestinal epithelial cell line, C. difficile enterotoxins initiate colonic inflammation in humans
by injuring epithelial cells and inducing production of IL8.169 Detachment of these cells from the base-
ment membrane leads to cell death by apoptosis. IL8 produced by the injured epithelial cells, as well as
responses by the exposed lamina propria cells (especially macrophages that lie just below the basement
membrane 43), to toxins A and B and other luminal contents would initiate a cascade of events character-
ized by migration of circulating polymorphonuclear cells into the mucosa to mediate tissue damage and
induce diarrhea. Most interestingly, colonic epithelial cells from some adults may be resistant to the effects
of even high concentrations of toxin A, possibly explaining why some people are resistant to disease.
In cells of the monocyte lineage, C. difficile toxins A and B stimulate inflammatory cytokine release,
including TNF-α, IL-1β, IL-6, and IL-8,171–173 and induce necrosis in human monocytes and in THP-1
human monocytic cells168 (Table 9.2).
Toxins A and B share 63% amino acid homology174 and possess similar domains. These toxins were
shown to block small GTP-binding proteins. In monocytes, toxins induce IL-8 production and necrosis
by unknown mechanisms. When these methods are used in foodborne investigation, confirmation of
toxin production is necessary.
9.6 Conclusions
Being the most widely distributed pathogenic species within the Clostridium genus, C. botulinum,
C. perfringens, and C. difficile are frequently involved in foodborne outbreaks. Accurate methods for
isolating and detecting these pathogenic clostridia in raw material and processed foods are necessary to
take corrective measures throughout food processing that reduce potential clostridial cases. For isolation,
Clostridium 167
detection, and characterization of C. botulinum, C. perfringens, and C. difficile from food samples, tradi-
tional microbiological culture techniques and molecular methods (PCR and qPCR) are available. When
these methods are used in foodborne investigation, confirmation of toxin production by rapid methods
such as ELISA, immuno-PCR, or ELISA based on protein antibody microarray is important. While these
methods reveal the presence of pathogenic Clostridium species and/or the corresponding toxins in food,
no information is available about the effect of these microorganisms in live cells, which is required in
most of the cases during clostridial foodborne investigation. Bioassays using laboratory animal or cells
culture methods help determine the effects of neurotoxin or enterotoxins in live organisms. Based on
the ability of a suspected food extracts to induce symptoms in laboratory animal models and/or supe-
rantigenic action in cell culture models, these methods offer a very valuable alternative for foodborne
clostridia investigation. The mouse neutralization bioassay, using either monovalent, toxin-type-specific
botulinum antitoxin or polyvalent antibotulinum antitoxin is the most valuable method for foodborne
botulism investigation. Rabbit intestinal loops, rat, and mice laboratory models are available for C. per-
fringens foodborne investigation. For C. difficile investigation, hamster, mouse, and pigs may be utilized.
In addition, cell lines provide an alternative to laboratory animal models for foodborne clostridial cases
investigation. For example, rat spinal cord, chick embryo neuronal, and neuroblastoma cell lines are avail-
able for accurate investigation of foodborne botulism. For the study of pathogenic C. perfringens and C.
difficile, human intestinal epithelial lines such as Caco-2 cells have been reported as sensitive method.
Acknowledgments
This work has been funded by the Spanish Instituto Nacional de Investigación Agraria y Agroalimentaria
(INIA) and the Spanish Comisión Interministerial de Ciencia y Tecnología with the projects RTA-2013-
00070-C03-03 and Carnisenusa CSD2007-00016, Consolider Ingenio 2010, and GRU09162 of the Junta
de Extremadura and FEDER.
REFERENCES
1. Peck, M.W. et al. Development and application of a new method for specific and sensitive enumeration
of spores of nonproteolytic Clostridium botulinum types B, E, and F in foods and food materials. Appl.
Environ. Microbiol., 76, 6607–14, 2010.
2. Carter, A.T. and Peck, M.W. Genomes, neurotoxins and biology of Clostridium botulinum group I and
group II. Res. Microbiol., 166, 303–17, 2015.
3. Barash, J.R. and Arnon, S.S. A novel strain of Clostridium botulinum that produces type B and type H
botulinum toxins. J. Infect. Dis., 209, 183–91, 2014.
4. Johnson, E.A. Validity of botulinum neurotoxin serotype H. J. Infect. Dis., 210, 992–3, 2014.
5. Dover, N., Barash, J.R., Hill, K.K., Xie, G. and Arnon, S.S. Molecular characterization of a novel botu-
linum neurotoxin type H gene. J. Infect. Dis., 209, 192–202, 2014.
6. Carter, A.T., Stringer, S.C., Webb, M.D. and Peck, M.W. The type F6 neurotoxin gene cluster locus of
Group II Clostridium botulinum has evolved by successive disruption of two different ancestral precur-
sors. Gen. Biol. Evol., 5, 1032–7, 2013.
7. Lindström, M. et al. Novel insights into the epidemiology of Clostridium perfringens type A food poi-
soning. Food Microbiol., 28, 192, 2011.
8. Gillers, S. et al. Microscale sample preparation for PCR of C. difficile infected stool. J. Microbiol.
Methods, 78, 203, 2009.
9. Li, J. et al. Toxin plasmids of Clostridium perfringens. Microbiol. Mol. Biol. Rev., 77, 208–33, 2013.
10. Heikinheimo, A., Lindstrom, M., Liu, D. and Korkeala, H. Clostridium. In: Molecular Detection of
Foodborne Pathogens, Ch. 11, Liu, D. (Ed.), CRC Press, Taylor & Francis Group, LLC, Boca Raton,
FL, 2009.
11. Hatheway, C.L. Toxigenic clostridia. Clin. Microbiol. Rev., 3, 66, 1990.
12. McClane, B.A. et al. The enterotoxic clostridia. In: The Prokaryotes, Dworkin, M. et al. (Eds.), Springer,
New York, pp. 688–752, 2006.
168 Laboratory Models for Foodborne Infections
13. Keyburn, A.L. et al. NetB, a pore-forming toxin from necrotic enteritis strains of Clostridium perfrin-
gens. Toxins, 2, 1913, 2008.
14. Smedley, J.G., Fisher, D.J., Sayeed, S., Chakrabarti, G. and McClane, B.A. The enteric toxins of
Clostridium perfringens. Rev. Physiol. Biochem. Pharmacol., 152, 183–204, 2004.
15. Songer, J.G. and Uzal, F.A. Clostridial enteric infections in pigs. J. Vet. Diagn. Invest., 17, 528, 2005.
16. McClane, B.A. and Rood, J.I. Clostridial toxins involved in human enteric and histotoxic infections. In:
Clostridia: Biotechnology and Medical Applications, Bahl, H. and Dürre, P. (Eds.), Wiley-VCH Verlag
GmbH, Weinheim, 2001.
17. Center for Disease Control and Prevention (CDC). National Botulism Surveillance, last updated May 3,
2016, accessed June 30, 2016, http://www.cdc.gov/botulism/surveillance.html.
18. Food and Drug Administration (FDA). Bad Bug Book: Foodborne Pathogenic Microorganisms and
Natural Toxins, 2nd ed., US Food and Drug Administration, Washington, DC, p. 108, 2012.
19. Aston, S. and Beeching, N. Botulism. In: Hunters Tropical Medicine and Emerging Infectious Diseases,
Magill, A., Ryan, E.T., Solomon, T. and Hill, D. (Eds.), 9th ed., vol. 1, Saunders, Philadelphia, PA,
pp. 511–3, 2012.
20. Therre, H. Botulism in the European Union. Euro Surveill., 4, 2, 1999.
21. Barash, J.R., Hsia, J.K. and Arnon, S.S. Presence of soil-dwelling clostridia in commercial powdered
infant formulas. J. Pediatr., 156, 402–8, 2010.
22. Arnon, S.S., Schechter, R., Maslanka, S.E., Jewell, N.P. and Hatheway, C.L. Human botulism immune
globulin for the treatment of infant botulism. N. Engl. J. Med., 354, 462–71, 2006.
23. Koepke, R., Sobel, J. and Arnon, S.S. Global occurrence of infant botulism, 1976–2006. Pediatrics,
122, 73–82, 2008.
24. Midura, T.F. and Arnon, S.S. Infant botulism: Identification of Clostridium botulinum and its toxins in
faeces. Lancet, 2, 934–6, 1976.
25. López-Laso, E. et al. Infant botulism in Andalusia (southern Spain). Eur. J. Paediatr. Neurol., 18, 321–
6, 2014.
26. Scallan, E. et al. Foodborne illness acquired in the United States—major pathogens. Emerg. Infect. Dis.,
17, 7–15, 2011.
27. Grass, J.E., Gould, L.H. and Mahon, B.E. Epidemiology of foodborne disease outbreaks caused by
Clostridium perfringens, United States, 1998–2010. Foodborne Pathog. Dis., 10, 131–6, 2013, doi:
10.1089/fpd.2012.1316..
28. Mage, D.T., Cohen, M. and Donner, M. Sudden infant death syndrome. N. Engl. J. Med., 24, 2581, 2009.
29. Lindström, M. and Korkeala, H. Laboratory diagnostics of botulism. Clin. Microbiol. Rev., 19, 298, 2006.
30. Reddy, N.R., Tetzloff, R.C. and Skinner, G.E. Effect of media, additives, and incubation conditions on the
recovery of high pressure and heat-injured Clostridium botulinum spores. Food Microbiol., 27, 613, 2010.
31. Mato Rodriguez, L. and Alatossava, T. Effects of copper on germination, growth and sporulation of
Clostridium tyrobutyricum. Food Microbiol., 27, 434, 2010.
32. Lenz, C. A. and Vogel, R.F. Effect of sporulation medium and its divalent cation content on the heat and
high pressure resistance of Clostridium botulinum type E spores. Food Microbiol., 44, 156–67, 2014.
33. Webb, M.D. et al. Does proximity to neighbours affect germination of spores of non-proteolytic
Clostridium botulinum? Food Microbiol., 32, 104, 2012.
34. Saeed, E.M.A. Studies on iolation and identification of Clostridium botulinum investigating field samples
specially from equine grass sickness cases. Doctoral thesis, University of Göttingen, Göttingen, 2005.
35. Stringer, S.C., Webb, M.D. and Peck, M.W. Lag time variability in individual spores of Clostridium
botulinum. Food Microbiol., 28, 228, 2011.
36. Hauschild, A.H.W. and Hilsheimer, R. Enumeration of Clostridium botulinum spores in meats by a
pour-plate procedure. Can. J. Microbiol., 23, 829, 1977.
37. Cato, E., George, W.L. and Finegold, S. Genus Clostridium. In: Bergey’s Manual of Systematic
Bacteriology, Sneath, P.H.A. et al. (Eds.), Williams and Wilkins, Baltimore, MD, 1986.
38. Mills, D.C., Midura, T.F. and Arnon, S.S. Improved selective medium for the isolation of lipase-positive
Clostridium botulinum from feces of human infants. J. Clin. Microbiol., 2, 947, 1985.
39. Silas, J.C. et al. Selective and differential medium for detecting Clostridium botulinum. Appl. Environ.
Microbiol., 50, 1110, 1985.
40. Smith, L.D.S. and Sugiyama, H. Botulism: The Organism, Its Toxins, the Disease, Charles C. Thomas,
Springfield, IL, 1988.
Clostridium 169
41. Fakruddin, M. et al. Nucleic acid amplification: Alternative methods of polymerase chain reaction. J.
Pharm. Bioallied Sci., 5(4), 245–52, 2013.
42. Lindström, M. et al. Multiplex PCR assay for detection and identification of Clostridium botulinum
types A, B, E, and F in food and fecal material. Appl. Environ. Microbiol., 67, 5694, 2001.
43. Kasai, Y. et al. Quantitative duplex PCR of Clostridium botulinum types A and B neurotoxin genes. J.
Food Hyg. Soc. Jpn., 48, 19, 2007.
44. Sakuma, T. et al. Rapid and simple detection of Clostridium botulinum types A and B by loop-mediated
isothermal amplification. J. Appl. Microbiol., 106, 1252, 2009.
45. De Medici, D. et al. Multiplex PCR for detection of botulinum neurotoxin-producing clostridia in clini-
cal, food, and environmental samples. Appl. Environ. Microbiol., 75, 6457–61, 2009.
46. Hyytiä, E. et al. Characterisation of Clostridium botulinum groups I and II by randomly amplified poly-
morphic DNA analysis and repetitive element sequence-based PCR. Int. J. Food Microbiol., 48, 179, 1999.
47. Paul, C. et al. A unique restriction site in the flaA gene allows rapid differentiation of group I and group
II Clostridium botulinum strains by PCR-restriction fragments length polymorphism analysis. J. Food
Prot., 70, 2133, 2007.
48. Dahlsten, E. et al. PCR assay for differentiating between group I (proteolytic) and group II (nonproteo-
lytic) strains of Clostridium botulinum. Int. J. Food Microbiol., 124, 108, 2008.
49. Janda, J.M. and Whitehouse, C.A. Usefulness of multilocus polymerase chain reaction followed by elec-
trospray ionization mass spectrometry to identify a diverse panel of bacterial isolates. Diagn. Microbiol.
Infect. Dis., 63, 403, 2009.
50. Fach, P. et al. An innovative molecular detection tool for tracking and tracing Clostridium botulinum
types A, B, E, F and other botulinum neurotoxin producing clostridia based on the GeneDisc cycler. Int.
J. Food Microbiol., 145, S145, 2011.
51. Hauser, D. et al. Botulinal neurotoxin C1 complex genes, clostridial neurotoxin homology, and genetic
transfer in Clostridium botulinum. Toxicon, 33, 515, 1995.
52. East, A.K. and Collins, M.D. Conserved structure of genes encoding components of botulinum neu-
rotoxin complex M and the sequence of the gene coding for the nontoxic component in nonproteolytic
Clostridium botulinum type F. Curr. Microbiol., 29, 69, 1994.
53. Aranda, E. et al. Detection of Clostridium botulinum types A, B, E, and F in foods by PCR and DNA
probe. Lett. Appl. Microbiol., 25, 186, 1997.
54. Raphael, B.H. and Andreadis, J.D. Real-time PCR detection of the nontoxic nonhemagglutinin gene as a
rapid screening method for bacterial isolates harboring the botulinum neurotoxin (A-G) gene complex.
J. Microbiol. Methods, 71, 343, 2007.
55. Dover, J.E. et al. Recent advances in peptide probe-based biosensors for detection of infectious agents.
J. Microbiol. Methods, 78, 10, 2009.
56. Yoo, M. et al. High-throughput identification of clinically important bacterial pathogens using DNA
microarray. Mol. Cell. Probe, 23, 171, 2009.
57. Fenicia, L. et al. Towards an international standard for detection and typing botulinum neurotoxin-
producing Clostridia types A, B, E and F in food, feed and environmental samples: A European ring
trial study to evaluate a real-time PCR assay. Int. J. Food Microbiol., 145, S152, 2011.
58. Raphael, B.H., Joseph, L.A., McCroskey, L.M., Luquez, C. and Maslanka, S.E. Detection and differen-
tiation of Clostridium botulinum type A strains using a focused DNA microarray. Mol. Cell Probes, 24,
146–53, 2010.
59. Ferreira, J.L. et al. Detection of botulinal neurotoxins A, B, E, and F by amplified enzyme-linked
immunosorbent assay: Collaborative study. J. AOAC Int., 86, 314, 2003.
60. Grate, J.W., Ozanich, R.M., Jr., Warner, M.G., Marks, J.D. and Bruckner-Lea, C.J. Advances in assays
and analytical approaches for botulinum-toxin detection. Trends Anal. Chem., 29, 1137–56, 2010.
61. Kalb, S.R. et al. The use of Endopep-MS for the detection of botulinum toxins A, B, E, and F in serum
and stool samples. Anal. Biochem., 351, 84, 2006.
62. Kalb, S.R. et al. Extraction of BoNT/A, /B, /E, and /F with a single, high affinity monoclonal antibody
for detection of botulinum neurotoxin by Endopep-MS. PLoS One, 5, 122–37, 2010.
63. Ladd, J. et al. Detection of botulinum neurotoxins in buffer and honey using a surface plasmon reso-
nance (SPR) sensor. Sens. Actuators, B, 130, 129, 2008.
64. Ferracci, G. et al. A label-free biosensor assay for botulinum neurotoxin B in food and human serum.
Anal. Biochem., 410, 281, 2011.
170 Laboratory Models for Foodborne Infections
65. Ching, K.H. et al. Rapid and selective detection of botulinum neurotoxin serotype-A and -B with a
single immunochromatographic test strip. J. Immunol. Methods, 380, 23, 2012.
66. Liu, Y.Y.B. et al. A functional dual-coated (FDC) microtiter plate method to replace the botulinum toxin
LD50 test. Anal. Biochem., 425, 28, 2012.
67. Ruge, D.R. et al. Detection of six serotypes of botulinum neurotoxin using fluorogenic reporters. Anal.
Biochem., 411, 200–9, 2011.
68. Raphael, B.H. et al. Ultrafiltration improves ELISA and Endopep MS analysis of botulinum neurotoxin
type A in drinking water. J. Microbiol. Methods, 90, 267, 2012.
69. Rajkovic, A. et al. Detection of Clostridium botulinum neurotoxins A and B in milk by ELISA and
immuno-PCR at higher sensitivity than mouse bio-assay. Food Anal. Methods, 5, 319, 2012.
70. Zhang, Y. et al. Simultaneous and sensitive detection of six serotypes of botulinum neurotoxin using
enzyme-linked immunosorbent assay-based protein antibody microarrays. Anal. Biochem., 430, 185, 2012.
71. Brunt, J., Webb, M.D. and Peck, M.W. Rapid affinity immunochromatography column-based tests for
sensitive detection of Clostridium botulinum neurotoxins and Escherichia coli O157. Appl. Environ.
Microbiol., 76, 4143–50, 2010.
72. Al-Khaldi, S.F. et al. Identification and characterization of Clostridium perfringens using single target
DNA microarray chip. Int. J. Food Microbiol., 91, 289, 2004.
73. Uzal, F.A. Diagnosis of Clostridium perfringens intestinal infections in sheep and goats. Anaerobe, 10,
135–43, 2004.
74. Córdoba, J.J. et al. Clostridium. In: Molecular Detection of Human Bacterial Pathogens, Liu, D. (Ed.),
CRC Press, Taylor & Francis Group, Boca Raton, FL, p. 367, 2011.
75. Borriello, S.P. and Aktories, K. Clostridium perfringens, Clostridium difficile, and other Clostridium
species. In: Topley and Wilson’s Microbiology and Microbial Infections, Mahy, B.W.J. et al. (Eds.), John
Wiley & Sons, Chichester, 2010.
76. Labbé, R. Clostridium perfringens. In: The Microbiological Safety and Quality of Food, Aspen
Publishers, Gaithersburg, MD, 2000.
77. Wu, J. et al. Detection and toxin typing of Clostridium perfringens in formalin-fixed, paraffin-embedded
tissue samples by PCR. J. Clin. Microbiol., 47, 807, 2009.
78. Nakamura, M. et al. PCR identification of the plasmid-borne enterotoxin gene (cpe) in Clostridium
perfringens strains isolated from food poisoning outbreaks. Int. J. Food Microbiol., 294, 261, 2004.
79. Tang, Y. et al. Detection, cloning, and sequencing of the enterotoxin gene of Clostridium perfringens
type C isolated from goat. Turkish J. Vet. Anim. Sci., 36, 153, 2012.
80. Shanmugasamy, M. and Rajeswar, J. Alpha toxin specific PCR for detection of toxigenic strains of
Clostridium perfringens in poultry. Vet. World, 5, 365, 2012.
81. Meer, R. and Songer, G. Multiplex polymerase chain reaction assay for genotyping Clostridium perfrin-
gens. Am. J. Vet. Res., 58, 702, 1997.
82. Heikinheimo, A. and Korkeala, H. Multiplex PCR assay for toxinotyping Clostridium perfringens iso-
lates obtained from Finnish broiler chickens. Lett. Appl. Microbiol., 40, 407, 2005.
83. Uzal, F.A. and Songer, J.G. Diagnosis of Clostridium perfringens intestinal infections in sheep and
goats. J. Vet. Diag. Invest., 20, 253, 2008.
84. Goncuoglu, E.M. et al. Molecular typing of Clostridium perfringens isolated from turkey meat by mul-
tiplex PCR. Lett. Appl. Microbiol., 47, 31, 2009.
85. Baums, C.G. et al. Diagnostic multiplex PCR for toxin genotyping of Clostridium perfringens isolates.
Vet. Microbiol., 100, 11, 2004.
86. Joshy, L., Chaurdhry, R. and Chandel, D.S. Multiplex PCR for the detection of Clostridium botulinum
and C. perfringens toxin genes. Indian J. Med. Res., 128, 206, 2008.
87. Feng, Y. et al. Identification of changes in the composition of ileal bacterial microbiota of broiler chick-
ens infected with Clostridium perfringens. Vet. Microbiol., 140, 116, 2010.
88. Gurjar, A.A. et al. Real-time multiplex PCR assay for rapid detection and toxintyping of Clostridium
perfringens toxin producing strains in feces of dairy cattle. Mol. Cell. Probe, 22, 90, 2008.
89. Albini, S. et al. Real-time multiplex PCR assays for reliable detection of Clostridium perfringens toxin
genes in animal isolates. Vet. Microbiol., 127, 179, 2008.
90. Abildgaard, L. et al. Sequence variation in the α-toxin encoding plc gene of Clostridium perfringens
strains isolated from diseased and healthy chickens. Vet. Microbiol., 136, 293, 2009.
Clostridium 171
91. Chon, J. et al. Development of real-time PCR for the detection of Clostridium perfringens in meats and
vegetables. J. Microbiol. Biotechnol., 22, 530, 2012.
92. Sterne, M. and Batty, I. Criteria for diagnosing clostridial infection. In: Pathogenic Clostridia, Sterne,
M. and Batty, L. (Eds.), Butterworths, London, p. 79, 1975.
93. Piyankarage, R.H. et al. Sandwich enzyme-linked immunosorbent assay by using monoclonal antibody
for detection of Clostridium perfringens enterotoxin. J. Vet. Med. Sci., 61, 45, 1999.
94. El Idrissi, A.H. and Ward, G.E. Development of double sandwich ELISA for Clostridium perfringens
beta and epsilon toxins. Vet. Microbiol., 31, 89, 1992.
95. Nagahama, M. et al. Enzyme-linked immunosorbent assay for rapid detection of toxins from Clostridium
perfringens. FEMS Microbiol. Lett., 84, 41, 1991.
96. Petit, L., Gibert, M. and Popoff, M.R. Detection of enterotoxin of Clostridium perfringens. In:
Encyclopedia of Food Microbiology, Batt, C.A., Patel, P. and Robinson, R.K. (Eds.), Academic Press,
London, 1999.
97. Marks, S.L. et al. Evaluation of methods to diagnose Clostridium perfringens associated diarrhea in
dogs. J. Am. Vet. Med. Assoc., 214, 357, 1999.
98. Berry, P.R. et al. Evaluation of ELISA, RPLA, and Vero cell assays for detecting Clostridium perfrin-
gens enterotoxin in faecal specimens. J. Clin. Pathol., 41, 458, 1988.
99. Seyer, A. et al. Rapid quantification of clostridial epsilon toxin in complex food and biological matrixes
by immunopurification and ultraperformance liquid chromatography-tandem mass spectrometry. Anal.
Chem., 84, 5103, 2012.
100. CDC (Center for Disease Control and Prevention). Botulism in the United States (1899–1996).
Handbook for Epidemiologists, Clinicians and Laboratory Workers. U.S. Department of Health and
Human Services, CDC, Atlanta, GA, 1998.
101. CFSAN (Center for Food Safety and Applied Nutrition). Bacteriological Analytical Manual (BAM).
U.S. Food and Drug Administration, Washington, DC, 2001.
102. Chun, C.L. et al. Prevalence of toxin-producing Clostridium botulinum associated with the macroalga
Cladophora in three Great Lakes: Growth and management. Sci. Total Environ., 511, 523–9, 2015.
103. Wohlfarth, K. et al. Foodborne botulism in 2 adults not treated by antiserum. Toxicon, 93, S65, 2015.
104. Cai, S., Lindo, P., Park, J.B., Vasa, K. and Singh, B.R. The identification and biochemical character-
ization of drug-like compounds that inhibit botulinum neurotoxin serotype A endopeptidase activity.
Toxicon, 55, 818–26, 2010.
105. Capkova, K., Hixon, M.S., Pellett, S., Barbieri, J.T., Johnson, E.A. and Janda, K.D. Benzylidene cyclo-
pentenediones: First irreversible inhibitors against botulinum neurotoxin A’s zinc endopeptidase.
Bioorg. Med. Chem. Lett., 20, 206–8, 2010.
106. Eichhorn, T., Dolimbek, B.Z., Deeg, K., Efferth, T. and Atassi, M.Z. Inhibition in vivo of the activity of
botulinum neurotoxin A by small molecules selected by virtual screening. Toxicon, 60, 1180–90, 2012.
107. Kumar, G., Agarwal, R. and Swaminathan, S. Discovery of a fluorine class of compounds as inhibitors
of botulinum neurotoxin serotype E by virtual screening. Chem. Commun., 48, 2412–4, 2012.
108. Pirazzini, M. et al. Thioredoxin and its reductase are present on synaptic vesicles, and their inhibition
prevents the paralysis induced by botulinum neurotoxins. Cell Rep., 8, 1870–8, 2015.
109. Montgomery, V.A. et al. Ex vivo inhibition of Clostridium botulinum neurotoxin types B, C, E, and F
by small molecular weight inhibitors. Toxicon, 8, 12–9, 2015.
110. Marinelli, S. et al. Botulinum neurotoxin type A counteracts neuropathic pain and facilitates functional
recovery after peripheral nerve injury in animal models. Neuroscience, 171, 316–28, 2010.
111. Eleopra, R. et al. Botulinum neurotoxin serotype D is poorly effective in humans: An in vivo electro-
physiological study. Clin. Neurophysiol., 124, 999–1004, 2013.
112. Mika, J. et al. The effect of botulinum neurotoxin A on sciatic nerve injury-induced neuroimmunologi-
cal changes in rat dorsal root ganglia and spinal cord. Neuroscience, 175, 358–66, 2011.
113. Cui, M., Khanijou, S., Rubino, J. and Aoki, K.R. Subcutaneous administration of botulinum toxin A
reduces formalin-induced pain. Pain, 107, 125–33, 2004.
114. Kato, K. et al. Botulinum neurotoxin A2 reduces incidence of seizures in mouse models of temporal
lobe epilepsy. Toxicon, 74, 109–11, 2013.
115. Broide, R.S. et al. The rat Digit Abduction Score (DAS) assay: A physiological model for assessing
botulinum neurotoxin-induced skeletal muscle paralysis. Toxicon, 71, 18–24, 2013.
172 Laboratory Models for Foodborne Infections
116. Zhao, R. Masayasu, H. and Holmgren, A. Ebselen: A substrate for human thioredoxin reductase strongly
stimulating its hydroperoxide reductase activity and a superfast thioredoxin oxidant. Proc. Natl. Acad.
Sci. USA, 99, 8579–84, 2002.
117. Pellett, S. et al. In vivo onset and duration of action varies for botulinum neurotoxin A subtypes 1–5.
Toxicon, 107, 37–42, 2015.
118. Sayeed, S. et al. Beta toxin is essential for the intestinal virulence of Clostridium perfringens type C
disease isolate CN3685 in a rabbit ileal loop model. Mol. Microbiol., 67, 15–30, 2008.
119. Garcia, J.P. et al. Comparative neuropathology of ovine enterotoxemia produced by Clostridium per-
fringens type D wild-type strain CN1020 and its genetically modified derivatives. Vet. Pathol., 52(3),
465–75, 2015.
120. Uzal, F.A. et al. Towards an understanding of the role of Clostridium perfringens toxins in human and
animal disease. Future Microbiol., 9, 361–77, 2014.
121. Sugimoto, N., Chen, Y.-M., Yue, S., Morihiro, L. and Lee, C.-Y. Pathodynamics of intoxication in rats
and mice by enterotoxin of Clostridium perfringens type A. Toxicon, 29, 751–9, 1991.
122. Caserta, J.A., Robertson, S.L., Saputo, J., Shrestha, A., McClane, B.A. and Uzal, F.A. Development
and application of a mouse intestinal loop model to study the in vivo action of Clostridium perfringens
enterotoxin. Infect. Immun., 79, 3020–7, 2011.
123. Bos, J. et al. Fatal necrotizing colitis following a foodborne outbreak of enterotoxigenic Clostridium
perfringens type A infection. Clin. Infect. Dis., 40, 78–83, 2005.
124. Fisher, D.J. et al. Dissecting the contributions of Clostridium perfringens type C toxins to lethality in
the mouse intravenous injection model. Infect. Immun., 74, 5200–10, 2006.
125. Smedley, J.G. et al. Noncytotoxic Clostridium perfringens enterotoxin (CPE) variants localize CPE
intestinal binding and demonstrate a relationship between CPE-induced cytotoxicity and enterotoxicity.
Infect. Immun., 76, 3793–800, 2008.
126. Katahira, J., Inoue, N., Horiguchi, Y., Matsuda, M. and Sugimoto, N. Molecular cloning and functional
characterization of the receptor for Clostridium perfringens enterotoxin. J. Cell. Biol., 136, 1239–47,
1997.
127. Katahira, J., Sugiyama, H., Inoue, N., Horiguchi, Y., Matsuda, M. and Sugimoto, N. Clostridium per-
fringens enterotoxin utilizes two structurally related membrane proteins as functional receptors in vivo.
J. Biol. Chem., 272, 26652–8, 1997.
128. Vidal, J.E., McClane, B.A., Saputo, J., Parker, J. and Uzal, F.A. Effects of Clostridium perfringens beta-
toxin on the rabbit small intestine and colon. Infect. Immun., 76, 4396–404, 2008.
129. Matsuda, T. et al. Enteritis necroticans “pigbel” in a Japanese diabetic adult. Pathol. Int., 57, 622–6, 2007.
130. Ma, M. et al. Synergistic effects of Clostridium perfringens enterotoxin and beta toxin in rabbit small
intestinal loops. Infect. Immun., 82, 2958–70, 2014.
131. Johannsen, U., Erwerth, W., Kunz, G. and Kohler, B. Clostridium perfringens type C enterotoxemia
(necrotizing enteritis) in suckling pigs. 1. Study of the experimental production of disease by Clostridium
perfringens type C poisoning and infection (experimental set-up, clinical aspects, pathological find-
ings). Arch. Exp. Vet. Med., 40, 811–25, 1986.
132. Johannsen, U., Menger, S., Erwerth, W. and Kohler, B. Clostridium perfringens type C enterotoxemia
(necrotizing enteritis) in suckling pigs. 2. Light and electron microscopic studies of the pathology and
pathogenesis of experimental Clostridium perfringens type C toxin poisoning. Arch. Exp. Vet. Med., 40,
881–94, 1986.
133. Johannsen, U., Menger, S., Erwerth, W. and Kohler, B. Clostridium perfringens type C enterotoxemia
(necrotizing enteritis) of suckling pigs. 3. Light and electron microscopic studies of the pathology and
pathogenesis of experimental Clostridium perfringens type C infection. Arch. Exp. Vet. Med., 40, 895–
909, 1986.
134. Schumacher, V.L., Martel, A., Pasmans, F., Van Immerseel, F. and Posthaus, H. Endothelial binding
of beta toxin to small intestinal mucosal endothelial cells in early stages of experimentally induced
Clostridium perfringens type C enteritis in pigs. Vet. Pathol., 50, 626–9, 2013.
135. Garcia, J.P. et al. The effect of Clostridium perfringens type C strain CN3685 and its isogenic beta toxin
null mutant in goats. Vet. Microbiol., 157, 412–9, 2012.
136. Uzal, F.A. et al. Development and application of new mouse models to study the pathogenesis of
Clostridium perfringens type C enterotoxemias. Infect. Immun., 77, 5291–9, 2009.
Clostridium 173
137. Hauschild, A.H.W., Hilsheimer, R. and Rogers, C.G. Experimental enteritis with food poisoning and
classical strains of Clostridium perfringens type A in lambs. J. Infect. Dis., 117, 379–86, 1967.
138. Niilo, L. Mechanism of action of the enteropathogenic factor of Clostridium perfringens type A. Infect.
Immunol., 3, 100–6, 1970.
139. Niilo, L. Clostridium perfringens type C enterotoxemia. Can. Vet. J., 29, 658–64, 1988.
140. Uemura, T., Sakaguchi, G., Ito, T., Okazawa, K. and Sakais, S. Experimental diarrhea in cynomolgus
monkeys by oral administration with Clostridium perfringens type A viable cells or enterotoxin. Jpn. J.
Med. Sci. Biol., 28, 165–77, 1975.
141. Hutton, M.L., Mackin, K.E., Chakravorty, A. and Lyras, D. Small animal models for the study of
Clostridium difficile disease pathogenesis. FEMS Microbiol. Lett., 352, 140–9, 2014.
142. Best, E.L., Freeman, J. and Wilcox, M.H. Models for the study of Clostridium difficile infection. Gut
Microbes, 3, 145–67, 2012.
143. Lawley, T.D. and Young, V.B. Murine models to study Clostridium difficile infection and transmission.
Anaerobe, 24, 94–7, 2013.
144. Hamm, E.E., Voth, D.E. and Ballard, J.D. Identification of Clostridium difficile toxin B cardiotoxicity
using a zebrafish embryo model of intoxication. Proc. Natl. Acad. Sci. USA, 103, 14176–81, 2006.
145. Lanis, J.M., Barua, S. and Ballard, J.D. Variations in TcdB activity and the hypervirulence of emerging
strains of Clostridium difficile. PLoS Pathog., 6, 1001–61, 2010.
146. Bartlett, J.G., Onderdonk, A.B., Cisneros, R.L. and Kasper, D.L. Clindamycin-associated colitis due to
a toxin-producing species of Clostridium in hamsters. J. Infect. Dis., 136, 701–5, 1977.
147. Price, A., Larson, H. and Crow, J. Morphology of experimental antibiotic-associated enterocolitis in the
hamster: A model for human pseudomembranous colitis and antibiotic-associated diarrhea. Gut, 20,
467–75, 1979.
148. Pawlowski, S.W., Calabrese, G. and Kolling, G.L. Murine model of Clostridium difficile infection with
aged gnotobiotic C57BL/6 mice and a BI/NAP1 strain. J. Infect. Dis., 202, 1708–12, 2010.
149. Reeves, A.E., Koenigsknecht, M.J., Bergin, I.L. and Young, V.B. Suppression of Clostridium diffi-
cile in the gastrointestinal tracts of germfree mice inoculated with a murine isolate from the family
Lachnospiraceae. Infect Immun., 80, 3786–94, 2012.
150. Chen, X. et al. A mouse model of Clostridium difficile-associated disease. Gastroenterology, 135,
1984–92, 2008.
151. Theriot, C.M., Koumpouras, C.C., Carlson, P.E., Bergin, I.I., Aronoff, D.M. and Young, V.B.
Cefoperazone-treated mice as an experimental platform to assess differential virulence of Clostridium
difficile strains. Gut Microbes, 2, 326–34, 2011.
152. Buffie, C.G., Jarchum, I. and Equinda, M. Profound alterations of intestinal microbiota following a
single dose of clindamycin results in sustained susceptibility to Clostridium difficile-induced colitis.
Infect. Immun., 80, 62–73, 2012.
153. Pellett, S., Tepp, W.H., Clancy, C.M., Borodic, G.E. and Johnson, E.A. A neuronal cell-based botulinum
neurotoxin assay for highly sensitive and specific detection of neutralizing serum antibodies. FEBS
Lett., 581, 4803–8, 2007.
154. Stahl, A.M., Ruthel, G., Torres-Melendez, E., Kenny, T.A., Panchal, R.G. and Bavari, S. Primary cul-
tures of embryonic chicken neurons for sensitive cell-based assay of botulinum eurotoxin: Implications
for therapeutic discovery. J. Biomol. Screen., 12, 370–7, 2007.
155. Eubanks, L.M. et al. An in vitro and in vivo disconnect uncovered through high-throughput identifica-
tion of botulinum neurotoxin A antagonists. Proc. Natl. Acad. Sci. USA, 104, 2602–7, 2007.
156. Hale, M., Oyler, G., Swaminathan, S. and Ahmed, S.A. Basic tetrapeptides as potent intracellular inhib-
itors of type A botulinum neurotoxin protease activity. J. Biol. Chem., 286, 1802–11, 2011.
157. Lyman, M.E., Tuznik, K.M. and McNutt, P. Methods to produce, culture and evaluate embryonic stem-
cell-derived neurons as a research tool for botulinum toxin and the black widow spider venom latro-
toxin. Toxicon, 68, 91, 2013.
158. Rasetti-Escargueil, C., Machado, C.B., Preneta-Blanc, R., Fleck, R.A. and Sesardic, D. Enhanced
sensitivity to botulinum type A neurotoxin of human neuroblastoma SH-SY5Y cells after d ifferentiation
into mature neuronal cells. Botulinum J., 2, 30, 2011.
159. Tegenge, M.A., Bohnel, H., Gessler, F. and Bicker, G. Neurotransmitter vesicle release from human
model neurons (NT2) is sensitive to botulinum toxin A. Cell Mol. Neurobiol., 32, 1021–9, 2012.
174 Laboratory Models for Foodborne Infections
160. Hatheway, C.L. Botulism. In: Laboratory Diagnosis of Infectious Diseases: Principles and Practice,
Balows, A., Hausler, W.H., Ohashi, M. and Turano, M.A. (Eds.), Springer-Verlag, New York, pp. 111–33,
1988.
161. Whitemarsh, R.C.M. et al. Model for studying Clostridium botulinum neurotoxin using differentiated
motor neuron-like NG108-15 cells. Biochem. Biophys. Res. Commun., 427, 426–30, 2012.
162. Fonfria, E. et al. Rodent- and human-derived cell-based models for the study of botulinum neurotoxin
A1 (BoNT/A1). Toxicon, 93, S27, 2015.
163. Delaflotte, S., Carré, D., Krupp, J. and Huchet, M. Development of an enriched and highly BoNT/
A1-sensitive motor neuron culture. Toxicon, 93, S20, 2015.
164. Borrmann, E., Schulze, F., Cussler, K., Hänel, I. and Diller, R. Development of a cell culture assay for
the quantitative determination of vaccination-induced antibodies in rabbit sera against Clostridium per-
fringens epsilon toxin and Clostridium novyi alpha toxin. Vet. Microbiol., 114, 41–50, 2006.
165. Allaart, J.G., van Asten, A.J., Vernooij, J.C. and Grone, A. Beta2 toxin is not involved in in vitro cell
cytotoxicity caused by human and porcine cpb2-harbouring Clostridium perfringens. Vet. Microbiol.,
171, 132–8, 2014.
166. Vidal, J.E., Saputo, M., Garcia, J., Uzal, F.A. and McClane, B.A. Evidence that the Agr-like quorum
sensing system regulates the toxin production, cytotoxicity and pathogenicity of Clostridium perfrin-
gens type C isolate CN3685. Mol. Microbiol., 83, 179–94, 2012.
167. Yasugi, M. et al. In vitro cytotoxicity induced by Clostridium perfringens isolate carrying a chromo-
somal cpe gene is exclusively dependent on sporulation and enterotoxin production. Microb. Pathog.,
85, 1–10, 2015.
168. Warny, M. et al. p38 MAP kinase activation by Clostridium difficile toxin A mediates monocyte necro-
sis, IL-8 production, and enteritis. J. Clin. Invest., 105, 1147–56, 2000.
169. Mahida, S., Makh, S., Hyde, T., Gray, S.P. and Borriello, Y.R. Effect of Clostridium difficile toxin A on
human intestinal epithelial cells: Induction of interleukin 8 production and apoptosis after cell detach-
ment. Gut, 38, 337–47, 1996.
170. Hecht, G., Pothoulakis, C., LaMont, J.T. and Madara, J.L. Clostridium difficile toxin A perturbs cyto-
skeletal structure and tight junction permeability of cultured human intestinal epithelial monolayers. J.
Clin. Invest., 82, 1516–24, 1988.
171. Miller, P.D., Pothoulakis, C., Baeker, T.R., LaMont, J.T. and Rothstein, T.L. Macrophage-dependent
stimulation of T cell depleted spleen cells by Clostridium difficile toxin A and calcium ionophore. Cell
Immunol., 126, 155–63, 1990.
172. Flegel, W.A. et al. Cytokine response by human monocytes to Clostridium difficile toxin A and toxin B.
Infect. Immun., 5, 3659–66, 1991.
173. Linevsky, J.K. et al. IL-8 release and neutrophil activation by Clostridium difficile toxin-exposed mono-
cytes. Am. J. Physiol., 273, G1333–40, 1997.
174. Eichel-Streiber, C., Laufenberg-Feldmann, R., Sartingen, S., Schulze, J. and Sauerborn, M. Cloning of
Clostridium difficile toxin B gene and demonstration of high N-terminal homology between toxin A and
B. Med. Microbiol. Immunol., 179, 271–9, 1990.
10
Enterococcus
Dongyou Liu
CONTENTS
10.1 Introduction....................................................................................................................................175
10.1.1 Classification, Morphology, and Genomics......................................................................175
10.1.1.1 Classification......................................................................................................175
10.1.1.2 Morphological and Biochemical Characteristics...............................................176
10.1.1.3 Genomics...........................................................................................................177
10.1.2 Biology and Epidemiology................................................................................................177
10.1.3 Clinical Features and Pathogenesis...................................................................................178
10.1.4 Identification......................................................................................................................178
10.1.5 Treatment and Prevention..................................................................................................179
10.2 Laboratory Models........................................................................................................................ 180
10.2.1 Animal Models................................................................................................................. 180
10.2.2 In Vitro Models..................................................................................................................181
10.3 Conclusion......................................................................................................................................181
References................................................................................................................................................181
10.1 Introduction
First observed by Thiercelin in 1899 as saprophytic Gram-positive diplococcus in the gastrointestinal
(GI) tract of humans, Enterococcus (initially named “Enterocoque” to emphasize its morphology and
its intestinal origin) was found to be associated with diarrhea, septicemia, and acute endocarditis.
In the early 1900s, similar organisms (then known as Streptococcus faecalis and Streptococcus fae-
cium) were described. In 1937, a classification scheme that separated streptococci into four groups—
pyogenic, viridans, lactic, and enterococcus—was proposed, with the “enterococcal group” covering
streptococci that grow between 10°C and 45°C, at pH 9.6, in 6.5% NaCl, and survive at 60°C for
30 min. This classification scheme correlated with a serological scheme developed by Lancefield in
the 1930s, in which the “enterococcal group” react with group D antisera (thus the so-called group
D streptococci), while nonenterococcal streptococci react with antiserum groups A, B, C, E, F, or G.
Following the description of additional members in the enterococcal group, a proposal to create the
genus Enterococcus was made in 1970 and formally accepted in 1984 upon DNA–DNA and DNA–
rRNA hybridization analyses [1].
175
176 Laboratory Models for Foodborne Infections
10.1.1.3 Genomics
Phylogentic comparison of ∼1.4 kb of the 16S rRNA gene demonstrates a closer genetic relatedness
of Enterococcus to Vagococcus, Tetragenococcus, and Carnobacterium than to Streptococcus and
Lactococcus.
Multilocus sequence typing (MLST) targeting the internal regions of seven housekeeping genes
(gdh, gyd, pstS, gki, aroE, xpt, and yqiL) permits assignment of E. faecalis strains into sequence types
(ST). MLST analysis of E. faecalis strains isolated worldwide from the early 1900s to 2006 suggested
that acquired antibiotic resistance is enriched in the clonal complex 2 (CC2), CC8, and CC9 lineages,
whereas examination of more recently isolated strains from Europe (between 2006 and 2009) indicated
that multidrug resistance is enriched in the CC2, CC16, and CC87 lineages. Similarly, MLST analysis
of internal gene fragments (between 395 and 583 nucleotides [nt] in size) of seven housekeeping genes
(atpA, ddl, gdh, purK, gyd, pstS, and adk) in E. faecium resulted in the identification of a large cluster of
clinical E. faecium isolates that was first termed lineage C1 and later renamed clonal complex 17 (CC17),
after ST17, which are resistant to ampicillin and ciprofloxacin and are enriched for several genes with
putative roles in virulence (e.g., the large surface protein esp, and carbohydrate metabolism), with the
presence of IS16 being the most prominent marker. In addition, Bayesian analysis of genetic population
structure (BAPS) analysis of 491 distinct STs found among 1720 E. faecium isolates identified 13 groups
of related E. faecium strains. Phylogenetic analysis based on concatenated MLST gene sequences of
isolates contained in the two largest BAPS groups (BAPS 2–1 and BAPS 3–3) revealed that currently
circulating clinical isolates belong to three different lineages (lineage-17, lineage-18, and lineage-78),
which stem from at least three different ancestral strains and have independently acquired genes that
characterize clinical isolates through convergent evolution [4].
The genomes of enterococci are found to range from 2.7 to 3.6 Mb across species, with the G + C content
of DNA between 37 and 45 mol% [5]. Examination of E. faecalis V583 genome (3.36 Mb)—a bloodstream
infection (BSI)-derived, vancomycin-resistant (vanB phenotype) ST6/CC2 lineage strain—revealed the
presence of a large amount of mobile DNA (accounting for >25% of the genomic content), including
seven predicted prophages, multiple integrated plasmids, IS elements, and genomic islands (including
the pathogenicity-associated island or PAI), a vanB-type transposon that confers vancomycin resistance,
and three extrachromosomal plasmids [pTEF1 (66.3 kb), pTEF2 (57.7 kb), and pTEF3 (18.0 kb)] [6,7].
This highlights the propensity of enterococci to acquire and disseminate mobile elements, such as those
that encode antibiotic resistance genes. Other notable traits identified include a capsule, a novel adhesin
termed enterococcal surface protein (Esp), a bile acid hydrolase, and an operon for the enterococcal
cytolysin [8].
The emergence and dissemination of MDR enterococci (especially E. faecium) exhibiting resistance to
ampicillin, vancomycin, and aminoglycosides have reduced the number of therapeutic options. There is
evidence that resistance is also emerging to newer agents used to treat vancomycin-resistant enterococci
(VRE) infections (e.g., linezolid, quinupristin/dalfopristin, and daptomycin) [1,11–13].
10.1.4 Identification
Enterococci grow slowly in culture media and require further conventional biochemical tests or com-
mercial test systems [e.g., the BD GeneOhm VanR (BD Diagnostics, Spark, MD) and Xpert vanA/vanB
(Cepheid, Sunnydale, CA)] for identification. As many enterococcal species vary by only one pheno-
typic trait, the biochemical approach is far from being straightforward. For VRE, chromogenic media
(e.g., CHROM-agar, chromID, and Spectra VRE media) reduce turnaround times through early visual
Enterococcus 179
identification of colonies, although use of standard broth macrodilution or disk diffusion method for
vancomycin-susceptibility testing requires additional time [23].
While 16S rRNA sequence analysis is valuable for identification of many Enterococcus species
(using 97% identity as the threshold), it does not provide adequate resolution between E. casselifla-
vus and E. gallinarum (which share 99.9% identity). Therefore, other molecular methods are required
for routine identification of enterococcal isolates. These include amplification and sequencing of the
domain V of the 23S rRNA gene, rRNA or tRNA intergenic spacers, the D-ala:D-ala ligase genes
(ddl), the manganese-dependent superoxide dismutase (sodA) genes, the chaperonin 60 (cpn60) gene,
the Enterococcus protein A (efaA) gene, genes encoding the RNA polymerase α-subunit (rpoA), the
phenylalanyl-tRNA synthase (pheS), and the elongation factor Tu (tufA), in addition to the application
of ribotyping, repetitive extragenic palindromic PCR (REP-PCR) or BOX-PCR, pulsed-field gel electro-
phoresis (PFGE), and multilocus sequence typing (MLST) [3].
PFGE represents the current standard in the clinical identification of Enterococcus spp. and strain
typing, while ribotyping techniques allow accurate discrimination among species with reduced cost.
Interestingly, MLST demonstrates an accuracy equivalent to that of PFGE for the identification of organ-
isms to the subspecies level. de Been et al. [24] developed a core genome MLST (cgMLST) scheme for
E. faecium that is capable of distinguishing between epidemiologically related and unrelated isolates,
even between those of identical ST. Hullahalli et al. [25] showed that clustered, regularly interspaced
short palindromic repeats (CRISPR) 2 analysis is an inexpensive alternative to MLST for assessing clon-
ality among E. faecalis isolates, and can be used in conjunction with MLST to identify recombination
events occurring between STs. Other novel approaches for enterococcal identification include quantita-
tive PCR (qPCR) targeting the esp gene of E. faecium (espfm), DNA microarrays, and use of bacterio-
phages specific to certain Enterococcus strains.
10.2 Laboratory Models
10.2.1 Animal Models
Rabbits, rats, and mice have proven to be useful for the investigation in the molecular mechanisms of an
antibiotic therapy against enterococcal infections [30].
Utilizing a rabbit subdermal abscess model, Frank et al. [31] identified three in vivo-activated genes
in the E. faecalis OG1RF chromosome that encode glutamate 5-kinase (proB [EF0038]), the transcrip-
tional regulator EbrA (ebrA [EF1809]), and the membrane metalloprotease Eep (eep [EF2380]). They
showed that the ΔebrA strain was fully virulent, the ΔproB strain was slightly attenuated, and the Δeep
strain was severely attenuated in a rabbit model of endocarditis. In a separate study, Frank et al. [32]
employed a rabbit model of endocarditis to test E. faecalis strains with transposon insertions or in-frame
deletions in biofilm-associated loci: ahrC, argR, atlA, opuBC, pyrC, recN, and sepF. They noted that
while only the ahrC mutant was significantly attenuated in endocarditis, the transcriptional regulator
AhrC and the protease Eep were also required for full virulence in murine catheter-associated urinary
tract infection (CAUTI), confirming AhrC and Eep as enterococcal biofilm-associated virulence factors.
More recently, Frank et al. [33] used Wistar rat model of acute foreign body osteomyelitis to examine
the relationship between biofilm formation and development of antimicrobial resistance, and found that
surface colonization alone is sufficient for E. faecalis cells to acquire the biofilm antimicrobial resistance
phenotype.
Eguchi et al. [34] assessed the pharmacodynamics of SMP-601 (also known as PTZ601, PZ-601,
or SM-216601, a novel parenteral carbapenem with potent activity against MDR Gram-positive
pathogens) against vancomycin-resistant E. faecium (VREF) in neutropenic murine thigh infec-
tion model, and found that SMP-601 had a sufficient therapeutic effect against VREF infections
at relatively low exposure conditions. In a rat model of plastic-catheter-induced left-sided entero-
coccal endocarditis, DAP monotherapy (20 mg/kg twice daily) demonstrated superior efficacy
for the treatment of penicillin-resistant E. faecalis in comparison with vancomycin. Similarly,
DAP (12 mg/kg every 8 h) was effective against vancomycin-resistant and gentamicin-susceptible
E. faecium [35,36].
Given the evolutionary conservation of both ancient innate host defenses and bacterial virulence
mechanisms, invertebrate hosts (e.g., the greater wax moth Galleria mellonella, the free-living bacte-
riovorus nematode Caenorhabditis elegans, and the common fruit fly Drosophila melanogaster) offer
valuable models to study innate immunity and host–pathogen interactions and help unravel the pathobio-
logic details in enterococcal infections [37–41].
Infection of the greater wax moth G. mellonella larvae with E. hirae led to the identification of sev-
eral novel enterococcal virulence factors in the insect hemolymph (e.g., GelE, Clp ATPase). Because of
its capacity to escape from the intestinal lumen to the body cavity during the larval to pupal transition,
E. hirae activates the host defense responses, yielding important clues in the pathogenic mechanisms
of enterococcal infection. As G. mellonella tolerates relatively high temperatures (37°C or higher), this
simple nonmammalian model is clearly more relevant to the study of human entercoccal infections than
C. elegans and D. melanogaster (maximum 25°C) [39].
The small roundworm C. elegans possesses 20 nonrenewing intestinal epithelial cells (IECs) that play
a key role in the mediation of intestinal immunity. As C. elegans IECs are morphologically similar to
mammalian IECs, including a “brush border” of microvilli, this worm represents an excellent model
for the study of gut immunity during enterococcal infections. Additionally, the relatively small size and
the ability to survive in liquid culture make C. elegans a cost-effective model for disease investigation
including enterococcal infections [42,43].
The common fruit fly D. melanogaster allows natural colonization of E. faecalis in the intes-
tine and thus offers a useful model for investigations related to its biology, immunity, and pathogen-
esis [44]. Indeed, examination of E. faecalis quorum regulatory system genes LrgAB and SprE, and
bacteriocin EF1097 in D. melanogaster confirmed their role in enhancing E. faecalis infectivity
and toxicity [45].
Enterococcus 181
10.3 Conclusion
The genus Enterococcus comprises over 50 species of Gram-positive, non-spore-forming bacteria that
mostly occur as commensal organisms in the GI tract of mammals and birds. However, when entero-
cocci escape from the gut and establish in other body sites, they may cause a diverse range of clinical
diseases, including UTIs, intra-abdominal, pelvic, and soft tissue infections, bacteremia, endocarditis, and
other uncommon infections. Further, with the ability to acquire and develop multidrug resistance traits,
enterococci have become a serious problem in health-care settings in recent decades, not only putting mil-
lions of people at risk, but also exerting a significant economic burden on the society. In order to devise
improved treatment and control strategies against enterococcal infections, it is important to clearly define
the molecular basis of disease progression and host–microbe interactions. Toward this end, application of
various laboratory models (both vertebrate and invertebrate; in vivo and in vitro) is invaluable.
REFERENCES
1. Cetinkaya Y, Falk P, Mayhall CG. Vancomycin-resistant enterococci. Clin Microbiol Rev
2000;13(4):686–707.
2. Byappanahalli MN, Nevers MB, Korajkic A, Staley ZR, Harwood VJ. Enterococci in the environment.
Microbiol Mol Biol Rev 2012;76(4):685–706.
3. Vu J, Carvalho J. Enterococcus: review of its physiology, pathogenesis, diseases and the challenges it
poses for clinical microbiology. Front Biol 2011;6:357.
4. Palmer KL, van Schaik W, Willems RJL, Gilmore MS. Enterococcal genomics. In: Gilmore MS,
Clewell DB, Ike Y, Shankar N, editors. Enterococci: From Commensals to Leading Causes of Drug
Resistant Infection. Boston: Massachusetts Eye and Ear Infirmary, 2014.
5. Bourgogne A, et al. Large scale variation in Enterococcus faecalis illustrated by the genome analysis of
strain OG1RF. Genome Biol 2008;9:R110.
6. Shankar N, Baghdayan AS, Gilmore MS. Modulation of virulence within a pathogenicity island in
vancomycin-resistant Enterococcus faecalis. Nature 2002;417:746–50.
7. Paulsen IT, et al. Role of mobile DNA in the evolution of vancomycin-resistant Enterococcus faecalis.
Science 2003;299:2071–4.
8. Vebo HC, Solheim M, Snipen L, Nes IF, Brede DA. Comparative genomic analysis of pathogenic and
probiotic Enterococcus faecalis isolates, and their transcriptional responses to growth in human urine.
PLoS One 2010;5:e12489.
9. Fisher K, Phillips C. The ecology, epidemiology and virulence of Enterococcus. Microbiology
2009;155:1749–57.
10. Hammerum AM, Lester CH, Heuer OE. Antimicrobial-resistant enterococci in animals and meat: a
human health hazard? Foodborne Pathog Dis 2010;7(10):1137–46.
11. Arias CA, Murray BE. Emergence and management of drug-resistant enterococcal infections. Expert
Rev Anti Infect Ther 2008;6(5):637–55.
12. Arias CA, Murray BE. The rise of the Enterococcus: beyond vancomycin resistance. Nat Rev Microbiol
2012;10:266–78.
13. Shenoy ES, Paras ML, Noubary F, Walensky RP Hooper DC. Natural history of colonization with
methicillin-resistant Staphylococcus aureus (MRSA) and vancomycin-resistant Enterococcus (VRE): a
systematic review. BMC Infect Dis 2014;14:177.
182 Laboratory Models for Foodborne Infections
14. Giard JC, et al. The stress proteome of Enterococcus faecalis. Electrophoresis 2001;22:2947–54.
15. Cox CR, Coburn PS, Gilmore MS. Enterococcal cytolysin: a novel two component peptide system
that serves as a bacterial defense against eukaryotic and prokaryotic cells. Curr Protein Peptide Sci
2005;6:77–84.
16. Park SY, Kim KM, Lee JH, Seo SJ, Lee IH. Extracellular gelatinase of Enterococcus faecalis destroys
a defense system in insect hemolymph and human serum. Infect Immun 2007;75:1861–9.
17. Heikens E, et al. Contribution of the enterococcal surface protein Esp to pathogenesis of Enterococcus
faecium endocarditis. Microbes Infect 2011;13:1185–90.
18. Michaux C. SlyA is a transcriptional regulator involved in the virulence of Enterococcus faecalis. Infect
Immun 2011;79:2638–45.
19. Lebreton F, et al. AsrR is an oxidative stress sensing regulator modulating Enterococcus faecium oppor-
tunistic traits, antimicrobial resistance, and pathogenicity. PLoS Pathog 2012;8:e1002834.
20. Van Tyne D, Martin MJ, Gilmore MS. Structure, function, and biology of the Enterococcus faecalis
cytolysin. Toxins 2013;5:895–911.
21. Sava IG, Heikens E, Huebner J. Pathogenesis and immunity in enterococcal infections. Clin Microb
Infect 2010;16:533–40.
22. Gaca AO, Abranches J, Kajfasz JK, Lemos JA. Global transcriptional analysis of the stringent response
in Enterococcus faecalis. Microbiology 2012;158:1994–2004.
23. Devriese LA, Pot B, Collins MD. Phenotypic identification of the genus Enterococcus and differentiation
of phylogenetically distinct enterococcal species and species groups. J Appl Bacteriol 1993;75:399–408.
24. de Been M, et al. Core genome multilocus sequence typing scheme for high-resolution typing of
Enterococcus faecium. J Clin Microbiol 2015;53(12):3788–97.
25. Hullahalli K, Rodrigues M, Schmidt BD, Li X, Bhardwaj P, Palmer KL. Comparative analysis of the
orphan CRISPR2 locus in 242 Enterococcus faecalis strains. PLoS One 2015;10(9):e0138890.
26. Crouzet L, Rigottier-Gois L, Serror P. Potential use of probiotic and commensal bacteria as non-antibiotic
strategies against vancomycin-resistant enterococci. FEMS Microbiol Lett 2015;362(8):fnv012.
27. Luther MK, Rice LB, LaPlante KL. Ampicillin plus ceftaroline, cefepime, or ceftriaxone demonstrate
equivalent activity in a high inoculum Enterococcus faecalis infection model. Antimicrob Agents
Chemother 2016;60(9):3178–82.
28. Ramos MC, Grayson ML, Eliopoulos GM, Bayer AS. Comparison of daptomycin, vancomycin, and
ampicillin–gentamicin for treatment of experimental endocarditis caused by penicillin-resistant entero-
cocci. Antimicrob Agents Chemother 1992;36:1864–9.
29. Gallagher JC, Pérez ME, Marino EA, LoCastro LG, Abrardo LA, MacDougall C. Daptomycin ther-
apy for vancomycin-resistant enterococcal bacteremia: a retrospective case series of 30 patients.
Pharmacotherapy 2009;29:792–9.
30. Gutschik E. The Enterococcus endocarditis model in experimental animals and its relevance to human
infection. J Antimicrob Chemother 1993;31(Suppl D):87–95.
31. Frank KL, Barnes AM, Grindle SM, Manias DA, Schlievert PM, Dunny GM. Use of recombinase-
based in vivo expression technology to characterize Enterococcus faecalis gene expression during infec-
tion identifies in vivo-expressed antisense RNAs and implicates the protease Eep in pathogenesis. Infect
Immun 2012;80(2):539–49.
32. Frank KL, et al. AhrC and Eep are biofilm infection-associated virulence factors in Enterococcus fae-
calis. Infect Immun 2013;81(5):1696–708.
33. Frank KL, et al. Evaluation of the Enterococcus faecalis biofilm-associated virulence factors AhrC and
Eep in rat foreign body osteomyelitis and in vitro biofilm-associated antimicrobial resistance. PLoS One
2015;10(6):e0130187.
34. Eguchi K, Kanazawa K, Eriguchi Y, Ueda Y. Pharmacodynamics of SMP-601 (PTZ601) against
vancomycin-resistant Enterococcus faecium and methicillin-resistant Staphylococcus aureus in neutro-
penic murine thigh infection models. Antimicrob Agents Chemother 2009;53(8):3391–8.
35. Vouillamoz J, Moreillon P, Giddey M, Entenza JM. Efficacy of daptomycin in the treatment of experi-
mental endocarditis due to susceptible and multidrug-resistant enterococci. J Antimicrob Chemother
2006;58:1208–14.
36. Munita JM, Murray BE, Arias CA. Daptomycin for the treatment of bacteraemia due to vancomycin-
resistant enterococci. Int J Antimicrob Agents 2014;44(5):387–95.
Enterococcus 183
37. Gaspar F, et al. Virulence of Enterococcus faecalis dairy strains in an insect model: the role of fsrB and
gelE. Microbiology 2009;155:3564–71.
38. Apidianakis Y, Rahme LG. Drosophila melanogaster as a model for human intestinal infection and
pathology. Dis Models Mech 2011;4:21–30.
39. Lebreton F, et al. Galleria mellonella as a model for studying Enterococcus faecium host persistence. J
Mol Microbiol Biotechnol 2011;21(3–4):191–6.
40. Panayidou S, Ioannidou E, Apidianakis Y. Human pathogenic bacteria, fungi, and viruses in Drosophila:
disease modeling, lessons, and shortcomings. Virulence 2014;5(2):253–69.
41. Yuen GJ, Ausubel FM. Enterococcus infection biology: lessons from invertebrate host models. J
Microbiol 2014;52(3):200–10.
42. Sifri CD, et al. Virulence effect of Enterococcus faecalis protease genes and the quorum-sensing locus
fsr in Caenorhabditis elegans and mice. Infect Immun 2002;70:5647–50.
43. Maadani A, Fox KA, Mylonakis E, Garsin DA. Enterococcus faecalis mutations affecting virulence in
the Caenorhabditis elegans model host. Infect Immun 2007;75:2634–37.
44. Teixeira N, et al. Drosophila host model reveals new Enterococcus faecalis quorum-sensing associated
virulence factors. PLoS One 2013;8:e64740.
45. Cox CR, Gilmore MS. Native microbial colonization of Drosophila melanogaster and its use as a model
of Enterococcus faecalis pathogenesis. Infect Immun 2007;75:1565–76.
46. Hall AD, Steed ME, Arias CA, Murray BE, Rybak MJ. Evaluation of standard- and high-dose daptomy-
cin versus linezolid against vancomycin-resistant Enterococcus isolates in an in vitro pharmacokinetic/
pharmacodynamic model with simulated endocardial vegetations. Antimicrob Agents Chemother
2012;56:3174–80.
11
Listeria monocytogenes
CONTENTS
11.1 Listeriosis Is a Significant Public Health Concern........................................................................185
11.2 L . monocytogenes Is a Facultative Intracellular Pathogen............................................................186
11.3 Cell Culture Models.......................................................................................................................186
11.4 Animal Models..............................................................................................................................187
11.4.1 Rabbits, Sheep, and Goats.................................................................................................187
11.4.2 Mice and Rats....................................................................................................................187
11.4.3 Guinea Pigs........................................................................................................................188
11.4.4 Gerbils...............................................................................................................................188
11.4.5 Nonhuman Primates..........................................................................................................189
11.5 L . monocytogenes Isolates Vary in Virulence...............................................................................189
11.6 M ethods of Oral Transmission...................................................................................................... 190
11.7 O ther Variables to Consider...........................................................................................................191
11.8 F uture Directions...........................................................................................................................191
Acknowledgments................................................................................................................................... 192
References............................................................................................................................................... 192
185
186 Laboratory Models for Foodborne Infections
when cantaloupes from Jensen Farms were contaminated during a washing procedure, and were then
widely distributed to at least 28 states.13 Since then, increased awareness of the risk of L. monocytogenes
contamination in processed forms of fresh produce has resulted in a significant increase in the number
of food recalls by the FDA, with an average of 50–60 due to L. monocytogenes per year (www.fda.gov/
Safety/Recalls/).
11.2
L . monocytogenes Is a Facultative Intracellular Pathogen
L. monocytogenes is a nonspore-forming, Gram-positive bacillus that is readily found in both soil and
water samples. A large portion of the L. monocytogenes genome is devoted to regulatory proteins, and
not surprisingly, the bacterium is able to adapt to changes in environmental temperature, pH, salt con-
centrations, and nutrient availability.14 Following transmission to a mammalian host, a large proportion
of L. monocytogenes continue to replicate extracellularly; however, a vital subset of the bacterium can
invade host cells and survive and replicate within this intracellular niche.15
L. monocytogenes mediates its own uptake into nonphagocytic cells via a family of surface-exposed
proteins called internalins. Internalin A (InlA) binds to E-cadherin and internalin B (InlB) binds to
c-Met, and engagement of these receptors results in bacterial internalization via clathrin-mediated endo-
cytosis.16 L. monocytogenes contained within host cell phagocytic or endocytic vacuoles produces lis-
teriolysin O (LLO), a cholesterol-dependent pore-forming toxin that lyses the vacuole, releasing the
bacterium into the cytosol.17–19 L. monocytogenes multiplies rapidly in the cytosol and begins to nucleate
actin filaments via the surface-exposed protein ActA. L. monocytogenes can move through the cytoplasm
by actin-based motility towards the cell membrane and can be enveloped in pseudopod-like structures
and then phagocytosed by neighboring cells.20,21 This results in a cell-to-cell spread of L. monocytogenes
without exposure to the extracellular environment.
11.4 Animal Models
An ideal small animal model of L. monocytogenes infection should closely mimic all phases of human
listeriosis. Oral challenge is the preferred route of transmission to allow investigation of environmental
factors that promote bacterial infectivity and the host innate resistance mechanisms that limit coloniza-
tion of the gut. Only a small subset of humans exposed to L. monocytogenes develops severe systemic
infections; thus, dissemination from the gut to peripheral tissues should be a rate-limiting step. Ideally,
resistant animals should be able to inhibit bacterial replication rapidly, and there should be a lag period
before L. monocytogenes cross the blood–brain barrier or the maternal–fetal interface in susceptible
animals.
Most of the existing models of L. monocytogenes infection have caveats that either present significant
technical hurdles or limit the physiologic relevance of the model. One such limitation is the large inocu-
lum size required to establish infection in most laboratory animals. The infectious dose in humans has
been estimated at 106 –107 CFU.29,30 At least 100-fold larger doses (108–1010 CFU) are typically needed
for experimental infections in small animal models. Differences in the stomach pH, bile content, or
gastric enzyme composition may account for some of the relative resistance of experimental animals.
However, it is also possible that in otherwise healthy individuals, systemic listeriosis only occurs when
very large doses of bacteria are ingested. In that case, the range of inocula used in immune competent
laboratory animals may, in fact, closely model human transmission of the disease.
Another issue is the species specificity of the interaction between the L. monocytogenes surface
proteins InlA and InlB and the receptors found on mammalian cells.31,32 Early studies indicated that
InlA was important for invasion of epithelial cells,24,33 and InlB was needed for uptake in endothelial
cells.34,35 However, more recent work suggests that these two proteins may work in conjunction to allow
for efficient internalization of L. monocytogenes in nonphagocytic cells.36,37 InlA has a high affinity for
human E-cadherin, and the human Met protein serves as a receptor for InlB, so that both ligand/receptor
interactions are functional during human infections.38 As described below, most small animal models
of listeriosis are hampered by a low affinity interaction for either InlA or InlB. Importantly, intestinal
infection and fetoplacental transfer do still occur in some of these animals, which suggest that other
uptake mechanisms may be able to compensate, but it is not yet clear how accurately these systems
mimic human infections.
The differential susceptibility of these mouse strains is not likely due to the intestinal microbiota, since
fecal transplantation between C57BL/6 and BALB/c/By mice did not change the infection outcomes.51
Two different “humanized” mouse strains have been developed in an effort to enhance the effi-
ciency of oral infection in mice. In both cases, modifications were designed to promote interaction
between InlA expressed on the bacterial surface and E-cadherin expressed on the intestinal epithe-
lium. In the first mouse strain, human E-cadherin was ectopically expressed under the control of
the iFABP promoter, resulting in dual expression of both mouse and human E-cadherin in the small
intestine.52 The second mouse strain is a “knock-in” strain that has a single amino acid substitution
(E16P) in murine E-cadherin that allows the protein to serve as a high affinity receptor for InlA.53
Orally infected E16P mice showed enhanced colonization in the gut compared to wild-type mice;
therefore, this mouse strain represents the best option for mimicking the InlA/InlB-mediated inva-
sion events that can occur in the human gastrointestinal tract. However, as with most transgenic mice,
the knock-in was generated on a C57BL/6 background, and these mice are innately more resistant to
L. monocytogenes infection compared to other mouse strains. Although it would be time consum-
ing and expensive, it would be very useful to cross the E16P mutation onto more susceptible strain
backgrounds such as BALB/c mice.
Intravenous or intraperitoneal injection of L. monocytogenes in rats was frequently used in the 1980s
as an infection model to study immune responses against an intracellular pathogen. However, very few
studies have examined the oral transmission of listeriosis in rats. Czuprynski and Balish first showed that
germfree rats were more readily colonized than conventionally housed rats offered L. monocytogenes
in their drinking water.54 Later, Schlech et al. used a feeding tube to deliver the bacteria and found that
doses of at least 109 CFU were needed to consistently establish intestinal infection in young Sprague–
Dawley rats.55 Rats have the same species barrier that limits InlA-mediated invasion of the gut mucosa
as is found in mice,38 and thus, offer no particular advantage for use as a model organism.
11.4.3 Guinea Pigs
Young guinea pigs have also been used to study L. monocytogenes infection, and like mice or rats, doses
of 108–1010 CFU are typically needed to facilitate intestinal colonization.56,57 Melton-Witt et al. infected
guinea pigs with a mixture of 20 signature-tagged strains and showed that the spread from the MLN to
the spleen was a rate-limiting step for systemic dissemination, with only one in every 100–1000 bacteria
getting beyond this bottleneck.56 Using this model, the total number of L. monocytogenes that reached
the spleen or liver was very low, suggesting that the guinea pig is not the ideal model to study the sys-
temic phases of listeriosis.
As with most other rodents, the use of guinea pigs does present a species barrier for internaliza-
tion of L. monocytogenes. Guinea pig E-cadherin does bind efficiently to InlA, but the Met protein
in guinea pigs has an amino acid substitution that prevents optimal interaction with InlB.58 In spite of
this limitation, guinea pigs are considered the model of choice to study maternal–fetal transmission of
L. monocytogenes, largely due to the architecture of their placenta. Both humans and guinea pigs have
a hemochorial placenta, with only a single layer of cells separating the fetal and maternal blood sup-
plies. Particularly during the last stages of pregnancy, this means that bloodborne L. monocytogenes
need to traverse just a single trophoblast to invade the placenta.59 Williams et al. showed that infection
of pregnant guinea pigs with oral doses ranging from 104 to 108 CFU led to invasion in approximately
half of the fetuses.60,61
11.4.4 Gerbils
The E-cadherin and Met proteins in gerbils efficiently bind InlA and InlB, respectively.53 The lack of a
species barrier for interaction with these key bacterial surface proteins makes gerbils perhaps the most
physiologically relevant rodent model to study listeriosis. Disson et al. orally inoculated gerbils and
found significant colonization of both the small and large intestines.53 Furthermore, they demonstrated
100% lethality in fetuses when pregnant females were infected orally.53 The inocula used in those studies
were 109–1010 CFU, but lower doses were not tested, so it is possible that the infectious dose in gerbils
Listeria monocytogenes 189
could be closer to that observed in humans. Blanot et al. also demonstrated that gerbils could develop
rhombencephalitis, a localized brainstem infection, that closely mimicked the type of brain infections
seen in about 20% of human cases.62 However, in those studies, the gerbils were infected via the middle
ear, so it is not yet clear whether brainstem infections can naturally occur following the oral ingestion of
L. monocytogenes in gerbils. Unfortunately, a lack of available tools and reagents specific for gerbils has
limited the enthusiasm for studying host responses in this model.
11.4.5 Nonhuman Primates
Nonhuman primate models have the distinct disadvantage of being expensive to use, and so only small
numbers of animals are usually available for any given study. The clinical course of sporadic listeriosis
appears to mimic human disease closely, as evidenced by occasional outbreaks of L. monocytogenes
infection in nonhuman primate colonies that resulted in either meningoencephalitis or spontaneous abor-
tion in pregnant females.63,64 The few primate infection studies performed to date have been most useful
for approximating the infectious dose of L. monocytogenes in humans. For example, Farber et al. found
that cynomolgus monkeys fed at least 107 CFU of strain Scott A shed L. monocytogenes in feces for
3 weeks, but only the animals given 109 CFU displayed signs of disease (septicemia and occasional diar-
rhea).65 More recently, Smith et al. used a monkey clinical isolate of L. monocytogenes to infect pregnant
rhesus macaques.30 In that study, the overall LD50 was estimated to be 107 CFU, but it was noted that
monkeys with a stillborn fetus needed a significantly lower dose to establish an intestinal infection than
monkeys that had normal deliveries. This suggests that host susceptibility factors are also likely to play
an important role in determining the clinical outcomes following ingestion of L. monocytogenes, and
the use of an inbred animal model (such as laboratory mice) may be the easiest way to identify these
genetic loci.
11.5
L . monocytogenes Isolates Vary in Virulence
Based on serological reactions between listerial antigens and specific antibodies, L. monocytogenes is
separated into at least 12 serovars (1/2a, 1/2b, 1/2c, 3a, 3b, 3c, 4a, 4b, 4c, 4d, 4e, and 7), which tend to
show varied virulence potential in human and animal hosts. It is notable that serovars 1/2a, 1/2b, 1/2c,
and 4b are responsible for >95% of human clinical listeriosis cases, and are also highly pathogenic to
animal hosts such as rodents via intragastric (i.g.) route. Most published studies of oral infection models
have utilized one of the two most common strains of L. monocytogenes: EGDe and 10403s. An advan-
tage of using one of these strains is that there are many isogenic derivatives (specific gene knockouts
and reporter strains) available, and it is easier to compare directly the results of unrelated experiments.
However, the choice of which L. monocytogenes isolate to use for oral challenge studies in small animal
models could greatly alter the course of the ensuing infection. Barbour et al. infected BALB/c mice
intragastrically with 66 different isolates of Listeria and found that the serovar 4b and 1/2a strains were
the most virulent, a pattern that has also been observed in humans.66 Both L. monocytogenes EGDe and
10403s are serovar 1/2a strains, but they have been extensively passaged in the lab. Each is probably a
good representative of pathogenic L. monocytogenes, but it is important to passage the bacteria through
an animal periodically to maintain virulence traits.
Alternatively, the use of strains that have been freshly isolated from particular sources may yield
results that more closely mimic specific types of human disease. For example, a recent microarray analy-
sis suggested there may be identifiable differences between strains that primarily cause febrile gas-
troenteritis and those that lead to more invasive disease.67 In support of this idea, Jensen et al. tested a
strain (La111) that persistently colonized a fish processing plant and demonstrated that the bacteria were
not more virulent in vitro and they did not persist well in the intestines of orally infected guinea pigs.
However, oral infection with strain La111 did result in a significantly greater rate of infected fetuses than
similar challenges with other clinical isolates.68,69 In another example, pregnant mice were significantly
more susceptible to oral challenge with L. monocytogenes 2203, a strain that was isolated during an
outbreak of listeriosis that affected primarily pregnant women.70,71
190 Laboratory Models for Foodborne Infections
Mouse-adapted strains of L. monocytogenes have also been developed to overcome the species bar-
rier to receptor-mediated invasion of epithelial cells in the gut mucosa. The modified InlA expressed
by these strains (InlAm) has a similar affinity for mouse E-cadherin as wild-type InlA has for human
E-cadherin.72,73 Foodborne transmission of InlAm-expressing L. monocytogenes can be achieved with
lower doses (106 –108 CFU) than wild-type strains and results in an infection course that closely mimics
all phases of human disease.49 However, the modified InlAm protein also acquired the ability to bind
N-cadherin in addition to E-cadherin, resulting in an enhanced uptake by villous M cells.74 Thus, the use
of an InlAm-expressing strain may result in altered tropism of bacterial invasion compared with human
infections.
of inoculation procedure may affect the route used by L. monocytogenes to invade the gut barrier and
spread systemically, caution should be used when interpreting the results of a study that does not specify
the exact technique used.
11.8 Future Directions
Orally transmitted models of L. monocytogenes infection have not been as widely used as intravenous or
intraperitoneal models due to a high degree of innate resistance in many species and significant pheno-
typic variability among infected animals. It is often difficult to directly compare the results obtained in
192 Laboratory Models for Foodborne Infections
previously published studies because of significant differences in bacterial strain choice, route of inocu-
lation, and various manipulations investigators have used to enhance the infectivity. Recent improve-
ments, particularly for the mouse model, have greatly improved the efficiency of intestinal infection
following oral transmission, and the use of a standardized model would help to greatly advance the field.
Future studies should focus on the natural pathways used by L. monocytogenes to disseminate from
the gut to peripheral tissues. Of particular relevance will be mechanisms used to cross the blood–brain
barrier and the placental barrier in pregnant females. Finally, the vast majority of human L. monocyto-
genes infections take place in the elderly.92 Studies that directly compare infections in young versus aged
animals may shed light on potential therapeutic strategies that could prevent these deadly infections.
Acknowledgments
S.E.F.D. was supported by a grant from the National Institutes of Health (R56 AI091918).
REFERENCES
1. Carpentier, B. & Cerf, O. Review—persistence of Listeria monocytogenes in food industry equipment
and premises. Int J Food Microbiol 145, 1–8 (2011).
2. Tasara, T. & Stephan, R. Cold stress tolerance of Listeria monocytogenes: a review of molecular adap-
tive mechanisms and food safety implications. J Food Prot 69, 1473–1484 (2006).
3. Rocourt, J., BenEmbarek, P., Toyofuku, H. & Schlundt, J. Quantitative risk assessment of Listeria
monocytogenes in ready-to-eat foods: the FAO/WHO approach. FEMS Immunol Med Microbiol 35,
263–267 (2003).
4. Wing, E.J. & Gregory, S.H. Listeria monocytogenes: clinical and experimental update. J Infect Dis 185,
S18–S24 (2002).
5. Scallan, E. et al. Foodborne illness acquired in the United States—major pathogens. Emerg Infect Dis
17, 7–15 (2011).
6. Dalton, C.B. et al. An outbreak of gastroenteritis and fever due to Listeria monocytogenes in milk. N Engl
J Med 336, 100–105 (1997).
7. Ooi, S.T. & Lorber, B. Gastroenteritis due to Listeria monocytogenes. Clin Infect Dis 40, 1327–1332
(2005).
8. Goulet, V., King, L.A., Vaillant, V. & de Valk, H. What is the incubation period for listeriosis? BMC
Infect Dis 13, 11 (2013).
9. Mead, P.S. et al. Food-related illness and death in the United States. Emerg Infect Dis 5, 607–625
(1999).
10. Allerberger, F. & Wagner, M. Listeriosis: a resurgent foodborne infection. Clin Microbiol Infect 16,
16–23 (2010).
11. Broome, C. Role of foods in sporadic and epidemic listeriosis—a retrospective. 16th International
Symposium on Problems of Listeriosis (Savannah, GA, 2007).
12. Gillespie, I.A. et al. Changing pattern of human listeriosis, England and Wales, 2001–2004. Emerg
Infect Dis 12, 1361–1366 (2006).
13. McCollum, J.T. et al. Multistate outbreak of listeriosis associated with cantaloupe. N Engl J Med 369,
944–953 (2013).
14. Xayarath, B. & Freitag, N.E. Optimizing the balance between host and environmental survival skills:
lessons learned from Listeria monocytogenes. Future Microbiol 7, 839–852 (2012).
15. Jones, G.S. et al. Intracellular Listeria monocytogenes comprise a minimal but vital fraction of the
intestinal burden following foodborne infection. Infect Immun 83, 3146–356 (2015).
16. Pizarro-Cerda, J., Kuhbacher, A. & Cossart, P. Entry of Listeria monocytogenes in mammalian epithe-
lial cells: an updated view. Cold Spring Harb Perspect Med 2, a010009 (2012).
17. Cossart, P., Pizarro-Cerda, J. & Lecuit, M. Invasion of mammalian cells by Listeria monocytogenes:
functional mimicry to subvert cellular functions. Trends Cell Biol 13, 23–31 (2003).
18. Portnoy, D.A., Auerbach, V. & Glomski, I.J. The cell biology of Listeria monocytogenes infection: the
intersection of bacterial pathogenesis and cell-mediated immunity. J Cell Biol 158, 409–414 (2002).
Listeria monocytogenes 193
19. Vazquez-Boland, J.A. et al. Listeria pathogenesis and molecular virulence determinants. Clin Microbiol
Rev 14, 584–640 (2001).
20. Cossart, P. & Bierne, H. The use of host cell machinery in the pathogenesis of Listeria monocytogenes.
Curr Opin Immunol 13, 96–103 (2001).
21. Tilney, L.G. & Portnoy, D.A. Actin filaments and the growth, movement, and spread of the intracellular
bacterial parasite, Listeria monocytogenes. J Cell Biol 109, 1597–1608 (1989).
22. Linden, S.K. et al. Listeria monocytogenes internalins bind to the human intestinal mucin MUC2. Arch
Microbiol 190, 101–104 (2008).
23. Mariscotti, J.F., Quereda, J.J., Garcia-Del Portillo, F. & Pucciarelli, M.G. The Listeria monocytogenes
LPXTG surface protein Lmo1413 is an invasin with capacity to bind mucin. Int J Med Microbiol 304,
393–404 (2014).
24. Lecuit, M., Ohayon, H., Braun, L., Mengaud, J. & Cossart, P. Internalin of Listeria monocytogenes with
an intact leucine-rich repeat region is sufficient to promote internalization. Infect Immun 65, 5309–5319
(1997).
25. Jensen, V.B., Harty, J.T. & Jones, B.D. Interactions of the invasive pathogens Salmonella typhimurium,
Listeria monocytogenes, and Shigella flexneri with M cells and murine Peyer’s patches. Infect Immun
66, 3758–3766 (1998).
26. Corr, S., Hill, C. & Gahan, C.G. An in vitro cell-culture model demonstrates internalin- and hemolysin-
independent translocation of Listeria monocytogenes across M cells. Microb Pathog 41, 241–250
(2006).
27. Sato, T. & Clevers, H. Growing self-organizing mini-guts from a single intestinal stem cell: mechanism
and applications. Science 340, 1190–1194 (2013).
28. Zhang, Y.G., Wu, S., Xia, Y. & Sun, J. Salmonella-infected crypt-derived intestinal organoid culture
system for host-bacterial interactions. Physiol Rep 2, e12147 (2014).
29. FAO/WHO. Risk assessment of Listeria monocytogenes in ready-to-eat foods. In Microbial Risk
Assessment Series No. 4 (Rome, Italy, 2004).
30. Smith, M.A. et al. Dose-response model for Listeria monocytogenes-induced stillbirths in nonhuman
primates. Infect Immun 76, 726–731 (2008).
31. Mengaud, J., Ohayon, H., Gounon, P., Mege, R.M. & Cossart, P. E-cadherin is the receptor for intern-
alin, a surface protein required for entry of L. monocytogenes into epithelial cells. Cell 84, 923–932
(1996).
32. Shen, Y., Naujokas, M., Park, M. & Ireton, K. InIB-dependent internalization of Listeria is mediated by
the Met receptor tyrosine kinase. Cell 103, 501–510 (2000).
33. Gaillard, J.L., Berche, P., Frehel, C., Gouin, E. & Cossart, P. Entry of L. monocytogenes into cells is
mediated by internalin, a repeat protein reminiscent of surface antigens from Gram-positive cocci. Cell
65, 1127–1141 (1991).
34. Greiffenberg, L. et al. Interaction of Listeria monocytogenes with human brain microvascular endo-
thelial cells: InlB-dependent invasion, long-term intracellular growth, and spread from macrophages to
endothelial cells. Infect Immun 66, 5260–5267 (1998).
35. Parida, S.K. et al. Internalin B is essential for adhesion and mediates the invasion of Listeria monocy-
togenes into human endothelial cells. Mol Microbiol 28, 81–93 (1998).
36. Grundler, T. et al. The surface proteins InlA and InlB are interdependently required for polar baso-
lateral invasion by Listeria monocytogenes in a human model of the blood-cerebrospinal fluid barrier.
Microbes Infect 15, 291–301 (2013).
37. Pentecost, M., Kumaran, J., Ghosh, P. & Amieva, M.R. Listeria monocytogenes internalin B activates
junctional endocytosis to accelerate intestinal invasion. PLoS Pathog 6, e1000900 (2010).
38. Lecuit, M. et al. A single amino acid in E-cadherin responsible for host specificity towards the human
pathogen Listeria monocytogenes. EMBO J 18, 3956–3963 (1999).
39. Murray, E., Webb, R. & Swann, M. A disease of rabbits characterized by large mononuclear leuko-
cytosis, caused by a hitherto undescribed bacillus Bacterium monocytogenes. J Pathol Bacteriol 29,
407–439 (1926).
40. Gates, G.A., Blenden, D.C. & Kintner, L.D. Listeric myelitis in sheep. J Am Vet Med Assoc 150,
200–204 (1967).
41. Gray, M.L., Singh, C. & Thorp, F., Jr. Abortion, stillbirth, early death of young in rabbits by Listeria
monocytogenes. II. Oral exposure. Proc Soc Exp Biol Med 89, 169–175 (1955).
194 Laboratory Models for Foodborne Infections
42. Osebold, J.W. & Inouye, T. Pathogenesis of Listeria monocytogenes infections in natural hosts. II. Sheep
studies. J Infect Dis 95, 67–78 (1954).
43. Belen Lopez, M. et al. Serological response in rabbits to Listeria monocytogenes after oral or intragas-
tric inoculation. FEMS Immunol Med Microbiol 7, 131–134 (1993).
44. Miettinen, A., Husu, J. & Tuomi, J. Serum antibody response to Listeria monocytogenes, listerial excre-
tion, and clinical characteristics in experimentally infected goats. J Clin Microbiol 28, 340–343 (1990).
45. Hoelzer, K., Pouillot, R. & Dennis, S. Animal models of listeriosis: a comparative review of the current
state of the art and lessons learned. Vet Res 43, 18 (2012).
46. Audurier, A., Pardon, P., Marly, J. & Lantier, F. Experimental infection of mice with Listeria monocy-
togenes and L. innocua. Ann Microbiol (Paris) 131B, 47–57 (1980).
47. Golnazarian, C.A., Donnelly, C.W., Pintauro, S.J. & Howard, D.B. Comparison of infectious dose of
Listeria-Monocytogenes F5817 as determined for normal versus compromised C57b1/6j mice. J Food
Prot 52, 696–701 (1989).
48. Czuprynski, C.J., Faith, N.G. & Steinberg, H. A/J mice are susceptible and C57BL/6 mice are resistant
to Listeria monocytogenes infection by intragastric inoculation. Infect Immun 71, 682–689 (2003).
49. Bou Ghanem, E.N. et al. InlA promotes dissemination of Listeria monocytogenes to the mesenteric
lymph nodes during food borne infection of mice. PLoS Pathog 8, e1003015 (2012).
50. Bergmann, S. et al. Influence of internalin A murinisation on host resistance to orally acquired listerio-
sis in mice. BMC Microbiol 13, 90 (2013).
51. Myers-Morales, T., Bussell, K.M. & D’Orazio, S.E.F. Fecal transplantation does not transfer either sus-
ceptibility or resistance to food borne listeriosis in C57BL/6 and BALC/c/By mice. F1000 Res 2, 177
(2013).
52. Lecuit, M. et al. A transgenic model for listeriosis: role of internalin in crossing the intestinal barrier.
Science 292, 1722–1725 (2001).
53. Disson, O. et al. Conjugated action of two species-specific invasion proteins for fetoplacental listeriosis.
Nature 455, 1114–1118 (2008).
54. Czuprynski, C.J. & Balish, E. Pathogenesis of Listeria monocytogenes for gnotobiotic rats. Infect
Immun 32, 323–331 (1981).
55. Schlech, W.F., III, Chase, D.P. & Badley, A. A model of food-borne Listeria monocytogenes infection in
the Sprague-Dawley rat using gastric inoculation: development and effect of gastric acidity on infective
dose. Int J Food Microbiol 18, 15–24 (1993).
56. Melton-Witt, J.A., Rafelski, S.M., Portnoy, D.A. & Bakardjiev, A.I. Oral infection with signature-tagged
Listeria monocytogenes reveals organ-specific growth and dissemination routes in guinea pigs. Infect
Immun 80, 720–732 (2012).
57. Roldgaard, B.B., Andersen, J.B., Hansen, T.B., Christensen, B.B. & Licht, T.R. Comparison of three
Listeria monocytogenes strains in a guinea-pig model simulating food-borne exposure. FEMS Microbiol
Lett 291, 88–94 (2009).
58. Khelef, N., Lecuit, M., Bierne, H. & Cossart, P. Species specificity of the Listeria monocytogenes InlB
protein. Cell Microbiol 8, 457–470 (2006).
59. Leiser, R. & Kaufmann, P. Placental structure: in a comparative aspect. Exp Clin Endocrinol 102,
122–134 (1994).
60. Williams, D., Irvin, E.A., Chmielewski, R.A., Frank, J.F. & Smith, M.A. Dose-response of Listeria
monocytogenes after oral exposure in pregnant guinea pigs. J Food Prot 70, 1122–1128 (2007).
61. Williams, D., Dunn, S., Richardson, A., Frank, J.F. & Smith, M.A. Time course of fetal tissue inva-
sion by Listeria monocytogenes following an oral inoculation in pregnant guinea pigs. J Food Prot 74,
248–253 (2011).
62. Blanot, S. et al. A gerbil model for rhombencephalitis due to Listeria monocytogenes. Microb Pathog
23, 39–48 (1997).
63. Lemoy, M.J., Lopes, D.A., Reader, J.R., Westworth, D.R. & Tarara, R.P. Meningoencephalitis due to
Listeria monocytogenes in a pregnant rhesus macaque (Macaca mulatta). Comp Med 62, 443–447
(2012).
64. Smith, M.A. et al. Nonhuman primate model for Listeria monocytogenes-induced stillbirths. Infect
Immun 71, 1574–1579 (2003).
65. Farber, J.M., Daley, E., Coates, F., Beausoleil, N. & Fournier, J. Feeding trials of Listeria monocyto-
genes with a nonhuman primate model. J Clin Microbiol 29, 2606–2608 (1991).
Listeria monocytogenes 195
66. Barbour, A.H., Rampling, A. & Hormaeche, C.E. Variation in the infectivity of Listeria monocytogenes
isolates following intragastric inoculation of mice. Infect Immun 69, 4657–4660 (2001).
67. Laksanalamai, P., Jackson, S.A., Mammel, M.K. & Datta, A.R. High density microarray analysis reveals
new insights into genetic footprints of Listeria monocytogenes strains involved in listeriosis outbreaks.
PLoS One 7, e32896 (2012).
68. Jensen, A., Williams, D., Irvin, E.A., Gram, L. & Smith, M.A. A processing plant persistent strain of
Listeria monocytogenes crosses the fetoplacental barrier in a pregnant guinea pig model. J Food Prot
71, 1028–1034 (2008).
69. Jensen, A. et al. Processing plant persistent strains of Listeria monocytogenes appear to have a lower
virulence potential than clinical strains in selected virulence models. Int J Food Microbiol 123, 254–
261 (2008).
70. MacDonald, P.D. et al. Outbreak of listeriosis among Mexican immigrants as a result of consumption of
illicitly produced Mexican-style cheese. Clin Infect Dis 40, 677–682 (2005).
71. Poulsen, K.P., Faith, N.G., Steinberg, H. & Czuprynski, C.J. Pregnancy reduces the genetic resistance
of C57BL/6 mice to Listeria monocytogenes infection by intragastric inoculation. Microb Pathog 50,
360–366 (2011).
72. Monk, I.R., Casey, P.G., Hill, C. & Gahan, C.G. Directed evolution and targeted mutagenesis to murinize
Listeria monocytogenes internalin A for enhanced infectivity in the murine oral infection model. BMC
Microbiol 10, 318 (2010).
73. Wollert, T. et al. Extending the host range of Listeria monocytogenes by rational protein design. Cell
129, 891–902 (2007).
74. Tsai, Y.H., Disson, O., Bierne, H. & Lecuit, M. Murinization of internalin extends its receptor reper-
toire, altering Listeria monocytogenes cell tropism and host responses. PLoS Pathog 9, e1003381 (2013).
75. Miller, J.K. & Burns, J. Histopathology of Listeria monocytogenes after oral feeding to mice. Appl
Microbiol 19, 772–775 (1970).
76. Hardy, J. et al. Extracellular replication of Listeria monocytogenes in the murine gall bladder. Science
303, 851–853 (2004).
77. Conte, M.P. et al. Acid tolerance in Listeria monocytogenes influences invasiveness of enterocyte-like
cells and macrophage-like cells. Microb Pathog 29, 137–144 (2000).
78. Gajendran, N. et al. Regional IFNγ expression is insufficient for efficacious control of food-borne bacte-
rial pathogens at the gut epithelial barrier. Int Immunol 19, 1075–1081 (2007).
79. Kursar, M. et al. Antigen-specific CD8 + T cell responses in intestinal tissues during murine listeriosis.
Microbes Infect 6, 8–16 (2004).
80. MacDonald, T.T. & Carter, P.B. Cell-mediated immunity to intestinal infection. Infect Immun 28, 516–
523 (1980).
81. Czuprynski, C.J., Faith, N.G., Steinberg, H. & Neudeck, B. Sodium pentobarbital anesthesia transiently
enhances the severity of infection following intragastric, but not intravenous, inoculation of Listeria
monocytogenes in mice. Microb Pathog 35, 81–86 (2003).
82. Sahaghian, R., Faith, N.G. & Czuprynski, C. Comparison of systemic Listeria monocytogenes infec-
tion in esophageally inoculated mice anesthetized with isoflurane or pentobarbital. Lab Anim (NY) 38,
126–130 (2009).
83. Manohar, M., Baumann, D.O., Bos, N.A. & Cebra, J.J. Gut colonization of mice with actA-negative
mutant of Listeria monocytogenes can stimulate a humoral mucosal immune response. Infect Immun 69,
3542–3549 (2001).
84. Bou Ghanem, E.N., Myers-Morales, T. & D’Orazio, S.E. A mouse model of foodborne Listeria mono-
cytogenes infection. Curr Protoc Microbiol 31, 9B.3.1–9B.3.16 (2013).
85. Bou Ghanem, E.N., Myers-Morales, T., Jones, G.S. & D’Orazio, S.E.F. Oral transmission of Listeria
monocytogenes in mice via ingestion of contaminated food. J Visual Exp 75, e50381, doi:10.3791/50381
(2013).
86. Ho, J.L., Shands, K.N., Friedland, G., Eckind, P. & Fraser, D.W. An outbreak of type 4b Listeria mono-
cytogenes infection involving patients from eight Boston hospitals. Arch Intern Med 146, 520–524
(1986).
87. Saklani-Jusforgues, H., Fontan, E. & Goossens, P.L. Effect of acid-adaptation on Listeria monocyto-
genes survival and translocation in a murine intragastric infection model. FEMS Microbiol Lett 193,
155–159 (2000).
196 Laboratory Models for Foodborne Infections
88. Czuprynski, C.J., Faith, N.G. & Steinberg, H. Ability of the Listeria monocytogenes strain Scott A to
cause systemic infection in mice infected by the intragastric route. Appl Environ Microbiol 68, 2893–
2900 (2002).
89. Czuprynski, C.J. & Faith, N.G. Sodium bicarbonate enhances the severity of infection in neutropenic
mice orally inoculated with Listeria monocytogenes EGD. Clin Diagn Lab Immunol 9, 477–481 (2002).
90. O’Driscoll, B., Gahan, C.G. & Hill, C. Adaptive acid tolerance response in Listeria monocytogenes:
isolation of an acid-tolerant mutant which demonstrates increased virulence. Appl Environ Microbiol
62, 1693–1698 (1996).
91. Ramalheira, R. et al. Survival of clinical and food isolates of Listeria monocytogenes through simulated
gastrointestinal tract conditions. Foodborne Pathog Dis 7, 121–128 (2010).
92. Centers for Disease Control and Prevention (CDC). Vital signs: Listeria illnesses, deaths, and outbreaks—
United States, 2009–2011. Morb Mortal Wkly Rep 62, 448–452 (2013).
12
Mycobacterium
CONTENTS
12.1 Introduction................................................................................................................................... 197
12.2 Etiology......................................................................................................................................... 198
12.3 Life Cycle...................................................................................................................................... 198
12.4 Virulence....................................................................................................................................... 199
12.4.1 Two M. bovis Strains and Their Virulence Genes........................................................... 199
12.4.2 PE and PPE Genes............................................................................................................ 199
12.4.3 Mycobactin....................................................................................................................... 199
12.4.4 Mce Genes........................................................................................................................ 200
12.5 Immunity....................................................................................................................................... 200
12.6 Foodborne Illnesses Associated with Mycobacterium Species.................................................... 200
12.7 Genomics....................................................................................................................................... 201
12.7.1 Genome Rearrangements................................................................................................. 203
12.7.2 Repeat Sequences............................................................................................................. 203
12.8 Conclusion..................................................................................................................................... 203
Acknowledgments................................................................................................................................... 204
References............................................................................................................................................... 204
12.1 Introduction
Mycobacterium spp. are intimately linked to human society. Leprosy, caused by Mycobacterium
leprae, has been described in historical texts since ancient times; archaeological and osteological findings
indicate its existence in human populations for thousands of years [1]. Evidences of Mycobacterium
tuberculosis complex (MTC) infection date from the Neolithic C period, more than 8000 years ago [2].
Evidences of human infection by Mycobacterium bovis (a bovine tuberculosis agent) have been described
in skeletons from South Siberia, with dates ranging from approximately 1761 to 2199 years before pres-
ent, placing the remains within the Iron Age period [3].
The zoonotic transmission of Mycobacterium spp. has been related, among other means, to the inges-
tion of contaminated food. Historically, M. bovis has been associated with extrapulmonary tuberculosis
in infants and children, usually occurring due to the consumption of milk, which had not been pasteur-
ized or boiled, from infected cattle [4].
This chapter presents an overview of foodborne Mycobacterium spp., including the background infor-
mation and recent findings relating to the etiology, life cycle, virulence, immunity, and clinical diseases,
as well as genomic aspects such as annotation, genome comparison of closely related organisms, and
virulence-related genes.
197
198 Laboratory Models for Foodborne Infections
12.2 Etiology
The genus Mycobacterium includes a group of high GC Gram-positive microorganisms, including
saprophytic species, and important human and animal pathogens. Pathogenic members are usually char-
acterized by their slow growth in culture, with generation times of 12–24 h, whereas nonpathogenic
members grow considerably faster [5]. In this section, we focus on mycobacteria species involved in
foodborne diseases.
Mycobacterial species causing tuberculosis in humans and animals belong to the Mycobacterium
tuberculosis complex (MTBC). The following organisms are considered members of the MTBC:
M. tuberculosis [6]; Mycobacterium africanum [7] and Mycobacterium canettii [8], which are mainly
human pathogens; M. bovis [9] and Mycobacterium caprae [10], which are mainly ruminant pathogens;
Mycobacterium microti, a pathogen of small rodents [11]; Mycobacterium pinnipedii [12] from marine
mammals; Mycobacterium mungi from mongooses [13]; and Mycobacterium orygis from oryx [14].
Members of the MTBC have been implicated in foodborne transmission to man. The consumption
of contaminated raw dairy products has been recognized as a major cause of transmission of M. bovis
to humans [15], generally associated with the development of extrapulmonary TB [16]. Another species
of the MTBC that infects man is M. caprae. Although the transmission to man by raw dairy products
has not been proven formally for M. caprae, the relatedness of the pathogens and the epidemiological
settings suggest that this is probably the case [17].
Interestingly, there are pre-Columbian evidences of zoonotic infection in Peruvian human skeletons,
revealing that a member of the M. tuberculosis complex caused human disease before contact. The
ancient strains are distinct from known human-adapted forms and are most closely related to those
adapted to seals and sea lions (M. pinnipedii) [18], raising the possibility of foodborne transmission by
ingestion of sea lions’ meat, hides, and bones, which was common practice in this region [19].
Nontuberculous mycobacteria (NTM) are ubiquitous organisms and were believed to represent envi-
ronmental contamination. These organisms are a significant cause of infection in both immunocompe-
tent [20] and immunocompromised humans [21].
M. avium subsp. paratuberculosis (MAP), which belongs to the Mycobacterium avium complex
(MAC), is a well-known NTM pathogen causing Johne’s disease (or paratuberculosis), a chronic
progressive, infectious granulomatous enteritis that principally affects ruminants [21]. There is a pos-
sible association between MAP and Crohn’s disease, a human inflammatory bowel disease. MAP is also
a potential human foodborne pathogen because it survives pasteurization treatments [22].
A series of other NTM have been found to be associated with food: M. nonchromogenicum,
M. peregrinum, M. smegmatis, M. neoaurum, M. fortuitum, M. chelonae, M. flavescens, M. kansasii,
M. scrofulaceum, M. genavense, M. simiae, and M. szulgai, which were detected in milk [23,24],
and M. chelonae and M. kansasii, which were detected in water [25,26].
12.3 Life Cycle
As reviewed by Scherr and Nguyen [27], the developmental stages of mycobacterial life cycle are better
defined in M. tuberculosis. This pathogen infects human macrophages within granulomatous structures
of the lung, where they may undergo morphological alterations into filamentous cells within the infected
host cells. Later, host immune responses suppress the vegetative growth, inducing the formation of
persisters that are able to survive in a dormant state for years (the latent TB stage). These dormant cells
of M. tuberculosis may also become shorter, and some transform into a spherical, ovoid morphology
characterized by a thickened cell wall with altered chemical characteristics. Probably after a weakness in
the host immune defense, M. tuberculosis reactivates vegetative growth from the dormant state to cause
active TB. In this state of the disease, many M. tuberculosis cells may escape the host cells to replicate
extracellularly in the TB cavity within the lung. These actively growing cells also become transmissible
through aerosolized droplets expelled from the lung of TB patients, and continue their life cycle in
the newly infected patients who inhale the droplets.
Mycobacterium 199
12.4 Virulence
Despite the high overall phylogenetic relationship among species of the MTBC, they show wide
variability in their phenotypes, especially in their virulence characteristics. The same occurs with MAP
members. This section points out some important, recent works on MTBC and MAP virulence issues
present in the literature.
12.4.3 Mycobactin
Because some Mycobacterium species secrete iron-chelating siderophores as virulence factors in the
iron-limiting environments of their hosts to compete for ferric iron, they need to produce mycobactin,
200 Laboratory Models for Foodborne Infections
a siderophore responsible for transporting iron into cells. One important phenotypic of MAP when
compared with the MTBC and other MAC species is its inability to produce mycobactin [34,35]. Iron
transport into cells is associated with a 10-gene cluster mbtA-J. In MAP species, the first gene of this
cluster is shorter than the corresponding one in the MTBC and MAC. Because MbtA is responsible for
initiating mycobactin production, this truncation reported suggests that the cascade leading to mycobac-
tin production may be attenuated or disrupted in MAP [35].
12.4.4 Mce Genes
Virulence genes that are important for the entry and persistence of bacteria in the host deserve our
attention. The mammalian cell entry (Mce) gene has this property and has been identified in the MTBC
species. Four copies of the Mce gene are present in M. tuberculosis, and there are eight homologs of the
Mce gene in M. avium subsp. paratuberculosis K-10 [35]. Because there are Mce operons in both patho-
genic and nonpathogenic mycobacteria, it implies that the mere presence of these genes does not give to
the species the ability to be virulent. However, the role of this operon in virulence may be determined by
its expression under specific conditions.
12.5 Immunity
In this section, we briefly review the model of immune response against M. bovis. Following infec-
tion, innate immune responses are important for recruiting immune cells and establishing early lesion
formation; however, these responses do little to limit infection [36]. On the other hand, the adaptive
cell-mediated immune response by Th1 CD4 T cells and their soluble mediators are essential for control-
ling the disease [37]. Additional cells and soluble mediators that contribute to immunity include CD8 T
cells [38], IL-17 [39], nitric oxide [40], and IL-2 [41].
Zoonotic TB has reemerged in some places of the world such as the Mexico–EUA border among
immigrants from regions where bovine TB is endemic, associated with the consumption of soft fresh
cheeses [50–53]. An epidemiological investigation conducted by the Centers for Disease Control and
Prevention (CDC) with 35 human cases (1.12% of all analyzed TB cases) of M. bovis infection in
New York City (NYC) also suggested that the fresh cheese brought to NYC from Mexico was a likely
source of infection [54]. In Brazil, this association has also been found. In a recent study, three of 189
patients (1.6%) diagnosed with TB exhibited a coinfection of M. bovis–M. tuberculosis associated with
the consumption of homemade cheeses, processed from raw milk [55].
Some artisanal cheeses are made with raw milk, followed by a ripening period, to ensure safety, since
the process of ripening can contribute significantly to the reduction of pathogens in such products [56].
Nevertheless, MAP could be cultured from Cheddar cheeses prepared from pasteurized milk artificially
contaminated with MAP strains after a 27-week ripening period [57]. The survival of M. bovis during
the ripening of cheeses has also been reported. This bacterium was shown to be viable in cheeses for dif-
ferent periods: in Camembert for 47 [58], 60 [59], and more than 180 [60] days; in Edam for 60 days [59];
in Cheddar for 220 days [60]; in Gruyere for more than 22 days; and in Swiss Tilsitier for more than
305 days [58]. During the ripening of blue cheese made from raw milk with tubercle bacilli (104/mL), a
decrease in numbers was observed during the first and second weeks, but bacilli were still present after
three to four months [61]. Guinea pigs developed TB when inoculated with a 3-month-old Emmental
cheese artificially contaminated with M. bovis [59].
12.7 Genomics
This section provides a short description and also important information related to genome annotation
and the whole genome comparison of some currently available sequenced strains of Mycobacterium spp.,
with special attention to MAP and MTBC. Table 12.1 shows Genbank information and basic features,
such as the chromosome length, %GC, and total number of CDS of the genomes used in this analysis.
M. tuberculosis H37Rv was included as an outgroup in the comparison, since it is closely related with
M. bovis and M. avium spp. and, on the other hand, branched from the parent group before any other one
in this set of genomes.
M. tuberculosis H37Rv and M. bovis AF2122/97 are genomes from the MTBC. The other ones
are genomes from MAP. Specifically, there are two major groups in MAP isolates, known as Type S
(Sheep Type) and Type C (Cattle Type). MapS5, MapS397, and Map104 are of Type S and MpaK10 and
MapMAP4 are of Type C.
Figure 12.1 shows a maximum-likelihood phylogeny of all seven genomes. Even using only MtH37Rv
as the outgroup, the tree clearly shows a separation of MbAF2122 and MtH37Rv from all other
TABLE 12.1
Genbank Information and Some Basic Features of the Genomes Used in This Comparative Analysis
Accession Chromosome %
Taxon Name Short Name Taxon ID Number Length CDS GC References
M. avium subsp. MapK10 262316 NC_002944 4,829,781 4350 69 [35]
paratuberculosis K-10
M. avium subsp. MapMAP4 1199187 CP005928 4,829,424 4326 69 [62]
paratuberculosis MAP4
M. avium subsp. MapS397 1010838 GCA_000219085 4,815,461 4619 69 [63]
paratuberculosis S397
M. avium subsp. MapS5 1247747 GCA_000330785 4,799,927 4288 69 [64]
paratuberculosis S5
M. avium 104 Ma104 243243 NC_008595 5,475,491 5120 69 TIGR
M. bovis AF2122/97 MbAF2122 233413 NC_002945 4,345,492 3918 66 [65]
M. tuberculosis H37Rv MtH37Rv 83332 NC_000962 4,411,532 3906 66 [66]
202 Laboratory Models for Foodborne Infections
M. avium taxa, ratifying the differences among genomes in MTBC and MAP. Besides, strains of Type
C and Type S could not be clearly clustered in the tree, showing that it is not easy to type these strains
using only sequence comparisons in the protein level. The tree was built using first OrthoMCL [67] to
find families with orthologous proteins. From all the 5599 families found by OrthoMCL, 2195 con-
tain exactly one representative member of each genome and at most one member of MtH37Rv (the
outgroup). Each one of these families was aligned by using MUSCLE [68] and filtered by Gblocks [69],
in order to remove noninformative sites. The alignments were concatenated, and the resulting alignment,
with 695,953 columns, was used as input in RAxML [70] software and the PROTCATWAGF evolution
model to build the tree. All bootstrap support values were 100% and were obtained with 100 replicates.
FigTree [71] was used to draw the tree.
In another whole genome comparison method, suggested in Ref. [72], one can calculate a genomic-
distance index, called MUMi, taking into account both criteria of diversity, which are based on DNA
maximal unique matches shared by two genomes. The matrix of MUMi indexes of our seven genomes
is shown in Table 12.2.
A second tree (data not shown) was obtained by using this matrix as input for a neighbor-joining
distance-based phylogeny method [73]. The distance-based topology obtained completely agrees with
the tree shown in Figure 12.1.
By using the software PanGP [74], based on the output families from OrthoMCL, core-genome and
pan-genome rarefaction curves of all five MAP genomes have been built. They are shown in Figure 12.2.
Ma104
MapS397
MapMAP4
MapK10
MapS5
MtH37Rv
MbAF2122
0.03
FIGURE 12.1 Phylogenetic whole genome tree, based on protein families of the seven genomes showed in Table 12.1.
TABLE 12.2
MUMi Indexes of the Seven Genomes Used in Our Comparison
Ma104 MapK10 MapMAP4 MapS397 MapS5 MbAF2122
MapK10 0.1520
MapMAP4 0.1521 0.0010
MapS397 0.1448 0.0252 0.0255
MapS5 0.1558 0.0112 0.0118 0.0296
MbAF2122 0.9060 0.9011 0.9011 0.9008 0.9015
MtH37Rv 0.9061 0.9013 0.9013 0.9010 0.9017 0.0208
Note: Pairs of closely related genomes have small indexes.
Mycobacterium 203
4800
4500
3900
3600
3300
0 1 2 3 4 5 6
Genome number
FIGURE 12.2 Core- and pan-genome sizes according to the number of genomes considered in the dataset. For each k in
the X-axis, all possible combinations of k genomes (among five) are taken and, for each one of these combinations, pan and
core numbers are plotted.
Both curves are far from reaching a plateau, suggesting that currently there is little data on the charac-
teristics of isolates of Type C and Type S. Thus, new genome-wide data would improve the identification
of genes involved in functional phenotypes.
12.7.1 Genome Rearrangements
Large genomic rearrangements, like long insertions, deletions, inversions, translocations, and duplica-
tions, are also mutational mechanisms that can cause phenotypic and genotypic differences among spe-
cies and subspecies. It is known that they may lead to distinct virulence and resistance characteristics.
Large inversions, for example, may be used by mycobacteria to modify the expression levels of specific
genes in order to acquire some advantages during infection [75].
12.7.2 Repeat Sequences
Another important source of variation in mycobacteria is repeat sequences. Variable number of tandem
repeat (VNTR) is based on the analysis of DNA segments containing tandem repeated sequences in
which the number of copies of the repeated sequence varies among strains. These repeats are 15–100 bp
long and dispersed at multiple locations in the genome sequence [76]. Mycobacterial Interspersed
Repetitive Units (MIRUs) are a particular case of VNTR. They are intergenic regions and are reported as
12-character designations, each character corresponding to the number of repeats at one of the 12 MIRU
loci. Short-sequence repeats (SSRs) are 2–5 bp long tandem repeats and are also present in mycobacteria.
All of these repeat sequences have been extensively used for typing [77–79] and also for inferring epide-
miologic issues of Mycobacterium isolates [80].
12.8 Conclusion
Several reasons make Mycobacterium an important bacterial model for foodborne infection. The
consumption of contaminated raw dairy products is recognized as a significant cause of transmission of
M. bovis to humans. M. caprae is probably also another species that infects humans by the same means.
M. avium subsp. paratuberculosis is a nontuberculosis mycobacterial human foodborne pathogen, with
the ability to survive pasteurization treatments. In addition, several other Mycobacterium spp. detected
in milk and water have been implicated in foodborne diseases.
204 Laboratory Models for Foodborne Infections
Acknowledgments
We thank the Conselho Nacional de Desenvolvimento Científico e Tecnológico (grants 443235/2014-7,
305857/2013-4), Fundect-MS (grants TO 0096/12, TO 0085/15, and TO 007/2015) and Embrapa
(03.14.00.054.00.00, 02.13.16.002.00.00) for partial funding.
REFERENCES
1. Inskip, S.A., et al. Osteological, biomolecular and geochemical examination of an early Anglo-Saxon
case of lepromatous leprosy. PLoS One 10, e0124282 (2015).
2. Hershkovitz, I., et al. Tuberculosis origin: The neolithic scenario. Tuberculosis 95, S122–S126 (2015).
3. Taylor, G.M., Murphy, E., Hopkins, R., Rutland, P. & Chistov, Y. First report of Mycobacterium bovis
DNA in human remains from the Iron Age. Microbiology 153, 1243–1249 (2007).
4. Thoen, C., Lobue, P. & de Kantor, I. The importance of Mycobacterium bovis as a zoonosis. Vet.
Microbiol. 112, 339–345 (2006).
5. Chacon, O., Bermudez, L.E. & Barletta, R.G. Johne’s disease, inflammatory bowel disease, and
Mycobacterium paratuberculosis. Annu. Rev. Microbiol. 58, 329–363 (2004).
6. Koch, R. Die Aetiologie der Tuberkulose. Berl. Klin. Wochenschr. 19, 221–230 (1882).
7. Castets, M. & Sarrat, H. Experimental study of the virulence of Mycobacterium africanum (prelimi-
nary note). Bull. Soc. Med. Afr. Noire Lang. Fr. 14, 693–696 (1969).
8. van Soolingen, D., et al. A novel pathogenic taxon of the Mycobacterium tuberculosis complex, canetti:
Characterization of an exceptional isolate from Africa. Int. J. Syst. Bacteriol. 47, 1236–1245 (1997).
9. Karson, A.G. & Lessel, E.F. Mycobacterium bovis nom. nov. Int. J. Syst. Evol. Microbiol. 20, 273–282
(1970).
10. Aranaz, A., Cousins, D., Mateos, A. & Domínguez, L. Elevation of Mycobacterium tuberculosis subsp.
caprae to species rank as Mycobacterium caprae comb. nov., sp. nov. Int. J. Syst. Evol. Microbiol. 53,
1785–1789 (2003).
11. Wells, A.Q. & Oxon, D.M. Tuberculosis in wild voles. Lancet 1, 1221 (1937).
12. Cousins, D.V., et al. Tuberculosis in seals caused by a novel member of the Mycobacterium tuberculosis
complex: Mycobacterium pinnipedii sp. nov. Int. J. Syst. Evol. Microbiol. 53, 1305–1314 (2003).
13. Alexander, K.A., et al. Novel Mycobacterium tuberculosis complex pathogen, M. mungi. Emerg. Infect.
Dis. 16, 1296–1299 (2010).
14. van Ingen, J., et al. Characterization of Mycobacterium orygis as M. tuberculosis complex subspecies.
Emerg. Infect. Dis. 18, 653–655 (2012).
15. Bezerra, A.V., dos Reis, E.M., Rodrigues, R.O., Cenci, A., Cerva, C. & Mayer, F.Q. Detection of
Mycobacterium tuberculosis and Mycobacterium avium complexes by real-time PCR in bovine milk
from Brazilian dairy farms. J. Food Prot. 78, 1037–1042 (2015).
16. Grange, J.M. & Yates, M.D. Zoonotic aspects of Mycobacterium bovis infection. Vet. Microbiol. 40,
137–151 (1994).
17. Prodinger, W.M., Indra, A., Koksalan, O.K., Kilicaslan, Z. & Richter, E. Mycobacterium caprae
infection in humans. Expert Rev. Anti Infect. Ther. 12, 1501–1513 (2014).
18. Bos, K.I., et al. Pre-Columbian mycobacterial genomes reveal seals as a source of New World human
tuberculosis. Nature 23, 494–497 (2015).
19. Standen, V.G., Santoro, C.M. & Arriaza, B.T. Síntesis y propuestas para el período arcaico en la costa
del extremo norte de Chile. Chungará 36, 201–212 (2004).
20. Chayakulkeeree, M. & Naksanguan, T. Epidemiology and clinical characteristic of mycobacterial
infections in human immunodeficiency virus-infected patients in Siriraj Hospital. J. Med. Assoc. Thai.
98, 238–244 (2015).
21. Rindi, L. & Garzelli, C. Genetic diversity and phylogeny of Mycobacterium avium. Infect. Genet. Evol.
21, 375–383 (2014).
22. Naser, A.S. & Thanigachalam, S. Mycobacterium avium subspecies paratuberculosis causes Crohn’s
disease in some inflammatory bowel disease patients. World J. Gastroenterol. 20, 7403–7415 (2014).
23. Sgarioni, S.A., et al. Occurrence of Mycobacterium bovis and non-tuberculous mycobacteria (NTM) in raw
and pasteurized milk in the northwestern region of Paraná, Brazil. Braz. J. Microbiol. 45, 707–711 (2014).
Mycobacterium 205
24. Aydın, F.E., Ulger, M., Emekdaş, G., Aslan, G. & Günal, S. Isolation and identification of Mycobacterium
bovis and non-tuberculous mycobacteria in raw milk samples in Mersin province. Mikrobiyol. Bul. 46,
283–289 (2012).
25. Cafri, U., Aslan, G., Direkel, S., Tarhan, G., Ceyhan, I. & Emekdaş, G. Identification and isolation of
non-tuberculous mycobacteria from environmental samples. Mikrobiyol. Bul. 44, 395–403 (2010).
26. Briancesco, R., et al. An Italian investigation on non-tuberculous mycobacteria in an urban water
supply. Ann. Ig. 26, 264–271 (2014).
27. Scherr, N. & Nguyen, L. Mycobacterium versus Streptomyces—We are different, we are the same. Curr.
Opin. Microbiol. 12, 699–707 (2009).
28. Aguilar León, D., et al. Mycobacterium bovis with different genotypes and from different hosts induce dis-
similar immunopathological lesions in a mouse model of tuberculosis. Clin. Exp. Immunol. 157, 139–147
(2009).
29. Blanco, F.C., et al. Differential transcriptome profiles of attenuated and hypervirulent strains of
Mycobacterium bovis. Microbes Infect. 11, 956–963 (2009).
30. Altschul, S.F., Gish, W., Miller, W., Myers, E.W. & Lipman, D.J. Basic local alignment search tool. J.
Mol. Biol. 215, 403–410 (1990).
31. Forrellad, M.A., et al. Virulence factors of the Mycobacterium tuberculosis complex. Virulence 4, 3–66
(2013).
32. Marri, P.R., Bannantine, J.P. & Golding, G.B. Comparative genomics of metabolic pathways in
Mycobacterium species: Gene duplication, gene decay and lateral gene transfer. FEMS Microbiol. Rev.
30, 906–925 (2006).
33. Mackenzie, N., Alexander, D.C., Turenne, C.Y., Behr, M.A. & De-Buck, J.M. Genomic comparison of
PE and PPE genes in the Mycobacterium avium complex. J. Clin. Microbiol. 47, 1002–1011 (2009).
34. Quadri, L.E., Sello, J., Keating, T.A., Weinreb, P.H. & Walsh, C.T. Identification of a Mycobacterium
tuberculosis gene cluster encoding the biosynthetic enzymes for assembly of the virulence-conferring
siderophore mycobactin. Chem. Biol. 5, 631–645 (1998).
35. Li, L., et al. The complete genome sequence of Mycobacterium avium subspecies paratuberculosis.
Proc. Natl. Acad. Sci. USA 102, 12344–12349 (2005).
36. McGill, J.L., Sacco, R.E., Baldwin, C.L., Telfer, J.C., Palmer, M.V. & Waters, W.R. The role of gamma
delta T cells in immunity to Mycobacterium bovis infection in cattle. Vet. Immunol. Immunopathol.
159, 133–143 (2014).
37. Pollock, J.M., Welsh, M.D. & McNair, J. Immune responses in bovine tuberculosis: Towards new
strategies for the diagnosis and control of disease. Vet. Immunol. Immunopathol. 108, 37–43 (2005).
38. Villarreal-Ramos, B., McAulay, M., Chance, V., Martin, M., Morgan, J. & Howard, C.J. Investigation of
the role of CD8 + T cells in bovine tuberculosis in vivo. Infect. Immun. 71, 4297–4303 (2003).
39. Aranday-Cortes, E., et al. Upregulation of IL-17A, CXCL9 and CXCL10 in early-stage granulomas
induced by Mycobacterium bovis in cattle. Transbound. Emerg. Dis. 60, 525–537 (2013).
40. Waters, W.R., Palmer, M.V., Whipple, D.L., Carlson, M.P. & Nonnecke, B.J. Diagnostic implications of
antigen-induced gamma interferon, nitric oxide, and tumor necrosis factor alpha production by periph-
eral blood mononuclear cells from Mycobacterium bovis infected cattle. Clin. Diagn. Lab. Immunol. 10,
960–966 (2003).
41. Buddle, B.M., Pollock, J.M., Skinner, M.A. & Wedlock, D.N. Development of vaccines to control
bovine tuberculosis in cattle and relationship to vaccine development for other intracellular pathogens.
Int. J. Parasitol. 33, 555–566 (2003).
42. Roswurm, J.D. & Ranney, A.F. Sharpening the attack on bovine tuberculosis. Am. J. Public Health 63,
884–886 (1973).
43. Bovine TB Eradication Programme for England. Defra publications. http://www.defra.gov.uk/publications/
2011/07/19/pb13601-bovine-tb-eradication-programme (2011).
44. Cosivi, O., et al. Zoonotic tuberculosis due to Mycobacterium bovis in developing countries. Emerg.
Infect. Dis. 4, 59–70 (1998).
45. Kleeberg, H.H. Human tuberculosis of bovine origin in relation to public health. Rev. Sci. Tech. Off. Int.
Epiz. 3, 11–32 (1984).
46. Hammer, P., Kiesner, C. & Walte, H.-G.C. Short communication: Effect of homogenization on heat
inactivation of Mycobacterium avium subspecies paratuberculosis in milk. J. Dairy Sci. 97, 2045–2048
(2014).
206 Laboratory Models for Foodborne Infections
47. Faria, A.C., et al. Short communication: Viable Mycobacterium avium subspecies paratuberculosis in
retail artisanal Coalho cheese from Northeastern Brazil. J. Dairy Sci. 97, 4111–4114 (2014).
48. Pereira-Suárez, A.L., et al. Detection of Mycobacterium tuberculosis complex by PCR in fresh cheese
from local markets in Hidalgo, Mexico. J. Food Prot. 77, 849–852 (2014).
49. Stephan, R., Schumacher, S., Tasara, T. & Grant, I.R. Prevalence of Mycobacterium avium subspecies
paratuberculosis in Swiss raw milk cheeses collected at the retail level. J. Dairy Sci. 90, 3590–3595
(2007).
50. Dankner, W.M., Waecker, N.J., Essey, M.A., Moser, K., Thompson, M. & Davis, C.E. Mycobacterium
bovis infections in San Diego: A clinic epidemiologic study of 73 patients and a historical review of a
forgotten pathogen. Medicine 72, 11–37 (1993).
51. Lobue, P.A., Betacourt, W., Peter, C. & Moser, K.S. Epidemiology of Mycobacterium bovis disease in
San Diego County, 1994–2000. Int. J. Tuberc. Lung Dis. 7, 180–185 (2003).
52. Harris, N.B., et al. Recovery of Mycobacterium bovis from soft fresh cheese originating in Mexico.
Appl. Environ. Microbiol. 73, 1025–1028 (2007).
53. Rodwell, T.C., et al. Tracing the origins of Mycobacterium bovis tuberculosis in humans in the USA to
cattle in Mexico using spoligotyping. Int. J. Infect. Dis. 14, e129–e135 (2010).
54. Centers for Disease Control and Prevention (CDC). Human tuberculosis caused by Mycobacterium
bovis—New York City, 2001–2004. Morb. Mortal. Wkly. Rep. 54, 605–608 (2005).
55. Silva, M., et al. Tuberculosis in patients co-infected with Mycobacterium bovis and Mycobacterium
tuberculosis in an urban area of Brazil. Mem. Inst. Oswaldo Cruz. 108, 321–327 (2013).
56. Martins, J.M., Galinari, É., Pimentel-Filho, N.J., Ribeiro, J.I., Furtado, M.M. & Ferreira, C.L.
Determining the minimum ripening time of artisanal Minas cheese, a traditional Brazilian cheese.
Braz. J. Microbiol. 46, 219–230 (2015).
57. Donaghy, J.A., Totton, N.L. & Rowe, M.T. Persistence of Mycobacterium paratuberculosis during
manufacture and ripening of Cheddar cheese. Appl. Environ. Microbiol. 70, 4899–4905 (2004).
58. Kastli, P. & Binz, M. Die Lebensfähigkeit von Mycobacterium tuberculosis in verschiedenen
Käsesorten. Milchwissenschaft 4, 391–394 (1949).
59. Hahn, H. Is manufacture of Emmental cheese from raw milk safe from the viewpoint of public health?
Tierärztl. Umsch. 14, 254–256 (1959).
60. Hammer, B.W. & Babel, F.J. Dairy Bacteriology, 4th ed. John Wiley & Sons Inc., New York (1957).
61. Lafont, J. & Lafont, P. Some modifications in Koch’s bacillus during ripening of blue cheese. Bull. Acad.
Vet. Fr. 53, 457–461 (1981).
62. Bannantine, J.P., Li, L., Mwangi, M., Cote, R., Garay, J.A. & Kapur, V. Complete genome sequence of
Mycobacterium avium subsp. paratuberculosis, isolated from human breast milk. Genome Announc. 2,
e01252-13 (2014).
63. Bannantine, J.P., et al. Genome sequencing of ovine isolates of Mycobacterium avium subspecies para-
tuberculosis offers insights into host association. BMC Genomics 13, 89 (2012).
64. Singh, S.V., et al. Genome sequence of the “Indian bison type” biotype of Mycobacterium avium subsp.
paratuberculosis strain S5. Genome Announc. 1, e00005-13 (2013).
65. Garnier, T. & Eiglmeier, K. The complete genome sequence of Mycobacterium bovis. Proc. Natl. Acad.
Sci. USA 100, 7877–7882 (2003).
66. Camus, J.C., Pryor, M.J., Médigue, C. & Cole, S.T. Re-annotation of the genome sequence of
Mycobacterium tuberculosis H37Rv. Microbiology 148, 2967–2973 (2002).
67. Li, L., Stoeckert, C.J. & Roos, D.S. OrthoMCL: Identification of ortholog groups for eukaryotic
genomes. Genome Res. 13, 2178–2189 (2003).
68. Edgar, R.C. MUSCLE: Multiple sequence alignment with high accuracy and high throughput. Nucleic
Acids Res. 32, 1792–1797 (2004).
69. Castresana, J. Selection of conserved blocks from multiple alignments for their use in phylogenetic
analysis. Mol. Biol. Evol. 17, 540–552 (2000).
70. Stamatakis, A. RAxML-VI-HPC: Maximum likelihood-based phylogenetic analyses with thousands of
taxa and mixed models. Bioinformatics 22, 2688–2690 (2006).
71. Rambaut, A. FigTree v1.4.0. http://tree.bio.ed.ac.uk/software/figtree (2014).
72. Deloger, M., El-Karoui, M. & Petit, M.A. A genomic distance based on MUM indicates discontinuity
between most bacterial species and genera. J. Bacteriol. 191, 91–99 (2009).
73. Felsenstein, J. PHYLIP—Phylogeny inference package (version 3.2). Cladistics 5, 164–166 (1989).
Mycobacterium 207
74. Zhao, Y., et al. PanGP: A tool for quickly analyzing bacterial pan-genome profile. Bioinformatics
30, 1297–1299 (2014).
75. Stevenson, K. Genetic diversity of Mycobacterium avium subspecies paratuberculosis and the influence
of strain type on infection and pathogenesis: A review. Vet. Res. 46, 64 (2015).
76. Centers for Disease Control and Prevention. Guide to the application of genotyping to tuberculo-
sis prevention and control—Division of tuberculosis elimination. http://www.cdc.gov/tb/programs/
genotyping (2012).
77. Allix-Béguec, C., Harmsen, D., Weniger, T., Supply, P. & Niemann, S. Evaluation and strategy for use of
MIRU-VNTRplus, a multifunctional database for online analysis of genotyping data and phylogenetic
identification of Mycobacterium tuberculosis complex isolates. J. Clin. Microbiol. 48, 2692–2699 (2008).
78. Weniger, T., Krawczyk, J., Supply, P., Niemann, S. & Harmsen, D. MIRU-VNTRplus: A web tool for
polyphasic genotyping of Mycobacterium tuberculosis complex bacteria. Nucleic Acids Res. 38, W326–
W331 (2010).
79. Zumárraga, M., et al. Understanding the relationship between Mycobacterium bovis spoligotypes
from cattle in Latin American countries. Res. Vet. Sci. 94, 9–21 (2013).
80. Barandiaran, S., Pérez, A., Gioffré, A., Vivot, M., Cataldi, A. & Zumárraga, M.M. Tuberculosis in
swine co-infected with Mycobacterium avium subsp. hominissuis and Mycobacterium bovis in a cluster
from Argentina. Epidemiol. Infect. 143, 966–974 (2015).
13
Staphylococcus
CONTENTS
13.1 Introduction................................................................................................................................... 209
13.2 Characteristics and Incidence of Foodborne Intoxications due to Enterotoxin-Producing
Staphylococci................................................................................................................................ 209
13.3 Diagnosis of Staphylococcal Foodborne Poisoning......................................................................211
13.4 Laboratory Models for Study of Staphylococcal Foodborne Poisoning........................................212
13.4.1 Animal Models..................................................................................................................212
13.4.2 Cell Culture Models..........................................................................................................215
13.5 Conclusions....................................................................................................................................216
Acknowledgments....................................................................................................................................217
References................................................................................................................................................217
13.1 Introduction
Staphylococci are Gram-positive and catalase-positive ubiquitous bacteria found on the skin and mucous
membranes of warm-blooded animals and humans. They can also be isolated from environmental
sources such as soil, air, and water and from a wide range of foodstuffs including dry-cured meat prod-
ucts and cheeses.1 To date, 52 species of staphylococci have been described.2,3 Staphylococci are grouped
into coagulase-positive (CPS) and coagulase-negative (CNS) staphylococci, according to their ability to
coagulate rabbit plasma.4 Staphylococcus aureus is the most frequently characterized CPS and a well-
known etiological factor of a variety of infections including superficial skin inflammations, systemic
infections, and septicemia.4,5 Furthermore, S. aureus is a recognized causative agent of staphylococcal
foodborne poisoning and has been considered the only representative of the genus Staphylococcus capa-
ble of producing enterotoxins (SEs).4,6,7 However, some CNS strains showing enterotoxigenic capacity
have been found in sheep and goat milk and cheeses,8,9 Spanish dry-cured hams,10,11 and even in starter
cultures.12 Enterotoxigenicity of these strains could pose considerable risk for food safety as, in some
cases, they are introduced as starter cultures at high levels in many animal-derived products due to their
positive impact on fermentation processes and sensory characteristics of products.4,13
209
210 Laboratory Models for Foodborne Infections
surveillance of clinical stool specimens for S. aureus or its enterotoxins have been reported as the cause
of the low incidence of S. aureus foodborne disease.17
Although enterotoxigenic staphylococci are thermally destroyed, the cooked food may contain
SEs because such toxins are thermostable and cannot be eliminated by heat processing.18 Besides
Staphylococcus spp. are usually very tolerant to NaCl and grow well in NaCl concentrations above 10%.19
In addition, Staphylococcus spp. are tolerant to water activity reduction, being able to survive in ripened
products when water activity is higher than 0.87.11 Thus, foods that have been frequently involved in
staphylococcal intoxication include meat and meat products, poultry and egg products, milk and dairy
products, salads, bakery products, particularly cream-filled pastries and cakes, and sandwich fillings.14,20
Symptoms of staphylococcal intoxication appear in patients between 2 and 8 h after food consumption,
and include nausea, vomiting, retching, and abdominal cramping, with or without diarrhea.21,22 Vomiting
is the most frequently observed symptom.23 The disease is usually self-limiting and typically resolves
within 24–48 h after onset.14 Occasionally, it can be severe enough to warrant hospitalization, particularly
when infants, elderly, or debilitated people are concerned.24 The amount of SEs required to produce food-
borne poisoning in humans is difficult to determine. Reliable results from the examination of food impli-
cated in food-poisoning outbreaks are difficult to obtain because normally the enterotoxin is not uniformly
distributed in the food and it is impossible to know how much food have been consumed by each person.23
TABLE 13.1
Biological Characteristics of Staphylococcal Enterotoxins and Enterotoxin-Like Toxins
Emetic Activity
Molecular Superantigenic
Toxin Genetic Element Weight (kDa) Activity Monkey House Musk Shrew
SEA Prophage 27.1 + + +
SEB Chromosome, plasmid, 28.3 + + +
pathogenicity island
SEC1 Pathogenicity island 27.5 + + NE
SEC2 Pathogenicity island 27.6 + NE +
SEC3 Pathogenicity island 27.6 + + NE
SED Plasmid 26.4 + NE +
SEE Prophage 26.4 + NE +
SEG egc, chromosome 27.0 + + +
SEH Transposon 25.2 + + +
SEI egc, chromosome 24.9 + + +
SElJ Plasmids 28.6 + NE NE
SEK Pathogenicity island 25.5 + + NE
SEL Pathogenicity island 25.2 + + NE
SEM egc, chromosome 24.8 + + NE
SEN egc, chromosome 26.1 + + NE
SEO egc, chromosome 26.8 + + NE
SEP Prophage 26.6 + + +
SEQ Pathogenicity island 25.1 + + NE
SER Plasmid 27.1 + + +
SES Plasmid 26.3 + + +
SET Plasmid 22.6 + + +
SElU egc, chromosome 27.2 + NE NE
SElV egc, chromosome 27.6 + NE NE
SElX Chromosome 19.3 + NE NE
Source: Adapted from Hennekinne, J.A., et al., FEMS Microbiol. Rev., 36, 815, 2012; Omoe, K., et al., Infect. Immun., 81,
3627, 2013; Hu, D.L. and Nakane, A. Eur. J. Pharmacol., 722, 95, 2014.
NE, not examined.
Staphylococcus 211
There are three types of methods to detect bacterial toxins in foods—immunological tools, mass
spectrometry-based methods, and bioassays.
Immunological tools are the most commonly used methods for detecting SEs in foods. Commercially
available kits have been developed according to two different principles: (1) enzyme immunoassay com-
prising ELISA (TECRA Kit, TRANSIA PLATE, and RIDASCREEN SET) and enzyme-linked fluo-
rescent assay (ELFA) (VIDAS SET and VIDASTM SET2), and (2) latex agglutination (SET-RPLA). It
is widely recognized that the use of immunological methods to detect contaminants in food matrices is
a difficult task, mainly because of the lack of specificity and sensitivity of the marketed kits. Moreover,
only antibodies against SEA to SEE, SEG, SEH, and SElQ were available until recently. The immuno-
logical tools do not detect the other SEs.25 The main drawback of current methods to detect enterotoxins
based on specific polyclonal or monoclonal antibodies remains their high cost.43
As an alternative to immunological tools, other strategies based on physicochemical techniques have
been developed. Among these, mass spectrometry method,44 based on the use of immunoaffinity capture
and matrix-assisted laser desorption/ionization time-of-flight mass spectrometry (MALDI-TOF-MS) or
electrospray ionization (ESI),45,46 biomolecular interaction analysis mass spectrometry (BIA-MS),47 and
high-performance liquid chromatography (HPLC)48 have been proposed. These methods allow an accu-
rate detection of SEs in foods but require investing in expensive HPLC, MALDI-TOF-MS, or BIA-MS
equipments.
Bioassays are based on the ability of a suspected food extract to induce symptoms such as vomiting or
gastrointestinal signs in laboratory animal models and/or superantigenic action in cell culture models.25
These methods offer a very valuable alternative for foodborne staphylococcal investigation and will be
explorated in a separate section.
TABLE 13.2
Animal Models Used for the Detection of Staphylococcal Enterotoxins
Animal Symptoms, Pathology SE Tested References
Cynomolgus monkey Emetic activity A, B, C, D, E, K, L, M, N, O, P, 28,52,55,107,108
(Macaca fascicularis) Q, R, S, T
Cytokine and chemokine B 109
response
Rhesus monkey (Macaca Emetic activity A, B, C, D, E, G, H, I 53–55,110,111
mulatta) Cytokine response A, B 112
Pigtail monkey (Macaca Emetic activity C1 80,81
nemestrina)
House musk shrew (Suncus Emetic activity A, B, C, C2, D, E, G, H, I, P, R, 52,59–61
murinus) S, T,
Intestinal loop assay A, C 61
Ferret (Mustela putorius furo) Emetic activity B, C2 66,67
Pig/piglet Emetic activity A, B 68,69
Cytokine response B 70
Superantigen activity B 71
Goat Clinical symptomatology B 72
Dog Acute hemodynamic and Unknown 73
gastrointestinal changes
Cat Emetic activity A, B, C2 74,75
Pyrogenic activity A, B 76
Rabbit Intestinal loop assay A, C 61
Pyrogenic activity A, C2, K, Q, L 75,78–81
Superantigenicity
Lethality K 79
Rat Gastrointestinal changes A, B 84,85
Superantigen activity B 86,87
Mouse Cytokine response A, B, C3, K, Q 89–93,95
Toxic shock B 82
the oral administration of the toxin in parallel with real-time recording using a video camera. The num-
ber of vomiting events, the time until the first vomiting event (latency period), and behavioral changes
are recorded. To minimize the effect of previous intoxication, there was at the minimum a 2-week inter-
val between the intoxication experiments. A response in at least two animals is accepted as a positive
reaction.28,51 Monkeys have been considered to be the primary animal model.
Regarding the sensitivity of this method, the emetic dose for monkeys is somewhat variable for the
different SEs. The least amount of SEA that was observed to produce an emetic reaction when given
intragastrically was 5 μg/kg of weight, with SEB, SEC, and SEE requiring 10 μg/kg and SED requiring
20 μg/kg. The minimum dose required for the other SEs was not determined.51 Studies with the new SEs
reveal that higher concentrations are needed to induced emetic reactions in monkeys, with values rang-
ing from 30 to 150 μg/kg.28,52–54
In addition, it has also been found that some monkeys responded only to some SEs, while other mon-
keys were sensitive to almost all the SEs, suggesting that the emetic responses of monkeys vary on an
individual basis.28,55
Other researchers have shown that monkeys have developed tolerance to the emetic effects of SEs if
repeated doses are given during a short period of time.51,56
Finally, this animal model has been used to demonstrate that the abdominal viscera is the site of
action for the induction of the emetic reaction and that the SEs stimulate the vomiting center of the
medulla oblongata through the vagus and sympathetic nerves.57 The use of monkeys in researching SEs
214 Laboratory Models for Foodborne Infections
is severely restricted by the high cost, availability of these animals, and ethical considerations.50,58 All
these reasons have limited their use for routine testing.
The house musk shrew has been described as a suitable small animal model for research on the emetic
response to various emetic SEs52,59–61 (Table 13.2). The emetic assays are performed with house musk
shrews weighing from 40 to 70 g. The shrews are housed under controlled conditions of illumination
(12/12 h light/dark cycle). Purified SEs are diluted in 0.01 M phosphate-buffered saline (PBS; pH 7.2),
and 200 μL volumes of SEs at an appropriate dilution were administered perorally or intraperitoneally to
the house musk shrews. The animals are observed for emesis for 3 h after the administration of SEs. The
number and times of vomiting, the time to the first vomiting episode, and any behavioral changes were
recorded.62 Vomiting occurred within 14–130 min after administration.
SEA provoked more potent emetic response in vivo. The 50% emetic dose of SEA by peroral adminis-
tration is 10 times higher than by intraperitoneal administration (32 and 3 μg/kg of body weight, respec-
tively).61,62 Studies with other SEs reveal that higher concentrations are needed to induce emetic reactions
in house musk shrews, with values ranging from 10 to 1000 μg/animal.52,59,60 It is noteworthy that differ-
ent types of SEs have different emetic activities in house musk shrews.50
Interestingly, in contrast to the emetic responses, diarrhea was observed in none of the animals tested,
although SEA and SEC induced emetic responses. The results showed that the administration of SEA
provoked a potent emetic response in vivo and showed high superantigenic activity in vitro, but did not
induce diarrheal symptom in the animal model.61
This model has been useful for vaccine development against SEA, using a recombinantly attenuated
SEA devoid of superantigen and emetic activity.63 Furthermore, sera from these vaccinated animals
inhibit SEA-induced proliferation of naïve shrew splenocytes (in vitro) as well as emesis.52
Another animal used as a model system for SEs detection has been the ferret (Table 13.2). Ferrets were
used as the emetic model for finding antagonists to treat emesis induced by anticancer therapy64 and
respond to the full spectrum of agents known to induce emesis in humans.65 An advantage of the ferret
for the investigation of foodborne poisoning is that the morphology and physiology of its gastrointestinal
tract have many features in common with the human gastrointestinal tract.66
For foodborne staphylococcal investigation, the emetic assays are performed with ferrets weighing
700–735 g. Prior to dosing with SEs, animals are deprived of food for 24 h, but are allowed free access
to water. SEs are given to groups of five animals at doses of 1–5 mg into the stomach via an oral dosing
tube.66,67 Changes in the body temperature and the activity and the incidence of retching, vomiting, and
defecation were monitored over a period of 3 h. This ferret model has been used to check oral SEB and
SEC2 intoxications.66,67 However, in the ferret, the dose of SEs required to elicit emesis is higher than
that required to elicit it in primates. Possible explanations for requiring this large dose of SEs include
receptor differences and/or more efficient degradation of SEs in the gut of ferrets versus humans or
monkeys.52
Pig68 and piglet69 models have also been used to reproduce some of the features associated with staph-
ylococcal intoxication in humans. However, it is not a widely used animal model. It has been only applied
for SEA and SEB foodborne investigation (Table 13.2). In this animal model, several breeds (Yorkshire
pig, Hampshire pig), crossbreeds, and pigs of different ages (piglets, weanling pigs—0.9 to 9.1-kg weight
range) have been used.68–71 For the analysis, each pig is placed in a separate cage with commercial pig
feed and water provided ad libitum. Pigs are maintained under controlled lighting (15-h light and 9-h
dark cycle) and temperature (20.5°C ± 0.75°C) conditions. The SE can be administered orally, by duode-
nal catheterization, or intravenously. In these conditions, vomiting occurs 90–180 min after emetic doses
of SEs. The 50% emetic dose of SEA was between 20 and 50 μg. The lowest dose inducing emesis in
weanling pigs was from 10 to 20 μg.68–71 Pigs are somewhat more resistant than monkeys to the emetic
effects of SEs.68 This animal model has been used to determine the efficacy of immunization with vac-
cine against SEB too.70,71
Goats are used for studying in vivo gastrointestinal effects caused by SEB72 (Table 13.2). This entero-
toxin induces colic and watery diarrhea and a more pronounced increase in blood urea nitrogen and tem-
perature in this animal.52,72 For analysis, goats weighing between 21 and 45 kg are kept indoors and fed a
diet of hay and pelleted concentrate. Water is provided ad libitum. Dose levels of SEB are between 0.02
Staphylococcus 215
and 0.5 μg/kg and are administered by nasal catheters into the rumen. This animal does not have emetic
activity, but SEB-induced inhibition of rumen contractions, which is a consequence of internal vomiting.72
Dogs and cats have been used too, but only in some of the staphylococcal toxin investigations
(Table 13.2). The dog model system has been used to study gastrointestinal and hemodynamic changes
caused by intravenous and intraintestinal administration of SEs. They were observed by an autopsy 24 h
after injection to make a histological study of injuries.73
The cat model has been widely used to study emetic74,75 and pyrogenic76 activities. For these analyses,
cats aged about 8–10 weeks old and, with a mean body weight of 500 g, are caged separately in 12-h light/
dark cycle at a temperature of 22°C–26°C with food and water available at all times. Two-milliliter vol-
umes of SEA, SEB, and SEC2 are administered intraperitoneally to the cats. The animals are observed
for emesis and diarrhea for up to 6 h after the intraperitoneal administration. The number and times of
vomiting and diarrhea and the time to the first response episode are recorded for the foodborne staphy-
lococcal investigation.75
The main disadvantage of using those models remain similar to the monkey model—high cost and
short supply in the available tools for the study of SEs-associated immunopathology.77
Other animals like rabbits, rats, and mice also have low susceptibility to SEs; their response to SEs are
not specific or they are not vomit-competent species.50 These animals are used mainly as a model for the
study of superantigenicity, pyrogenicity, capacity to enhance endotoxin shock, and lethality (Table 13.2).
The rabbit model was used to compare in vivo toxicity induced by SEC2 using intravenous injection
to test the pyrogenic activity. Rectal temperatures of rabbits were measured with indwelling rectal ther-
mometers and recorded for 4 h after pyrogen administration.75 Pyrogen assays have also been used in
rabbits that are administered with SEA,78 SEK,79 SEL,80 and SEQ.81
Rabbit intestinal loop assay has been used to test the diarrheagenic activity of SEA and SEC.61 New
Zealand white and Dutch belted are the most frequently utilized rabbit strains for testing SEs.52
Rodents are frequently used as models because of their inbred homogeneity. In addition, large num-
bers of animals with the corresponding results are available in a relatively short time. However, mice are
poor responders to SEs as the affinity of these toxins to mouse major histocompatibility complex class II
(MHC class II) is much lower than that for human MHC class II.82 Moreover, rat- and mouse-based
models are regarded as being substantially less sensitive to SEB intoxication than monkey models.83
Therefore, the use of potentiating agents such as d-galactosamine, actinomycin D, lipopolysaccharide
(LPS), viruses, or even protozoa is required. Thus, lower amounts of these protein toxins elicit a quantifi-
able form of toxic shock useful for therapeutic and vaccine developments.52
Rat has been reported in a model of intestinal inflammation postweaning, based on the systemic
administration of SEB84 and SEA85 (Table 13.2) and for evaluating the SEB superantigenic activity by
intravenous administration.86,87
Mice are useful for basic toxin studies and discovery of therapeutics/vaccines for combating staphy-
lococcal superantigen-induced shock. However, mice lack an emetic response and are thus not very
appropriate for investigation of SEs foodborne poisoning.82
Transgenic mice with human MHC class II were found to be an ideal animal model for examining the
biological effects of superantigens, as they responded to much lower doses of toxins due to the higher
affinity binding of SEs to human MHC class II molecules.82,88,89 For this purpose, several mouse strains
(BALB/c, NMRI, C57BL/6, C3H/HeJ, C3H/OuJ, HLA-DR3, knockout mouse, etc.) weighing 18–20 g
are used.52
Major studies using mice as animal model have focused on comparing the responses of T-cells in the
gut-associated lymphoid tissue to different SEs administered by oral, intragastric, or intraperitoneal routes.
SEA, SEB, SEC3 and SEK, and SEQ have been assessed by using this animal model89–95 (Table 13.2).
There are several advantages of cell culture models over animal models: (1) it is a simple and con-
trolled model to study host–SEs interaction, (2) it is easy to run experiments, (3) cells are able to
multiply rapidly—thus the experiment can be conducted quickly, (4) the secondary cells are immortal
if nutrients and proper culturing conditions are provided, (5) it is relatively inexpensive compared
to animal model, specially secondary cells, and (6) it is ethically more acceptable than assays using
animals.49
However, there are several limitations: (1) cultured mammalian cells are generally derived from tumor
cells, and therefore, genetic aberrations have occurred in these cells, may lose traits of original tissue and
lose tissue-specific receptor, (2) cultured cells consist of only one type of cell; therefore, interaction with
concerted host cell cannot be studied with cell culture models, and (3) lack of mucus and other secretory
components, which normally interact. Thus, animal models are often used to confirm or verify the find-
ings from the in vitro cell culture models.49
The gastrointestinal injuries associated with SEs foodborne poisoning have been extensively studied
for a number of years using various animal models (Section 13.4.1), but in particular, insight into how
SEs breach the epithelial barrier is scarce and has been mainly studied in epithelial cell lines.33 The bind-
ing of SEA and SEB toxins onto the surface of the enterocyte microvillus, mediated by binding to diga-
lactosylceramide residues, has been demonstrated. This research further confirmed that the toxin entry
into the enterocytes via apical endocytosis within the endosomes and the pathological events following
SEs introduction into the intestinal area are due to the combined effect of SEs that disrupt the epithelial
barrier by inducing enterocyte-cytopathic toxins produced by S. aureus.96,97
SEs act as superantigens that target the immune system, inducing massive T-cell activation, cyto-
kine release, and systemic shock.98 Examination of these SEs for the ability of causing mammalian cell
damage provides a means to assay these toxins. Recognition of the superantigen–MHC II complex by
T-cell receptor (TCR) results in cell signaling, proliferation, and subsequent release of cytokines/chemo-
kines.33,52 Immune cell activation by superantigens and subsequent cellular changes are similar to those
of conventional antigens and require three important signals: (1) from superantigen interaction with
TCR and activation of protein tyrosine kinases; (2) engagement of costimulatory molecules on antigen-
presenting cells and T-cells, upon superantigen binding that optimizes T-cell activation; and (3) inter-
leukin (IL)-1, tumor necrosis factor α (TNFα), interferon gamma (IFNγ), IL-2, IL-6, and chemokines,
specifically monocyte chemoattractant protein-1 (MCP-1), which are induced directly by superantigens
and represent the third signal for T-cell activation.52 Clearly, SEs-based activation of cells involves a
multifactorial event encompassing multiple host molecules.
Several types of cells are available for superantigenic activity bioassays induced by SEs. Thus, human
peripheral blood mononuclear cells (PBMCs) and human B lymphoblastoid cells have been used for SEB,
SEC2, SEP, SER, SES, SET, and SElX-induced immune response analysis.31,52,60,67,83,99–101 Superantigenic
effect was explored by treating T-lymphocytes isolated from thymus of rats or mice using various doses
of SEA, SEG, SEI, SEK, SEM, SEN, SEO, and SEQ.30,95,102,103 Splenocyte assays have been used for
measuring the superantigenic activity of SEA, SEH, and SEL.32,80,104,105
Moreover, the interaction between SEs and MHC II molecules was studied using secondary cell mod-
els as the human B cell lymphoma Raji and human colorectal cancer cells.67,106
13.5 Conclusions
SEs, produced by the Staphylococcus genus, mainly S. aureus, belong to the family of superantigens
that induce potent emesis and are involved in foodborne poisoning. Staphylococcal foodborne poisoning
occurs as a consequence of consumption of food containing enough amounts of preformed enterotoxins.
Vomiting is the most frequently observed symptom.
Enterotoxin-producing staphylococci have been reported as producers of 23 types of SEs. They are
characterized by superantigenic activity and by induction of emesis. Emetic activity is the main factor that
has to be met so as to be considered in the “category of SEs.” The diagnosis of staphylococcal foodborne
poisoning could be confirmed by detecting bacterial toxins in foods by immunological tools or mass
spectrometry-based methods. However, additional laboratory models are needed to evaluate the emetic
Staphylococcus 217
activity (just animal models) as superantigenic activity (both animal and cell models). Lack of progress
in elucidating the mechanism of the emetic activity of SEs can be attributed to the lack of convenient and
appropriate animal models. Thus, the commonly used animal models are relatively insensitive to the SEs.
The most susceptible animal species are monkeys, followed by house musk shrews and ferrets.
Moreover, pigs, piglets, goats, dogs, and cats have been used. The main disadvantage of using these later
models remains similar to the monkey models—high cost and short supply in the available tools for the
study of SE-associated immunopathology.
Others animals such as rabbits, rats, and mice are less susceptible to SEs or their response to SEs are not
specific or they are not vomit-competent species. These animals are mainly used as a model for the study of
superantigenicity, pyrogenicity, capacity to enhance endotoxin shock, and lethality. The house musk shrew
and the ferret appear to be valuable animal models for studying the emetic activity of SEs. Comparative
studies using monkeys, house musk shrews, and ferrets to assess the emetic activity of SEs will lead to an
understanding of the molecular basis of the emesis caused by SEs.59
Cells derived from animal tissue are attractive models for studying superantigenic activity. There are
several advantages of cell culture over animal model: simple host–SEs interaction, easy, rapid, relatively
inexpensive compared to animal models, and ethically more acceptable to assays using animals. But this
is not an appropriate model for testing the emetic activity of SEs.
Finally, animal models could be useful for additional identification of SEs or the identification of
staphylococcal strains that produce an unidentified enterotoxin. For this reason, when other methods
become available for all SEs involved in staphylococcal foodborne poisoning, experimental models will
not be employed for routine analysis, but only in special cases to confirm outbreak due to SEs.
Acknowledgments
This work has been funded by the Spanish Instituto Nacional de Investigación Agraria y Agroalimentaria
(INIA) and the Spanish Comisión Interministerial de Ciencia y Tecnología with the projects RTA-2013-
00070-C03-03 and Carnisenusa CSD2007-00016, Consolider Ingenio 2010, and GRU09162 of the Junta
de Extremadura and FEDER.
REFERENCES
1. Irlinger, F., Safety assessment of dairy microorganisms: Coagulase-negative staphylococci, Int. J. Food
Microbiol., 126, 302, 2008.
2. National Center for Biotechnology Information (NCBI), http://www.ncbi.nlm.nih.gov/Taxonomy/
Browser/wwwtax.cgi?mode=Root. Accessed April 30, 2015.
3. Tong, S.Y.C. et al., Novel staphylococcal species that form part of a Staphylococcus aureus-related
complex: The non-pigmented Staphylococcus argenteus sp. nov. and the non-human primate-associated
Staphylococcus schweitzeri sp. nov., Int. J. Syst. Evol. Microbiol., 65, 15, 2015.
4. Podkowik, M. et al., Enterotoxigenic potential of coagulase-negative staphylococci, Int. J. Food
Microbiol., 163, 34, 2013.
5. Wertheim, H.F. et al., The role of nasal carriage in Staphylococcus aureus infections, Lancet Infect.
Dis., 5, 751, 2005.
6. Hennekinne, J.A., De Buyser, M.L. and Dragacci, S., Staphylococcus aureus and its food poisoning
toxins: Characterization and outbreak investigation, FEMS Microbiol. Rev., 36, 815, 2012.
7. Le Loir, Y., Baron, F. and Gautier, M., Staphylococcus aureus and food poisoning, Genet. Mol. Res., 2,
63, 2003.
8. Bautista, L. et al., A quantitative study of enterotoxin production by sheep milk staphylococci, Appl.
Environ. Microbiol., 54, 566, 1988.
9. Vernozy-Rozand, C. et al., Enterotoxin production by coagulase-negative staphylococci isolated from
goats’ milk and cheese, Int. J. Food Microbiol., 30, 271, 1996.
10. Marín, M.E., de la Rosa, M.C. and Cornejo, I., Enterotoxigenicity of Staphylococcus strains isolated
from Spanish dry-cured hams, Appl. Environ. Microbiol., 58, 1067, 1992.
218 Laboratory Models for Foodborne Infections
11. Rodríguez, M. et al., Gram-positive, catalase-positive cocci from dry cured Iberian ham and their
enterotoxigenic potential, Appl. Environ. Microbiol., 62, 1897, 1996.
12. Zell, C. et al., Characterization of toxin production of coagulase-negative staphylococci isolated from
food and starter cultures, Int. J. Food Microbiol., 127, 246, 2008.
13. Even, S. et al., Low occurrence of safety hazards in coagulase negative staphylococci isolated from
fermented foodstuffs, Int. J. Food Microbiol., 139, 87, 2010.
14. Argudin, M.A., Mendoza, M.C. and Rodicio, M.R., Food poisoning and Staphylococcus aureus entero-
toxins, Toxins, 2, 1751, 2010.
15. EFSA, The European Union summary report on trends and sources of zoonoses, zoonotic agents and
food-borne outbreaks in 2013, EFSA J., 13, 3991, 2015.
16. Scallan, E., Foodborne illness acquired in the United States—Major pathogens, Emerg. Infect. Dis., 17,
7, 2011.
17. Kadariya, J., Smith, T.C. and Thapaliya, D., Staphylococcus aureus and staphylococcal food-borne
disease: An ongoing challenge in public health, BioMed Res. Int., 2014, 9, Article ID 827965, 2014.
18. Schmitt, M., Schuler-Schmid, U. and Scmidt-Lorenz, W., Temperature limits of growth, TNase, and
enterotoxin production of Staphylococcus aureus strains isolated from foods, Int. J. Food Microbiol.,
11, 1, 1990.
19. Bergdoll, M.S., Staphylococcus aureus, in Foodborne Bacterial Pathogens, p. 463, Doyle, M.P. (Ed.),
Marcel Dekker, New York, 1989.
20. Aydin, A., Sudagidan, M. and Muratoglu, K., Prevalence of staphylococcal enterotoxins, toxin genes
and genetic-relatedness of foodborne Staphylococcus aureus strains isolated in the Marmara region of
Turkey, Int. J. Food Microbiol., 148, 99, 2011.
21. Balaban, N. and Rasooly, A., Staphylococcal enterotoxins, Int. J. Food Microbiol., 61, 1, 2000.
22. Tranter, H.S., Foodborne staphylococcal illness, Lancet, 336, 1044, 1990.
23. Bergdoll, M.S. and Wong, A.C.L., Staphylococcal intoxications, in Infections and Intoxications, p. 523,
Riemann H.P. and Cliver, D.O. (Eds.), Academic Press, San Diego, CA, 2006.
24. Murray, R.J., Recognition and management of Staphylococcus aureus toxin-mediated disease, Intern.
Med. J., 2, S106, 2005.
25. Hennekinne, J.A. and Dragacii, S., Staphylococcal enterotoxins, in Security Sensitive Microbes and
Toxins, Liu, D. (Ed.), p. 533, CRC Press Taylor & Francis Group, Boca Raton, FL, 2014.
26. Otto, M., Staphylococcus aureus toxins, Curr. Opin. Microbiol., 17, 32, 2014.
27. Lina, G. et al., International Nomenclature Committee for Staphylococcal Superantigens, Standard
nomenclature for the superantigens expressed by Staphylococcus, J. Infect. Dis., 189, 2334, 2004.
28. Omoe, K. et al., Emetic potentials of newly identified staphylococcal enterotoxin-like toxins, Infect.
Immun., 81, 3627, 2013.
29. Ortega, E. et al., Multiple roles of Staphylococcus aureus enterotoxins: Pathogenicity, superantigenic
activity, and correlation to antibiotic resistance, Toxins, 2, 2117, 2010.
30. Thomas, D.Y. et al., Staphylococcal enterotoxin-like toxins U2 and V, two new staphylococcal superan-
tigens arising from recombination within the enterotoxin gene cluster, Infect. Immun., 74, 4724, 2006.
31. Wilson, G.J. et al., A novel core genome-encoded superantigen contributes to lethality of community-
associated MRSA necrotizing pneumonia, PLoS Pathog., 7, e1002271, 2011.
32. Rasooly, R. and Hernlem, B.J., CD154 as a potential early molecular biomarker for rapid quantification
analysis of active Staphylococcus enterotoxin A, FEMS Immunol. Med. Microbiol., 64, 169, 2012.
33. Principato, M.A. and Qian, B.F., Staphylococcal enterotoxins in the etiopathogenesis of mucosal auto-
immunity within the gastrointestinal tract, Toxins, 6, 1471, 2014.
34. Hennekinne, J.A. et al., Intralaboratory validation according to the EN ISO 16 140 Standard of the
Vidas SET2 detection kit for use in official controls of staphylococcal enterotoxins in milk products, J.
Appl. Microbiol., 102, 1261, 2007.
35. Nagaraj, S. et al., Development and evaluation of a novel combinatorial selective enrichment and mul-
tiplex PCR technique for molecular detection of major virulence-associated genes of enterotoxigenic
Staphylococcus aureus in food samples, J. Appl. Microbiol., 116, 435, 2013.
36. Johler, S. et al., Further evidence for staphylococcal food poisoning outbreaks caused by egc-encoded
enterotoxins, Toxins, 7, 997, 2015.
37. Baumgartner, A., Niederhauser, I. and Johler, S., Virulence and resistance gene profiles of Staphylococcus
aureus strains isolated from ready-to-eat foods, J. Food Prot., 77, 1232, 2014.
Staphylococcus 219
38. Bryan, F.L., Guzewich, J.J. and Todd, E.C.D., Surveillance of foodborne disease II. Summary and pre-
sentation of descriptive data and epidemiologic patterns; their value and limitations, J. Food Prot., 60,
567, 1997.
39. Kérouanton, A. et al., Characterization of Staphylococcus aureus strains associated with food poisoning
outbreaks in France, Int. J. Food Microbiol., 115, 369, 2007.
40. Anonymous, EN ISO 6888-1, Microbiology of food and animal feeding stuffs—Horizontal method for
the enumeration of coagulase-positive staphylococci (Staphylococcus aureus and other species)—Part 1:
Technique using Baird-Parker agar medium, European Committee for Standardization, Brussels, 1999.
41. Anonymous, EN ISO 6888-2, Microbiology of food and animal feeding stuffs—Horizontal method for
the enumeration of coagulase-positive staphylococci (Staphylococcus aureus and other species)—Part
2: Technique using rabbit plasma fibrinogen agar medium, European Committee for Standardization,
Brussels, 1999.
42. Anonymous, EN ISO 6888-3, Microbiology of food and animal feeding stuffs—Horizontal method for
the enumeration of coagulase-positive staphylococci (Staphylococcus aureus and other species)—Part 3:
Detection and MPN technique for low numbers, European Committee for Standardization, Brussels, 2003.
43. Sospedra, I., Soriano, J.M. and Mañés, J., Enterotoxinomics: The omic sciences in the study of staphy-
lococcal toxins analyzed in food matrices, Food Res. Int., 54, 1052, 2013.
44. Dupuis, A. et al., Protein Standard Absolute Quantification (PSAQ) for improved investigation of staph-
ylococcal food poisoning outbreaks, Proteomics, 8, 4633, 2008.
45. Schlosser, G. et al., Coupling immunomagnetic separation on magnetic beads with matrix-assisted laser
desorption ionization-time of flight mass spectrometry for detection of staphylococcal enterotoxin,
Appl. Environ. Microbiol., 73, 6945, 2007.
46. Sospedra, I. et al., Analysis of staphylococcal enterotoxin A in milk by matrix-assisted laser desorption/
ionization-time of flight mass spectrometry, Anal. Bioanal. Chem., 400, 1525, 2011.
47. Nedelkov, D., Rasooly, A., and Nelson, R.W., Multitoxin biosensor mass spectrometry analysis: A new
approach for rapid, real-time, sensitive analysis of staphylococcal toxins in food, Int. J. Food Microbiol.,
60, 1, 2000.
48. Sospedra, I. et al., Rapid whole protein quantification of staphylococcal enterotoxin B by liquid chroma-
tography, Food Chem., 133, 163, 2012.
49. Bhunia, A.K., Animal and cell culture models to study foodborne pathogen interaction, in Foodborne
Microbial Pathogens, Mechanisms and Pathogenesis, Heldman, D.R. (Ed.), p. 113, Springer, New York,
2008.
50. Hu, D.L. and Nakane, A., Mechanisms of staphylococcal enterotoxin-induced emesis, Eur. J.
Pharmacol., 722, 95, 2014.
51. Bergdoll, M.S. and Wong, A.C.L. Staphylococcal intoxications, in Foodborne Infection and Intoxication,
p. 523, Riemann, H.P. and Cliver, D.O. (Eds.), Academic Press, London, UK, 2006.
52. Ono, H.K. et al., Identification and characterization of two novel staphylococcal enterotoxins, types S
and T, Infect. Immun., 76, 4999, 2008.
53. Munson, S.H. et al., Identification and characterization of staphylococcal enterotoxin types G and I from
Staphylococcus aureus, Infect. Immun., 66, 3337, 1998.
54. Su, Y.C. and Wong, A.C.L., Identification and purification of a new staphylococcal enterotoxin, H.,
Appl. Environ. Microbiol., 61, 1438, 1995.
55. Bergdoll, M.S., Monkey feeding test for staphylococcal enterotoxin, Methods Enzymol., 165, 324, 1988.
56. Sugiyama, H., Bergdoll, M.S. and Dack, G.M., Early development of a temporary resistance to the
emetic action of staphylococcal enterotoxin, J. Infect. Dis., 111, 233, 1962.
57. Ono, H.K. et al., Submucosal mast cells in the gastrointestinal tract are a target of staphylococcal entero-
toxin type A, FEMS Immunol. Med. Microbiol., 64, 392, 2012.
58. Krakauer, T. and Stiles, B.G., The staphylococcal enterotoxin (SE) family SEB and siblings, Virulence,
4, 759, 2013.
59. Hu, D.L. et al., Induction of emetic response to staphylococcal enterotoxins in the house musk shrew
(Suncus murinus), Infect. Immun., 71, 567, 2003.
60. Omoe, K. et al., Characterization of novel staphylococcal enterotoxin-like toxin type P, Infect. Immun.,
73, 5540, 2005.
61. Maina, E.K. et al., Staphylococcal enterotoxin A has potent superantigenic and emetic activities but not
diarrheagenic activity, Int. J. Med. Microbiol., 302, 88, 2012.
220 Laboratory Models for Foodborne Infections
62. Hu, D.L. et al., Emesis in the shrew mouse (Suncus murinus) induced by peroral and intraperitoneal
administration of staphylococcal enterotoxin A, J. Food Protect., 62, 1350, 1999.
63. Hu, D.L. et al., Immunization with a nontoxic mutant of staphylococcal enterotoxin A, SEAD227A,
protects against enterotoxin-induced emesis in house musk shrews, J. Infect. Dis., 199, 302, 2009.
64. Andrews, P.L.R., and Davies C.J., Physiology of emesis induced by anti-cancer therapy, in Serotonin
and the Scientific Basis of Anti-emetic Therapy, Reynolds, D.J.M., Andrews, P.L.R., and Davis, C.J.
(Eds.), p. 25, Oxford Clinical Communications, Oxford, UK, 1995.
65. King, G.L., Animal models in the study of vomiting, Can. J. Physiol. Pharmacol., 68, 260, 1990.
66. Wright, A., Andrews, P.L.R. and Titball, R.W., Induction of emetic, pyrexic, and behavioral effects of
Staphylococcus aureus enterotoxin B in the ferret, Infect. Immun., 68, 2386, 2000.
67. Hui, J. et al., Staphylococcus aureus enterotoxin C2 mutants: Biological activity assay in vitro, J. Ind.
Microbiol. Biotechnol., 35, 975, 2008.
68. Taylor, S.L. et al., Emetic action of staphylococcal enterotoxin A on weanling pigs, Infect. Immun., 36,
1263, 1982.
69. Van Gessel, Y.A. et al., Functional piglet model for the clinical syndrome and post-mortem findings
induced by staphylococcal enterotoxin B, Exp. Biol. Med. (Maywood), 229, 1061, 2004.
70. Hudson, L.C. et al., Sublethal staphylococcal enterotoxin B challenge model in pigs to evaluate protec-
tion following immunization with a soybean-derived vaccine, Clin. Vaccine Immunol., 20, 24, 2013.
71. Inskeep, T.K. et al., Oral vaccine formulations stimulate mucosal and systemic antibody responses
against staphylococcal enterotoxin B in a piglet model, Clin. Vaccine Immunol., 17, 1163, 2010.
72. Van Miert, A.S., Van Duin, C.T. and Schotman, A.J., Comparative observations of fever and associated
clinical hematological and blood biochemical changes after intravenous administration of staphylococ-
cal enterotoxins B and F (toxic shock syndrome toxin-1) in goats, Infect. Immun., 46, 354, 1984.
73. Kocandrle, V., Houttuin, E. and Prohaska, J.V., Acute hemodynamic and gastrointestinal changes pro-
duced by staphylococcal exotoxin and enterotoxin in dogs, J. Surg. Res., 6, 50, 1966.
74. Clark, W.G., Vanderhooft, G.F. and Borison, H.L., Emetic effect of purified staphylococcal enterotoxin
in cats, Proc. Soc. Exp. Biol. Med., 111, 205, 1962.
75. Wang, X. et al., Enhancement of superantigen activity and antitumor response of staphylococcal entero-
toxin C2 by site-directed mutagenesis, Cancer Immunol. Immunother., 58, 677, 2009.
76. Clark, W.G. and Page, J.S., Pyrogenic responses to staphylococcal enterotoxins A and B in cats, J.
Bacteriol., 96, 1940, 1968.
77. Pinchuk, I.V., Beswick, E.J. and Reyes, V.E., Staphylococcal enterotoxins, Toxins, 2, 2177, 2010.
78. Huang, W.T., Lin, MT. and Won, S.J., Staphylococcal enterotoxin A-induced fever is associated with
increased circulating levels of cytokines in rabbits, Infect. Immun., 65, 2656, 1997.
79. Orwin, P.M. et al., Biochemical and biological properties of staphylococcal enterotoxin K, Infect.
Immun., 69, 360, 2001.
80. Orwin, P.M. et al., Characterization of Staphylococcus aureus enterotoxin L, Infect. Immun., 71, 2916,
2003.
81. Orwin, P.M. et al., Characterization of a novel staphylococcal enterotoxin-like superantigen, a member
of the group V subfamily of pyrogenic toxins, Biochemistry, 41, 14033, 2002.
82. Krakauer, T., Buckley, M. and Fisher, D., Murine models of staphylococcal enterotoxin B-induced toxic
shock, Mil. Med., 175, 917, 2010.
83. Lindsay, C.D. and Griffiths, G.D., Addressing bioterrorism concerns: Options for investigating the
mechanism of action of Staphylococcus aureus enterotoxin B, Hum. Exp. Toxicol., 32, 606, 2013.
84. Pérez-Bosque, A. and Moretó, M., A rat model of mild intestinal inflammation induced by Staphylococcus
aureus enterotoxin B, Proc. Nutr. Soc., 69, 447, 2010.
85. Beery, J.T. et al., Effects of staphylococcal enterotoxin A on the rat gastrointestinal tract, Infect. Immun.,
44, 234, 1984.
86. Yang, W.X. et al., Decreased Vβ8.2 T-cells in neonatal rats exposed prenatally to staphylococcal entero-
toxin B are further deleted by restimulation in an in vitro cultured thymus, Mol. Med. Rep., 10, 989, 2014.
87. Zhang, T. et al., Staphylococcal enterotoxin B administration during pregnancy imprints the increased
CD4:CD8 T-cell ratio in the peripheral blood from neonatal to adult offspring rats, J. Med. Microbiol.,
64, 1, 2015.
88. Roy, C.J. et al., Human leukocyte antigen-DQ8 transgenic mice: A model to examine the toxicity of
aerosolized staphylococcal enterotoxin B, Infect. Immun., 73, 2452, 2005.
Staphylococcus 221
89. Tilahun, M.E. et al., Chimeric anti-staphylococcal enterotoxin B antibodies and lovastatin act synergis-
tically to provide in vivo protection against lethal doses of SEB, PLoS One, 6, e-27203, 2011.
90. Spiekermann, G.M. and Nagler-Anderson, C., Oral administration of the bacterial superantigen staphy-
lococcal enterotoxin B induces activation and cytokine production by T cells in murine gut-associated
lymphoid tissue, J. Immunol., 161, 5825, 1998.
91. Stiles, B.G. et al., Correlation of temperature and toxicity in murine studies of staphylococcal enterotox-
ins and toxic shock syndrome toxin 1, Infect. Immun., 67, 1521, 1999.
92. Chen, J.Y. et al., Increased susceptibility to staphylococcal enterotoxin B intoxication in mice primed
with actinomycin D, Infect. Immun., 62, 4626, 1994.
93. Miethke, T. et al., T cell-mediated lethal shock triggered in mice by the superantigen staphylococcal
enterotoxin B: Critical role of tumor necrosis factor, J. Exp. Med., 175, 91, 1992.
94. Huzella, L.M. et al., Central roles for IL-2 and MCP-1 following intranasal exposure to SEB: A new
mouse model, Res. Vet. Sci., 86, 241, 2009.
95. Kang, H. et al., Superantigenicity analysis of staphylococcal enterotoxins SElK and SElQ in a mouse
model, RSC Adv., 5, 29684, 2015.
96. Danielsen, E.M., Hansen, G.H. and Karlsdottir, E., Staphylococcus aureus enterotoxins A and B:
Binding to the enterocyte brush border and uptake by perturbation of the apical endocytic membrane
traffic, Histochem. Cell Biol., 139, 513, 2013.
97. Edwards, L.A. et al., Enterotoxin-producing staphylococci cause intestinal inflammation by a combina-
tion of direct epithelial cytopathy and superantigen-mediated T cell activation, Inflamm. Bowel Dis., 18,
624, 2012.
98. Fraser, J. et al., Superantigens-powerful modifiers of the immune system, Mol. Med. Today, 6, 125, 2000.
99. Krakauer, T., Sulfasalazine attenuates staphylococcal enterotoxin B-induced immune responses, Toxins,
7, 553, 2015.
100. Boles, J.W. et al., Generation of protective immunity by inactivated recombinant staphylococcal entero-
toxin B vaccine in nonhuman primates and identification of correlates of immunity, Clin. Immunol.,
108, 51, 2003.
101. Omoe, K. et al., Biological properties of staphylococcal enterotoxin-like toxin type R, Infect. Immun.,
72, 3664, 2004.
102. René-Trouillefou, M. et al., Staphylococcal enterotoxin A: Partial unfolding caused by high pressure or
denaturing agents enhances superantigenicity, Biochim. Biophys. Acta, 1804, 1322, 2010.
103. Jarraud, S. et al., egc, a highly prevalent operon of enterotoxin gene, forms a putative nursery of supe-
rantigens in Staphylococcus aureus, J. Immunol., 166, 669, 2001.
104. Rasooly, R. and Hernlem, B.J., Quantitative analysis of Staphylococcus enterotoxin a by differential
expression of IFN-γ in splenocyte and CD4 + T-cells, Sensors, 14, 8869, 2014.
105. Liu, Y. et al., Staphylococcal enterotoxin H induced apoptosis of bovine mammary epithelial cells in
vitro, Toxins, 6, 3552, 2014.
106. Herrmann, T., Accolla, R.S. and MacDonald, H.R., Different staphylococcal enterotoxins bind prefer-
entially to distinct major histocompatibility complex class II isotypes, Eur. J. Immunol., 19, 2171–1989.
107. Reck, B. et al., Protection against the staphylococcal enterotoxin-induced intestinal disorder in the mon-
key by anti-idiotypic antibodies, Proc. Natl. Acad. Sci. USA, 85, 3170, 1988.
108. Reiser, R.F. et al., Identification, purification, and some physicochemical properties of staphylococcal
enterotoxin C3, Infect. Immun., 45, 625, 1984.
109. He, C., Narayanan, P.K. and Fort, M.M., Assessment of the performance of three multiplex array panels
for the detection of circulating cytokines and chemokines in naive, LPS, and SEB-treated cynomolgus
macaques, Toxicol. Pathol., 42, 286, 2014.
110. Stiles, J.W. and Denniston, J.C., Response of the rhesus monkey, Macaca mulatta, to continuously
infused staphylococcal enterotoxin B, Lab. Invest., 25, 617, 1971.
111. Sheahan, D.G. et al., The effect of staphylococcal enterotoxin on the epithelial mucosubstances of the
small intestine of rhesus monkeys, Am. J. Pathol., 60, 1, 1970.
112. Krakauer, T., et al., Superantigen-induced cytokine release from whole-blood cell culture as a functional
measure of drug efficacy after oral dosing in nonhuman primates, Res. Vet. Sci., 83, 182, 2007.
14
Streptococcus
Dongyou Liu
CONTENTS
14.1 Introduction................................................................................................................................... 223
14.1.1 Classification and Morphology......................................................................................... 224
14.1.1.1 Classification..................................................................................................... 224
14.1.1.2 Morphology....................................................................................................... 226
14.1.1.3 Genomics.......................................................................................................... 226
14.1.2 Biology and Epidemiology............................................................................................... 226
14.1.3 Clinical Features and Pathogenesis.................................................................................. 227
14.1.3.1 Pharyngitis........................................................................................................ 227
14.1.3.2 Scarlet Fever...................................................................................................... 228
14.1.3.3 Impetigo............................................................................................................ 228
14.1.3.4 Acute Rheumatic Fever (ARF)......................................................................... 228
14.1.3.5 Acute Poststreptococcal Glomerulonephritis (APSGN).................................. 228
14.1.3.6 Pediatric Autoimmune Neuropsychiatric Disorders Associated with
Streptococcal Infections (PANDAS)��������������������������������������������������������������� 228
14.1.4 Diagnosis.......................................................................................................................... 229
14.1.5 Treatment and Prevention................................................................................................. 229
14.2 Laboratory Models........................................................................................................................ 230
14.2.1 Animal Models................................................................................................................. 230
14.2.1.1 Rodents............................................................................................................. 230
14.2.1.2 Chinchilla.......................................................................................................... 232
14.2.1.3 Nonhuman Primates......................................................................................... 232
14.2.1.4 Insects............................................................................................................... 232
14.2.1.5 Caenorhabditis elegans Nematode.................................................................. 232
14.2.1.6 Zebrafish........................................................................................................... 232
14.2.2 In Vitro Models................................................................................................................. 232
14.3 C onclusion..................................................................................................................................... 233
References............................................................................................................................................... 233
14.1 Introduction
The genus Streptococcus comprises a large group of Gram-positive bacteria that have been known to
cause human diseases from ancient times. A quick review of historical records indicates that in 1553, a
rash (then termed “rossalia,” with the whole body covered by numerous spots of large and small, fiery,
and red) that differs from measles was first described by Giovanni Filippo Ingrassias of Italy; in 1565, a
sore throat epidemic was mentioned by Johann Weyer of the Netherlands; in 1578, scarlet fever (show-
ing general weariness, headache, redness of the eyes, sore throat, and fever) was noted by Jean Cottyar;
in 1628, an epidemic showing scarlatinal desquamation, arthritis, and postscarlatinal dropsy and asci-
tes was documented by Daniel Sennert; in 1675, the term “scarlatina” was first used by Sydenham to
223
224 Laboratory Models for Foodborne Infections
describe a disease that is distinct from other exanthemas (e.g., measles); in the 1840s, childbed fever
was linked by Ignac Semmelweis of Hungary to medical personnel failing to wash their hands and who
then transmitted the disease to patients; in 1874, an organism isolated from patients with erysipelas and
wound infections was named “streptococcus” (Greek streptos, a chain; coccos, a berry) by Theodor
Billroth of Austria due to its formation of short chains; in 1879, streptococcus was confirmed by Louis
Pasteur as the etiological agent of puerperal fever, which caused the highest mortality rates of women
and newborns at that time; in 1884, the bacterium isolated from suppurative lesions was defined by
Friedrich Julius Rosenbach as Streptococcus pyogenes (Greek pyo, pus, and genes, forming); shortly
afterward, the previously proposed species names of pyogenes, eryespaltis, scarlatinae, and puerperalis
were united under the single name Streptococcus pyogenes by Andrews and Christie; in 1909, serotype-
specific immunity against streptococci was reported by Meakins; in the 1920s, hemolytic streptococci
that produced a secreted toxin (known as scarlet fever toxin or Dick toxin) were identified by George and
Gladys Dick as the causative agent of sore throat that is accompanied by scarlet fever [1].
To date, more than 50 species have been recognized in the genus Streptococcus, of which nine
(S. pyogenes, S. agalactiae, S. equisimilis, S. bovis, S. anginosus, S. sanguinis, S. mitis, S. mutans,
and S. pneumoniae) have been implicated in human infections. In particular, S. pyogenes (commonly
referred to as group A Streptococcus or GAS) is responsible for a majority of human streptococcal dis-
eases, with clinical manifestations ranging from pharyngitis, impetigo, cellulitis, scarlet fever, puerperal
sepsis, bacteremia, pneumonia, streptococcal toxic shock syndrome (STSS), necrotizing fasciitis, acute
rheumatic fever (ARF), rheumatic heart disease (RHD), to acute poststreptococcal glomerulonephritis
(APSGN). All together, over half a million deaths per year worldwide are attributable to S. pyogenes [2].
This chapter will focus on S. pyogenes, beginning with a brief overview on its classification, morphology,
genomics, biology, epidemiology, clinical features, pathogenesis, diagnosis, treatment, and prevention,
followed by discussion on laboratory models applied to S. pyogenes research.
proteins with high levels of antigenic heterogeneity and are direct determinants of host tissue site prefer-
ences of infection [4].
Developed by Lancefield in 1933, serological typing scheme using group-specific antisera to M pro-
tein (a surface protein that is responsible for colony’s matte appearance) differentiates Streptococcus
β-hemolytic species into Lancefield groups A to X (excluding I and J), with strains from human diseases
classified as group A, those from bovine and dairy sources as group B, those from other animal sources
as group C, and so on. GAS consists of S. pyogenes, which causes both noninvasive and invasive infec-
tions in humans. The noninvasive infections are more common but less severe, as exemplified by strepto-
coccal pharyngitis (strep throat), impetigo, and scarlet fever. The invasive infections are more severe but
less common, as exemplified by STSS, necrotizing fasciitis, pneumonia, and bacteremia. Complications
of GAS infections include ARF and acute glomerulonephritis. Affecting the joints, kidneys, and heart
valves, rheumatic fever results from damages caused by the antibodies that are generated by the host
immune system against the untreated GAS infection. These antibodies cross-react with other proteins
in the body, leading to self-inflicted attack and damage. Indeed, the group A S. pyogenes strains can be
further subdivided into >200 M-types using a combination of serological and molecular typing methods.
Group B Streptococcus (GBS) consists of S. agalactiae, which causes pneumonia, meningitis, and occa-
sional systemic bacteremia in neonates and the elderly. As it also colonizes the intestines and the female
reproductive tract, this bacterium may contribute to premature rupture of membranes during pregnancy
and subsequent transmission to the infant. Group C Streptococci include S. equi (causing strangles in
horses), S. zooepidemicus (infecting cattle and horses, with S. equi being a clonal descendent or biovar
of the ancestral S. zooepidemicus), and S. dysgalactiae (causing pharyngitis and other pyogenic infec-
tions similar to GAS). Group D streptococci consist of enterococcal and nonenterococcal strains. The
former have been reclassified and placed in the genus Enterococcus (including E. faecalis, E. faecium,
E. durans, and E. avium, with E. faecalis being sometimes α-hemolytic and E. faecium being sometimes
β-hemolytic) (see Chapter 10); the latter include S. bovis and S. equinus. Group F streptococci (so-called
“minute hemolytic streptococci”) are represented by S. anginosus or the S. milleri group. Group G strep-
tococci are usually, but not exclusively, β-hemolytic and represented by S. canis that typically occurs in
animals, but may cause infection in humans. Group H streptococci are found in canines and rarely cause
illness unless humans have direct contact with a canine (mouth-to-mouth or canine licking a human
hand) [3].
The serotyping scheme based on T-antigens (trypsin-resistant surface antigens, which are contained
within extended surface pili composed of covalently linked polymers of two or three distinct gene prod-
ucts) divides S. pyogenes strains into ∼20 T-serotypes. However, many S. pyogenes strains have multiple
T-types (e.g., T3/13/B, T8/25/Imp19) [4].
Another classification scheme proposed by Sherman in 1937 incorporating Lancefield grouping and
other criteria divides streptococci into four groups: pyogenic, viridans, lactic, and enterococci. The pyo-
genic division comprises β-hemolytic strains of Lancefield groups A, B, C, E, F, and G. The viridans
division consists of non-β-hemolytic streptococci that are not tolerant to high-pH growth conditions and
salt and do not grow at 10°C. The lactic division includes strains of dairy origin, which are nonhuman
pathogenic. This group differs from the pyogenic group by being non-β-hemolytic, growing at 10°C
but not at 45°C, and failure to grow in broth with 6.5% NaCl. The lactic division was reclassified as
the Lactococcus genus in the mid-1980s. The enterococci division includes Lancefield group D strains,
which are now known as the genus Enterococcus. Members of this genus have the capacity to grow in
broths at high pH (9.6), high salt concentrations (6.5% NaCl), and a wide temperature range (10°C–45°C),
with some enterococci demonstrating β-hemolytic property (see Chapter 10) [3].
Examination of Streptococcus 16S rRNA sequences also permits discrimination of streptococci into
six groups: S. anginosus, S. bovis, S. mitis (including S. pneumoniae), S. mutans, S. pyogenes, and
S. salivarius.
Further, sequencing analysis of the emm gene that encodes the mature M protein molecule provides
additional confirmation on the validity of the traditional M serological typing scheme. Specifically, the
emm sequence-based typing divides S. pyogenes strains into five pattern groups (A–E), with emm pattern
A–C groups accounting for 47% of pharyngitis isolates, but only 8% of impetigo isolates; emm pattern D
group accounting for 50% of impetigo isolates, but only <2% of pharyngitis isolates; and emm pattern E
226 Laboratory Models for Foodborne Infections
group accounting for almost equal fractions of throat and skin infections (52% and 42%, respectively).
Not surprisingly, emm pattern A–C groups are referred to as “throat specialists,” emm pattern D group
as “skin specialists,” and emm pattern E group as “generalists” [4].
Moreover, genetic analysis of pilus genes (particularly the FCT region, with FCT standing for
Fibronectin- and Collagen-binding proteins and T-antigen) uncovers the molecular basis of the serotyp-
ing scheme targeting T-antigens. However, not all S. pyogenes strains express pili, and those with a nega-
tive regulator may be T-nontypable [4].
Multilocus sequence typing (MLST) targeting seven core housekeeping genes (glucose kinase, gki;
glutamine transporter protein, gtr; glutamate racemase, murI; DNA mismatch repair protein, mutS;
transketolase, recP; xanthine phosphoribosyl transferase, xpt; and acetyl coenzyme A acetyltransfer-
ase, yqiL) offers another valuable approach for determination of S. pyogenes strains, with clones of
S. pyogenes being defined by their sequence type (ST) (http://pubmlst.org/spyogenes/). Interestingly,
while most emm pattern A–C strains correspond to same clone (ST) or clonal complex, emm patterns D
and E strains correlate with distant ST [4].
14.1.1.2 Morphology
Streptococci are Gram-positive, facultative anaerobic cocci that often grow in chains or in pairs (due to
the fact that cell division occurs along a single axis). Streptococcus colonies on agar plates are small,
smooth, and moist in appearance. Streptococcal cell wall is largely composed of group-specific carbohy-
drate (the M protein), with Lancefield group A being a polymer of rhamnose and N-acetylglucosamine.
Apart from the M protein, other proteins present on the cell surface include T-protein, SOF, C5a pepti-
dase, collagen-like protein Scl1, GRAB, and protein F. In GAS, the pili (fimbriae) appear as long, flexible
rods protruding up to 3 μm from the cell surface. Structurally, GAS pili are heteropolymers forming a
pilus shaft, which is composed of the major pilin protein subunit (Spy0128) and assembled through a
series of transpeptidase reactions catalyzed by a class B accessory sortase, SrtC (Spy0129) [3].
14.1.1.3 Genomics
The genomes of Streptococcus species are of 1.8–2.3 Mb in size with GC content of about 38.5%,
1700–2300 protein-coding sequences (CDSs), 5–6 rRNA, and 57–67 tRNA encoding genes. Specifically,
S. pyogenes, S. agalactiae, S. pneumoniae, and S. mutans possess genomes of 1,852,442, 2,211,488,
2,160,837, and 2,030,921 bp, respectively. The genome of GAS contains genes that encode proteins
secreted into the extracellular fluid during growth (e.g., erythrogenic toxin, streptolysin O, streptoly-
sin S, proteinase, streptokinase, DNase, RNase, hyaluronidase, CAMP factor, streptococcal inhibitor
of complement, immunogenic secreted protein, and superantigens). These proteins are mostly virulence
factors with important roles in colonization, invasion, spreading, and pathogenesis. The genome of
pathogenic GAS strains also harbors a pathogenicity island located in the FCT region, which encodes
fibronectin-binding proteins, collagen-binding proteins, and T-antigens (pilus subunit genes). Among
GAS isolates, the FCT region displays considerable genetic diversity, with nine different FCT variants
identified [4]. Additionally, the S. pyogenes genome consists of some metabolic pathways (e.g., complete
glycolytic pathway, fatty acid synthesis, nucleotide synthesis and transport, and carbohydrate transport
and metabolism), but lacks a complete tricarboxylic acid cycle pathway. Many Streptococcus species
contain bacteriophages, with 18 prophages (ranging from 38 to 41 kb in size, encoding from 42 to 66
genes each) being described in S. pneumoniae.
S. pyogenes (GAS) is not considered part of normal flora in humans, despite its presence in 5%–20%
of healthy/asymptomatic individuals. As an exclusively human pathogen, GAS enters into human host
via oral cavity, skin, and wounds, and typically attaches to the epithelial surfaces of the throat and
skin, as well as the vagina and rectum, on which it forms large microcolony aggregates. Additionally,
GAS may organize as biofilms and hide inside nonphagocytic cells. These enhance its survival under
unfavorable or stressful conditions including antimicrobial therapy, and contribute to its spread to other
individuals [5,6].
GAS is commonly transmitted through two routes: foodborne or airborne. Foodborne GAS epidemics
often result from consumption of inappropriately prepared/cooked raw milk, cold salads, eggs, mayon-
naise, tuna, potatoes, cheese, conch, and other ingredients that are inadvertently contaminated by GAS
from infected animals (e.g., cows with streptococcal mastitis) or food handlers (who have sore throat, or
have infected skin lesions on hands/arms, or are asymptomatic carriers) [7–9]. Airborne GAS epidem-
ics are due to inhalation of respiratory/saliva droplets from carriers who disseminate the bacterium via
sneeze or cough. Another means for GAS to spread is through person-to-person skin contact. External
factors favoring GAS disease outbreaks include crowded settings (e.g., military training centers), mass
consumption of contaminated foods, and hospital acquisition (e.g., puerperal sepsis). Intrinsic factors
include the emergence of dominant clones [e.g., M1T1 clone that acquires three regions of heterologous
DNA: a 36-kb chromosomal region encoding the toxins SLO and NAD-glycohydrolase and two bacte-
riophages encoding the DNase Sda1 and the superantigen SpeA (streptococcal pyrogenic exotoxin A);
M3 clone that acquires prophages encoding phospholipase A2 and SpeA and the duplication of four
amino acids in the N-terminal region of the M3 protein].
Interestingly, S. pyogenes pharyngitis tends to predominate in temperate regions, with seasonal
peak in winter, whereas S. pyogenes impetigo is mostly present in tropical and subtropical regions,
with seasonal peak in summer. In addition, there are notable links of certain emm types to particular
clinical diseases, including the association of emm types 1, 3, 5, 6, 12, 14, 17, 19, and 24 with pharyn-
gitis; emm types 33, 41, 42, 52, 53, and 70 with impetigo; emm types 1, 3, 5, 6, 11, 12, 14, 17, 18, 19,
24, 27, 29, 30, 32, and 41 with ARF; emm types 1, 4, 12, 49, 55, 57, and 60 with APSGN; emm type 28
with puerperal sepsis; emm types 1, 3, and 28 with necrotizing fasciitis; and emm types 1 and 3 with
STSS [2].
14.1.3.1 Pharyngitis
Commonly known as “strep sore throat,” GAS pharyngitis presents with sudden-onset fever accompa-
nied by sore throat, and obvious inflammation in the pharynx and tonsils. Other symptoms may include
general malaise, headache, nausea, abdominal pain, vomiting, patchy exudates, and cervical lymph node
adenopathy. Patients with uncomplicated GAS pharyngitis usually recover within 7–14 days [2].
228 Laboratory Models for Foodborne Infections
14.1.3.2 Scarlet Fever
When pharyngitis involves a GAS strain that generates bacteriophage-encoded streptococcal pyogenic
exotoxins (notably SpeA), scarlet fever (also known as scarlatina) may emerge, with a deep red, finely
papular, erythematous rash (strawberry tongue) and exudative pharyngitis. Scarlet fever tends to occur in
children of 4–8 years but rarely in adults. Pharyngitis and soft tissue infection at a surgical site (surgical
scarlet fever) are most commonly associated with scarlet fever [2].
14.1.3.3 Impetigo
As a contagious GAS infection of the skin, impetigo (or pyoderma, which is a term for a localized
purulent infection of the skin and is used synonymously with streptococcal impetigo and impetigo con-
tagiosa) shows pustules that enlarge and rupture to form thick, honey-colored scabs. Spread through
direct skin contact, impetigo mainly affects children with poor hygiene and crowded living conditions
in tropical and subtropical climate. When GAS moves to deeper layers of the skin, erysipelas and cel-
lulitis may appear. Further spreading of GAS to the fascia may lead to necrotizing fasciitis (flesh-eating
disease), which is severely invasive, potentially fatal disease in the absence of immediate surgical and
medical intervention. Another severe invasive disease due to GAS is STSS, which often develops within
24–72 h of minor nonpenetrating trauma resulting in hematoma, deep bruise to the calf, or following
muscle strain, in addition to suction lipectomy, hysterectomy, vaginal delivery, bunionectomy, and bone
pinning [2].
activity for dysregulation of coagulation; generates SLS, SLO, streptococcal pyrogenic exotoxins A
and C (SpeA and SpeC, which are superantigens responsible for scarlet fever and STSS), and Nga for
cytotoxic and cytolytic activity toward various host cell types, including neutrophils, platelets, and
subcellular organelles [15,16].
14.1.4 Diagnosis
Isolation of GAS from throat, food specimens, or swabs from kitchen utensils and surfaces represents a
useful initial step for the diagnosis of streptococcal pharyngitis and hemolytic streptococci.
GAS may be grown in a highly nutritious medium consisting of meat extract, peptones, dextrose, and
salts (e.g., 5% horse-blood agar), with the formation of colonies showing β-hemolysis. The medium may
be modified to enhance the production of some GAS proteins. For instance, addition of a digested RNA
fraction increases streptolysin S production; supplement with reducing agents (e.g., glutathione) favors
streptolysin O production; a slightly acidic pH strengthens the production of the cysteine proteinase pre-
cursor; and addition of hyaluronate improves the production of hyaluronidase [3].
The resulting GAS isolates are then typed by using serotype-specific antisera raised against the M pro-
tein (an immunodominant surface antigen and key virulence determinant), T-antigens (trypsin-resistant
surface antigens, which are contained within extended surface pili), or SOF (an LPXTG-anchored,
multifunctional surface protein that binds fibronectin and enzymatically disrupts the structure of high-
density lipoproteins present in blood). Further analysis with pulsed-field gel electrophoresis (PFGE)
using SmaI is also valuable.
More recently, typing of GAS strains is conducted according to the sequence of the 5′ variable region
of the emm gene encoding the M protein, leading to the identification of >200 emm types. Of these, emm
pattern A–C strains are considered as “throat specialists” (causing pharyngitis), emm pattern D strains
as “skin specialists” (causing impetigo), and emm pattern E strains as “generalists” (causing both phar-
yngitis and impetigo) [4].
Use of MLST technique allows definition of S. pyogenes clones by their ST (http://pubmlst.org/
spyogenes/). Interestingly, most emm pattern A–C strains closely correspond to a single clone (ST) or
clonal complex [4].
14.2 Laboratory Models
As a strictly human pathogen, S. pyogenes is implicated in a range of clinical diseases, including
(1) local, lesional diseases (inflammation) in soft tissue, (2) both local and systemic diseases associated
with streptococcal toxins, and (3) immune dysfunction due to streptococcal antigens. To complicate the
situation further, S. pyogenes encompasses a diversity of strains with varied virulence potential. In spite
of our long-standing efforts to uncover the secrets about this ever-present pathogen, our understanding
of GAS and its pathogenesis and immune evasion is far from being adequate. Use of laboratory models
is vital in helping address this conundrum. Indeed, over the past two decades, a number of in vivo and
in vitro models based on both laboratory animals and cell lines have been exploited, with the ultimate
goal of elucidating the molecular basis of S. pyogenes pathogenesis and other perplexing issues [17–20].
14.2.1 Animal Models
14.2.1.1 Rodents
Being small, relatively inexpensive to maintain, easy to handle, along with rapid reproduction and short
life span, rodents (e.g., mice and rats) represent ideal animal models for disease investigations includ-
ing GAS infections. It is no surprise that a diverse range of model systems have been established using
rodents.
airway colonization and allows quick development of clinical signs in infected mice. S. pyogenes viru-
lence is determined by lethality and the kinetics of clearance in the spleen, liver, and lungs (via CFU
enumeration) [5].
In addition, sera from Lewis rats immunized with a whole cell lysate of S. pyogenes (showing quantifi-
able alterations in behavioral and motor functions) cross-react with brain tissue in vivo and with dopa-
mine and serotonin receptors in vitro. Direct perfusion of purified IgG from these cross-reacting sera
into the striatum region of the brain could reproduce symptomatology of PANDAS [17,18,21].
14.2.1.2 Chinchilla
Chinchilla otitis media model involves inoculation of 105 CFU S. pyogenes via transbullar injection into
the ear of a chinchilla, with tympanic membrane and inner ear inflammation as well as serous middle ear
effusion evident by day 2 as examined by otoscopy. The infection can be monitored through enumera-
tion of CFUs recovered from macroscopic structures and also serous effusion, and observation of animal
mortality over the 7-day period [20].
14.2.1.3 Nonhuman Primates
Nonhuman primate pharyngitis model offers a more reliable approach for assessing S. pyogene-induced
pharyngitis than other animals such as rodents. Several nonhuman primate species (e.g., baboon, rhesus
monkey, chimpanzee, and cynomolgus macaque) appear to be susceptible to S. pyogenes colonization in
the oropharynx, with the production of type-specific M protein antibodies in serum [20].
Nonhuman primate sepsis model involves intravenous infusion of baboons (Papio cynocephalus cyno-
cephalus) under light anesthesia with 1010 CFU S. pyogenes over a 2-h period followed by analyses of
physiology and blood chemistry over a 10-h period. The infection is evaluated by body temperature,
heart rate, mean systemic arterial blood pressure, serum chemistry, and cytokine profiles. It appears that
blocking the cytokine TNF-α with a therapeutic monoclonal antibody helps improve both mean arterial
blood pressure and survival and reduce hypotension and multiorgan failure. This model is valuable for
the study of streptococcal toxic-shock syndrome [20].
14.2.1.4 Insects
Silkworm model involves injection of approximately 9 × 108 CFU S. pyogenes into the hemolymph (com-
parable to the bloodstream) of invertebrate silkworm (Bombyx mori) followed by monitoring survival for
5 days at 27°C. Wax worm model differs from silkworm model in that wax worm (Galleria mellonella)
allows infection to be conducted at 37°C [20].
14.2.1.6 Zebrafish
Zebrafish myonecrosis model involves inoculation with S. pyogenes via IM injection into the dorsal
muscle of zebrafish (Danio rerio), leading to the formation of a hypopigmented lesion at the site of injec-
tion within 24 h (indicative of extensive muscle necrosis as revealed by histology) and the death (resulting
from toxic shock) of the animal at 36–96 h postinjection. Colonization may be evaluated by dissecting
the muscle tissue and the spleen for serial dilution plating [22].
In a separate study, human keratinocytes are shown to allow in vitro formation of GAS biofilms that
have superior capacity to colonize the oropharynx in vivo compared to broth-grown GAS and provide an
effective means to evaluate GAS colonization, invasive disease, and natural transformation [5].
14.3 Conclusion
Constituting a key member of the Gram-positive bacterial genus Streptococcus, S. pyogenes (commonly
called group A Streptococcus or GAS) is renowned for its serotype diversity, virulence variation, and
ferocious infectivity. Adapted exclusively to human host, pathogenic S. pyogenes strains are capable of
inducing localized inflammatory lesions (e.g., pharyngitis, impetigo), secreting streptococcal toxins that
cause both local and systemic diseases (e.g., pneumonia, sepsis, STSS), and producing streptococcal
antigens that confuse host immune systems, leading to autoimmune diseases (e.g., ARF, RHD, APSGN).
Given its medical and public health significance, S. pyogenes has been extensively studied, and a plethora
of laboratory models (both in vitro and in vivo) have been applied to elucidate the molecular mechanisms
of its pathogenicity and to aid in the design of innovative measures for its control and prevention.
REFERENCES
1. Ferretti J, Köhler W. History of streptococcal research. In: Ferretti JJ, Stevens DL, Fischetti VA, editors.
Streptococcus pyogenes: Basic Biology to Clinical Manifestations. Oklahoma City, OK: University of
Oklahoma Health Sciences Center; 2016.
2. Walker MJ, et al. Disease manifestations and pathogenic mechanisms of group A Streptococcus. Clin
Microbiol Rev. 2014;27(2):264–301.
3. Facklam R. What happened to the streptococci: Overview of taxonomic and nomenclature changes. Clin
Microbiol Rev. 2002;15(4):613–30.
4. Bessen DE. Molecular basis of serotyping and the underlying genetic organization of Streptococcus
pyogenes. In: Ferretti JJ, Stevens DL, Fischetti VA, editors. Streptococcus pyogenes: Basic Biology to
Clinical Manifestations. Oklahoma City, OK: University of Oklahoma Health Sciences Center; 2016.
5. Marks LR, Mashburn-Warren L, Federle MJ, Hakansson AP. Streptococcus pyogenes biofilm
growth in vitro and in vivo and its role in colonization, virulence, and genetic exchange. J Infect Dis.
2014;210(1):25–34.
6. Fiedler T, Köller T, Kreikemeyer B. Streptococcus pyogenes biofilms-formation, biology, and clinical
relevance. Front Cell Infect Microbiol. 2015;5:15.
7. Katzenell U, Shemer J, Bar-Dayan Y. Streptococcal contamination of food: An unusual cause of epi-
demic pharyngitis. Epidemiol Infect. 2001;127(2):179–84.
8. Falkenhorst G, et al. Outbreak of group A streptococcal throat infection: Don’t forget to ask about food.
Epidemiol Infect. 2008;136(9):1165–71.
9. Okamoto F, et al. A foodborne outbreak of group A streptococcal infection in Fukuoka Prefecture,
Japan. Jpn J Infect Dis. 2014;67(4):321–2.
10. van der Mee-Marquet N, et al. Molecular characterization of temporally and geographically matched
Streptococcus agalactiae strains isolated from food products and bloodstream infections. Foodborne
Pathog Dis. 2009;6(10):1177–83.
11. Lemos JA, Quivey RG Jr, Koo H, Abranches J. Streptococcus mutans: A new Gram-positive paradigm?
Microbiology. 2013;159(3):436–45.
12. Leung V, Dufour D, Lévesque CM. Death and survival in Streptococcus mutans: Differing outcomes of
a quorum-sensing signaling peptide. Front Microbiol. 2015;6:1176.
13. Watanabe S, Takemoto N, Ogura K, Miyoshi-Akiyama T. Severe invasive streptococcal infection
by Streptococcus pyogenes and Streptococcus dysgalactiae subsp. equisimilis. Microbiol Immunol.
2016;60(1):1–9.
14. Cunningham MW. Rheumatic fever, autoimmunity, and molecular mimicry: The streptococcal connec-
tion. Int Rev Immunol. 2014;33(4):314–29.
15. Fieber C, Kovarik P. Responses of innate immune cells to group A Streptococcus. Front Cell Infect
Microbiol. 2014;4:140.
234 Laboratory Models for Foodborne Infections
16. LaRock CN, Nizet V. Inflammasome/IL-1β responses to streptococcal pathogens. Front Immunol.
2015;6:518.
17. Hornig M, Lipkin WI. Immune-mediated animal models of Tourette syndrome. Neurosci Biobehav Rev.
2013;37(6):1120–38.
18. Macrì S, Onori MP, Roessner V, Laviola G. Animal models recapitulating the multifactorial origin of
Tourette syndrome. Int Rev Neurobiol. 2013;112:211–37.
19. Brosnahan AJ. Animal models used to study superantigen-mediated diseases. Methods Mol Biol.
2016;1396:1–17.
20. Watson ME Jr, Neely MN, Caparon MG. Animal models of Streptococcus pyogenes infection. In:
Ferretti JJ, Stevens DL, Fischetti VA, editors. Streptococcus pyogenes: Basic Biology to Clinical
Manifestations [Internet]. Oklahoma City, OK: University of Oklahoma Health Sciences Center; 2016.
21. Rush CM, Govan BL, Sikder S, Williams NL, Ketheesan N. Animal models to investigate the pathogen-
esis of rheumatic heart disease. Front Pediatr. 2014;2:116.
22. Saralahti A, Rämet M. Zebrafish and streptococcal infections. Scand J Immunol. 2015;82(3):174–83.
23. Miyoshi-Akiyama T, Zhao J, Uchiyama T, Yagi J, Kirikae T. Positive correlation between low adhesion
of group A Streptococcus to mammalian cells and virulence in a mouse model. FEMS Microbiol Lett.
2009;293(1):107–14.
Section III
Dongyou Liu
CONTENTS
15.1 Introduction................................................................................................................................... 237
15.1.1 Classification, Morphology, and Genomics..................................................................... 238
15.1.1.1 Classification..................................................................................................... 238
15.1.1.2 Morphology....................................................................................................... 238
15.1.1.3 Genomics.......................................................................................................... 239
15.1.2 Biology and Epidemiology............................................................................................... 240
15.1.3 Clinical Features and Pathogenesis.................................................................................. 240
15.1.3.1 Gastrointestinal Infections................................................................................ 240
15.1.3.2 Extraintestinal Infections.................................................................................. 240
15.1.4 Diagnosis.......................................................................................................................... 241
15.1.5 Treatment and Prevention................................................................................................. 242
15.2 Laboratory Models........................................................................................................................ 242
15.2.1 Animal Models................................................................................................................. 242
15.2.1.1 Rodents............................................................................................................. 242
15.2.1.2 Zebrafish........................................................................................................... 243
15.2.1.3 Caenorhabditis elegans Nematode.................................................................. 243
15.2.1.4 Tetrahymena Protozoa...................................................................................... 243
15.2.2 In Vitro Models................................................................................................................. 243
15.3 Conclusion..................................................................................................................................... 244
References............................................................................................................................................... 244
15.1 Introduction
Aeromonas was first described in 1890 by Zimmermann as part of the genus Bacillus (Bacillus punc-
tatus). In the following year, an Aeromonas strain was isolated (then named Bacillus hydrophilus fus-
cus, which is now known as Aeromonas hydrophila) by Sanarelli from the blood of infected frog with
hemorrhagic exudation in the abdominal and peritoneal cavities (so-called “red leg” disease). In 1936,
the genus Aeromonas (Greek aer aeros, air, gas; Greek monas, unit, monad; Aeromonas, gas-producing
unit) was created by Kluyver and van Neil to cover this group of bacteria. In 1951, association of the
genus Aeromonas with human infection (fulminant metastatic myositis) was confirmed with the recov-
ery of Aeromonas from autopsy samples. In 1968, involvement of the genus Aeromonas in other human
diseases (e.g., septicemia associated with Laennec’s cirrhosis) was documented. In 1986, the genus
Aeromonas was shown to be phylogenetically distinct from vibrios, and was transferred from the family
Vibrionaceae to a newly established family Aeromonadaceae. To date, about 30 species have been iden-
tified in the genus Aeromonas, and some of them are responsible for a variety of infections in humans
and animals.
237
238 Laboratory Models for Foodborne Infections
15.1.1.2 Morphology
Aeromonas is a Gram-negative, cocco bacillary or rod-shaped bacterium 0.3–1.0 μm × 1.0–3.5 μm
in size, which appears singly, in pairs, and occasionally in short chains. On blood agar, Aeromonas
forms round, raised, opaque colonies of 1–3 mm in diameter, with color changing from grayish (due to
β-hemolysis) to dark green after 3 days (except A. caviae). Being a facultative anaerobe, Aeromonas is
oxidase and catalase positive (best tested on media without a fermentable sugar, such as MacConkey
agar).
Aeromonas 239
Other morphological features include the possession of a single polar flagellum (especially motile
strains, although some species may form peritrichous or lateral flagella or no flagella in solid media) and
pili (fimbriae, which are surface appendages that facilitate attachment to host cells), which are either
short rigid (S/R type) or long wavy flexible (L/W type). Most motile aeromonads do not have capsule,
although A. hydrophila serotypes O:11 and O:34 are known to produce capsule when grown in a glucose-
rich medium, which has a potential role in virulence.
15.1.1.3 Genomics
The genome sequences of a number of Aeromonas species are available in GenBank. The sizes of
Aeromonas genomes appear to be in the range of 3.9–5.18 Mb, with GC contents of 57.6%–63.1% and
3609–4794 genes (Table 15.1). Many Aeromonas species harbor pseudogenes, while a few also possess
plasmids (e.g., A. salmonicida subsp. salmonicida strain A449). In addition, the genomes of Aeromonas
spp. encode a variety of virulence factors (including structural components, extracellular factors, secre-
tion systems, iron-acquisition, and quorum-sensing mechanisms) that actively participate in host adher-
ence, colonization, and infection [5–8]. Phylogenomic network analysis of three clinically important
Aeromonas species (A. hydrophila, A. veronii, A. caviae) highlights the influence of homologous recom-
bination and lateral gene transfer in the evolution of Aeromonas spp. [9].
TABLE 15.1
Genomic Features of Aeromonas Species
Species Strain HG Genome (Mb) GC Content (%) Genes Plasmid (kb)
A. aquatica 4.58 61.2 4163
A. australiensis 4.11 58.1 3735
A. bestiarum HG2 4.69 60.6 4153
A. bivalvium 4.3 62.2 3912
A. caviae 8LM HG4 4.47 61.8 4076 30
A. dhakensis AAK1 4.76 61.8 4268
A. encheleia HG16 4.47 61.9 4065
A. eucrenophila HG6 4.54 61.1 4081
A. finlandiensis 4.72 58.6 4376
A. fluvialis 3.9 58.3 3609
A. hydrophila ATCC 7966 HG1 4.74 61.5 4284
A. lacus 4.39 59 4005
A. media WS HG5 4.78 60.7 4340
A. molluscorum 848 4.24 59.2 4033
A. piscicola 5.18 59.2 4794
A. popoffii HG17 4.76 58.6 4329
A. rivuli 4.52 60 4230
A. salmonicida A449 HG3 4.7 58.5 4320 168 and 175
subsp.
salmonicida
A. sanarellii 4.19 63.1 3823
A. schubertii HG12 4.4 61.5 4287
A. simiae 3.99 61.3 3756
A. sobria HG7 4.68 57.6 4212
A. tecta 4.76 60.1 4329
A. veronii B565 HG8/HG10 4.66 58.6 4057
240 Laboratory Models for Foodborne Infections
15.1.3.1 Gastrointestinal Infections
The most common gastrointestinal infection is gastroenteritis, with symptoms ranging from fever,
vomiting, and abdominal cramp to diarrhea (secretory, dysentery, chronic, and choleric). The secretory
form of Aeromonas gastroenteritis is most common and manifests with low-grade fever and abdomi-
nal pain, and watery diarrhea, with mild to moderate dehydration. The watery diarrhea is generally
a self-limiting illness, lasting a few days to a few weeks. The dysenteric form of Aeromonas gas-
troenteritis is less common and shows cramping abdominal pain and mucus (leukocytes) and blood
in stools (similar to shigellosis). The chronic form of Aeromonas gastroenteritis lasts for more than
2 months and is f requently associated with A. caviae and A. hydrophila infections. The choleraic form
of Aeromonas gastroenteritis is a rare, cholera-like disease. With the spread of gastrointestinal infec-
tions, appendicitis, peritonitis, pancreatitis, and acute cholangitis may emerge [19]. Complications
linked to Aeromonas gastroenteritis include small bowel obstruction, ileal ulceration, intramural intes-
tinal hemorrhage with small bowel obstruction, refractory inflammatory bowel disease, acute renal
failure, and hemolytic-uremic syndrome (HUS). Among 14 Aeromonas species involved in human
illness, six (A. hydrophila, A. caviae, A. veronii, A. schubertii, A. jandaei, and A. trota) are shown to
cause human diarrhea [1].
15.1.3.2 Extraintestinal Infections
Extraintestinal infections include wound and soft tissue infections (cellulitis, abscesses), blood-borne
infections (bacteremia/septicemia, pneumonia, empyema, septic arthritis, necrotizing fasciitis, myo-
necrosis, endocarditis, meningitis), and miscellaneous infections (hepatobiliary tract infections,
Aeromonas 241
osteomyelitis, endophthalmitis, keratitis, corneal ulcer, infections of bones and joints, the respiratory and
urogenital tracts) [20–23]. Application of leeches (which may harbor aeromonads symbiotically) to tis-
sue flaps or replantation areas during plastic or reconstructive surgery to relieve venous congestion may
be a potential cause of Aeromonas infections (e.g., cellulitis) [24,25]. Aeromonas bacteremia/septicemia
(attributed mainly to A. hydrophila, A. dhakensis, A. veronii, or A. caviae) often displays fever, jaundice,
abdominal pain, septic shock, and dyspnea and tends to occur in severely immunocompromised individ-
uals (e.g., those with myeloproliferative disorders, Laennec’s cirrhosis, chronic liver disease, neoplasia,
biliary disease, acute myeloid leukemia, myelodysplastic syndromes, non-Hodgkin’s lymphoma, acute
lymphocytic leukemia, diabetes mellitus, renal problems, cardiac anomalies, aplastic anemia, thalas-
semia, multiple myeloma, and Waldenstrom’s macroglobulinemia) [26]. Aeromonas biliary tract infec-
tions (due to A. hydrophila and to a lesser extent, A. caviae and A. veronii) often occur in patients with
biliary tract obstruction or stasis due to hepatobiliary cancer or stones, and most of them are polymicro-
bial in nature (e.g., Escherichia coli, Klebsiella pneumoniae, Enterococcus, and Staphylococcus aureus).
A. hydrophila and A. veronii may be also a cause of hemorrhagic septicemia in carp, tilapia, perch,
catfish, and salmon; red sore disease in bass and carp; ulcerative infections in catfish, cod, carp, and
goby; ulcerative stomatitis in snakes and lizards; “red leg” disease in frogs; septicemia in dogs; and
septic arthritis in calves. A. salmonicida sensu stricto is linked to fish furunculosis, particularly in sal-
monids, with presentation ranging from septicemia, hemorrhages at the bases of fins, inappetence, mela-
nosis, lethargy, and slight exophthalmia to hemorrhaging in muscle and internal organs.
Aeromonas spp. produce a number of virulence factors and proteins that enhance their pathogenicity.
These include invasins, adhesins, outer membrane proteins, S-layer proteins, lipopolysaccharide (LPS),
proteases [thermostable metalloprotease (TSMP) and thermolabile serine protease (TLSP)], lipases,
glycerophospholipid cholesterol acyl-transferase (GCAT), superoxide dismutase, hemolysins, cytotoxic
and cytotonic enterotoxins [Aeromonas cytotoxic enterotoxin (Act), Aeromonas heat-labile (56°C) cyto-
tonic enterotoxin (Alt), and Aeromonas heat-stable cytotonic enterotoxin (Ast)], as well as type III secre-
tion system (AscV and AscF-G) [27–31].
15.1.4 Diagnosis
Laboratory identification of aeromonads is facilitated by in vitro isolation of Aeromonas spp. from stool or
other gastrointestinal samples. Aeromonads grow readily on routine enteric isolation media (MacConkey,
xylose lysine deoxycholate (XLD), Hektoen enteric (HE), Salmonella-Shigella (SS), and deoxycholate-
citrate (DC) agars). Most (90%) of the Aeromonas species from humans produce β-hemolysis on sheep
blood agar, with the exception of A. popoffii and A. trota (0% and 50%, respectively) [32]. Identification at
the genus level is achieved by positive oxidase test, fermentation of d-glucose, motility (most Aeromonas
species are motile apart from A. salmonicida and A. media), the absence of growth in 6.5% sodium chlo-
ride, and resistance to the vibriostatic agent O/129 (150 μg). However, phenotypic identification to species
level may require additional work involving the use of selective and differential media [33].
A number of selective media (supplemented with ampicillin and/or inhibitors such as bile salts, brilliant
green, and sodium lauryl sulfate) have been used for recovery of Aeromonas species. For example, ampi-
cillin dextrin agar (ADA) is useful for isolating most Aeromonas spp. apart from A. trota and certain
strains of A. caviae, due to their sensitivity to ampicillin. Comparative analysis indicated that enrichment
in alkaline peptone water (APW) and consecutive plating in two selective media [ampicillin–sheep-blood
agar supplemented with 30 μg/mL of ampicillin ASBA 30 or bile salts–Irgasan–brilliant green (BIBG)
agar] enable qualitative isolation of Aeromonas species from meat and fish. In BIBG, bile salts and brilliant
green inhibit the growth of Gram-positive bacteria while Irgasan inhibits the growth of Gram-negative
bacteria, which possess a type A nitratase. In addition, BIBG also detects the fermentation of xylose in
some aeromonads, as production of acid during xylose fermentation retains BIBG’s original, purple-red
color (instead of green-yellow color). Other selective media include Aeromonas agar (AA, containing
irgasan and d-xylose, which aeromonads do not ferment), and cefsulodin–irgasan–novobiocin (CIN) agar
(in which Aeromonas forms a bull’s-eye-like colony due to fermentation of d-mannitol) [33].
Aeromonas isolates surviving the selective process can be further distinguished via differentiation
media (e.g., detection of amylase and pattern of carbohydrate fermentation in Aeromonas species). For
242 Laboratory Models for Foodborne Infections
example, the modified BIBG medium (mBIBG) has a higher pH (up to 8.7), together with the replace-
ment of xylose by soluble starch as a carbon source, allowing identification of mesophilic aeromonads
that are not able to ferment xylose. Further differentiation of aeromonads from vibrios and plesiomo-
nads is their resistance to the vibriostatic agent O/129 (2,4-diamino-6,7-diisopropylpteridine), growth on
nutrient agar without salt supplementation, and inability to grow on media containing 6%–6.5% sodium
chloride. A combination of selective and differential media enables to classify clinical Aeromonas
isolates into the A. hydrophila group (A. hydrophila, A. bestiarum, and A. salmonicida), the A. caviae
group (A. caviae, A. media, and A. eucrenophila), and the A. sobria group (A. veronii, A. jandei,
A. schubertii, and A. trota). Although use of the Aerokey II system allows an accurate and reliable
phenotypic identification of A. hydrophila, A. caviae, A. veronii biovar sobria, A. veronii biovar veronii,
A. schubertii, A. jandaei, and A. trota, it is unable to detect many newly described taxa. Similarly, the
Vitek 2 does not differentiate between A. hydrophila and A. caviae [33].
Molecular methods targeting 16S rRNA, hemolysin (ahh1), aerolysin (aerA), and asa1 genes allow
precise determination of Aeromonas species identity and virulence potential [34–39]. Analysis of house-
keeping genes (e.g., dnaK, gltA, gyrB, radA, rpoB, tsf, and zipA) provides an effective tool for study
of taxonomic and phylogenetic relationships among Aeromonas species [4,5,40–43]. Indeed, use of a
multiplex PCR directed toward the gyrB and rpoB genes enabled simultaneous identification of four
Aeromonas species (A. hydrophila, A. media, A. veronii, and A. caviae) [44].
15.2 Laboratory Models
15.2.1 Animal Models
15.2.1.1 Rodents
Intramuscular inoculation into BALB/c mice provides a useful way to assess the pathogenicity of
Aeromonas isolates (e.g., A. hydrophila, A. veronii, and A. caviae) in soft tissue infections. It appears
that A. hydrophila is capable of causing more severe muscle damage (e.g., fragmentation of muscle fibers,
Aeromonas 243
edema of myocytes, and infiltration of inflammatory cells) than A. caviae. Additionally, mice undergoing
streptomycin pretreatment allow transient Aeromonas colonization and enable determination of the colo-
nization rates of different isolates. Indeed, A. hydrophila, A. veronii, and A. caviae exhibit relatively high
rates of mouse colon tissue colonization than A. salmonicida, A. encheleia, and A. allosaccharophila [53].
Furthermore, intraperitoneal injection of immunocompromised mice or gastric lavage of neonatal mice
offer alternative approach for investigating of Aeromonas pathogenicity relating to Aeromonas-induced
gastroenteritis as well as extraintestinal infections [54,55].
Rats (Rattus norvegicus) undergoing clindamycin pretreatment develop a self-limited, loose stool
(evidence of enteritis) after oral feeding with A. hydrophila, suggesting that antibiotic usage represents a
predisposing risk factor to Aeromonas infection [56].
15.2.1.2 Zebrafish
Zebrafish represents a useful model for assessing the virulence of and host immune responses against
Aeromonas strains, and its value for examining the host–pathogen interactions at the molecular level
may be limited. Zebrafish succumbing to Aeromonas infection often displays clinical signs typical of
hemorrhagic septicemia [57].
15.3 Conclusion
The genus Aeromonas encompasses a large group of Gram-negative, rod-shaped bacteria that are com-
monly present in aquatic environments (marine water, fresh water, and sediments), vegetation, and
various animals. Besides causing diseases in fish and other animals, Aeromonas species are also respon-
sible for opportunistic foodborne or wound-related infections in humans, including gastroenteritis and
extraintestinal infections (e.g., serious wound infections in healthy individuals, primary and secondary
septicemia in immunocompromised individual, as well as peritonitis, meningitis, and infections of the
eye, joints, and bones). Pathogenic Aeromonas species are known to generate a number of virulence
factors that aid their invasion of host cells and sabotage host immune surveillance. Molecular assays
targeting the virulence genes, housekeeping genes, and 16S rRNA genes of Aeromonas spp. provide a
rapid and precise means for their detection, identification, and epidemiological tracking and contribute
to the early implementation of antimicrobial therapy. Nonetheless, despite our concerted efforts in the
past, an effective vaccine against Aeromonas infections is still unavailable. Therefore, further research
is urgently required to reveal the intricacy of host–bacterial interactions, and pinpoint the weak links in
Aeromonas physiobiology and pathogenicity. Undoubtedly, laboratory models will form an indispens-
able part of this endeavor.
REFERENCES
1. Janda JM, Abbott SL. The genus Aeromonas: taxonomy, pathogenicity, and infection. Clin Microbiol
Rev 2010;23(1):35–73.
2. List of Prokaryotic Names with Standing in Nomenclature. Genus Aeromonas. Available at http://www.
bacterio.net/aeromonas.html, Last Updated 2015, Accessed May 31, 2016.
3. Beaz-Hidalgo R, et al. Aeromonas aquatica sp. nov., Aeromonas finlandiensis sp. nov. and Aeromonas
lacus sp. nov. isolated from Finnish waters associated with cyanobacterial blooms. Syst Appl Microbiol
2015;38:161–8.
4. Figueras MJ, Soler L, Chacón MR, Guarro J, Martínez-Murcia AJ. Extended method for discrimination
of Aeromonas spp. by 16S rDNA RFLP analysis. Int J Syst Evol Microbiol 2000;50(6):2069–73.
5. Roger F, Marchandin H, Jumas-Bilak E, Kodjo A, colBVH study group, Lamy B. Multilocus genetics to
reconstruct aeromonad evolution. BMC Microbiol 2012;12:62.
6. Ruiz-Ruiz JM, Aguilera-Arreola MG, Castro-Escarpulli G. Markers of pathogenicity islands
in strains of Aeromonas species of clinical and environmental origin. Indian J Med Microbiol
2012;30(4):467–9.
7. Beaz-Hidalgo R, Figueras MJ. Aeromonas spp. whole genomes and virulence factors implicated in fish
disease. J Fish Dis 2013;36(4):371–88.
8. Dallaire-Dufresne S, Tanaka KH, Trudel MV, Lafaille A, Charette SJ. Virulence, genomic features, and
plasticity of Aeromonas salmonicida subsp. salmonicida, the causative agent of fish furunculosis. Vet
Microbiol 2014;169(1–2):1–7.
9. Ghatak S, et al. Pan-genome analysis of Aeromonas hydrophila, Aeromonas veronii and Aeromonas
caviae indicates phylogenomic diversity and greater pathogenic potential for Aeromonas hydrophila.
Antonie van Leeuwenhoek 2016;109(7):945–56.
10. Zhang Q, Shi GQ, Tang GP, Zou ZT, Yao GH, Zeng G. A foodborne outbreak of Aeromonas hydrophila
in a college, Xingyi City, Guizhou, China, 2012. Western Pac Surveill Response J 2012;3(4):39–43.
11. Fontes MC, Martins C, Martínez-Murcia AJ, Saavedra MJ. Phylogenetic diversity of Aeromonas from
“alheira,” a traditional Portuguese meat product. Foodborne Pathog Dis 2012;9(8):713–8.
12. Jahid IK, Ha SD. Inactivation kinetics of various chemical disinfectants on Aeromonas hydrophila
planktonic cells and biofilms. Foodborne Pathog Dis 2014;11(5):346–53.
13. Ghenghesh KS, Ahmed SF, El-Khalek RA, Al-Gendy A, Klena J. Aeromonas-associated infections in
developing countries. J Infect Dev Ctries 2008;2(2):81–98.
14. Igbinosa IH, Igumbor EU, Aghdasi F, Tom M, Okoh AI. Emerging Aeromonas species infections and
their significance in public health. Sci World J 2012;2012:625023.
Aeromonas 245
15. Diaz JH. Skin and soft tissue infections following marine injuries and exposures in travelers. J Travel
Med 2014;21(3):207–13.
16. Chao CM, Gau SJ, Lai CC. Empyema caused by Aeromonas species in Taiwan. Am J Trop Med Hyg
2012;87(5):933–5.
17. Chao CM, Lai CC, Tang HJ, Ko WC, Hsueh PR. Skin and soft-tissue infections caused by Aeromonas
species. Eur J Clin Microbiol Infect Dis 2013;32(4):543–7.
18. Chen PL, et al. Aeromonas stool isolates from individuals with or without diarrhea in southern Taiwan:
predominance of Aeromonas veronii. J Microbiol Immunol Infect 2015;48(6):618–24.
19. Huang D, Zhao Y, Jiang Y, Li Z, Yang W, Chen G. Spontaneous bacterial peritonitis caused by Aeromonas
caviae in a patient with cirrhosis. Zhong Nan Da Xue Xue Bao Yi Xue Ban 2015;40(3):341–4.
20. Cui H, Hao S, Arous E. A distinct cause of necrotizing fasciitis: Aeromonas veronii biovar sobria. Surg
Infect (Larchmt) 2007;8(5):523–8.
21. Danaher PJ, Mueller WP. Aeromonas hydrophila septic arthritis. Mil Med 2011;176(12):1444–6.
22. Liakopoulos V, et al. Aeromonas hydrophila as a causative organism in peritoneal dialysis-related peri-
tonitis: case report and review of the literature. Clin Nephrol 2011;75(Suppl 1):65–8.
23. Issa N, Napolitano LM. Aeromonas pneumonia in a trauma patient requiring extracorporeal membrane
oxygenation for severe acute respiratory distress syndrome: case report and literature review. Surg Infect
(Larchmt) 2011;12(3):241–5.
24. Tena D, et al. Surgical site infection due to Aeromonas species: report of nine cases and literature
review. Scand J Infect Dis 2009;41(3):164–70.
25. Patel KM, Svestka M, Sinkin J, Ruff P, 4th. Ciprofloxacin-resistant Aeromonas hydrophila infec-
tion following leech therapy: a case report and review of the literature. J Plast Reconstr Aesthet Surg
2013;66(1):e20–2.
26. Turner P, Willemse C, Phakaudom K, Zin TW, Nosten F, McGready R. Aeromonas spp. bacteremia in
pregnant women, Thailand–Myanmar border, 2011. Emerg Infect Dis 2012;18(9):1522–3.
27. Edberg SC, Browne FA, Allen MJ. Issues for microbial regulation: Aeromonas as a model. Crit Rev
Microbiol 2007;33(1):89–100.
28. Chopra AK, Graf J, Horneman AJ, Johnson JA. Virulence factor-activity relationships (VFAR) with
specific emphasis on Aeromonas species (spp.). J Water Health 2009;7(Suppl 1):S29–54.
29. Singh V, Chaudhary DK, Mani I, Jain R, Mishra BN. Development of diagnostic and vaccine markers
through cloning, expression, and regulation of putative virulence-protein-encoding genes of Aeromonas
hydrophila. J Microbiol 2013;51(3):275–82.
30. Gashgari RM, Selim SA. Detection and characterization of antimicrobial resistance and putative viru-
lence genes in Aeromonas veronii biovar Sobria isolated from gilthead sea bream (Sparus aurata L.).
Foodborne Pathog Dis 2015;12(9):806–11.
31. Ji Y, et al. Contribution of nuclease to the pathogenesis of Aeromonas hydrophila. Virulence
2015;6(5):515–22.
32. Chen PL, Ko WC, Wu CJ. Complexity of β-lactamases among clinical Aeromonas isolates and its clini-
cal implications. J Microbiol Immunol Infect 2012;45(6):398–403.
33. Abbott SL, Cheung WK, Janda JM. The genus Aeromonas: biochemical characteristics, atypical reac-
tions, and phenotypic identification schemes. J Clin Microbiol 2003;41(6):2348–57.
34. Balakrishna K, Murali HS, Batra HV. Detection of toxigenic strains of Aeromonas species in foods by
a multiplex PCR assay. Indian J Microbiol 2010;50(2):139–44.
35. Mendes-Marques CL, Nascimento LM, Theophilo GN, Hofer E, Melo Neto OP, Leal NC. Molecular
characterization of Aeromonas spp. and Vibrio cholerae O1 isolated during a diarrhea outbreak. Rev
Inst Med Trop Sao Paulo 2012;54(6):299–30.
36. Mendes-Marques CL, Hofer E, Leal NC. Development of duplex-PCR for identification of Aeromonas
species. Rev Soc Bras Med Trop 2013;46(3):355–7.
37. Senderovich Y, Ken-Dror S, Vainblat I, Blau D, Izhaki I, Halpern M. A molecular study on the preva-
lence and virulence potential of Aeromonas spp. recovered from patients suffering from diarrhea in
Israel. PLoS One 2012;7(2):e30070.
38. Sreedharan K, Philip R, Singh IS. Characterization and virulence potential of phenotypically diverse
Aeromonas veronii isolates recovered from moribund freshwater ornamental fishes of Kerala, India.
Antonie van Leeuwenhoek 2013;103(1):53–67.
246 Laboratory Models for Foodborne Infections
39. Meng S, Wang Y, Wang Y, Liu D, Ye C. Development of cross-priming amplification assays for rapid
and sensitive detection of Aeromonas hydrophila. Lett Appl Microbiol 2015;61(2):171–8.
40. Parker JL, Shaw JG. Aeromonas spp. clinical microbiology and disease. J Infect 2011;62(2):109–18.
41. Ye Y, et al. Resistance characterization, virulence factors, and ERIC-PCR fingerprinting of Aeromonas
veronii strains isolated from diseased Trionyx sinensis. Foodborne Pathog Dis 2012;9(11):1053–5.
42. Rather MA, Willayat MM, Wani SA, Munshi ZH, Hussain SA. A multiplex PCR for detection of entero-
toxin genes in Aeromonas species isolated from foods of animal origin and human diarrhoeal samples.
J Appl Microbiol 2014;117(6):1721–9.
43. Fernandes AM, et al. Development of highly sensitive electrochemical genosensor based on multiwalled
carbon nanotubes-chitosan-bismuth and lead sulfide nanoparticles for the detection of pathogenic
Aeromonas. Biosens Bioelectron 2015;63:399–406.
44. Persson S, Al-Shuweli S, Yapici S, Jensen JN, Olsen KE. Identification of clinical Aeromonas species
by rpoB and gyrB sequencing and development of a multiplex PCR method for detection of Aeromonas
hydrophila, A. caviae, A. veronii, and A. media. J Clin Microbiol 2015;53(2):653–6.
45. Ko WC, Chiang SR, Lee HC, Tang HJ, Wang YY, Chuang YC. In vitro and in vivo activities of fluoro-
quinolones against Aeromonas hydrophila. Antimicrob Agents Chemother 2003;47(7):2217–22.
46. Figueira V, Vaz-Moreira I, Silva M, Manaia CM. Diversity and antibiotic resistance of Aeromonas spp.
in drinking and waste water treatment plants. Water Res 2011;45(17):5599–611.
47. Kadlec K, et al. Molecular basis of sulfonamide and trimethoprim resistance in fish-pathogenic
Aeromonas isolates. Appl Environ Microbiol 2011;77(20):7147–50.
48. Carvalho MJ, Martínez-Murcia A, Esteves AC, Correia A, Saavedra MJ. Phylogenetic diversity, antibi-
otic resistance and virulence traits of Aeromonas spp. from untreated waters for human consumption.
Int J Food Microbiol 2012;159(3):230–9.
49. Shakir Z, et al. Molecular characterization of fluoroquinolone-resistant Aeromonas spp. isolated from
imported shrimp. Appl Environ Microbiol 2012;78(22):8137–41.
50. Aires A, Dias CS, Rosa EA, Saavedra MJ. Antimicrobial susceptibility of Aeromonas spp. isolated from
pig ileum segments to natural isothiocyanates. Med Chem 2013;9(6):861–6.
51. Jones BL, Wilcox MH. Aeromonas infections and their treatment. J Antimicrob Chemother
1995;35(4):453–61.
52. Chen PL, Wu CJ, Ko WC. Aeromonas species. Available at http://www.antimicrobe.org/b74.asp,
Accessed May 31, 2016.
53. Lye DJ. A mouse model for characterization of gastrointestinal colonization rates among environmental
Aeromonas isolates. Curr Microbiol 2009;58(5):454–8.
54. Lye DJ, Rodgers MR, Stelma G, Vesper SJ, Hayes SL. Characterization of Aeromonas virulence using
an immunocompromised mouse model. Curr Microbiol 2007;54(3):195–8.
55. Lye DJ. Gastrointestinal colonization rates for human clinical isolates of Aeromonas veronii using a
mousemodel. Curr Microbiol 2011;63(4):332–6.
56. Haberberger RL Jr, Yonushonis WP, Daise RL, Mikhail IA, Ishak EA. Re-examination of Rattus nor-
vegicus as an animal model for Aeromonas-associated enteritis in man. Experientia 1991;47(5):426–9.
57. Pang MD, Lin XQ, Hu M, Li J, Lu CP, Liu YJ. Tetrahymena: an alternative model host for evaluating
virulence of Aeromonas strains. PLoS One 2012;7(11):e48922.
58. Chen PL, et al. Virulence diversity among bacteremic Aeromonas isolates: ex vivo, animal, and clinical
evidences. PLoS One 2014;9(11):e111213.
59. Li J, Zhang XL, Liu YJ, Lu CP. Development of an Aeromonas hydrophila infection model using the
protozoan Tetrahymena thermophila. FEMS Microbiol Lett 2011;316(2):160–8.
60. Noonin C, Jiravanichpaisal P, Söderhäll I, Merino S, Tomás JM, Söderhäll K. Melanization and patho-
genicity in the insect, Tenebrio molitor, and the crustacean, Pacifastacus leniusculus, by Aeromonas
hydrophila AH-3. PLoS One 2010;5(12):e15728.
61. Hayes SL, Waltmann M, Donohue M, Lye DJ, Vesper SJ. Predicting virulence of Aeromonas isolates
based on changes in transcription of c-jun and c-fos in human tissue culture cells. J Appl Microbiol
2009;107(3):964–9.
16
Bacteroides
CONTENTS
16.1 Introduction................................................................................................................................... 247
16.2 Taxonomic and Ecological Aspects.............................................................................................. 249
16.3 Biology and Virulence Factors...................................................................................................... 249
16.3.1 Capsule............................................................................................................................ 250
16.3.2 Evasion from the Host’s Immune Response................................................................... 250
16.3.3 Enzymes.......................................................................................................................... 250
16.3.4 Enterotoxin.......................................................................................................................251
16.3.5 Endotoxin/LPS.................................................................................................................251
16.4 P athogenesis.................................................................................................................................. 252
16.5 D iagnosis....................................................................................................................................... 252
16.6 S usceptibility to Antimicrobials................................................................................................... 253
16.6.1 Mechanisms of Antimicrobial Resistance...................................................................... 253
16.6.2 β-Lactam Agents............................................................................................................. 253
16.6.3 Carbapenems................................................................................................................... 253
16.6.4 PBP.................................................................................................................................. 253
16.6.5 Outer Membrane Proteins or Porins............................................................................... 255
16.6.6 Aminoglycosides............................................................................................................. 255
16.6.7 M acrolides, Lincosamides, and Chloramphenicol......................................................... 255
16.6.8 Tetracycline..................................................................................................................... 255
16.6.9 Nitroimidazoles............................................................................................................... 255
16.6.10 Quinolones...................................................................................................................... 255
16.6.11 P lasmids.......................................................................................................................... 256
16.6.12 Multidrug Resistance...................................................................................................... 256
16.7 Laboratory Models........................................................................................................................ 256
16.8 C onclusions................................................................................................................................... 257
References............................................................................................................................................... 257
16.1 Introduction
Taxonomically, the genus Bacteroides belongs to the phylum Bacteroidetes, class Bacteroidia, order
Bacteroidales, and family Bacteroidaceae. This genus covers a large number of species pathogenic to
humans and animals. Bacteroides species are strictly anaerobes and some are aerotolerant, and are
considered the most important constituent of the human resident microbiota. Species of this genus are
Gram-negative, non-spore-forming rods, living in intestines and giving the host several nutrients pro-
duced by the breaking down of food. In disequilibrium in the gastrointestinal ecosystem, they can pro-
duce several infections of endogenous nature, such as periodontal diseases, lung and brain infections,
urinary or genital infections, and intraabdominal abscesses, and recently they have been linked to colon
247
248
TABLE 16.1
Phenotypic Characteristics of the Bacteroides fragilis Group and Parabacteroides
Fermentation
Species Growth in 20% Bile Indol Catalase Esculin Hydrolysis Arabinose Cellobiose Rhamnose Sucrose Trehalose
B. fragilis group + V V + −+ + V + +
B. fragilis + − + + − +− − + −
B. caccae + − + + + +− +− + +
B. ovatus + + + + + + + + +
B. stercoris + + − + −+ −+ + + −
+ + + + + +− + + +
cancer. Bacteroides spp. show a great variability to the antimicrobial susceptibility, and several species
are resistant to various antimicrobial drugs expressing resistance genes or other mechanisms, such as
efflux pumps.1 The key phenotypic characteristics of the species of the B. fragilis group are summarized
in Table 16.1.
TABLE 16.2
Occurrence of the Bacteroides fragilis Group in 170 Stool Samples from Children
with and without Acute Diarrhea
Diarrhea No Diarrhea
Bacterial Isolates No. % No. %
Non-ETBF 67 72.82 42 53.8
ETBF 2 2.17 0 0
B. uniformis 7 7.6 1 1.3
B. vulgatus 4 4.3 13 16.7
B. thetaiotaomicron 1 1.1 1 1.3
B. ovatus 0 0 4 5.1
Parabacteroides distasonis 11 11.9 17 21.8
250 Laboratory Models for Foodborne Infections
FIGURE 16.1 Bacteriocin-producing B. fragilis against B. vulgatus. Arrow shows the inhibition halo.
16.3.1 C apsule
The capsule of B. fragilis is largely responsible for abscess formation due to its protection afforded to the
bacteria against the host’s immune response. These abscesses can produce intestinal obstruction, fistula,
bacteremia, and disseminated infection. Encapsulated B. fragilis has been used to produce experimental
abscess in animal model, and it has been reported that responses to other polysaccharide antigens are
T-cell independent, but abscess formation induced by B. fragilis is dependent on the involvement of
T cells. B. fragilis produces a polysaccharide capsule of varying molecular weights (PS-A, PS-B, and
PS-C). By using electron microscopy, three capsule variants within an individual strain of B. fragilis
might be observed: large and small capsules, and noncapsulate strain.
16.3.3 Enzymes
Histolytic enzymes, such as hyaluronidase and chondroitin sulfatase present in some B. fragilis, can
attack the host’s extracellular matrix. Proteases of B. fragilis have also been implicated in destroying
Bacteroides 251
brush border enzymes acting on the microvillus membranes for the final digestion of food and absorp-
tion of nutrients. Other enzymes, hemolysins (HlyA and HlyB), and neuraminidase encoded by the nanH
gene are also important. Neuraminidase is found in many pathogenic bacteria and is generally con-
sidered as a virulence factor; this enzyme catalyzes the removal of the sialic acid from epithelial cells
and immunoactive proteins such as IgG. Several B. fragilis strains have neuraminidase activity, and it
has been suggested that this activity plays a role in the bacterial attachment to animal cells and to the
hemagglutination.5
16.3.4 Enterotoxin
The B. fragilis enterotoxin (BFT) is a zinc metalloprotease that destroys the adherence tight junctions in
intestinal epithelium by cleaving E-cadherin, resulting in rearrangements of the actin cytoskeleton of the
epithelial cells, which causes diarrhea. Enterotoxigenic B. fragilis (ETBF) strains encode three isotypes
of BFT on distinct bft loci, carried on a 6-kb genome segment that is unique and called the B. fragilis
pathogenicity island, and the presence of the bft gene is generally detected by PCR techniques.6–9 This
pathogenicity island is flanked by genes encoding mobilization proteins and may be transmissible to
nontoxigenic strains (non-ETBF) (Figure 16.2). Recent studies have shown that the BFT has a possible
role as a carcinogen in colorectal cancer.10
16.3.5 Endotoxin/LPS
LPS in B. fragilis is 10–1000 times less toxic than that in Escherichia coli. This endotoxin displays
toxicity, and the exposure to antibiotics enhances its production many times higher in B. fragilis than in
the other species of the B. fragilis group. This may partly explain its association with clinical infections
and mortality.
18-kb region
6-kb region
Pattern I (ETBF)
bft (isotypes bft-1, bft-2, and bft-3)
12-kb region
Pattern II (NTBF)
FIGURE 16.2 Pathogenicity island (6-kb region) anchored in chromosomal DNA from enterotoxigenic B. fragilis, and
their genetic profile. (Adapted from Franco, A.A. et al., Mol. Microbiol., 45, 1067–1077, 2002.)
252 Laboratory Models for Foodborne Infections
16.4 Pathogenesis
Capsule present in anaerobes is considered an important virulence factor. Other factors related to the
virulence of anaerobes include mucosal damage and resistance against the host immune response. ETBF
has been implicated in inflammatory intestinal diseases.11–13 Studies show that intestinal colonization
with ETBF leads to acute or chronic intestinal inflammations explaining its potential pathogenicity. The
BFT alters the F-actin structure, resulting in disruption of the epithelial barrier-producing diarrhea.14,15
Studies have implicated the presence of E. coli and Bacteroides vulgatus in the development of Crohn’s
disease and ulcerative colitis; however, the relationship of this bacterial association with these diseases
remains unclear.
Adhesion to the epithelial surface is considered a prerequisite for pathogenicity in most bacteria, and
this attachment may be selective for different cell types.5,16 Establishing a site in the host is critical to the
role of Bacteroides spp. and Parabacteroides spp., both as commensals on the mucosal surfaces of the
intestinal epithelium and as pathogens causing abscesses or other infectious processes. The adhesion and
invasion processes of intestinal Bacteroidales isolated from fecal microbiota of children with diarrhea
are shown in Table 16.3.
These organisms contain a variety of cell surface molecules that are either critical or advantageous
for colonization, including adhesins, hemagglutinins, a polysaccharide capsule, fimbriae, and proteases.
B. fragilis is one of the most important species of intestinal Bacteroidales and possesses a complex
capsular polysaccharide composed of at least eight different polysaccharides, which appear to be anti-
genically diverse. Also, B. fragilis may express three different types of capsules, large or small, and an
electron-dense layer. This heterogeneous nature of encapsulating structures within individual strains
could explain the controversial observations in the literature with respect to the B. fragilis surface struc-
tures related to adhesion. Similar surface structures have been seen in other intestinal species, such as
B. thetaiotaomicron.
16.5 Diagnosis
Species of Bacteroides are considered resident members in intestinal microbiota, and they are mostly
recovered in intra- and extra-abdominal infections. Most anaerobic infections originate from the host’s
resident microbiota and are considered infections of an endogenous nature. Conditions such as low blood
supply into an affected site can predispose the host to anaerobic infection, as well as trauma, foreign
body, malignancy, surgery, edema, shock, colitis, and vascular disease. Mixed infections with aerobic
or facultative organisms make the local tissue conditions favorable for the growth of anaerobic bacteria.
Anaerobic bacteria are the most common residents of the skin and mucous membrane surfaces and
natural cavities’ microbiota. Anaerobes belonging to the resident microbiota of the oral cavity can be
recovered from various infections adjacent to that area, such as cervical lymphadenitis, subcutaneous
TABLE 16.3
Adhesion and Invasion Assays of 114 Strains of the Intestinal Bacteroidales Isolated
from Children with Acute Diarrhea
Adhesion-Positive Invasion-Positive
Species No. of Isolates (%) (%)
Bacteroides fragilis 39 30 (77) 13 (33.3)
B. vulgatus 8 6 (75) 4 (50)
B. uniformis 6 5 (83.3) 5 (83.3)
B. ovatus 5 4 (80) 0
B. eggerthii 0 0 0
B. thetatiotaomicron 0 0 0
Parabacteroides distasonis 6 4 (66.6) 2 (33.3)
Bacteroides 253
abscesses, and burns in proximity to the oral cavity, human and animal bites, tonsillar and retropharyn-
geal abscesses, chronic sinusitis, chronic otitis media, and periodontal abscess. Species of anaerobic
Gram-negative rods including pigmented Prevotella, Porphyromonas, B. fragilis group, Fusobacterium,
and Gram-positive anaerobic cocci are associated with several infections, such as peritonitis, liver
abscess, intra-abdominal abscesses, neonatal infections, and recently to colorectal cancer.17,18 In addi-
tion, quantitative real-time PCR has provided a convenient, dependable, and rapid method to study the
diversity of the presence of the bft subtypes and the significance of ETBF in clinical infections.12
16.6 Susceptibility to Antimicrobials
Because Bacteroides spp. are most commonly found in mixed infections, β-lactams added or not with
β-lactamase inhibitors, carbapenems, clindamycin, and metronidazole are often the antibiotics of
choice.19,20 Clindamycin and metronidazole along with or in combination with some fluoroquinolones
are also used. These species are highly resistant to a broad range of antibiotics, including heavy metals,21
and their resistance to antimicrobials can vary in accordance with geographical locations. The resistance
to active drugs, such as imipenem, piperacillin-tazobactam, ampicillin-sulbactam, and metronidazole, is
observed in some strains.17,22 Considering that a distinct profile of drug resistance is observed in different
countries, it is possible that this resistance is geographically dependent. The resistance profiles of some
Bacteroidales species are shown in Table 16.4, and the distribution of resistance genes and β-lactamase
production in Bacteroides spp. and P. distasonis are shown in Table 16.5.
16.6.2 β
-Lactam Agents
Most Bacteroides spp. are resistant to β-lactam agents as they produce β-lactamase commonly medi-
ated by the chromosomal cepA gene. However, this enzyme is inhibited by the most commonly used
β-lactamase inhibitors (sulbactam, clavulanic acid, and tazobactam).24
16.6.3 Carbapenems
Resistance to carbapenems remains rare, and the genes cfiA and ccrA, both encoding class B metallo-β-
lactamases, are able to degrade these agents. Some strains of Bacteroides contain “silent” carbapenemase
genes, and expression levels are dependent on promoter-containing insertion sequences (IS). Alterations
in penicillin-binding proteins (PBP) or porins, and efflux pumps are other bacterial resistance mecha-
nisms for carbapenems.
16.6.4 PBP
Penicillin and other β-lactams are cell-wall-active drugs. The structure of the β-lactam antibiotics facili-
tates their binding to the active site of PBP in an irreversible inhibition of proteins blocking the trans-
peptidation of the peptidoglycan layer. In B. fragilis, the sequenced genome showed the presence of
seven putative PBP genes, and it was difficult to correlate the MIC50 since this is not the only mechanism
responsible for the resistance.25
254
TABLE 16.4
Resistance Profiles of Intestinal Bacteroidales Species to Eight Antibiotics
% Resistanceb
Bacteroides spp. and
B. fragilis B. vulgatus B. uniformis B. ovatus B. thetaiotaomicron P. distasonis Parabacteroides sp.
Antibioticsa (n = 66) (n = 14) (n = 7) (n = 7) (n = 2) (n = 16) (n = 144)
Amoxicillin 92.4 85.7 100 100 100 93.7 93
Amoxicillin/clavulanic acid 40.9 42.8 0 85.7 100 68.7 47.3
Ampicillin 98.4 92.8 100 100 100 87.7 96.4
Cephalexin 100 92.8 100 100 100 100 99
TABLE 16.5
Distribution of Resistance Genes and β-Lactamase Production in Intestinal Bacteroides spp. and
Parabacteroides distasonis
Genes
β-Lactamase
cfiA cepA ermF tetQ nim Production
Species (No.) No. (%) No. (%) No. (%) No. (%) No. (%) No. (%)
B. fragilis (64) 51 (77.2) 53 (80.3) 16 (24.2) 54 (81.8) 5 (7.5) 60 (90.9)
B. vulgatus (14) 5 (35.7) 11 (78.5) 5 (35.7) 7 (50) 1 (7.14) 13 (92.8)
B. uniformis (7) 6 (85.7) 7 (100) 0 (0) 5 (71.4) 2 (28.5) 7 (100)
B. ovatus (7) 1 (14.2) 1 (14.2) 1 (14.2) 6 (85.7) 0 (0) 7 (100)
B. eggerthii (2) 0 (0) 2 (100) 1 (50) 2 (100) 0 (0) 2 (100)
B. thetaiotaomicron (2) 2 (100) 2 (100) 2 (100) 2 (100) 0 (0) 1 (50)
P. distasonis (16) 6 (37.5) 11 (68.7) 6 (37.5) 15 (93.7) 1 (6.25) 15 (93.7)
16.6.6 Aminoglycosides
Bacteroides species are intrinsically resistant to aminoglycosides since the uptake of this drug requires
free oxygen or a nitrate-dependent electron transport chain, and it is lacking in anaerobes.
16.6.8 Tetracycline
Tetracyclines inhibit bacterial protein synthesis by blocking the attachment of the tRNA-amino acid
to the ribosome. Studies show that 80%–90% of the clinical isolates of Bacteroides are resistant to
this drug, due to the presence of the gene tetQ that is the most commonly involved one in this resis-
tance. Also, the efflux pump for tetracycline has also been described as a resistance mechanism in this
microorganism.
16.6.9 Nitroimidazoles
Metronidazole is the most commonly used drug for anaerobic infections, and resistance to this drug is
rare. Metronidazole-resistant B. fragilis strains have been reported, and their resistance is due to the
presence of nim genes or associated genes to IS; however, this gene can be “silent” unless activated by
IS elements.26,27 In Bacteroides strains, seven nim genes (nimA to nimG) can be found, and each one is
associated with both a distinct mobile genetic element and a specific activating IS element.28
16.6.10 Quinolones
Quinolones inhibit DNA gyrase and DNA topoisomerase IV, which act in bacterial DNA replication,
and gene mutations producing these enzymes are the most common causes of quinolone resistance.29
256 Laboratory Models for Foodborne Infections
Mutations in gyrA causing fluoroquinolone resistance in B. fragilis have been identified. Studies have
shown that the quinolone resistance in clinical isolates and in mutants is due to the increased activity of
the efflux pumps belonging to the RND family.
16.6.11 Plasmids
These genetic elements are very common in Bacteroides species and are found in 20%–50% of strains.
Genes conferring resistance to different classes of antibiotics have been found on plasmids in Bacteroides.
Resistance genes nimA to nim-F and cfiA have been observed in clinical isolates and identified on trans-
ferable plasmids. Intestinal Bacteroidales species often display genetic elements, such as plasmids and/
or transposons, because of their predominant number and a close contact with other intestinal bacteria.
Clinical isolates of Bacteroidales often harbor cryptic plasmids (from 3 to 7 kb of size) with no defined
phenotypic or genotypic association.25
16.6.12 Multidrug Resistance
Little is known about efflux pumps in anaerobic bacteria. In Bacteroides spp., particularly in B. fra-
gilis, 16 homologs of RND pumps (bmeABC1 to bmeABC 16) called B. fragilis multidrug efflux have
been described. A MATE-type efflux system has also been characterized in B. thetaiotaomicron.28 This
system (B. fragilis multidrug efflux) shows that (1) each operon is formed for three components, (2) the
bmeC10 outer membrane component forms part of a contiguous gene with the bmeB10 pump gene,
(3) there are two functional pump genes (bmeB11 and bmeB11) which are transcribed separately in
bme11, and (4) at least 15 of the 16 genes are transcribed.1
16.7 Laboratory Models
Laboratories studying anaerobic bacteria use some principles for isolating and identifying Bacteroides
species. Most of the laboratories refer to the Wadsworth-KTL Anaerobic Bacteriology Manual and the
Manual of Clinical Microbiology. The processing for clinical specimens is important to the success of
isolation, and they are as follows: (1) aseptic collection of the specimen avoiding contamination with resi-
dent microbiota, (2) oxygen-free transport medium system, (3) species of Bacteroides grown on selective
medium Bacteroides bile esculin agar, and (4) B. fragilis is resistant to 20% bile, kanamycin, vancomy-
cin, and colistin. Several PCR schemes are also used to identify Bacteroides species according to the
genus and/or species level, such as using group-specific primers to the β-isopropylmalate dehydrogenase
gene leuB for rapid diagnosis from infections.30 In addition, a multiplex PCR assay with group- and
species-specific primers to identify rapidly 10 species of the B. fragilis group has also been developed.31
This bacterium can cause a wide range of human diseases including abscesses, diabetic foot, diarrhea,
and sepsis. Studies in vivo have shown that B. fragilis toxin (BFT) produced by ETBF induces both a
fluid secretion and exfoliation of intestinal epithelial cells. In certain human intestinal carcinoma cell
lines, particularly HT29/C1, this toxin causes the rounding-up of cells and the rearrangement of the
F-actin cytoskeleton.32,33
Inoculation of ETBF strains harboring different gene subtypes in germ-free mice produced histo-
pathological alterations mainly in the distal ileum–cecum–colon transition area. Moreover, inoculation
in the cecum can produce a superficial erosion and inflammatory infiltration in the lamina propria. It is
known that BFT toxin stimulates morphological changes in intestinal epithelial cell lines, such as HT29,
HT29/C1, Caco-2, T84, MDCK, and HCT-8.
Studies have suggested that strains carrying different bft gene subtypes may have a different patho-
genic potential; however, more studies are needed to evaluate the pathogenesis of each subtype in differ-
ent animal hosts.34 Certainly, they would provide a better understanding of these organisms in ecological
and pathogenic terms.
Bacteroides 257
16.8 Conclusions
Species of Bacteroides, including Bacteroides fragilis, in an unbalanced gastrointestinal ecosystem, can
produce several infections of endogenous nature, and recently have been linked to colon cancer. The
B. fragilis group is considered as the most commonly isolated anaerobic pathogen. Several virulence
factors are produced by B. fragilis. The presence of adhesins allows its colonization of the host’s tissues,
and Bacteroides spp. are able to produce bacteriocin-like substances that inhibit the growth of other
bacterial species or genera.
ETBF has been implicated in inflammatory intestinal diseases, and its intestinal colonization results
in acute or chronic intestinal inflammations. Species of Bacteroides are considered resident intestinal
microbiota, but they are often recovered from intra- and extra-abdominal infections. Bacteroides spp.
show great resistance to various antimicrobial drugs expressing resistance genes. B. fragilis is intrin-
sically resistant to several classes of antibiotics, but the mechanisms are poorly understood. Most
Bacteroides spp. are resistant to β-lactam by producing β-lactamase commonly mediated by the chro-
mosomal cepA gene.
Studies in vivo have shown that the inoculation of ETBF strains harboring different gene subtypes in
germ-free mice produced histopathological alterations, since the BFT toxin stimulates morphological
changes in different intestinal epithelial cells. The in vivo evaluation of pathogenic potentials of the sub-
types of B. fragilis could uncover important details about the biology and pathogenesis of this important
anaerobic bacterial group.
REFERENCES
1. Wexler, H. M. 2007. Bacteroides: the good, the bad, and the nitty-gritty. Clin. Mirobiol. Rev. 20: 593–621.
2. Song, Y.; Liu, C.; McTeague, M.; Finegold, S. M. 2004. “Bacteroides nordii” sp. nov. and “Bacteroides
salyersae” sp. nov. isolated from clinical specimens of human intestinal origin. J. Clin. Microbiol. 42:
5565–5570.
3. Song, Y.; Liu, C.; Bolanos, M.; Lee, J.; McTeague, M.; Finegold, S. M. 2005. Evaluation of 16S rRNA
sequencing and reevaluation of a short biochemical scheme for identification of clinically significant
Bacteroides species. J. Clin. Microbiol. 43: 1531–1537.
4. Sakamoto, M.; Benno, Y. 2006. Reclassification of Bacteroides distasonis, Bacteroides goldsteinii and
Bacteroides merdae as Parabacteroides distasonis gen. nov., comb. nov., Parabacteroides goldsteinii
comb. nov. and Parabacteroides merdae comb. nov. Int. J. Syst. Evol. Microbiol. 56: 1599–1605.
5. Nakano, V.; Piazza, R. M. F.; Avila-Campos, M. J. 2006. A rapid assay of the sialidase activity in spe-
cies of the Bacteroides fragilis group by using peanut lectin hemagglutination. Anaerobe 12: 238–241.
6. Almeida, F. S.; Nakano, V.; Avila-Campos, M. J. 2007. Occurrence of enterotoxigenic and nonentero-
toxigenic Bacteroides fragilis in calves and evaluation of their antimicrobial susceptibility. FEMS
Microbiol. Lett. 272: 15–21.
7. Avila-Campos, M. J.; Liu, C.; Song, Y.; Rowlinson, M.-C.; Finegold, S. M. 2007. Determination of bft
gene subtypes in Bacteroides fragilis clinical isolates. J. Clin. Microbiol. 45: 1336–1338.
8. Bressane, M. A.; Durigon, L. E.; Avila-Campos, M. J. 2001. Prevalence of the Bacteroides fragilis
group and enterotoxigenic Bacteroides fragilis in immunodeficient children. Anaerobe 7: 277–281.
9. Claros, M. C.; et al. 2006. Characterization of the Bacteroides fragilis pathogenicity island in human
blood culture isolates. Anaerobe 12: 17–22.
10. Toprak, N. U.; et al. 2006. A possible role of Bacteroides fragilis enterotoxin in the aetiology of colorec-
tal cancer. Clin. Microbiol. Infect. 12: 782–786.
11. Kryzanowsky, F.; Avila-Campos, M. J. 2003. Detection of non-enterotoxigenic and enterotoxigenic
Bacteroides fragilis in stool samples from children in São Paulo, Brazil. Rev. Inst. Med. Trop. São Paulo
45: 1–4.
12. Merino, V. R. C.; Nakano, V.; Liu, C.; Song, Y.; Finegold, S. M.; Avila-Campos, M. J. 2011. Quantitative
detection of enterotoxigenic Bacteroides fragilis subtypes isolated from children with and without diar-
rhea. J. Clin. Microbiol. 49: 416–418.
258 Laboratory Models for Foodborne Infections
13. Nakano, V.; Gomes, T. A. T.; Vieira, M. A. M.; Ferreira, R. C.; Avila-Campos, M. J. 2007. bft gene
subtyping in enterotoxigenic Bacteroides fragilis isolated from children with acute diarrhea. Anaerobe
13: 1–5.
14. Franco, A. A.; Cheng, R. K.; Goodman, A.; Sears, C. L. 2002. Modulation of bft expression of the
Bacteroides fragilis pathogenicity island and its flanking region. Mol. Microbiol. 45: 1067–1077.
15. Wexler, H. M.; Read, E. K.; Tomzynski, T. J. 2002. Identification of an OmpA protein from Bacteroides
fragilis: ompA gene sequence, OmpA amino acid sequence and predictions of protein structure.
Anaerobe 8: 180–191.
16. Nakano, V.; et al. 2008. Adherence and invasion of Bacteroidales isolated from the human intestinal
tract. Clin. Microbiol. Infect. 14: 955–963.
17. Diniz, C. G.; Farias, L. M.; Carvalho, M. A.; Rocha, E. R.; Smith, C. J. 2004. Differential gene expres-
sion in a Bacteroides fragilis metronidazole resistant mutant. J. Antimicrob. Chemother. 54: 100–108.
18. Fukugaiti, M. H.; Ignacio, A.; Fernandes, M. R.; Júnior, U. R.; Nakano, V.; Avila-Campos, M. J. 2015.
High occurrence of Fusobacterium nucleatum and Clostridium difficile in the intestinal microbiota of
colorectal carcinoma patients. Braz. J. Microbiol. 46(4): 1135–1140.
19. Behra-Miellet, J.; Calvet, L.; Dubreuil, L. A. 2004. A Bacteroides thetaiotamicron porin that could take
part in resistance to β-lactams. Int. J. Antimicrob. Agents 24: 135–143.
20. Brazier, J. S.; Stubbs, S. L.; Duerden, B. I. 1999. Metronidazole resistance among clinical isolates belong-
ing to the Bacteroides fragilis group: time to be concerned? J. Antimicrob. Chemother. 44: 580–581.
21. Ignacio, A.; Nakano, V.; Avila-Campos, M. J. 2015. Intestinal Bacteroides vulgatus showing resistance
to metals. Appl. Med. Res. 1: 43–47.
22. Hecht, D. W. 2004. Prevalence of antibiotic resistance in anaerobic bacteria: worrisome developments.
Clin. Infect. Dis. 39: 92–97.
23. Schapiro, J. M.; Gupta, R.; Stefansson, E.; Fang, F. C.; Limaye, A. P. 2004. Isolation of metronidazole-
resistant Bacteroides fragilis carrying the nimA nitroreductase gene from a patient in Washington State.
J. Clin. Microbiol. 42: 4127–4129.
24. Nakano, V.; Nascimento-Silva, A.; Merino, V. R. C.; Wexler, H. M.; Avila-Campos, M. J. 2011.
Antimicrobial resistance and prevalence of resistance genes in intestinal Bacteroidales strains. Clinics
66: 543–547.
25. Nakano, V.; Padilla, G.; Valle, M. M.; Avila-Campos, M. J. 2004. Plasmid-related β-lactamase produc-
tion in Bacteroides fragilis strains. Res. Microbiol. 155: 843–846.
26. Pumbwe, L.; Chang, A.; Smith, R. L.; Wexler, H. M. 2007. BmeRABC5 is a multidrug efflux system that
can confer metronidazole resistance in Bacteroides fragilis. Microb. Drug Resist. 13: 96–101.
27. Pumbwe, L.; Glass, D.; Wexler, H. M. 2006. Efflux pumps over expression in multiple-antibiotic-
resistant mutants of Bacteroides fragilis. Antimicrob. Agents Chemother. 50: 3150–3153.
28. Ueda, O.; Wexler, H. M.; Hirai, K.; Shibata, Y.; Yoshimura, F.; Fujimura, S. 2005. Sixteen homologs of
the mex-type multidrug resistance efflux pump in Bacteroides fragilis. Antimicrob. Agents Chemother.
49: 2807–2815.
29. Miyamae, S.; Ueda, O.; Yoshimura, F.; Hwang, J.; Tanaka, Y.; Nikaido, H. 2001. A MATE family
multidrug efflux transporter pumps out fluoroquinolones in Bacteroides thetaiotaomicron. Antimicrob.
Agents Chemother. 45: 3341–3346.
30. Miki, T.; et al. 2005. Simultaneous detection of Bacteroides fragilis group species by leuB-directed
PCR. J. Med. Invest. 52: 101–108.
31. Liu, C.; Song, Y.; McTeague, M.; Vu, A. W.; Wexler, H.; Finegold, S. M. 2003. Rapid identification of
the species of the Bacteroides fragilis group by multiplex PCR assays using group- and species-specific
primers. FEMS Microbiol. Lett. 222: 9–16.
32. Chambers, F. G.; Koshy, S. S.; Saidi, R. F.; Clark, D. P.; Moore, R. D.; Sears, C. L. 1997. Bacteroides fra-
gilis toxin exhibits polar activity on monolayers of human intestinal epithelial cells (T84 cells) in vitro.
Infect. Immun. 67: 3561–3570.
33. Donelli, G.; Fabbri, A.; Fiorentini, C. 1996. Bacteroides fragilis enterotoxin induces cytoskeletal
changes and surfaces blebbing in HT-29 cells. Infect. Immun. 64: 113–119.
34. Nakano, V.; Gomes, D. A.; Arantes, R. M.; Nicoli, J. R.; Avila-Campos, M. J. 2006. Evaluation of the
pathogenicity of the Bacteroides fragilis toxin gene subtypes in gnotobiotic mice. Curr. Microbiol. 53:
113–117.
17
Brucella
CONTENTS
17.1 The Pathogen................................................................................................................................. 259
17.2 Brucella Species Associated with Foodborne Infections............................................................. 260
17.3 Microbiological Characteristics of Brucella..................................................................................261
17.4 Invasion and Cellular Pathogenesis...............................................................................................261
17.5 Clinical Disease in Humans.......................................................................................................... 263
17.6 Immunity....................................................................................................................................... 263
17.7 Foodborne Brucellosis.................................................................................................................. 264
17.7.1 Inactivation in Food Products.......................................................................................... 264
17.7.2 Transmission by Milk or Cheese Products....................................................................... 264
17.7.3 Meat or Fish Products....................................................................................................... 264
17.7.4 Laboratory Models........................................................................................................... 265
17.8 Conclusion..................................................................................................................................... 265
References............................................................................................................................................... 266
Clinical disease caused by infection with bacteria in the genus Brucella, brucellosis, remains one of
the most important zoonotic diseases worldwide and is considered to be reemerging in some countries.
Although the bacteria causing “Malta fever” was reported in human spleens by Bruce in 1887,1 the link
between human disease and infection in animal reservoirs was not correlated until 1918.2 The highest
prevalence of human disease is currently found in areas of Africa, Asia, Latin America, and the Middle
East and is associated with significant public health costs. Although transmission to humans can occur
through direct contact with abortions or birth materials from infected animals, indirect contact through
consumption of nonpasteurized dairy products is a common route for human infection.
Over half a million human cases are reported worldwide annually. Due to the wide-ranging and non-
specific nature of clinical signs, the true number may be much higher.3 Estimates of disease prevalence
range from <1 per 100,000 people in the United Kingdom, United States, and Australia, to >70 per
100,000 people in some countries in the Middle East and Central Asia.4 Of particular concern is the
observation that children appear to represent a high proportion of human brucellosis cases. Multiple
studies have demonstrated that addressing brucellosis in animal reservoirs is the most cost-efficient
mechanism for controlling human brucellosis.5–7 Therefore, most control strategies involve control of
livestock brucellosis through vaccination and use of test and removal strategies. Livestock vaccina-
tion has proved to successfully decrease human brucellosis and can be especially helpful in developing
nations. After initiation of a vaccination in Mongolia,8,9 the incidence rate of human brucellosis declined
from 4.8/10,000 to 0.23/100,000.
17.1 The Pathogen
Members of the genus Brucella are small (0.4–3 μm), Gram-negative, coccobacillary organisms within
alpha Proteobacteria that exist as intracellular pathogens in mammalian hosts. Most species of Brucella
259
260 Laboratory Models for Foodborne Infections
are obligate pathogens in that they do not exist as commensals nor are they found free-living in the envi-
ronment. However, a recently identified species of Brucella in voles, Brucella microti, may have genetic
differences that support a free-living as compared to a host-associated life style.10 Although traditional
species of Brucella may temporarily be recovered from environmental samples associated with infected
animals, environmental persistence is generally not believed to be of epidemiologic importance. Because
direct or close contact is generally believed to be required for transmission, maintenance of brucellosis in
animal populations generally requires continual infection of susceptible hosts. As humans are essentially
dead-end hosts, infection in humans requires exposure to brucellosis from animal hosts.
The Brucella genus has traditionally been characterized as having six species: Brucella abortus,
Brucella melitensis, Brucella suis, Brucella canis, Brucella ovis, and Brucella neotomae. The high
degree of homology at the genomic level has led to the proposition that the genus is actually composed
of only one species, B. melitensis, with the other classical species proposed as strains of B. meliten-
sis.11 However, host preference, epidemiologic features, and diagnostic benefits obtained by assigning
Brucella strains to separate “nomenspecies” based on their distinctive phenotypic characteristics is more
compelling and has supported retention of the current nomenclature. In the past 20 years, new Brucella
species from sea mammals,12–14 voles,15,16 and a prosthetic breast implant17 have been added to the genus.
Recent isolations from Austrian foxes,18 African bullfrogs (Pyxicephalus edulis),19 and baboons (Papio
spp.)20 may eventually further expand the number of species in the Brucella genus. Some of the classical
species (B. melitensis, B. abortus, and B. suis) are divided into biovars based on biochemical, pheno-
typic, and antigenic properties. Although division into biovars has been used for epidemiologic purposes,
biotyping can be somewhat subjective because it is based on subtle differences such as requirements for
higher CO2 tensions for growth, production of hydrogen sulfide, growth on media containing dyes (thio-
nin or basic fuchsin), and agglutination with monospecific A and M antisera.
One of the important characteristics of Brucella is the number of species in the genus that are capable
of causing zoonotic infections in humans. Most virulent strains of Brucella express the O-polysaccharide
(homopolymer of 4,6-dideoxy-4-formamido-α-d-mannopyranosyl units) on their lipopolysaccharide (LPS)
and are known as “smooth” strains. With the exception of B. canis which does not express the O side-chain
(a “rough” strain), most zoonotic strains of Brucella are smooth strains. Cumulative data suggests dosages
of virulent Brucella species required for 50% infection rates across mucosal surfaces of hosts other than
guinea pigs or mice are most likely in the 103–104 CFU range,21,22 Although virulent strains of Brucella
have a predilection for lymphoreticular tissues and the reproductive tract, most also frequently localize in
mammary tissues and are shed in milk.
rodents in the former USSR. Porcine brucellosis (biovars 1, 2, and 3) has widespread distribution in
domestic and feral swine globally, but prevalence is higher in some areas such as South-East Asia and
South America. It should be noted that unlike biovars 1, 3, and 4, biovar 2 of B. suis is not considered to
be important for causing human brucellosis. The increasing isolations of B. suis in cattle in the south-
eastern U.S. has public health significance due to the shedding of high numbers of bacteria within milk
of infected cattle.31
Isolates of marine mammal Brucella have been recovered from the Mysticeti and Odontoceti sub-
orders of cetaceans. Genomic data have led to taxonomic classification of these isolates into Brucella
ceti (predominantly porpoises and dolphins) and Brucella pennipedalis (predominantly seals) strains,
each of which has several subgroups.32 Bacteriologic studies have indicated these strains are phenotypi-
cally smooth; like the classical Brucella strains. Seropositive responses to marine brucellae have been
observed in 53 cetaceans, with isolation or identification of the bacteria having occurred in samples from
18 marine mammal species.14 It is suspected that cetacean brucellosis may be distributed worldwide in
the oceans. The ST27 genotype in Pacific B. ceti and B. pinnipedalis strains, characterized by a specific
genetic location for the mobile genetic element IS711, has been associated with zoonotic infections in
Peru and New Zealand.33
tissues and organs such as the spleen, reproductive tract, and/or mammary gland. The bacteremia
associated with most Brucella species is short, and live bacteria are not readily isolated from blood
samples.
For cellular entry, smooth Brucella that have not been opsonized by antibodies use the cytoskeleton of
the host cell and interact with cholesterol-rich microdomains (lipid rafts) within the plasma membrane
that facilitate contact with the host cell and mediate internalization. Lipid rafts contain glycosphin-
golipids, cholesterol, and glycosyl-phosphatidylinositol-anchored proteins38 and facilitate membrane-
associated sorting events such as the formation of multi-subunit membrane complexes and signaling
across membranes and membrane fusion. Besides the plasma membrane, lipid rafts are also found in
intracellular organelles and vesicles. The Brucella LPS O-polysaccharide appears to be a key molecule
for interaction with lipid rafts on host cells, but also prevents complement-mediated bacterial lysis and
prevents host cell apoptosis.39 Opsonization of smooth strains of Brucella increases entry 10-fold and
occurs through IgG (Fc) and complement (C3b and 4b) receptors on the surface of phagocytes, which
diverts smooth bacteria from lipid rafts and targets entry to the phagolysosomal compartment. Receptor-
mediated phagocytosis leads to greater killing of internalized Brucella by monocytes. In contrast to
smooth Brucella, rough strains of Brucella cannot sustain interactions with lipid rafts and are phago-
cytosed by either toll-like receptor 4 or mannose receptor recognition of the LPS-deficient bacterial
surface leading to rapid targeting to the phagolysosomal compartment where they are generally unable
to replicate.
Smooth Brucella that enter via lipid rafts quickly traffic through the early endosomal compartment
and depart the phagosome to form the modified phagosome (termed brucellosome) by acquiring com-
ponents of endoplasmic reticulum in a manner similar to autophagosome biogenesis. Brucella initially
localize within acidified phagosomes40 where they are exposed to free oxygen radicals generated by the
respiratory burst. Brucella require acidification of the phagosomal compartment to a pH < 4.5 before
they display wild-type intracellular replication. The requirement for low pH is transient and only extends
through the initial stages of intracellular infection. Localization in an acidified environment induces
expression of the VirB operon (virB 1–10), which controls expression of genes associated with a type IV
secretion system. The VirB operon interacts with the endoplasmic reticulum to neutralize the pH of the
phagosome.41 Brucella-induced modifications of the phagosome leads to inhibition of phagosome matu-
ration and prevention of fusion with lysosomes. The brucellosome environment provides Brucella with
conditions of nutrient depletion and limited oxygen availability. It should be noted that under in vitro
conditions, up to 90% of virulent Brucella, and 99% of nonvirulent Brucella, may be killed following
intracellular entry.42
Brucella have multiple mechanisms to detoxify free radicals produced by host phagocytes, cationic
peptides, and oxygen metabolites. Brucella probably use both stationary and exponential stages for
intracellular survival, with stationary-phase physiology providing Brucella with benefits for adapting
to the harsh conditions encountered in the phagosome and exponential stages associated with repli-
cation under favorable conditions. The presence of cytochromes (cytochrome bc1 complex or quinol
oxidase) with a high oxygen affinity may represent an important adaptation of Brucella to their intra-
cellular survival. Several Brucella have been shown to utilize heme as an iron source in vitro, and have
a critical need for heme during residence in the phagosomal compartment43 and during replication
in trophoblasts.44 Brucella scavenge iron through siderophores such as 2,3-dihydroxybenzoic acid or
brucebactin.45
In accordance with its stealthy nature, Brucella has developed mechanisms to minimize stimulation of
pattern recognition receptors (PRRs) by the innate immune system of the host. The Brucella cell enve-
lope has high hydrophobicity, and its LPS has a noncanonical structure that elicits a reduced and delayed
inflammatory response as compared to other Gram-negative bacteria,40 and has lower stimulatory activity
on TLR4 receptors.46 The O side-chain on the LPS can form complexes with the MHC Class II molecules
that interfere with the ability of macrophages to present exogenous proteins. Brucella ornithine-containing
lipids and lipoproteins in the outer membrane are poor activators of innate immunity. Brucella bacteria
are also devoid of many classical structures involved in virulence such as pili, fimbria, capsules, and
plasmids that stimulate PRRs. The ability of Brucella to prevent phagosome maturation and fusion with
Brucella 263
lysosomes may interfere with other innate and adaptive immune processes. As proteins have been identi-
fied in Brucella that demonstrate significant homology with toll-like receptor (TLR) adaptor molecules,
these peptides may be a mechanism to interfere with, or subvert TLR signaling.47 Compared to other
Gram-negative bacteria, Brucella induces a reduced innate immune response, and a lower rate of matu-
ration and activation of dendritic cells, which may impair development of adaptive immune responses.
17.6 Immunity
Currently, no vaccines are available to protect humans from brucellosis. Currently available livestock
vaccines are composed of attenuated live bacteria, which can cause clinical disease in humans in
the event of inadvertent infection. Long-term protection against brucellosis in livestock is associ-
ated with stimulation of cellular immunity, while antibodies are considered to play a minor role
in protection. Although specific correlates of protective immunity are currently not known, it is
believed that protection is mediated by the Th1 subset of CD4+ lymphocytes and is associated with
the production of IFN-γ and other cytokines associated with cellular immunity. CD4+ cells play a
central role in coordinating and culminating the adaptive immune response by differentiation into
functional subsets, such as the Th1 type, which is associated with the production of interferon-γ
(IFN-γ), interleukin-2 (IL-2), and tumor necrosis factor-α (TNF-α). CD4+ cells also provide growth
factors and signals for the generation and maintenance of CD8+ T cells. CD8+ cells are considered to
be of importance for protective immunity because of their ability to lyse or kill infected cells, thereby
releasing Brucella from intracellular hiding and exposing it to extracellular bactericidal mechanisms.
Cytokines released by CD4+ and CD8+ cells may activate macrophages and dendritic cells, thereby
increasing their bactericidal activity against Brucella.58 Although many studies have used qualita-
tive measurements of IFN-γ production as an indication that vaccination induces protective Th1
responses, recent data with other pathogens have suggested that a better indicator of the quality of
the immune response after immunization may be through the measurement of increases in antigen-
specific polyfunctional T cells capable of producing a traid of relevant cytokines.59 In addition, the
type of memory cell produced may influence the capability for protective responses, cytokine pro-
duction, and cytotoxic activity.60
264 Laboratory Models for Foodborne Infections
17.7 Foodborne Brucellosis
17.7.1 Inactivation in Food Products
In regard to survival in food materials, Brucella is killed by pasteurization61 or heat treatment (up to
71°C), but environmental studies indicate the bacteria can survive drying, freezing, or long-term stor-
age at cold temperatures.62 Souring of milk has negligible effects on the survival of Brucella63 and the
bacteria can survive in milk for up to 87 days, water for up to 60 days, less than a week in yogurt, and in
ripened cheese for up to 50 days despite the reduction in pH associated with ripening.64–67 Ripening of
cheese at 24°C for 20 days was associated with the inability to recover B. melitensis, whereas the bacteria
were recovered when cheese was ripened at 4°C.68 In meat products, processes for preparation of cured/
smoked meat can reduce, but not completely kill, Brucella.69
Ingestion of fish or marine products may also be a source for foodborne infection with Brucella.
Three cases of human infection with the ST27 genotype of Pacific B. ceti and B. pinnipedalis strains
have occurred in individuals with a history of consumption of raw shellfish or fish.33,91 The isolation of
B. melitensis biovar 3 from Nile catfish may also indicate a possible route for foodborne transmission
to humans. The presence of B. melitensis in this species was hypothesized to have spilled over from
endemic infection in ruminants in the area.29
17.7.4 Laboratory Models
Due to their high susceptibility to infection with Brucella, guinea pigs have historically been used for
isolation. For some samples such as cheeses, animal inoculation may be the only means for detecting
the presence of brucellae.92 Guinea pigs are also an accepted model for testing the virulence of Brucella
strains by assessment of spleen pathology and quantification of infection at 8–12 weeks after inocula-
tion. Lesions of brucellosis are observed in the liver, spleen, lungs, and lymph nodes.93 Guinea pigs have
been used to evaluate effectiveness of antibiotic treatments, and for evaluation of efficacy of vaccines.94
Currently, inbred mice are the most common laboratory animal model used to study chronic infec-
tion or disease pathogenesis.95 This is partly because Brucella colonize and induce pathologic lesions in
liver and spleen tissues of mice that to some extent mimics human infection. Inbred mouse strains differ
in susceptibility to infection, with BALB/c and CBA/H being considered relatively more susceptible
to infection and C57BL/6 and C57BL/10 more resistant.95 Intraperitoneal is the most common route of
infection for mouse models, but aerosol, oral, and intranasal routes have also been used. It should be
noted that murine models generally do not accurately replicate disease pathogenesis in natural hosts
where Brucella spp. more frequently localize in lymphatic tissues, mammary gland, and reproductive
organs, and reproductive losses (i.e., abortion in females and orchitis/epididymitis in males) are common.
With the exception of abortion, natural hosts generally do not demonstrate clinical disease. In compari-
son, humans appear to be aberrant hosts for Brucella and often have significant symptoms of clinical
disease that are frequently chronic.
Other laboratory models that have been used include rabbits, rats, and nonhuman primates although
each has limitations regarding the study of Brucella pathogenesis. Brucella-induced abortion is rare,
but rats can venerally transmit B. abortus and offspring can be latently infected.93 Lesions of necrosis
in periplacentomal chorionic epithelium and metritis have been observed in the uterus of pregnant rats
infected with B. abortus. Rabbits are partially susceptible to Brucella, with susceptibility increasing
with pregnancy. Nonhuman primate models (Macaca arctoides and Macaca mulatta) have been infected
through oral, subcutaneous, and aerosol routes with virulent strains of Brucella. Bacteremia was dem-
onstrated for up to 8 weeks, and lesions of focal granulomatous hepatitis, splenitis, lymphadenitis, and
occasional lesions of granulomatous orchitis, epididymitis, and endometritis were reported, similar to
lesions in humans infected with Brucella reviewed by Silva et al.95 Current knowledge would suggest
that primate models most closely mimic pathogenesis of disease in humans. Although laboratory models
continue to have value for basic research, they do not accurately replicate disease pathogenesis in natural
hosts of Brucella.
17.8 Conclusion
Brucellosis in humans remains a significant zoonosis in many parts of the world and frequently occurs
through international travel. In the absence of direct contact with infected animal hosts, transmission of
infection to humans almost exclusively occurs through products made from nonpasteurized milk. Brucella
appear to have a long shelf life in these types of products. In some regions or countries, human brucellosis
is associated with certain ethnic groups and may be related to dietary preferences. Although addressing
the disease in its natural hosts is the most economic approach, regulatory efforts to prevent the sale or con-
sumption of nonpasteurized milk products would also reduce human brucellosis and benefit public health.
266 Laboratory Models for Foodborne Infections
REFERENCES
1. Bruce, D. Note on the discovery of a microorganism in Malta fever. Practitioner (London) 39, 161, 1887.
2. Evans, A.C. Further studies on bacterium abortus and related bacteria. II. A comparison of bacterium
abortus with bacterium bronchisepticus and with the organism which causes Malta fever. J. Infect. Dis.
22, 580, 1918.
3. Godfroid, J., et al. A “One Health” surveillance and control of brucellosis in developing countries: mov-
ing away from improvisation. Comp. Immunol. Microbiol. Infect. Dis. 36, 241, 2013.
4. Cutler, S.J., Whatmore, A.M., & Commander, N.J. Brucellosis—new aspects of an old disease. J. Appl.
Microbiol. 98, 1270, 2005.
5. Bernués, A., Manrique, E., & Maza, M.T. Economic evaluation of bovine brucellosis and tuberculosis
eradication programmes in a mountain area of Spain. Prev. Vet. Med. 30, 137, 1997.
6. Jelastopulu, E., Bikas, C., Petropoulos, C., & Leotsinidis, M. Incidence of human brucellosis in a rural
area in Western Greece after the implementation of a vaccination programme against animal brucel-
losis. BMC Public Health 8, 241, 2008.
7. Roth, F., et al. Human health benefits from livestock vaccination for brucellosis: case study. Bull. World
Health Organ. 81, 867, 2003.
8. Kolar, J. Control of Brucella melitensis brucellosis in developing countries. Ann. Inst. Pasteur Microbiol.
138, 122, 1987.
9. Roth, F., et al. Human health benefits from livestock vaccination for brucellosis: case study. Bull. World
Health Organ. 81, 876, 2003.
10. Scholz, H.C., et al. Isolation of Brucella microti from soil. Emerg. Infect. Dis. 14, 1316, 2008.
11. Vizcaino, N., Cloeckaert, A., Verger, J., Grayon, M., & Fernandez-Lago, L. DNA polymorphism in the
genus Brucella. Microbes Infect. 2, 1089, 2000.
12. Ewalt, D.R., Payeur, J.B., Martin, B.M., Cummins, D.R., & Miller, W.G. Characteristics of a Brucella
species from a bottlenose dolphin (Tursiops truncatus). J. Vet. Diagn. Invest. 6, 448, 1994.
13. Foster, G., Osterman, B.S., Godfroid, J., Jacques, I., & Cloeckaert, A. Brucella ceti sp. nov. and Brucella
pinnipedialis sp. nov. for Brucella strains with cetaceans and seals as their preferred hosts. Int. J. Syst.
Evol. Microbiol. 57, 2688, 2007.
14. Hernandez-Mora, G., Palacios-Alfaro, J.D., & Gonzalez-Barrientos, R. Wildlife reservoirs of brucel-
losis: Brucella in aquatic environments. Rev. Sci. Tech. 32, 89, 2013.
15. Hofer, E., et al. Isolation of Francisella tularensis and Brucella suis from red foxes (Vulpes vulpes).
Tierarztl. Umsch. 65, 229, 2010.
16. Scholz, H.C., et al. Isolation of Brucella microti from mandibular lymph nodes of red foxes, Vulpes
vulpes, in lower Austria. Vector Borne Zoonotic Dis. 9, 153, 2009.
17. Scholz, H.C., et al. Brucella inopinata sp. nov., isolated form a breast implant infection. Int. J. Syst.
Evol. Microbiol. 60, 801, 2010.
18. Hofer, E., et al. A potential novel Brucella species isolated from mandibular lymph nodes of red foxes
in Austria. Vet. Microbiol. 155, 93, 2012.
19. Eisenberg, T., et al. Isolation of potentially novel Brucella spp. from frogs. Appl. Environ. Microbiol. 78,
3753, 2012.
20. Schlabritz-Loutsevitch, N.E., et al. A novel Brucella isolate in association with two cases of stillbirth in
non-human primates: first report. J. Med. Primatol. 38, 70, 2009.
21. Manthei, C.A. Application of research to bovine brucellosis control and eradication programs. J. Dairy
Sci. 51, 1115, 1968.
22. Teske, S.S., Huang, Y., Tamrakar, S.B., Bartrand, T.A., Weir, M.H., & Haas, C.N. Animal and human
dose-response models for Brucella species. Risk Anal. 31, 1576, 2011.
23. Olsen, S.C., & Palmer, M.V. Advancement of knowledge of Brucella over the past 50 years. Vet. Pathol.
51, 1076, 2014.
24. Bouvier, G. Distribution geographique de quelques maladies du gibier et des animaux sauvages de la
Suisse. Bull. Off. Int. Epizoot. 61, 67, 1964.
25. Rollinson, D.H.L. Brucella agglutinins in East African game animals. Vet. Rec. 74, 904, 1962.
26. Samaha, H., Al-Towaily, M., Khoudair, R.M., & Ashour, H.M. Multicenter study of brucellosis in
Egypt. Emerg. Infect. Dis. 14, 1916, 2008.
27. Wernery, U. Camelid brucellosis: a review. Rev. Sci. Tech. 33, 839, 2014.
Brucella 267
28. Gastellu, J., Garin-Bastuji, B., & Oudar, J. Pathologic features of a case of systemic brucellosis due to
Brucella melitensis biovar 3 in a chamois (Rupicapra rupicapra). Bull. Acad. Natl. Med. 177, 575, 1993.
29. El-Tras, W.F., Tayell, A.A., Eltholth, M.M., & Guitian, J. Brucella infection in fresh water fish: evidence
of natural infection of the Nile catfish Clarias gariepinus, with Brucella melitensis. Vet. Microbiol. 141,
321, 2010.
30. Godfroid, J., Garin-Bastuji, B., Saegerman, C., & Blasco, J.M. Brucellosis in terrestrial wildlife. Rev.
Sci. Tech. 32, 27, 2013.
31. Ewalt, D.R., Payeur, J.B., Rhyan, J.C., & Geer, P.L. Brucella suis biovar 1 in naturally infected cattle: a
bacteriological, serological and histological study. J. Vet. Diagn. Invest. 4, 417, 1997.
32. Foster, G., Osterman, B.S., Godfroid, J., Jacques, I., & Cloeckaert, A. Brucella ceti sp. nov. and Brucella
pinnipedialis sp. nov. for Brucella strains with cetaceans and seals as their preferred hosts. Int. J. Syst.
Evol. Microbiol. 57, 2688, 2007.
33. Cloeckaert, A., Bernardet, N., Koylass, M.S., Whatmore, A.M., & Zygmunt, M.S. Novel IS711 chromo-
somal location useful for identification of marine mammal Brucella genotype ST27, which is associated
with zoonotic infection. J. Clin. Microbiol. 49, 3954, 2011.
34. Corbel, M.J. Brucella phages: advances in the development of a reliable phage typing system for smooth
and non-smooth Brucella isolates. Ann. Inst. Pasteur Microbiol. 138, 70, 1987.
35. Olsen, S.C. Recent developments in livestock and wildlife brucellosis vaccination. Rev. Sci. Tech. 32,
207, 2013.
36. Giamartolomei, G.H., & Cassataro, J. Confronting the barriers to develop novel vaccines against brucel-
losis. Expert Rev. Vaccines 10, 1291, 2011.
37. Kaufmann, A., et al. Smooth and rough lipopolysaccharide phenotypes of Brucella induce different
intracellular trafficking and cytokine/chemokine release in human monocytes. J. Leukoc. Biol. 74,
1045, 2003.
38. Dawson, C.E., et al. Isolation and characterization of Brucella from the lungworms of a harbor porpoise
(Phocoena phocoena). J. Wildl. Dis. 44, 237, 2008.
39. Jimenez de Bagüés, M.P., Terraza, A., Gross, A., & Domand, J. Different responses of macrophages to
smooth and rough Brucella spp.: relationship to virulence. Infect. Immun. 72, 2429, 2004.
40. Rittig, M.G., Alvarez-Martinez, M.T., Porte, F, Liautard, J.P., & Rouot, B. Intracellular survival
of Brucella spp. in human monocytes involves conventional uptake but special phagosomes. Infect.
Immun. 69, 3995, 2001.
41. Celli, J. Surviving inside a macrophage: the many ways of Brucella. Res. Microbiol. 157, 93, 2006.
42. Porte, F., Liautard, J.P., & Kohler, S. Early acidification of phagosomes containing Brucella suis is
essential for intracellular survival in murine macrophages. Infect. Immun. 67, 4041, 1999.
43. Arellano-Reynoso, B., et al. Cyclic β-1,2-glucan is a Brucella virulence factor required for intracellular
survival. Nat. Immunol. 6, 618, 2005.
44. Cutler, S.J., Whatmore, A.M., & Commander, N.J. Brucellosis—new aspects of an old disease. J. Appl.
Microbiol. 98, 1270, 2005.
45. Bellaire, B.H., Elzer, P.H., Baldwin, C.L., & Roop, R.M. Production of the siderophore 2,3-dihydroxybenzoic
acid is required for wild-type growth of Brucella abortus in the presence of erythritol under low-iron con-
ditions in vitro. Infect. Immun. 71, 2927, 2003.
46. Rittig, M.G., et al. Smooth and rough lipopolysaccharide phenotypes of Brucella induce different intra-
cellular trafficking and cytokine/chemokine release in human monocytes. J. Leukoc. Biol. 74, 1045,
2003.
47. Oliveira, S.C., Giamartolomei, G.H., & Cassataro, J. Confronting the barriers to develop novel vaccines
against brucellosis. Expert Rev. Vaccines 10, 1291, 2011.
48. Franco, M.P., Mulder, M., Gilman, R.H., & Smits, H.L. Human brucellosis. Lancet Infect. Dis. 7, 775,
2007.
49. Mantur, B.G., Amarnath, S.K., & Shinde, R.S. Review of clinical and laboratory features of human
brucellosis. Indian J. Med. Microbiol. 25, 188, 2007.
50. Baldi, P.C., & Giambartolemi, G.H. Pathogenesis and pathobiology of zoonotic brucellosis in humans.
Rev. Sci. Tech. 32, 117, 2013.
51. Ariza, J., et al. Perspectives for the treatment of brucellosis in the 21st century: the Ioannina recommen-
dations. PLoS Med. 4, e317, 2007.
268 Laboratory Models for Foodborne Infections
52. Corbel, M.J. Brucellosis in humans and animals. World Health Organization. http://www.who.int/csr/
resources/publications/Brucellosis.pdf, 2006.
53. Celebi, G., Külah, C., Kilic, S., & Ustündağ, G. Asymptomatic Brucella bacteraemia and isolation of
Brucella melitensis biovar 3 from human breast milk. Scand. J. Infect. Dis. 39, 205, 2007.
54. Al-Mafada, S.M., Al-Eissa, Y.A., Saeed, E.S., & Kambal, A.M. Isolation of Brucella melitensis from
human milk. J. Infect. 26, 346, 1993.
55. Arroyo Carrera, I., Lopez Rodriguez, M.J., Sapina, A.M., Lopez Lafuenta, A., & Sacristan, A.R.
Probable transmission of brucellosis by breast milk. J. Trop. Pediatr. 52, 380, 2006.
56. Doyle, T.J., & Bryan, R.T. Infectious disease morbidity in the US region bordering Mexico, 1990–1998.
J. Infect. Dis. 182, 1503, 2000.
57. Troy, S.B., Rickman, L.S., & Davis, C.E. Brucellosis in San Diego: epidemiology and species-related
differences in acute clinical presentations. Medicine (Baltimore) 84, 174, 2005.
58. Olsen, S.C. Recent developments in livestock and wildlife brucellosis vaccination. Rev. Sci. Tech. 32,
207, 2013.
59. Orkhon, D., Chimed-Ochir, G., Nansalmaa, M., Kolar, J., & Vounatsou, P. A model of animal-human
brucellosis transmission in Mongolia. Prev. Vet. Med. 69, 77, 2005.
60. Thakur, A., Pedersen, L.E., & Jungersen, G. Immune markers and correlates of protection for vaccine-
induced immune responses. Vaccine 30, 4907, 2012.
61. Van den Heever, L.W, Katz, K.W., & Te Brugge, L.A. On the inactivation of Brucella abortus in natu-
rally contaminated milk by commercial pasteurisation procedures. J. South Afr. Vet. Assoc. 53, 233,
1982.
62. CFSPH-YSU. Porcine and rangiferine brucellosis: Brucella suis. Enzootic abortion, contagious abor-
tion, undulant fever. Disease factsheets. B. suis. The Centre of Food Security and Public Health, Iowa
State University, 2007.
63. Kangumba, J.G., Venter, E.H., & Coetzer, J.A. The effect of activation of the lactoperoxidase system and
souring on certain potential human pathogens in cows milk. J. South Afr. Vet. Assoc. 68, 130, 1997.
64. Mendez-Gonzalez, K., et al. Brucella melitensis survival during manufacture of ripened goat cheese at
two temperatures. Foodborne Pathog. Dis. 12, 11257, 2011.
65. Plommet, M., Fensterbank, R., Vassal, L., Auclair, J., & Mocquot, G. Survival of Brucella abortus in
ripened soft cheese made from naturally infected cow’s milk. Le Lait 68, 115, 1988.
66. Santiago-Rodiquez Mdel, R., et al. Survival of Brucella abortus aqpX mutant in fresh and ripened
cheeses. Foodborne Pathog. Dis. 12, 170, 2015.
67. Falenski, A., Mayer-Scholl, A., Filter, M., Göllner, C., Appel, B., & Nöckler, K. Survival of Brucella
spp. in mineral water, milk and yogurt. Int. J. Food Microbiol. 145, 326, 2011.
68. Mendez-Gonzalez, K.Y., et al. Brucella melitensis survival during manufacture of ripened goat cheese
at two temperatures. Foodborne Path. Dis. 8, 1257, 2011.
69. Hutchings, L.M., McCullough, N.B., Donham, C.B., Eisele, C.W., & Bounnell, D.F. The viability of
Brucella melitensis in naturally infected cured hams. Public Health Rep. 66, 1402, 1951.
70. Smith, J. Excretion of Br. Abortus in milk. J. Compar. Pathol. Ther. 47, 123, 1934.
71. Fleischner, E.C., Vecki, M., Shaw, E.B., & Meyer, K.F. The pathogenicity of B. abortus and B. meliten-
sis for monkeys. J. Infect. Dis. 29, 663, 1921.
72. Capparelli, R., et al. Heterogeneous shedding of Brucella abortus in milk and its effect on the control
of animal brucellosis. J. Appl. Microbiol. 106, 2041, 2009.
73. Brinley Morgan, W.J., & McDiarmid, A. The excretion of Brucella abortus in the milk of experimen-
tally infected cattle. Res. Vet. Sci. 1, 53, 1960.
74. Beattie, C.P., & Rice, R.M. Undulant fever due to Brucella of the porcine type—Brucella suis. J. Am.
Med. Assoc. 201, 1934, 1970.
75. Borts, I.H., Harris, D.M., Joynt, M.F., Jennings, J.R., & Jordan, C.F. A milk borne epidemic of brucel-
losis: caused by the porcine type of Brucella (Brucella suis) in a raw milk supply. J. Am. Med. Assoc.
121, 319, 1943.
76. Committee on Infectious Diseases, Committee on Nutrition, American Academy of Pediatrics.
Consumption of raw or unpasteurized milk and milk products by pregnant women and children.
Pediatrics 133, 175, 2014.
77. Arnow, P.M., Smaron, M., & Ormiste, V. Brucellosis in a group of travelers to Spain. J. Am. Med. Assoc.
251, 505, 1984.
Brucella 269
78. Gautret, P., Benkouiten, S., Gaillard, C., Parola, P., & Brouqui, P. Camel milk-associated infection risk
perception and knowledge in French Haji pilgrims. Vector Borne Zoonotic Dis. 13, 425, 2013.
79. Di Pierdomenico, A., Borgia, S.M., Richardson, D., & Baqi, M. Brucellosis in a returned traveller. Can.
Med. Assoc. J. 183, E690, 2011.
80. Chommel, B.B., et al. Changing trends in the epidemiology of human brucellosis in California from
1973 to 1992. A shift toward foodborne transmission. J. Infect. Dis. 178, 1216, 1994.
81. Taylor, J.P., & Perdue, J.N. The changing epidemiology of human brucellosis in Texas 1977–1986. Am.
J. Epidemiol. 130, 160, 1989.
82. Al Dahouk, S., et al. Human brucellosis in a nonendemic country: a report from Germany, 2002 and
2003. Eur. J. Clin. Microbiol. Infect. Dis. 24, 450, 2005.
83. Galbraith, N.S., Ross, M.S., DeMowbray, R.R., & Payne, D.J. Outbreak of Brucella melitensis type 2
infection in London. Br. Med. J. 1, 612, 1969.
84. Thaper, M.K., & Young, E.J. Urban outbreak of goat cheese brucellosis. Pediatr. Infect. Dis. 640, 1986.
85. Beutlich, J., et al. Characterization of illegal food items and identification of foodborne pathogens
brought into the European Union via two major German airports. Int. J. Food Microbiol. 209, 13–19,
2015, doi: 10.1016/j.ijfoodmicro.2014.10.017.
86. Fosgate, G.T., Carpenter, T.E., Chomel, B.B., Case, J.T., DeBess, E.E., & Reilly, K.F. Time-space clus-
tering of human brucellosis, California, 1973–1992. Emerg. Infect. Dis. 8, 672, 2002.
87. Hassan Ali, N., Farooqui, A., Khan, A., Khan, A.Y., & Kazmi, S.U. Microbial contamination of raw
meat and its environment in retail shops in Karachi, Pakistan. J. Infect. Dev. Countries 4, 382, 2010.
88. Pappas, G., Akritidis, N., Bosilkovski, M., & Tsianos, E. Brucellosis. New Engl. J. Med. 352, 2325,
2005.
89. Mohd, M.G. Brucellosis in the Gezira area, Central Sudan. J. Trop. Med. Hyg. 92, 86, 1989.
90. Chan, J., Baxter, C., & Wenman, W.M. Brucellosis in an inuit child, probably related to caribou meat
consumption. Scand. J. Infect. Dis. 21, 337, 1989.
91. Whatmore, A.M., et al. Marine mammal Brucella genotype associated with zoonotic infection. Emerg.
Infect. Dis. 14, 517, 2008.
92. Alton, G.G., Jones, L.M., Angus, R.D., & Verger, J.M. Techniques for the brucellosis laboratory. Institut
National de la Recherche Agronomique, Paris, pp. 33, 154, 1988.
93. Islam, M.A., Khatun, M.M., & Baek, B.K. Male rats transmit Brucella abortus biotype 1 through sexual
intercourse. Vet. Microbiol. 165, 475, 2013.
94. Ivanov, A.V., Salmakov, K.M., Olsen, S.C., & Plumb, G.E. A live vaccine from Brucella abortus strain
82 for control of cattle brucellosis in the Russian Federation. Anim. Health Res. Rev. 12, 113, 2011.
95. Silva, T.M., Costa, E.A., Paixão, T.A., Tsolis, R.M., & Santos, R.L. Laboratory animal models for bru-
cellosis research. J. Biomed. Biotechnol. 2011, 518323, 2011.
18
Burkholderia
CONTENTS
18.1 Introduction................................................................................................................................... 271
18.2 B. mallei........................................................................................................................................ 272
18.2.1 Overview.......................................................................................................................... 272
18.2.2 Laboratory Infection Models............................................................................................ 272
18.3 B. pseudomallei............................................................................................................................. 273
18.3.1 Overview.......................................................................................................................... 273
18.3.2 Laboratory Infection Models.............................................................................................274
18.4 Burkholderia cepacia Complex (Bcc).......................................................................................... 275
18.4.1 Overview.......................................................................................................................... 275
18.4.2 Laboratory Infection Models............................................................................................ 276
18.5 B. gladioli pv. cocovenenans........................................................................................................ 278
18.5.1 Overview.......................................................................................................................... 278
18.5.2 Laboratory Infection Models............................................................................................ 278
18.6 Conclusions................................................................................................................................... 279
References............................................................................................................................................... 279
18.1 Introduction
The genus Burkholderia covers a diverse group of Gram-negative β-proteobacteria. Although currently
at least 60 species and proposed species exist in the genus Burkholderia, very few have been studied
extensively. Much of the research to date has focused on the bacteria of the Burkholderia cepacia com-
plex (Bcc), Burkholderia mallei, Burkholderia pseudomallei, and more recently, Burkholderia gladioli.
The bacteria of the Bcc are pathogens that typically cause serious infections in plants, animals, and
humans.1–3 However, they can also be beneficial in the environment as they fix nitrogen symbiotically
for plants, produce antibiotics and antifungals, and have the capacity to degrade organic and xenobiotic
compounds.4–6 B. mallei causes “glanders,” a rare condition usually associated with horses, but that can
also affect humans, whereas B. pseudomallei, endemic in Southeast Asia, causes “melioidosis,” a serious
disease in humans with a wide variety of symptoms.7 The species B. gladioli has been divided into four
pathovars: gladioli, alliicola, agaricicola, and cocovenenans.8 The first three B. gladioli pathovars listed
are primarily plant pathogens,9–11 but members of these pathovars can occasionally also infect immu-
nocompromised patients with chronic granulomatous disease (CGD), cystic fibrosis (CF), or acquired
immune deficiency syndrome (AIDS).8,12,13 The taxonomic description of B. gladioli pvs. gladioli, alli-
icola, and agaricicola, published in 2003, suggests that they do not produce toxins that are harmful to
humans, although some strains have since been shown to synthesize toxoflavin.8,14 The fourth B. gladioli
pathovar, B. gladioli pv. cocovenenans, is distinct from the other pathovars with regards to its epidemiol-
ogy and pathogenicity. Although the other Burkholderia species can be found as contaminants in food
and water supplies (including the Bcc, B. mallei, and B. pseudomallei), B. gladioli pv. cocovenenans is
the only bacterium of the Burkholderia genus that is traditionally characterized as a foodborne pathogen.
271
272 Laboratory Models for Foodborne Infections
Strains of B. gladioli pv. cocovenenans do not cause disease directly, but instead produce toxins that
contaminate foods before ingestion by humans, similar to Clostridium botulinum. The laboratory infec-
tion models used to test Burkholderia pathogenicity and virulence are generally useful across the genera,
whereas specific models have been devised to measure B. gladioli toxin activity. This chapter will briefly
present the literature regarding the presence of B. mallei, B. pseudomallei, the Bcc, and B. gladioli in
food and water supplies, and subsequently review the laboratory infection models that have been devel-
oped for each group of Burkholderia pathogenic bacteria.
18.2 B . mallei
18.2.1 Overview
B. mallei causes glanders, a zoonosis disease of horses that can also be transmitted to humans. The
symptoms vary depending on the route of transmission, but can include pneumonia, skin lesions, and
septic shock.15 Horses generally become infected following ingestion of B. mallei introduced into their
food and water by other infected horses.15 Humans may also become infected via ingestion, typically due
to drinking untreated water.16 Glanders is difficult to diagnose, there is no vaccine available to protect
against the bacterium, and any potentially successful therapy requires prolonged antibiotic treatment.
However, as a result of stringent measures implemented to control the spread of glanders, the disease
has not been seen in the Western hemisphere since 1939 (except following accidental laboratory expo-
sure).17,18 However, because both B. mallei and B. pseudomallei have been identified as potential food
contaminants, there is concern that these species could be released as bioterrorism agents into a coun-
try’s food supply.19,20
typical clinical signs and tissue histopathology, whereas intranasal inoculation at this dosage did not
produce any obvious symptoms of disease. The differences observed in bacterial dosage and survival
reflect the different routes of inoculation used: subcutaneous injection likely circumvents physical bar-
riers used to combat infection, such as the respiratory system’s mucosal barrier, and allows immediate
access to host tissues. These results suggest that, depending on the site of inoculation and dosage used,
marmosets can be used to model effectively different aspects of B. mallei infection and pathogenesis,
producing results similar to infections observed in humans and equids.
Also recently, researchers have begun to utilize low-cost alternative infection models in order to study
various aspects of B. mallei infection. For example, in order to evaluate 650 potential gene targets in
the B. mallei “virulome,” Schell et al.35 utilized Galleria mellonella (wax moth) larvae as a surrogate
host. B. mallei was highly pathogenic in this host, and importantly, four previously identified mutants
avirulent in hamster and mouse models also proved to be avirulent in the wax moth larvae, thus vali-
dating this approach.35 Similarly, Madagascar hissing (MH) cockroaches possess a number of qualities
that make them desirable for use as B. mallei surrogate hosts, including low cost, ease of breeding and
handling, a competent innate immune system, and the ability to thrive at 37°C.36 MH cockroaches are
highly susceptible to B. mallei infection, the LD50 being <10 CFU. Similar to the results obtained for
G. mellonella, B. mallei avirulent Type VI secretion system mutants originally identified in rodents were
also attenuated in MH cockroaches.36 Again, this indicates that alternative infection models can be effec-
tive and may be useful as an alternative to mammals to study some aspects of host–pathogen interactions
and bacterial virulence determinants.
18.3 B . pseudomallei
18.3.1 Overview
B. pseudomallei causes melioidosis, a potentially fatal condition with a variety of symptoms including
pneumonia, skin lesions, and septic shock.7 Most melioidosis cases are seen in Southeast Asia (especially
Thailand) and northern Australia, although incidents also occur in nonendemic areas such as Brazil.7,37 In
Southeast Asia, approximately 80% of the population tests seropositive by the age of 4 years,38 although
only a small percentage of these seropositive individuals go on to develop melioidosis. This finding sug-
gests that the dose and frequency of inoculum influences the infectivity and severity of the symptoms. In
support of this idea, increased rates of melioidosis follow the monsoon and typhoon seasons,39,40 when
prolonged inhalation of storm-generated aerosols of B. pseudomallei-contaminated particles would be
highest.7
Although the first cases of melioidosis were described in 1913, there remains some debate as to how
B. pseudomallei is transmitted to humans.41,42 It has been established that infections can occur following
inoculation of broken skin or inhalation of water aerosols containing B. pseudomallei.43 Although there
is some evidence of infection occurring following ingestion, it is by no means conclusive. Historically,
it was thought that melioidosis was spread through the consumption of contaminated food and water,
as it can be transmitted in this manner to guinea pigs, rabbits, and rats.41,44 However, B. pseudomallei
has not been definitively shown to spread to humans by this route, although a report published in 1998
described the development of melioidosis in five adults in northwestern Australia.45 Using pulsed-field
gel electrophoresis (PFGE) typing, it was determined that all five patients were infected with the same
PFGE type and that this type was identical to that of B. pseudomallei isolates found in the area’s drink-
ing water, suggesting that these cases developed due to ingestion of a single source of B. pseudomallei.45
B. pseudomallei can also be isolated from drinking water in areas where melioidosis is not endemic.
Zanetti et al.46 showed that B. pseudomallei could be isolated from 7.1% of drinking water samples
taken in Bologna, Italy, and that the levels of B. pseudomallei were relatively high, ranging from 1020
to 15,000 CFU/100 mL sample. Treatment of water with chlorine will kill B. pseudomallei (as well as
B. mallei) in an experimental setting, but various factors in the environment (including attachment and
nutrient limitation) may affect this susceptibility.47 Antibiotic treatment for acute and chronic melioi-
dosis includes ceftazidime followed by trimethoprim–sulfamethoxazole (TMP–SMX) with or without
274 Laboratory Models for Foodborne Infections
doxycycline.48 Despite this, melioidosis has a high mortality rate and is the third most common cause of
death from an infectious disease in endemic areas.49 Probable causes for this include late diagnosis due
to nonspecific symptomatology and a high recurrence rate when antibiotics are discontinued early. Given
the severity of the disease, it is understandable that there is significant interest in the development of a
B. pseudomallei vaccine, and the identification of an accurate laboratory infection model for melioidosis.
T6SS-1 Hcp proteins, integral membrane proteins of the T6SS mechanism that are expressed dur-
ing functional operation of the system, were found to be important in the intracellular behavior of
B. pseudomallei in RAW 264.7 macrophages. Mutants unable to express the Hcp1 protein were only
weakly cytotoxic toward murine macrophages, exhibited a growth defect inside of macrophages, and
were unable to form multinucleated macrophage cells like wild-type B. pseudomallei.58 In a different
murine macrophage cell line (J774A.1), it was shown that different strains of B. pseudomallei exhibit
different levels of pathogenicity and that these differing strain-dependent levels of pathogenicity were
conserved across mice infection models, murine macrophages, and a G. mellonella (wax moth larvae)
alternative infection model.63 It was also shown that in these same models, related species B. oklaho-
mensis and B. thailandensis were significantly less virulent than B. pseudomallei strains.63 Interestingly,
another study demonstrated an inverse relationship between B. pseudomallei virulence in a murine mac-
rophage J774 A1 phagocytosis assay as compared to a mouse infection model.54 However, in this case,
the mouse model used was an intraperitoneal infection model, which, as mentioned above, does not
accurately mimic the human condition, even though it is reproducible.54
Besides G. mellonella, which has also been used to identify virulence factors of and antibiotic phar-
macokinetics against B. pseudomallei,64,65 another invertebrate alternative infection model that has been
successfully used to characterize B. pseudomallei infection is Caenorhabditis elegans.66 At least five dif-
ferent bacterial mechanisms of killing C. elegans have been described, including toxin-mediated killing,
persistent or transient killing, intestinal infection, direct invasion, and biofilm formation.67 Originally,
studies suggested that a B. pseudomallei toxin was involved in C. elegans killing68; however, more recent
studies suggest that no toxin-mediated killing is involved, but rather direct prolonged contact with the
bacterium.69 How these putative B. pseudomallei virulence mechanisms identified in C. elegans relate
to the human infection condition remains to be determined. Unlike the G. mellonella infection model,
both C. elegans and the tomato leaf 70 infection models are unable to differentiate between B. pseudo-
mallei and B. thailandensis virulence levels, which may be an important consideration with respect to
accurately modeling human melioidosis.
18.4 B
urkholderia cepacia Complex (Bcc)
18.4.1 Overview
The Bcc is currently made up of 20 closely related Burkholderia species.71,72 These bacteria cause poten-
tially fatal infections in immunocompromised patients, particularly those with CF and CGD.73,74 Whereas
both melioidosis and glanders can have a wide range of symptoms that affect many different systems of
the body, Bcc infections tend to be limited to the lungs. Bcc infections may be asymptomatic, may cause
a patient’s condition to worsen over time, or may develop into a rapidly fatal pneumonia accompanied by
damage to the lung tissue and septicemia.3
The bacteria of the B. cepacia complex are found more commonly in food and water supplies than
either B. pseudomallei or B. mallei. They can be isolated from the rhizospheres of rice, corn, wheat, soy-
bean, and alfalfa plants.75,76 Bcc bacteria have also been found associated with Pleurotus ostreatus, the
oyster mushroom, which is the second-most cultivated mushroom in the United States.77,78 Bcc organisms
may cause spoilage in foods as diverse as onions, Swiss cheese, and Parma ham.1,79,80 However, these
bacteria can also prevent food spoilage: a Bcc strain isolated from Washington navel oranges was found
to prevent fungal infection of these fruits during storage.81 Because of the ability of the Bcc to produce
novel antifungals and antibiotic compounds and fix nitrogen for certain crops, there has been a great deal
of interest in developing these species into agricultural product growth promoters. However, because of
the potential risk to immunocompromised persons, the United States Environmental Protection Agency
(EPA) has placed a moratorium upon the use of Bcc bacteria for agricultural purposes since 2003.82
Despite their occasional identification in food products, including bakery items, onions, cheese, ham,
and oranges, in an extensive examination of different foods, Moore et al.83 were only able to isolate Bcc
from unpasteurized milk. Similarly, Bcc species were isolated from unpasteurized milk by Uraz and
Çitak84,85 and Munsch-Alatossava and Alatossava.86,87 It is unknown how milk becomes contaminated,
276 Laboratory Models for Foodborne Infections
but possibly it is through transfer of Bcc organisms from soil to the cow udder and/or to milk stor-
age tanks.88 However, the Bcc are not found in commercially available dairy products and are effec-
tively killed by pasteurization.83,88 Berriatua et al.2 found that the Bcc species Burkholderia cenocepacia
and Burkholderia vietnamiensis were responsible for causing subclinical mastitis (inflammation of the
udder) in milking sheep. However, when Moore et al.88 examined milk samples taken from cows with
mastitis to determine whether this condition was responsible for the contamination, they were unable to
isolate Bcc organisms.
Like B. pseudomallei, Bcc organisms can also be isolated from drinking water. Zanetti et al.46 found
that 3.5% of drinking water samples in Bologna, Italy, contained Bcc bacteria. The counts in these
samples were very low compared to B. pseudomallei, containing between 1 and 19 CFU/100 mL sample.
Pegues et al.89 examined a Bcc isolate from a water jug at a CF summer camp and found that the ribo-
type of this isolate was different from the ribotypes taken from patients infected at the camp, indicating
that this water was not the source of infection. In contrast to these findings, samples isolated by Moore
et al.83 and Vermis et al.90 from a variety of drinking water samples, including bottled, tap, well, and
spring water, were found to not contain Bcc organisms. However, the major contaminants of the NASA
space shuttle water systems for more than 24 missions have been bacteria of the Bcc, with more than
20 CFU/100 mL detected.91
To date, only one study has suggested a link between food consumption and Bcc infection. Fisher
et al.92 found that Bcc isolates taken from cucumber, lettuce, and cauliflower purchased from salad bars
and food stores near two CF centers were of the same ribotype that was most commonly isolated from
the patients in these centers. However, it is not possible to discern from these results whether or not these
foods acted as vectors of Bcc infection.
virulence factors evolved to combat natural predators in the environment and that different factors
might be effective against more than one type of organism. Because the innate immune systems of
invertebrates and vertebrates share many common well-conserved features, a bacterium able to infect
and/or cause disease in one type of host may use similar mechanisms and strategies to infect and/or
cause disease in another.106,107
Because the Bcc are natural pathogens of onions, causing soft rot,1 one of the earliest alternative infec-
tion models developed for the Bcc permitted the observation of the maceration of onion tissue.108–110 Such
studies identified a plasmid-encoded pectate hydrolase as the enzyme responsible for maceration in type
strain B. cenocepacia ATCC 25416.108 But B. cepacia and B. cenocepacia strains lacking this enzyme
can also produce a tissue water soaking phenotype through expression of a plasmid-encoded Type IV
secretion system gene cluster.111 It was also discovered that quorum sensing appears to be involved in
onion tissue maceration, as the quorum sensing signal synthase gene cepI and regulator gene cepR
mutants are attenuated for maceration.112
Another simple model used to assess Bcc virulence against plants is the alfalfa seedling infection
model,103 in which the yellowing of leaves (chlorosis), stunted root development, and brown tissue
destruction on leaves and roots were used as a measure of bacterial virulence. This model has been used
with success to ascertain the role of various virulence factors expressed by the Bcc.103 More recently,
the common duckweed (Lemna minor) was used for similar purposes, the advantage of duckweed
being that it can be grown axenically. Several different virulent factors discovered in other Bcc hosts
were also found to be virulent factors in duckweed,113 confirming duckweed’s validity as an infection
model. Interestingly, duckweed was also found to be an effective infection model for enteropathogenic
Escherichia coli.113
A large number of Bcc researchers are also now using insect animal models such as wax moth larvae
(G. mellonella) or fruit flies (Drosophila melanogaster) as substitutes for mammalian infection mod-
els.114,115 Such “lower” animal infection models hold the advantage that they typically possess at least
an innate immunity system, but do not involve all of the regulatory and ethical requirements needed for
“higher” animal experimentation. Although each alternative infection host may not exactly model the
same types of tissue found in humans, it is thought that for each different host, a subset of the bacterium’s
total armamentarium is expressed to overcome that host’s defenses. Therefore, at least some of the cel-
lular weapons used by the Bcc to defeat G. mellonella would also be expressed when the Bcc bacterium
finds itself in the human lung. Since its development,114 the G. mellonella infection model has been
used to identify a novel hemolytic toxin,116 as well as several other Bcc virulence factors,117–119 and also
show that the larvae can be rescued from lethal bacterial infection by the action of bacteriophages.120
Similarly, D. melanogaster has been successfully employed to demonstrate that different strains of the
Bcc can kill by infection. Unlike results found for Pseudomonas aeruginosa in D. melanogaster, species
of the B. cepacia complex were only able to kill flies by infection after pricking, rather than through
ingestion.115 This suggests that the Bcc are less toxic to fruit flies than other opportunistic CF bacterial
pathogens. Nevertheless, the results obtained with the fruit fly model are comparable with those obtained
using mammalian infection models, and B. cenocepacia K56–2 mutants less virulent in murine hosts or
other alternative infection models than wild type are also less virulent in the fruit fly, even when tested
using competitive index analyses.115
Caenorhabditis elegans, a roundworm nematode, has also been used successfully as a Bcc model
for mammalian infection.121 These studies have shown that there are two modes of killing, a fast mode
due to paralysis by an extracellular Bcc toxin and a slow mode due to ingestion and accumulation of
bacteria in the nematode intestine.121 The Bcc quorum sensing system appears to be required for both
types of killing,121 but only the slow mode was shown to be partially due to certain Bcc virulence fac-
tors such as AidA.122 Several different environmental parameters can influence the performance of this
model, including worm age,123 prior diet,124 diet,125 Bcc strain,102,126,127 and Bcc strain mucoidy status.128
Nevertheless, a number of Bcc virulence factors effective against C. elegans have been discovered or
confirmed using this model, including the type III secretion system,129 the phenylacetic acid catabolic
pathway,130 two different Hfq proteins,1321,132 the pleiotropic regulator of phenazines Pbr,133 a small regu-
latory RNA MtvR,134 a Type 2 secretion system pseudopilin GspJ,135 and the third chromosome/virulence
plasmid pC3 in many Bcc species.136,137
278 Laboratory Models for Foodborne Infections
The final Bcc alternative infection model to be discussed is perhaps the most recently developed
zebrafish embryos.138 Bcc bacteria survive and multiply in zebrafish macrophages, later disseminating
and causing bacteremia and death within 3 days. B. cenocepacia strains that are highly virulent in
other animal models are also highly virulent for the embryos, whereas less virulent Bcc species were
controlled by the fish innate immune system.138 This model was used to confirm that both the quorum
sensing system and the third chromosome are required for full virulence of B. cenocepacia in the zebraf-
ish embryos.136,138
BA toxin is one of the most potent respiratory poisons known. Following human consumption of food
contaminated with B. gladioli pv. cocovenenans, intoxication occurs rapidly, usually within 1–10 h.164
The classical symptoms are hyperglycemia followed by hypoglycemia, spasms, loss of consciousness,
and death.165 Following autopsy, the liver, kidneys, and brain all show signs of damage, and in some
cases, there is also damage to the heart, lungs, gastrointestinal tract, and spleen.164 Laboratory animal
models of infection are generally too sensitive to BA toxin to be of much experimental value. When food
contaminated with B. gladioli pv. cocovenenans is fed to dogs, they develop spasms, enter comas, and
die within 2–3 h.164 Dogs and rhesus monkeys fed with B. gladioli pv. cocovenenans culture supernatants
die within 6–33 h and 15.5–35 h, respectively.146 When mice were given B. gladioli pv. cocovenenans
culture supernatants (0.4 mL/mouse) partially purified by the method of Hu et al.,166,167 all mice died
within 45 min with stiff tail and feet.8
With respect to a mechanism of action, BA inhibits oxidative phosphorylation by binding to the ade-
nine nucleotide translocator, which normally shuttles ADP and ATP across the inner mitochondrial
membrane.168 The activity of this transporter is thought to inhibit cellular apoptosis.169 Because BA
inhibits oxidative phosphorylation, the only way for cells to generate ATP is through anaerobic glycoly-
sis.170 This metabolic changeover is responsible for the symptoms of intoxication. Glycogen stores are
broken down to allow for an increased level of glycolysis, leading to hyperglycemia and increased lactic
acid levels, but these stores are soon depleted, resulting in hypoglycemia and an inability to regenerate
ATP.171 It is the ATP depletion that is fatal and not the hypoglycemia, as the injection of glucose is insuf-
ficient to prevent death.171
18.6 Conclusions
The major pathogenic bacteria of the genus Burkholderia include the species B. mallei, B. pseudomal-
lei, the B. cepacia complex, and B. gladioli. Surprisingly, the diseases that each of these bacteria causes
are diverse in terms of host, mechanism of action, prognosis, and outcome. Coordinately, the laboratory
systems used to model these diseases and assess the virulence and toxicity of each of these bacterial spe-
cies are diverse as well. For B. mallei, the causative agent of glanders, the primary hosts are equine, and
thus, a variety of equine animal models can be used to study glanders disease progression. For B. pseu-
domallei, the modeling of the disease melioidosis is much more difficult, as the disease progression is
more complex, including variable human immune interactions, a latency period, and multifactorial local
and systemic virulence effects. Moreover, as humans appear to be a primary host, different aspects of
the disease are best studied in different animal models. Similarly, the B. cepacia complex is best studied
using a variety of animal models, due to its multifactorial virulence factor production. In contrast to
B. pseudomallei, however, modeling B. cepacia complex disease typically requires the animals to be
immunocompromised in some way, as healthy animals (and humans) are generally able to resist Bcc
infections. Finally, nontoxigenic B. gladioli disease modeling can be performed with systems similar to
those used for the Bcc, whereas toxigenic B. gladioli can be studied at the organismal level, or through
the purification of the toxin and with the use of specialized cellular or molecular assays. Historically,
murine and equine animal models have been used for the study of Burkholderia infections and toxicol-
ogy. In recent years, however, there has been a great deal of interest in alternative host infection models
to analyze all or specific components of the diseases caused by Burkholderia species. The development
of new and better laboratory models of disease caused by Burkholderia will bring about a better under-
standing of the disease progression and virulence factors utilized by each individual Burkholderia spe-
cies, leading to the identification of drug targets and the development of antibacterial strategies against
pathogenic Burkholderia species.
REFERENCES
1. Burkholder, W. H. 1950. Sour skin, a bacterial rot of onion bulbs. Phytopathology. 40:115–117.
2. Berriatua, E., et al. 2001. Outbreak of subclinical mastitis in a flock of dairy sheep associated with
Burkholderia cepacia complex infection. J. Clin. Microbiol. 39:990–994.
280 Laboratory Models for Foodborne Infections
3. Isles, A., I. Maclusky, and M. Corey. 1984. Pseudomonas cepacia infection in cystic fibrosis: an emerg-
ing problem. J. Pediatr. 104:206–210.
4. Estrada-De Los Santos, P., R. Bustillos-Cristales, and J. Caballero-Mellado. 2001. Burkholderia, a
genus rich in plant-associated nitrogen fixers with wide environmental and geographic distribution.
Appl. Environ. Microbiol. 67:2790–2798.
5. El-Banna, N., and G. Winkelmann. 1998. Pyrrolnitrin from Burkholderia cepacia: antibiotic activity
against fungi and novel activities against streptomycetes. J. Appl. Microbiol. 85:69–78.
6. Folsom, B. R., P. J. Chapman, and P. H. Pritchard. 1990. Phenol and trichloroethylene degradation
by Pseudomonas cepacia G4: kinetics and interactions between substrates. Appl. Environ. Microbiol.
56:1279–1285.
7. Wiersinga, W. J., T. van der Poll, N. L. White, N. P. Day, and S. J. Peacock. 2006. Melioidosis: insights
into the pathogenicity of Burkholderia pseudomallei. Nat. Rev. Microbiol. 4:272–282.
8. Jiao, Z., et al. 2003. Need to differentiate lethal toxin-producing strains of Burkholderia gladioli, which
cause severe food poisoning: description of B. gladioli pathovar cocovenenans and an emended descrip-
tion of B. gladioli. Microbiol. Immunol. 47:915–925.
9. Ballard, R. W., N. J. Palleroni, M. Doudoroff, R. Y. Stanier, and M. Mandel. 1970. Taxonomy of the
aerobic pseudomonads: Pseudomonas cepacia, P. marginata, P. alliicola and P. caryophylli. J. Gen.
Microbiol. 60:199–214.
10. Ko, S. S., W. N. Chang, J. F. Wang, S. J. Cherng, and S. Shanmugasundaram. 2002. Storage variability
among short-day onion cultivars under high temperature and high relative humidity, and its relationship
with disease incidence and bulb characteristics. J. Am. Soc. Hortic. Sci. 127:848–854.
11. Gill, W. M., and A. Tsuneda. 1997. The interaction of the soft rot bacterium Pseudomonas gladioli pv.
agaricicola with Japanese cultivated mushrooms. Can. J. Microbiol. 43:639–648.
12. Ross, J. P., S. M. Holland, V. J. Gill, E. S. DeCarlo, and J. I. Gallin. 1995. Severe Burkholderia
(Pseudomonas) gladioli infection in chronic granulomatous disease: report of two successfully treated
cases. Clin. Infect. Dis. 21:1291–1293.
13. Graves, M., T. Robin, A. M. Chipman, J. Wong, S. Khashe, and J. M. Janda. 1997. Four additional cases of
Burkholderia gladioli infection with microbiological correlates and review. Clin. Infect. Dis. 25:838–842.
14. Ura, H., N. Furuya, K. Iiyama, M. Hidaka, K. Tsuchiya, and N. Matsuyama. 2006. Burkholderia gladi-
oli associated with symptoms of bacterial grain rot and leaf-sheath browning of rice plants. J. Gen.
Plant Pathol. 72:98–103.
15. Whitlock, G. C., D. Mark Estes, and A. G. Torres. 2007. Glanders: off to the races with Burkholderia
mallei. FEMS Microbiol. Lett. 277:115–122.
16. Gilad, J., I. Harary, T. Dushnitsky, D. Schwartz, and Y. Amsalem. 2007. Burkholderia mallei and
Burkholderia pseudomallei as bioterrorism agents: national aspects of emergency preparedness. Israel
Med. Assoc. J. 9:499–503.
17. Wilson, G. S., and A. Miles. 1975. Topley and Wilson’s Principles of Bacteriology, Virology and
Immunity, 6th Edition. Edward Arnold, London.
18. Srinivasan, A., et al. 2001. Glanders in a military research microbiologist. New Engl. J. Med.
345:256–258.
19. Rotz, L. D., A. S. Khan, S. R. Lillibridge, S. M. Ostroff, and J. M. Hughes. 2002. Public health assess-
ment of potential biological terrorism agents. Emerg. Infect. Dis. 8:225–230.
20. Takhistov, P., and C. M. Bryant. 2006. Protecting the food supply. Food Technol. 60:34–43.
21. Schadewaldt, H. 1975. Discovery of glanders bacillus. Dtsch. Med. Wochenschr. 100:2292–2295.
22. Miller, W. R., L. Pannell, L. Cravitz, W. A. Tanner, and T. Rosebury. 1948. Studies on certain biological
characteristics of Malleomyces mallei and Malleomyces pseudomallei, II: virulence and infectivity for
animals. J. Bacteriol. 55:127–135.
23. Bondi, S. K., and J. B. Goldberg. 2008. Strategies towards vaccine against Burkholderia mallei and
Burkholderia pseudomallei. Expert Rev. Vaccines. 7:1357–1365.
24. Dembek, Z. F. 2007. Medical Aspects of Biological Warfare. Borden Institute, Washington, DC.
25. Wagg, D. M., and D. DeShazer. 2004. Glanders: new insights into an old disease, pp. 209–237. In L. E.
Lindler, F. J. Lebeda, and G. W. Korch (eds.), Biological Weapons Defense: Infectious Diseases and
Counterbioterrorism. Humana Press, Totowa, NJ.
26. Brett, P. J., D. DeShazer, and D. E. Woods. 1997. Characterization of Burkholderia pseudomallei and
Burkholderia pseudomallei-like strains. Epidemiol. Infect. 118:137–148.
Burkholderia 281
27. DeShazer, D., D. M. Waag, D. L. Fritz, and D. E. Woods. 2001. Identification of a Burkholderia mallei
polysaccharide gene cluster by subtractive hybridization and demonstration that the encoded capsule is
an essential virulence determinant. Microb. Pathog. 30:253–269.
28. Fritz, D. L., P. Vogel, D. R. Brown, and D. M. Waag. 1999. The hamster model of intraperitoneal
Burkholderia mallei (glanders). Vet. Pathol. 36:276–291.
29. Russell, P., et al. 2000. Comparison of efficacy of ciprofloxacin and doxycycline against experimental
melioidosis and glanders. J. Antimicrob. Chemother. 45:813–818.
30. Ulrich, R. L., and D. DeShazer. 2004. Type III secretion: a virulence factor delivery system essential for
the pathogenicity of Burkholderia mallei. Infect. Immun. 72:1150–1154.
31. Fritz, D. L., P. Vogel, D. R. Brown, D. DeShazer, and D. M. Waag. 2000. Mouse model of sublethal and
lethal intraperitoneal glanders (Burkholderia mallei). Vet. Pathol. 37:626–636.
32. Lever, M. S., et al. 2003. Experimental aerogenic Burkholderia mallei (glanders) infection in the
BALB/c mouse. J. Med. Microbiol. 52:1109–1115.
33. Jelesijevic, T., et al. 2015. Use of the common marmoset to study Burkholderia mallei infection. PLoS
One. 10:e0124181.
34. Nelson, M., et al. 2014. Comparative experimental subcutaneous glanders and melioidosis in the com-
mon marmoset (Callithrix jacchus). Int. J. Exp. Pathol. 95:378–391.
35. Schell, M. A., L. Lipscomb, and D. DeShazer. 2008. Comparative genomics and an insect model rapidly
identify novel virulence genes of Burkholderia mallei. J. Bacteriol. 190:2306–2313.
36. Fisher, N. A., W. J. Ribot, W. Applefeld, and D. DeShazer. 2012. The Madagascar hissing cockroach as
a novel surrogate host for Burkholderia pseudomallei, B. mallei and B. thailandensis. BMC Microbiol.
12:117.
37. Rolim, D. B., et al. 2005. Melioidosis, northeastern Brazil. Emerg. Infect. Dis. 11:1458–1460.
38. Leelarasamee A. 1998. Burkholderia pseudomallei: the unbeatable foe? Southeast Asian J. Trop. Med.
Public Health. 29:410–415.
39. Currie, B. J., and S. P. Jacups. 2003. Intensity of rainfall and severity of melioidosis, Australia. Emerg.
Infect. Dis. 9:1538–1542.
40. Ko, W. C., et al. 2007. Melioidosis outbreak after typhoon, southern Taiwan. Emerg. Infect. Dis.
13:896–898.
41. Whitmore, A. 1913. An account of a glanders-like disease occurring in Rangoon. J. Hyg. 13:1–34.
42. Dance, D. A. B. 2000. Ecology of Burkholderia pseudomallei and the interactions between environ-
mental Burkholderia spp. and human-animal hosts. Acta Trop. 74:159–168.
43. Suputtamongkol, Y., et al. 1994. The epidemiology of melioidosis in urban Ratchatani, northeast
Thailand. Int. J. Epidemiol. 23:1082–1090.
44. Stanton, A. T., and W. Fletcher. 1932. Studies from the Institute for Medical Research, Federated Malay
States, No. 21: Melioidosis. John Bale, Sons & Danielsson, Ltd., London.
45. Inglis, T. J. J., S. C. Garrow, C. Adams, M. Henderson, and M. Mayo. 1998. Dry-season outbreak of
melioidosis in Western Australia. Lancet. 352:1600.
46. Zanetti, F., G. De Luca, and S. Stampi. 2000. Recovery of Burkholderia pseudomallei and B. cepacia
from drinking water. Int. J. Food. Microbiol. 59:67–72.
47. Rose, L. J., et al. 2005. Chlorine inactivation of bacterial bioterrorism agents. Appl. Environ. Microbiol.
71:566–568.
48. White, N. J. 2003. Melioidosis. Lancet. 361:1715–1722.
49. Limmathurotsakul, D., et al. 2010. Increasing incidence of human melioidosis in northeast Thailand.
Am. J. Trop. Med. Hyg. 82:1113–1117.
50. Warawa, J. M. 2010. Evaluation of surrogate animal models of melioidosis. Front Microbiol. 1:141.
51. Leakey, A. K., G. C. Ulett, and R. G. Hirst. 1998. BALB/c and C57Bl/6 mice infected with virulent
Burkholderia pseudomallei provide contrasting animal models for the acute and chronic forms of
human melioidosis. Microb. Pathog. 24:269–275.
52. Lever, M. S., M. Nelson, A. J. Stagg, R. J. Beedham, and A. J. H. Simpson. 2009. Experimental acute
respiratory Burkholderia pseudomallei in BALB/c mice. Int. J. Exp. Pathol. 90:16–25.
53. Liu, B., G. C. Koo, E. H. Yap, K. L. Chua, and Y. H. Gan. 2002. Model of differential susceptibility to
mucosal Burkholderia pseudomallei infection. Infect. Immun. 70:504–511.
54. Welkos, S. L., et al. 2015. Characterization of Burkholderia pseudomallei strains using a murine intra-
peritoneal infection model and in vitro macrophage assays. PLoS One. 10:e0124667.
282 Laboratory Models for Foodborne Infections
55. Tan, G. Y., et al. 2008. Burkholderia pseudomallei aerosol infection results in differential inflammatory
responses in BALB/c and C57Bl/6 mice. J. Med. Microbiol. 57:508–515.
56. West, T. E., N. D. Myers, H. D. Liggitt, and S. J. Skerrett. 2012. Murine pulmonary infection and inflam-
mation induced by inhalation of Burkholderia pseudomallei. Int. J. Exp. Pathol. 93:421–428.
57. Woods, D. E., A. L. Jones, and P. J. Hill. 1993. Interaction of insulin with Pseudomonas pseudomallei.
Infect. Immun. 61:4045–4050.
58. Burtnick, M. N., et al. 2011. The cluster 1 type VI secretion system is a major virulence determinant in
Burkholderia pseudomallei. Infect. Immun. 79:1512–1525.
59. Manzeniuk, I. N., E. A. Galina, V. V. Dorokhin, I. Kalachev IIa, V. N. Borzenkov, and E. A. Svetoch.
1999. Burkholderia mallei and B. pseudomallei, study of immuno- and pathogenesis of glanders and
melioidosis, heterologous vaccines. Antibiot. Khimioter. 44:21–26.
60. Nelson, M., et al. 2011. Development of an acute model of inhalational melioidosis in the common mar-
moset (Callithrix jacchus). Int. J. Exp. Pathol. 92:428–435.
61. Yeager, J. J., et al. 2012. Natural history of inhalation melioidosis in rhesus macaques (Macaca mulatta)
and African green monkeys (Chlorocebus aethiops). Infect. Immun. 80:3332–3340.
62. Shalom, G., J. G. Shaw, and M. S. Thomas. 2007. In vivo expression technology identifies a type VI
secretion system locus in Burkholderia pseudomallei that is induced upon invasion of macrophages.
Microbiology. 153:2689–2699.
63. Wand, M. E., C. M. Mueller, R. W. Titball, and S. L. Michell. 2011. Macrophage and Galleria mel-
lonella infection models reflect the virulence of natural isolates of B. pseudomallei, B. thailandensis,
and B. oklahomensis. BMC Microbiol. 11:11.
64. Muller, C. M., L. Conjero, N. Spink, M. E. Wand, G. J. Bancroft, and R. W. Titball. 2012. Role of RelA
and SpoT in Burkholderia pseudomallei virulence and immunity. Infect. Immun. 80:3247–3255.
65. Thomas, R. J., et al. 2013. Galleria mellonella as a model system to test the pharmacokinetics and effi-
cacy of antibiotics against Burkholderia pseudomallei. Int. J. Antimicrob. Agents. 41:330–336.
66. O’Quinn, A. L., E. M. Wiegand, and J. A. Jeddeloh. 2001. Burkholderia pseudomallei kills the nema-
tode Caenorhabditis elegans using an endotoxin-mediated paralysis. Cell Microbiol. 3:381–393.
67. Sifri, C. D., J. Begun, and F. M. Ausubel. 2005. The worm has turned—microbial virulence modeled in
Caenorhabditis elegans. Trends Microbiol. 13:119–127.
68. Gan, Y. H., et al. 2002. Characterization of Burkholderia pseudomallei infection and identification of
novel virulence factors using a Caenorhabditis elegans host system. Mol. Microbiol. 44:1185–1197.
69. Lee, S. H., S. K. Oui, N. M. Mahadi, M. W. Tan, and S. Nathan. 2011. Complete killing of Caenorhabditis
elegans by Burkholderia pseudomallei is dependent on prolonged direct association with the viable
pathogen. PLoS One. 6:e16707.
70. Lee, Y. H., Y. Chen, X. Ouyang, and Y. H. Gan. 2010. Identification of tomato plant as a novel host for
Burkholderia pseudomallei. BMC Microbiol. 10:28.
71. Mahenthiralingam, E., T. A. Urban, and J. B. Goldberg. 2005. The multifarious, multireplicon
Burkholderia cepacia complex. Nat. Rev. Microbiol. 3:144–156.
72. De Smet, B., et al. 2015. Burkholderia stagnalis sp. nov. and Burkholderia territorii sp. nov., two novel
Burkholderia cepacia complex species from environmental and human sources. Int. J. Syst. Evol.
Microbiol. 65:2265–2271.
73. Bottone, E. J., S. D. Douglas, A. R. Rausen, and G. T. Keusch. 1975. Association of Pseudomonas cepa-
cia with chronic granulomatous disease. J. Clin. Microbiol. 1:425–428.
74. Rosenstein, B. J., and D. E. Hall. 1980. Pneumonia and septicemia due to Pseudomonas cepacia in a
patient with cystic fibrosis. Johns Hopkins Med. J. 147:188–189.
75. Ramette, A., and J. M. Tiedje. 2007. Multiscale responses of microbial life to spatial distance and
environmental heterogeneity in a patchy ecosystem. Proc. Natl. Acad. Sci. USA 104:2761–2766.
76. Zhang, L., and G. Xie. 2007. Diversity and distribution of Burkholderia cepacia complex in the rhizo-
sphere of rice and maize. FEMS Microbiol. Lett. 266:231–235.
77. Cohen, R., L. Persky, and Y. Hadar. 2002. Biotechnological applications and potential of wood-
degrading mushrooms of the genus Pleurotus. Appl. Microbiol. Biotechnol. 58:582–594.
78. Yara, R., W. Maccheroni Jr., J. Horii, and J. L. Azevedo. 2006. A bacterium belonging to the Burkholderia
cepacia complex associated with Pleurotus ostreatus. J. Microbiol. 44:263–268.
79. Smith, D., E. Mikolajcik, and J. Lindamood. 1987. Causative organisms and chemical nature of the
Swiss cheese rind rot defect. Cult. Dairy Prod. J. 22:9–12.
Burkholderia 283
80. Blanco, D., G. Barbieri, P. Mambriani, E. Spotti, and I. Barbuti. 1994. Study of “potato defect” of raw
dry-cured ham. Ind. Conserve. 69:230–236.
81. Huang, Y., B. J. Deverall, S. C. Morris, and B. L. Wild. 1993. Biocontrol of postharvest orange diseases
by a strain of Pseudomonas cepacia under semi-commercial conditions. Postharvest Biol. Technol.
3:293–304.
82. Environmental Protection Agency. 2003. Burkholderia cepacia complex; significant new use rule
[online]. Available from http://www.epa.gov/fedrgstr/EPA-TOX/2003/June/Day-13/t15010.htm.
83. Moore, J. E., B. McIlhatton, A. Shaw, P. G. Murphy, and J. S. Elborn. 2001. Occurrence of Burkholderia
cepacia in foods and waters: clinical implications for patients with cystic fibrosis. J. Food Protect.
64:1076–1078.
84. Uraz, G., and S. Çitak. 1998. An investigation about the distribution and isolation of Pseudomonas from
raw milk samples obtained from different areas. Turk. J. Agric. Forest. 22:469–474.
85. Uraz, G., and S. Çitak. 1998. The isolation of Pseudomonas and other Gram(−) psychrotrophic bacteria
in raw milks. J. Basic Microbiol. 38:129–134.
86. Munsch-Alatossava, P., and T. Alatossava. 2006. Phenotypic characterization of raw milk-associated
psychrotrophic bacteria. Microbiol. Res. 161:334–346.
87. Munsch-Alatossava, P., and T. Alatossava. 2007. Antibiotic resistance of raw-milk-associated psychro-
trophic bacteria. Microbiol. Res. 162:115–123.
88. Moore, J. E., B. C. Millar, T. Buckley, M. Crowe, and J. S. Elborn. 2002. Effect of high-temperature
short-time (HTST) laboratory pasteurization on the survival of Burkholderia cepacia complex organ-
isms in whole, low fat and skimmed milks. J. Dairy Res. 69:483–490.
89. Pegues, D. A., et al. 1994. Acquisition of Pseudomonas cepacia at summer camps for patients with
cystic fibrosis. J. Pediatr. 124:694–702.
90. Vermis, K., M. Brachkova, P. Vandamme, and H. Nelis. 2003. Isolation of Burkholderia cepacia com-
plex genomovars from waters. Syst. Appl. Microbiol. 26:595–600.
91. Koenig, D. W., and D. L. Pierson. 1997. Microbiology of the space shuttle water system. Water Sci.
Technol. 35:59–64.
92. Fisher, M. C., et al. 1993. Source of Pseudomonas cepacia: ribotyping of isolates from patients and from
the environment. J. Pediatr. 123:745–747.
93. Chu, K. K., D. J. Davidson, T. K. Halsey, J. W. Chung, and D. P. Speert. 2002. Differential persistence
among genomovars of the Burkholderia cepacia complex in a murine model of pulmonary infection.
Infect. Immun. 70:2715–2720.
94. Chu, K. K., K. L MacDonald, D. J. Davidson, and D. P. Speert. 2004. Persistence of Burkholderia mul-
tivorans within the pulmonary macrophage in the murine lung. Infect. Immun. 72:6142–6147.
95. Sokol, P. A., U. Sajjan, M. B. Visser, S. Gingues, J. Forstner, and C. Kooi. 2003. The CepIR quorum-
sensing system contributes to the virulence of Burkholderia cenocepacia respiratory infections.
Microbiology. 149:3649–3658.
96. Semler, D. D., A. D. Goudie, W. H. Finlay, and J. J. Dennis. 2014. Aerosol phage therapy efficacy in
Burkholderia cepacia complex respiratory infections. Antimicrob. Agents Chemother. 58:4005–4013.
97. Cieri, M. V., N. Mayer-Hamblett, A. Griffith, and J. L. Burns. 2002. Correlation between an in vitro invasion
assay and a murine model of Burkholderia cepacia lung infection. Infect. Immun. 70:1081–1086.
98. O’Grady, E. P., and P. A. Sokol. 2011. Burkholderia cenocepacia differential gene expression during
host-pathogen interactions and adaptation to the host environment. Front. Cell. Infect. Microbiol. 1:15.
99. Sokol, P. A., P. Darling, D. E. Woods, E. Mahenthiralingam, and C. Kooi. 1999. Role of ornibactin
biosynthesis in the virulence of Burkholderia cepacia: characterization of pvdA, the gene encoding
l-ornithine N(5)-oxygenase. Infect. Immun. 67:4443–4455.
100. Subramoni, S., D. T. Nguyen, and P. A. Sokol. 2011. Burkholderia cenocepacia ShvR-regulated genes
that influence colony morphology, biofilm formation, and virulence. Infect. Immun. 79:2984–2997.
101. Cash, H. A., D. E. Woods, B. McCullough, W. G. Johanson, Jr., and J. A. Bass. 1979. A rat model of
chronic respiratory infection with Pseudomonas aeruginosa. Am. Rev. Respir. Dis. 119:453–459.
102. Uehlinger, S., et al. 2009. Identification of specific and universal virulence factors in Burkholderia
cenocepacia strains by using multiple infection hosts. Infect. Immun. 77:4102–4110.
103. Bernier, S. P., L. Silo-Suh, D. E. Woods, D. E. Ohman, and P. A. Sokol. 2003. Comparative analysis
of plant and animal models for characterization of Burkholderia cepacia virulence. Infect. Immun.
71:5306–5313.
284 Laboratory Models for Foodborne Infections
104. Hunt, T. A., C. Kooi, P. A. Sokol, and M. A. Valvano. 2004. Identification of Burkholderia cenocepacia
genes required for bacterial survival in vivo. Infect. Immun. 72:4010–4022.
105. Visser, M. B., S. Majumdar, E. Hani, and P. A. Sokol. 2004. Importance of the ornibactin and pyo-
chelin siderophore transport systems in Burkholderia cenocepacia lung infections. Infect. Immun.
72:2850–2857.
106. Hoffmann, J. A., F. C. Kafatos, C. A. Janeway, and R. A. Ezekowitz. 1999. Phylogenetic perspectives in
innate immunity. Science. 284:1313–1318.
107. Mahajan-Miklos, S., L. G. Rahme, and F. M. Ausubel. 2000. Elucidating the molecular mechanisms of
bacterial virulence using non-mammalian hosts. Mol. Microbiol. 37:981–988.
108. Gonzalez, C. F., and A. K. Vidaver. 1979. Bacteriocin, plasmid and pectolytic diversity in Pseudomonas
cepacia of clinical and plant origin. J. Gen. Microbiol. 110:161–170.
109. Yohalem, D. S., and J. W. Lorbeer. 1994. Intraspecific metabolic diversity among strains of Burkholderia
cepacia isolated from decayed onions, soils, and the clinical environment. Antonie Van Leeuwenhoek.
65:111–131.
110. Wigley, P., and N. F. Burton. 1999. Genotypic and phenotypic relationships in Burkholderia cepacia
isolated from cystic fibrosis patients and the environment. J. Appl. Microbiol. 86:460–468.
111. Engledow, A. S., E. G. Medrano, E. Mahenthiralingam, J. J. LiPuma, and C. F. Gonzalez. 2004.
Involvement of a plasmid-encoded type IV secretion system in the plant tissue watersoaking phenotype
of Burkholderia cenocepacia. J. Bacteriol. 186:6015–6024.
112. Aguilar, C., I. Bertani, and V. Venturi. 2003. Quorum-sensing system and stationary-phase sigma fac-
tor (rpoS) of the onion pathogen Burkholderia cepacia genomovar I type strain, ATCC 25416. Appl.
Environ. Microbiol. 69:1739–1747.
113. Thomson, E. L., and J. J. Dennis. 2013. Common duckweed (Lemna minor) is a versatile high-
throughput infection model for the Burkholderia cepacia complex and other pathogenic bacteria. PLoS
One. 8:e80102.
114. Seed, K. D., and J. J. Dennis. 2008. Development of Galleria mellonella as an alternative infection
model for the Burkholderia cepacia complex. Infect. Immun. 76:1267–1275.
115. Castonguay-Vanier, J., L. Vial, J. Tremblay, and E. Déziel. 2010. Drosophila melanogaster as a model
host for the Burkholderia cepacia complex. PLoS One. 5:e11467.
116. Thomson, E. L., and J. J. Dennis. 2012. A Burkholderia cepacia complex non-ribosomal peptide-
synthesized toxin is hemolytic and required for full virulence. Virulence. 3:286–298.
117. Ferreira, A. S., et al. 2013. Comparative transcriptomic analysis of the Burkholderia cepacia tyrosine
kinase bceF mutant reveals a role in tolerance to stress, biofilm formation, and virulence. Appl. Environ.
Microbiol. 79:3009–3020.
118. Silva, I. N., et al. 2011. Mucoid morphotype variation of Burkholderia multivorans during chronic cystic
fibrosis lung infection is correlated with changes in metabolism, motility, biofilm formation and viru-
lence. Microbiology. 157:3124–3137.
119. Silva, I. N., A. C. Tavares, A. S. Ferreira, and L. M. Moreira. 2013. Stress conditions triggering mucoid
morphotype variation in Burkholderia species and effect on virulence in Galleria mellonella and bio-
film formation in vitro. PLoS One. 8:e82522.
120. Seed, K. D., and J. J. Dennis. 2009. Experimental bacteriophage therapy increases survival of Galleria
mellonella larvae infected with clinically relevant strains of the Burkholderia cepacia complex.
Antimicrob. Agents Chemother. 53:2205–2208.
121. Köthe, M., et al. 2003. Killing of Caenorhabditis elegans by Burkholderia cepacia is controlled by the
cep quorum-sensing system. Cell Microbiol. 5:343–351.
122. Huber, B., et al. 2004. Identification of a novel virulence factor in Burkholderia cenocepacia H111
required for efficient slow killing of Caenorhabditis elegans. Infect. Immun. 72:7220–7230.
123. Laws, T. R., S. V. Harding, M. P. Smith, T. P. Atkins, and R. W. Titball. 2004. Age influences resistance
of Caenorhabditis elegans to killing by pathogenic bacteria. FEMS Microbiol. Lett. 234:281–287.
124. Cooper, V. S., W. A. Carlson, and J. J. Lipuma. 2009. Susceptibility of Caenorhabditis elegans to
Burkholderia infection depends on prior diet and secreted bacterial attractants. PLoS One. 4:e7961.
125. Ellis, C. N., and V. S. Cooper. 2010. Experimental adaptation of Burkholderia cenocepacia to onion
medium reduces host range. Appl. Environ. Microbiol. 76:2387–2396.
126. Cardona, S. T., J. Wopperer, L. Eberl, and M. A. Valvano. 2005. Diverse pathogenicity of Burkholderia
cepacia complex strains in the Caenorhabditis elegans host model. FEMS Microbiol. Lett. 250:97–104.
Burkholderia 285
127. Springman, A. C., et al. 2009. Genetic diversity and multihost pathogenicity of clinical and environmen-
tal strains of Burkholderia cenocepacia. Appl. Environ. Microbiol. 75:5250–5260.
128. Reddy, K. C., R. C. Hunter, N. Bhatla, D. K. Newman, and D. H. Kim. 2011. Caenorhabditis elegans
NPR-1-mediated behaviors are suppressed in the presence of mucoid bacteria. Proc. Natl. Acad. Sci.
USA. 108:12887–12892.
129. Markey, K. M., K. J. Glendinning, J. A. Morgan, C. A. Hart, and C. Winstanley. 2006. Caenorhabditis
elegans killing assay as an infection model to study the role of type III secretion in Burkholderia ceno-
cepacia. J. Med. Microbiol. 55:967–969.
130. Law, R. J., J. N. Hamlin, A. Sivro, S. J. McCorrister, G. A. Cardama, and S. T. Cardona. 2008. A func-
tional phenylacetic acid catabolic pathway is required for full pathogenicity of Burkholderia cenocepa-
cia in the Caenorhabditis elegans host model. J. Bacteriol. 190:7209–7218.
131. Sousa, S. A., C. G. Ramos, L. M. Moreira, and J. H. Leitão. 2010. The hfq gene is required for stress
resistance and full virulence of Burkholderia cepacia to the nematode Caenorhabditis elegans.
Microbiology. 156:896–908.
132. Ramos, C. G., S. A. Sousa, A. M. Grilo, J. R. Feliciano, and J. H. Leitão. 2011. The second RNA chap-
erone, Hfq2, is also required for survival under stress and full virulence of Burkholderia cenocepacia
J2315. J. Bacteriol. 193:1515–1526.
133. Ramos, C. G., S. A. Sousa, A. M. Grilo, L. Eberl, and J. H. Leitão. 2010. The Burkholderia cenocepa-
cia K56–2 pleiotropic regulator Pbr, is required for stress resistance and virulence. Microb. Pathog.
48:168–177.
134. Ramos, C. G., A. M. Grilo, P. J. da Costa, J. R. Feliciano, and J. H. Leitão. 2013. MtvR is a global small
noncoding regulatory RNA in Burkholderia cenocepacia. J. Bacteriol. 195:3514–3523.
135. Somvanshi, V. S., P. Viswanathan, J. L. Jacobs, M. H. Mulks, G. W. Sundin, and T. A. Ciche. 2010. The
type 2 secretion pseudopilin, gspJ, is required for multihost pathogenicity of Burkholderia cenocepacia
AU1054. Infect. Immun. 78:4110–4121.
136. Agnoli, K., et al. 2012. Exposing the third chromosome of Burkholderia cepacia complex strains as a
virulence plasmid. Mol. Microbiol. 83:362–378.
137. Agnoli, K., et al. 2014. The third replicon of members of the Burkholderia cepacia complex, plasmid
pC3, plays a role in stress tolerance. Appl. Environ. Microbiol. 80:1340–1348.
138. Vergunst, A. C., A. H. Meijer, S. A. Renshaw, and D. O’Callaghan. 2010. Burkholderia cenocepacia
creates an intramacrophage replication niche in zebrafish embryos, followed by bacterial dissemination
and establishment of systemic infection. Infect. Immun. 78:1495–1508.
139. Ko, S. D. 1985. Growth and toxin production of Pseudomonas cocovenenans, the so-called “bongkrek
bacteria”. ASEAN Food J. 1:78–84.
140. Zhao, N., C. Qu, E. Wang, and W. Chen. 1995. Phylogenetic evidence for the transfer of Pseudomonas
cocovenenans (van Damme et al. 1960) to the genus Burkholderia as Burkholderia cocovenenans (van
Damme et al. 1960) comb. nov. Int. J. Syst. Bacteriol. 45:600–603.
141. van Veen, A. G., and W. K. Mertens. 1933. On the isolation of a toxic bacterial pigment. Proc. Acad. Sci.
Amsterdam. 36:666–670.
142. Nugteren, D. H., and W. Berends. 1957. Investigations on bongkrekic acid, the toxine from Pseudomonas
cocovenenans. Rec. Trav. Chim. Pays-Bas. 76:13–27.
143. van Damme, P. A., A. G. Johannes, H. C. Cox, and W. Berends. 1960. On toxoflavin, the yellow poison
of Pseudomonas cocovenenans. Rec. Trav. Chim. Pays-Bas. 79:255–267.
144. Cox, J., E. Kartadarma, and K. Buckle. 2000. Burkholderia cocovenenans, pp. 1871–1875. In R. K.
Robinson, C. A. Batt, and P. D. Patel (eds.), Encyclopedia of Food Microbiology. Academic Press, San
Diego.
145. Jin, J. X. 1963. A preliminary study on the pathogenesis of fermented corn meal food poisoning. Prev.
Hyg. 41:21–26. Cited in Zhao, N. X., M. S. Ma, Y. P. Zhang, and D. C. Xu. 1990. Comparative descrip-
tion of Pseudomonas cocovenenans (van Damme, Johannes, Cox, and Berends 1960) NCIB 9450T and
strains isolated from cases of food poisoning caused by consumption of fermented corn flour in China.
Int. J. Syst. Bacteriol. 40:452–455.
146. Meng, Z., et al. 1988. Studies on fermented corn flour food poisoning in rural areas of China. II. Isolation
and identification of causal microorganisms. Biomed. Environ. Sci. 1:105–114.
147. Zhao, N. X., M. S. Ma, N. F. Miao, and Y. P. Zhang. 1988. Comparative study on Pseudomonas coco-
venenans and Pseudomonas farinofermentans. Chin. J. Microbiol. Immunol. 8:151–156.
286 Laboratory Models for Foodborne Infections
148. Zhao, N. X., M. S. Ma, Y. P. Zhang, and D. C. Xu. 1990. Comparative description of Pseudomonas
cocovenenans (van Damme, Johannes, Cox, and Berends 1960) NCIB 9450T and strains isolated from
cases of food poisoning caused by consumption of fermented corn flour in China. Int. J. Syst. Bacteriol.
40:452–455.
149. Yabuuchi, E., et al. 1992. Proposal of Burkholderia gen. nov. and transfer of seven species of the
genus Pseudomonas homology group II to the new genus, with the type species Burkholderia cepacia
(Palleroni and Holmes 1981) comb. nov. Microbiol. Immunol. 36:1251–1275.
150. Gillis, M., et al. 1995. Polyphasic taxonomy in the genus Burkholderia leading to an emended descrip-
tion of the genus and proposition of Burkholderia vietnamiensis sp. nov. for N2-fixing isolates from rice
in Vietnam. Int. J. Syst. Bacteriol. 45:274–289.
151. Vandamme, P., et al. 1997. Occurrence of multiple genomovars of Burkholderia cepacia in cystic
fibrosis patients and proposal of Burkholderia multivorans sp. nov. Int. J. Syst. Evol. Microbiol.
47:1188–1200.
152. Coenye, T., B. Holmes, K. Kersters, J. R. W. Govan, and P. Vandamme. 1999. Burkholderia cocoven-
enans (van Damme et al. 1960) Gillis et al. 1995 and Burkholderia vandii Urakami et al. 1994 are junior
synonyms of Burkholderia gladioli (Severini 1913) Yabuuchi et al. 1993 and Burkholderia plantarii
(Azegami et al. 1987) Urakami et al. 1994, respectively. Int. J. Syst. Bacteriol. 49:37–42.
153. Christenson, J. C., D. F. Welch, G. Mukwaya, M. J. Muszynski, R. E. Weaver, and D. J. Brenner. 1989.
Recovery of Pseudomonas gladioli from respiratory tract specimens of patients with cystic fibrosis.
J. Clin. Microbiol. 27:270–273.
154. Kennedy, M. P., et al. 2007. Burkholderia gladioli: five year experience in a cystic fibrosis and lung
transplantation center. J. Cyst. Fibros. 6:267–273.
155. Segonds, C., T. Heulin, N. Marty, and G. Chabanon. 1999. Differentiation of Burkholderia species by
PCR-restriction fragment length polymorphism analysis of the 16S rRNA gene and application to cystic
fibrosis isolates. J. Clin. Microbiol. 37:2201–2208.
156. Khan, S. U., S. M. Gordon, P. C. Stillwell, T. J. Kirby, and A. C. Arroliga. 1996. Empyema and blood-
stream infection caused by Burkholderia gladioli in a patient with cystic fibrosis after lung transplanta-
tion, Pediatr. Infect. Dis. J. 15:637.
157. Shin, J. H., S. H. Kim, M. G. Shin, S. P. Suh, D. W. Ryang, and M. H. Jeong. 1997. Bacteremia due to
Burkholderia gladioli: case report, Clin. Infect. Dis. 25:1264.
158. Machlowitz, R. A., W. P. Fisher, B. S. McKay, A. A. Tytell, and J. Charney. 1954. Xanthothricin, a new
antibiotic. Antibiot. Chemother. 4:259–261.
159. Sato, Z., Y. Koiso, S. Iwasaki, I. Matsuda, and A. Shirata. 1989. Toxins produced by Pseudomonas
glumae. Ann. Phytopathol. Soc. Jpn. 55:353–356. Cited in Maeda, Y., et al. 2006. Phylogenetic study
and multiplex PCR-based detection of Burkholderia plantarii, Burkholderia glumae and Burkholderia
gladioli using gyrB and rpoD sequences. Int. J. Syst. Evol. Microbiol. 56:1031–1038.
160. Jeong, Y., J. Kim, S. Kim, Y. Kang, T. Nagamatsu, and I. Hwang. 2003. Toxoflavin produced by
Burkholderia glumae causing rice grain rot is responsible for inducing bacterial wilt in many field crops.
Plant Dis. 87:890–895.
161. Lijmbach, G. W. M., H. C. Cox, and W. Berends. 1970. Elucidation of the chemical structure of bong-
krekic acid. I. Isolation, purification and properties of bongkrekic acid. Tetrahedron. 26:5993–5999.
162. Buckle, K. A., and E. Kartadarma. 1990. Inhibition of bongkrek acid and toxoflavin production in tempe
bongkrek containing Pseudomonas cocovenenans. J. Appl. Bacteriol. 68:571–576.
163. Moebius, N., C. Ross, K. Scherlach, B. Rohm, M. Roth, and C. Hertweck. 2012. Biosynthesis of the
respiratory toxin bongkrekic acid in the pathogenic bacterium Burkholderia gladioli. Chem. Biol.
19:1164–1174.
164. Meng, Z., et al. 1988. Studies on fermented corn flour poisoning in rural areas of China. I. Epidemiology,
clinical manifestations, and pathology. Biomed. Environ. Sci. 1:101–104.
165. van Veen, A. G. 1966. The bongkrek toxins, pp. 43–50. In R. I. Mateles, and G. N. Wogan (eds.),
Biochemistry of Some Foodborne Microbial Toxins. MIT Press, Cambridge, UK.
166. Hu, W. J., G. S. Zhang, and F. S. Chu. 1984. Purification and partial characterization of flavotoxin A.
Appl. Environ. Microbiol. 48:690–693.
Burkholderia 287
167. Hu, W. J., X. M. Chen, H. D. Meng, and Z. H. Meng. 1989. Fermented corn flour poisoning in rural areas
of China. III. Isolation and identification of main toxin produced by causal microorganisms. Biomed.
Environ. Sci. 2:65–71.
168. Henderson, P. J., and H. A. Lardy. 1970. Bongkrekic acid: an inhibitor of the adenine nucleotide trans-
locase of mitochondria. J. Biol. Chem. 245:1319–1326.
169. Zamzami, N., et al. 1996. Inhibitors of permeability transition interfere with the disruption of the mito-
chondrial transmembrane potential during apoptosis. FEBS Lett. 384:53–57.
170. Schwerdt, G., R. Freudinger, C. Schuster, S. Silbernagl, and M. Gekle. 2003. Inhibition of mitochon-
dria prevents cell death in kidney epithelial cells by intra- and extracellular acidification. Kidney Int.
63:1725–1735.
171. Welling, W., J. A. Cohen, and W. Berends. 1960. Disturbance of oxidative phosphorylation by an antibi-
oticum produced by Pseudomonas cocovenenans. Biochem. Pharmacol. 3:122–135.
19
Campylobacter
CONTENTS
19.1 Introduction................................................................................................................................... 289
19.2 General Features of C. jejuni Colonization of Animal Models.................................................... 290
19.2.1 Cellular Models of Infection............................................................................................ 292
19.2.2 Three-Day-Old Chick Model........................................................................................... 292
19.2.3 Colostrum-Deprived Piglets............................................................................................. 294
19.2.4 Ferret Model..................................................................................................................... 294
19.2.5 Galleria mellonella.......................................................................................................... 295
19.2.6 Mouse Models.................................................................................................................. 295
19.3 Conclusions................................................................................................................................... 298
References............................................................................................................................................... 299
19.1 Introduction
Campylobacter jejuni is a Gram-negative, curved-rod shaped, microaerophilic, enteric bacterial patho-
gen, and one of the leading causes of gastroenteritis in both the developing and the developed world. This
highly infectious bacterium is often acquired through the consumption of contaminated water, raw milk,
or meat products (particularly contaminated poultry).1 Once infected, an individual’s symptoms can vary
significantly. Milder cases can be largely asymptomatic or involve watery diarrhea. In more severe cases,
fever, bloody diarrhea, and abdominal pain can be evident. In most cases, the disease resolves within
a few days or in up to 2 weeks; however, under rare and poorly understood circumstances, the infec-
tion can trigger the development of autoimmune disorders such as reactive arthritis or Guillain–Barré
syndrome,1 or other complications such as postinfective irritable bowel syndrome.2 Despite the broad
prevalence of this pathogen, the mechanisms by which this organism causes disease in humans remain
poorly understood.
Aside from assessing the severity of symptoms, the details of how C. jejuni triggers disease symp-
toms remain poorly defined. We know that a very small infectious dose in humans is capable of tran-
siting the stomach and establishing infection, largely within the colon. Pathogen burdens can be very
high, although they drop significantly as the adaptive immune response becomes engaged. The infection
usually responds to antibiotic treatment, such as either fluoroquinolones (e.g., ciprofloxacin) or macro-
lides (e.g., erythromycin), although antibiotic resistance rates in C. jejuni have been steadily increasing
in recent years.3 The nature of intestinal colonization by C. jejuni in humans is poorly characterized,
although numerous in vitro studies, as well as in vivo animal work, would suggest it is primarily local-
ized to intestinal crypts as well as to the mucus layer that overlies the mucosa. Moreover it is capable
of adhering to the intestinal epithelial surface and translocating across the epithelium.1 Cell invasion by
C. jejuni has been indicated in vitro, in vivo, and in clinical samples1,4; however, what role cell invasion
plays in C. jejuni pathogenesis remains unknown. The spread of C. jejuni into the blood stream and to
systemic sites has been documented in some patients, but it remains relatively atypical.5 Additionally,
long-term complications such as reactive arthritis and Guillain–Barré syndrome are rare and result from
289
290 Laboratory Models for Foodborne Infections
antigen mimicry between C. jejuni lipooligosaccharides and host gangliosides. To date, no proven ani-
mal models exist that mimic these long-term complications.
One of the difficulties in improving our understanding of campylobacteriosis has been the fact that
C. jejuni behaves quite differently depending on its surroundings. In the wider environment, birds are the
main reservoirs for the organism and its preferred hosts1,6; however, C. jejuni remains a relatively harm-
less commensal in avian hosts, even as it is capable of growing to high numbers in birds’ cecum.6 Various
Campylobacter species, including those that are infectious to humans such as C. jejuni, are commonly
recovered from wild birds,7,8 although the main reservoirs that serve as a source for human infection
are poultry farms.9 In the densely packed environment of a modern poultry barn, any Campylobacter
contamination can quickly spread between birds until the entire flock becomes colonized, making
Campylobacter contamination a significant health concern within the poultry industry.10 Precisely how
C. jejuni is able to spread so rapidly and so easily has been somewhat of a mystery, especially consider-
ing its rather fastidious growth requirements, including warm, low-oxygen conditions. Evidence suggests
that flies11 or wild birds6 may serve as sources of contamination, while other sources include runoff from
other livestock such as pigs or cattle,10,12 which themselves can become asymptomatically colonized.12
Coprophagous behavior of chickens, along with a high density of birds can quickly spread contamina-
tion throughout a flock.13 It has also been suggested that amoeba may serve as a temporary host for
Campylobacter, allowing for its prolonged survival within contaminated water sources.14,15 Additional
survival mechanisms within the C. jejuni cells themselves, such as their observed ability to convert to a
nonculturable coccoid form, may also aid in prolonging their survival in the low-nutrient, high-oxygen
conditions commonly found in the environment outside of a host,16 but these mechanisms remain poorly
understood.
Besides the commensal-like colonization of poultry, few other animals are susceptible to C. jejuni
colonization and infection in a fashion similar to human campylobacteriosis.17 Following exposure to
C. jejuni, most animals either exhibit only brief, transient colonization, or are colonized asymptomati-
cally. An exception to this involves a connection between spontaneous abortion in sheep and other live-
stock, and infection by certain Campylobacter species, including Campylobacter fetus and C. jejuni.18
Humans remain one of the relatively few hosts that exhibit enteric disease in response to exposure to
C. jejuni, making the use of animal models in the study of C. jejuni infection more problematic. Those
animal models that are employed usually come with the caveats that they do not entirely replicate the
human disease (e.g., poultry and insect models), or that they require some significant modification of the
host, such as the construction of mice deficient in key genes designed to make them more susceptible to
infection. Some of the more promising infection models currently being used include neonatal piglets,
ferrets, and certain knockout mouse strains, whereas models of commensal colonization are typically
newly hatched chicks and wild-type mice. Table 19.1 summarizes the current animal models available,
along with a brief description of each. In this chapter, we will outline the animal models currently
employed for the study of C. jejuni in published research, and discuss the advantages and drawbacks of
each one, while providing an overview as to what the research community has learned about C. jejuni
infection from these research models.
TABLE 19.1
Animal Models for the Study of Campylobacter jejuni Infections
Animal Model Colonization Pathology References
Chick Strong commensal-like Little to no pathology 21,35,49,50–56,60
colonization of the cecum and
colon, with a very small
inoculum
Piglet Colonization of the small Significant variation in pathology. 37,42,64,6–71
intestine, cecum, and colon Inflammation, distension with gas, and
immune cell infiltration
Ferret Colonization principally in the Significant inflammation and immune cell 22,72,75–78
colon infiltration in colonized areas
Galleria No real colonization, although The mortality and melanization of infected 80,81
C. jejuni may replicate and larvae is an indicator of pathology
spread following injection
Wild-type mouse Inconsistent colonization Minimal and inconsistent pathology 84–91
Antibiotic-pretreated Consistent colonization of the Minimal pathology in wild-type mice 26,86,88
or germfree mouse cecum and colon
IL-10−/− mouse Consistent colonization of the Chronic infection, severe inflammation, and 99,102–107
cecum and colon immune cell infiltration in the colon and
cecum
SIGIRR−/− mouse Consistent colonization of the Acute infection, moderate inflammation, 26
cecum and colon and immune cell infiltration
the microbes reaching very high numbers (often over 109) of colony forming units (CFUs) per gram of
luminal content.21 In animals without a developed cecum (ferrets) and in humans, the primary site of
colonization becomes the colon.22,23
Focusing more closely, current research suggests that C. jejuni’s preferred location within the intes-
tine is the mucus layer,24,25 which segregates the host epithelium from the bacteria and other contents
of the lumen. Histological images of a number of C. jejuni animal models, including both chicks21
and mice,26 have identified C. jejuni as being localized primarily within the mucus layer, particularly
within the mucus-filled crypts that line the intestine. Mechanistically, this observation fits with several
known aspects of C. jejuni colonization and cell biology. Firstly, C. jejuni is microaerophilic, meaning
it requires low levels of oxygen to grow. This reflects it having only a single class I-type ribonucleotide
reductase, which requires oxygen to function.27 However, C. jejuni is also dependent on numerous
metabolic enzymes which contain oxygen-sensitive iron–sulfur complexes.28 Together, these make it
unable to survive for extended periods under anaerobic conditions (such as the intestinal lumen), or
under higher oxygen conditions. This makes the low oxygen conditions of the intestinal lining its ideal
environment as it is distant from the anaerobic lumen, where the commensal anaerobes thrive, yet close
enough to be able to access the diffuse oxygen from the highly vascularized epithelium. Furthermore,
early research on C. jejuni chemotaxis indicated that it was attracted to mucins,29 which are the primary
glycoprotein component of intestinal mucus. Additionally, at least some strains are chemotactic toward
l-fucose,29 a common terminal glycan on mucins. The current hypothesis is that C. jejuni recognizes
mucin 2 as a chemoattractant to find its way to the mucus layer, using its flagellar motility and cork-
screw shape to push its way through intestinal contents to reach its destination.30,31 In all animal models
where the intestine is colonized, mutants in chemotaxis or flagella are either completely or severely
impaired for colonization, making motility toward the mucus layer one of the most critical factors for
colonization.32,33
The second critical factor that governs C. jejuni colonization, in almost any host it encounters, is how it
deals with the common stresses it almost universally encounters. For example, in a mammalian or avian
digestive system, orally inoculated C. jejuni would be exposed to the low pH of the stomach, antimi-
crobial effects of bile salts, oxidative stress, nitrosative stress, competition with the host and microbiota
292 Laboratory Models for Foodborne Infections
for resources, not to mention the innate immune defenses of the host organism. While many of these
stressors may depend on the specific host and situation in which C. jejuni finds itself, and thus require
more of an explanation than can be addressed here, a few common mechanisms appear to be present in
most potential animal hosts that C. jejuni can colonize. For example, mutants in the CmeABC multidrug
efflux pump, as well as several related pumps and regulators are more sensitive to a number of factors,
including bile salts, heavy metals, antimicrobials, and antibiotics, and are significantly impaired for
survival inside a living host.34 Oxidative stress is also a major factor at several stages of colonization,
and deletion mutants in key oxidative-stress-related genes including katA, cj1386, sodB, ahpC, as well as
strains lacking other defense and regulatory genes are severely impaired in gut colonization in a number
of C. jejuni animal models.35,36
Finally, the acquisition of nutrients is a common feature of C. jejuni’s survival in almost any environ-
ment. A curious feature of C. jejuni is its relatively limited repertoire of potential carbon sources. With
the exception of certain strains that have acquired an l-fucose metabolic pathway,37 C. jejuni is limited
to only a handful of amino acids, 4C-dicarboxylates, and a few molecules that can be easily plugged into
the citric acid cycle.38,39 This makes mutants in almost any of these key metabolic pathways exceptionally
poor colonizers, due to a lack of alternate metabolic pathways to draw upon.40,41 In particular, mutants
impaired in the acquisition or metabolism of two of its preferred amino acids, glutamate and serine,
are exceptionally poor colonizers.40,41 Furthermore, the acquisition of a number of metals and other key
micronutrients, especially iron, is critical for C. jejuni, particularly in a competitive environment such as
an animal host. In the case of iron, which is tightly regulated by the host and required by the microbiota,
the loss of any of C. jejuni’s major iron acquisition pathways or their regulators can result in a substantial
decrease in its colonization potential.42,43
CFUs per gram of luminal contents remains relatively constant over this period in the absence of any
external manipulation.21 Any limitations on the duration of an experiment are dependent on the facility
and the growth of the birds. While infected chicks are quite small at the beginning of the experiment,
they will grow and gain weight rapidly, making housing and handling more problematic over time.
While it has been demonstrated that C. jejuni will quickly establish itself within the GI tract of an
infected bird, our knowledge of exactly how C. jejuni colonizes chicks is relatively limited. The main
site of colonization is the cecum and to a lesser extent the cloaca,54 with relatively low numbers found
throughout the rest of the avian GI tract, although there has been little research performed to determine
whether other locations of the GI tract can be colonized, or if C. jejuni recovered from these regions is
just a transient population. Interestingly, several early studies were conducted to better characterize how
C. jejuni established itself within the cecum. Beery et al. compared histological sections of infected and
uninfected 8-day-old chicks.21 Their primary observation was the presence of large numbers of C. jejuni
at the base of the chick cecal crypts. A closer examination using transmission electron microscopy
clearly enabled the visualization of the helical C. jejuni cells in the mucus layer filling the crypts, but
they were not associated closely with the microvilli or glycocalyx, suggesting that they were not adher-
ing to the intestinal epithelial cells themselves. In contrast, other studies have identified cell adhesion not
only taking place in the chick cecum, but also being an important step in proper colonization.55 Whether
or not cell invasion takes place in the chicken cecum in vivo remains uncertain owing to conflicting
studies and evidence53,56; however, it has been observed to occur in chicken cells in vitro.57 This has led
to the belief that C. jejuni has the capacity to invade chick cecal epithelial cells; however, additional
factors may intervene to prevent this from occurring routinely in an in vivo situation.58 Notwithstanding
the lack of conclusive evidence, the cecal mucus layer has been implicated as a modulator of C. jejuni
pathogenicity. In cell culture experiments, the addition of chicken cecal mucus has been found to reduce
cell invasion of both chicken and human epithelial cells, although a mechanism to explain this obser-
vation remains unknown.57,58 It has been suggested that cell invasion may still take place in vivo, but
infrequently, as an explanation for observations regarding the stimulation of the avian immune response;
however, this remains only a hypothesis.56
Conflicting information also exists as to whether C. jejuni can be found outside of the chick intestinal
tract. Some studies have not recovered any C. jejuni from other sites throughout the body following
inoculation,21 however, other studies have been able to recover C. jejuni from other organs, including the
spleen and liver up to 7 days postinoculation.47 Despite the presence of C. jejuni at systemic sites, the
chick immune system remains largely unresponsive to C. jejuni, with virtually no signs of disease devel-
opment.53 The degree to which the chick immune system reacts to C. jejuni remains largely unknown,
despite the number of studies that have attempted to answer this question. It is of particular interest for
researchers attempting to develop a vaccine against C. jejuni chicken colonization, which would rely
heavily on the ability to stimulate the chicken’s own immune system to recognize and clear the coloniz-
ing C. jejuni, something the immune system does not seem inclined to do under normal circumstances.59
Despite the seeming lack of an immune response to C. jejuni, evidence would suggest that the chick
immune system is not entirely oblivious to the presence of C. jejuni. A transcriptomic study identified
270 genes differentially regulated in chicks inoculated by C. jejuni, as compared to uninfected controls,
with some of these genes involved in inflammatory pathways.60 Furthermore, studies have identified
chTLR4 and chTLR21 expressions as being particularly sensitive to C. jejuni colonization.61,62 Evidence
suggests that prior to exposure to C. jejuni, the avian innate immune system remains at least somewhat
sensitive to C. jejuni. Upon initial exposure to C. jejuni, there is a measurable spike in production of the
chicken equivalent of the human chemokine IL-8 (CXCLi1 and CXCLi2), accompanied by an increase
in the numbers of monocytes/macrophages within the cecal tissues.63 Curiously, this does not lead to an
increase in heterophil numbers (roughly the equivalent of a human neutrophil), as one would expect with
IL-8. This increase in a key proinflammatory chemokine appears to be temporary, and immune toler-
ance to C. jejuni appears to develop,53 allowing for a high pathogen burden, but virtually no immune/
inflammatory response.
In spite of the lack of immunological responses that resemble human disease, the chick has been
widely used as a model for C. jejuni colonization. This remains quite relevant due to similarities in basic
colonization factors, even in the absence of an immune response, as well as the primary role of poultry
294 Laboratory Models for Foodborne Infections
as a common source of human infection, making the colonization and spread of C. jejuni in chickens
relevant in its own right.
19.2.3 Colostrum-Deprived Piglets
The use of colostrum-deprived piglets as a model for C. jejuni infection was first defined by Babakhani
et al.64 Although conventionally reared piglets may be susceptible to Campylobacter infection early
in life, maternal antibodies are usually sufficient to prevent infection. Adult pigs are also known to be
frequent carriers of both C. jejuni and Campylobacter coli65; however, the development of symptoms
of gastroenteritis in response to these pathogens is rare. Colostrum-deprived piglets lack the protection
of maternal antibodies that usually helps prevent infection in piglets. Additionally, at very young ages,
they lack a fully mature immune response or a protective, fully developed intestinal microbiota, making
them particularly susceptible to infections by enteric pathogens. This vulnerability has been previously
exploited for the study of Escherichia coli66 and Salmonella67 infection, among others, and in a number
of studies, it has been successfully used as a model for C. jejuni infection.37,42,68
In the initial characterization of the model by Babakhani et al., the M129 strain of C. jejuni was orally
inoculated into newborn, colostrum-deprived piglets.64 Symptomology was somewhat varied, but all the
piglets became colonized, and exhibited symptoms including diarrhea, with blood and mucus in their
stool. These symptoms were often evident within a day of inoculation, and pathogen burden and disease
severity peaked within 3 days postinoculation. This first study is also the only study to undertake a
histological analysis of infected piglets. In their analyses, they found signs of inflammation, goblet cell
depletion, and immune cell infiltration primarily in the cecum and colon, with minimal signs of damage
in the small intestine. Electron microscopy also successfully identified C. jejuni cells adherent to the
epithelial surface as well as bacteria internalized within epithelial cells.
Continuing studies using the colostrum-deprived model have focused primarily on pathogen burdens,
with less attention paid to histology or immune responses within the host piglets. The most common
application has been to compare changes in pathogen burden between wild-type C. jejuni and isogenic
mutants lacking key pathogenicity genes. These have included genes related to nutrient uptake,37,42
stress-response regulators,68,69 capsule-modifying enzymes,70 as well as other genes of interest.71 In most
cases, the strain of choice has been the commonly used NCTC11168 lab strain, which induces limited
pathology and mortality in infected piglets.42 Impaired pathogenesis in a mutant strain is usually con-
cluded based on a decrease in pathogen burdens either by comparison to control piglets infected with
the wild-type strain, or through a competitive index from a piglet infected with both the mutant and
wild-type strains. In contrast to the NCTC11168 strain, the 81-176 strain is much more pathogenic in the
piglet model,70 and is less frequently used. On the occasions it has been studied, piglets often suffer high
mortality within 1–2 days postinoculation.
19.2.4 Ferret Model
Although they are a relatively uncommon and difficult animal species to use as a laboratory animal
infection model, domestic ferrets have been used in several studies as a model for human C. jejuni
infection.22,72 The first use of these animals as a model for C. jejuni infection was published by Fox
et al.22 Prior to this, Campylobacter-like organisms (likely Helicobacter), had been recovered from fer-
ret gastric lesions,73 leading to their common use as an animal model in the early study of Helicobacter
infection.74 Campylobacter infection was further described using domesticated ferrets, and has subse-
quently been used in several studies to assess the pathogenicity of isogenic C. jejuni mutant strains.
Recently, a more detailed evaluation of the immune responses and histopathology of C. jejuni infection
in ferrets has been published.75
Despite the difficulty in using ferrets as an animal model of infection, including cost, availability, and
a relative lack of experimental tools and expertise relating to ferrets, the ferret model is attractive due
to the ability of C. jejuni to trigger an acute, self-limiting diarrheal disease without substantial manipu-
lations of the host to render them more susceptible to infection.22 The exception to this is the need to
anaesthetize the animals prior to inoculation, and in some studies treatment with sodium bicarbonate
Campylobacter 295
was used prior to inoculation to reduce stomach acidity, and thereby increase the survival of C. jejuni
through the stomach.72,75–78 Additionally, some studies used a postinoculation treatment with opium to
reduce intestinal peristalsis to give more time for the infection to take hold.
As an infection model, the ferret remains somewhat understudied due to the relatively few researchers
who have actually employed it. The initial characterization of the model described an acute diarrheal
disease, with mild to moderate diarrhea observed within the first few days postinoculation.72 Symptoms
resolved within a few days, and the infection was cleared within 1–2 weeks. A more detailed descrip-
tion of the ferret immune response to C. jejuni was published in 2009.75 Researchers found a relatively
high pathogen burden (107–1010 CFUs/g) and more pronounced pathology in the colon relative to the
small intestine. The pathology observed included substantial increases in immune cell infiltration into
the mucosa, and the detection of lactoferrin and blood in the stool. Elevated fecal and systemic IgG
levels were detected and the authors linked their presence to the successful clearance of the infection.
Furthermore, immunohistochemistry and electron microscopy detected C. jejuni adherent to the surface
of epithelial cells, with possible evidence of epithelial cell invasion.
19.2.6 Mouse Models
For researchers using animals to model human infections, mice have long been the species of choice.
Mice have the advantage of being relatively small, thereby allowing one to house large numbers in a rela-
tively small space, and thus keeping maintenance costs manageable. Mice breed and mature relatively
quickly, allowing for the establishment of whole colonies within a matter of months, and for mice to be
generated for new experiments relatively quickly. Finally, in many respects, mice are physiologically
similar to humans, making them fairly relevant for studying human biology and diseases.
With these advantages in their favor, a variety of research tools have sprung up surrounding mouse
research, which are unmatched in any other animal model. Commercial companies have been established
to breed mice, providing them on order, thereby eliminating the need for all researchers to maintain their
296 Laboratory Models for Foodborne Infections
own mouse colonies. Genetic tools have allowed for the creation of mouse strains lacking almost any
gene that can be feasibly knocked out. Finally, commercial reagents and assays have been developed
specifically for use with mouse models. Many antibodies and diagnostic tools are tailored to mouse biol-
ogy, allowing for accurate tests and experiments. Although many of the other animal models have some
of these tools available for them, none comes close to matching what is available for mice.
With mice already firmly established as research tools, very early experiments on Campylobacter
sought to apply them to existing mouse infection models. As early as the 1970s, when Campylobacter
was just starting to be identified as a distinct genus of its own, some initial characterizations of
Campylobacter spp. colonization in mice were performed.84 Results from these experiments were often
limited and conflicting due largely to a lack of standard tools and techniques at the time. Field et al.
reported susceptibility in neonatal mice to intragastric Campylobacter infection;85 however, adult mice
were found to be resistant to colonization and infection, unless pretreated with antibiotics.86 Conversely,
using adult HA-ICR mice and a high inoculating dose, Blaser et al. successfully colonized the mice and
observed some degree of colonic inflammation and anti-Campylobacter IgG production.87 As further
studies were performed, results remained mixed using mice as models for C. jejuni infection. Germfree
mice, mice with limited flora, or antibiotic pretreated mice were found to be highly susceptible to colo-
nization.26,86,88 Untreated, conventional mice carrying a full microbiota were found to be colonized in
some studies, but in others, colonization either failed or was sporadic, making reproducibility problem-
atic.89 Symptomology was equally inconsistent between studies. Some studies reported the development
of mild to severe diarrhea, and even mortality, in infected mouse strains90; however, in other studies,
colonization was largely asymptomatic with no significant signs of diarrhea or overt pathology.86,88 These
significant inconsistencies between studies dampened the enthusiasm for the use of mice as a model for
C. jejuni infection, and by the end of the 1980s, conventional, wild-type mice were used only infrequently
as an infection model for C. jejuni. Some researchers with better success at colonization continued to
use mice as a simple colonization model,91 but, the success of chicks as an animal model for colonization
made them more attractive to those able to procure them. Mouse models, however, remained tempting,
once again due to the plethora of tools available, so researchers began trying more creative means of
infection, or began testing genetically modified knockout mouse strains.
With regards to the route of infection, IP injection and intranasal challenge have both been employed
in mice. IP injection is a common technique, often applied for the study of Salmonella Typhimurium,
or other pathogens that can cause systemic infections in mice. This technique with Campylobacter
has been applied on several occasions in mice and it serves the purpose of exposing C. jejuni directly
to the mouse’s immune system.92,93 Intravenous injection of C. jejuni in mice has also been tried, with
a similar effect.92,94 While the relevance of these infection routes has been questioned, they are not
entirely artificial, as C. jejuni has been found on occasion to cause systemic infection in humans.95
However, Campylobacter-associated bacteremia is rare, and not typical for human infection. Despite
this, these models can prove useful in understanding interactions between C. jejuni and the mouse
immune system.
The intranasal challenge model is perhaps more unusual in concept since C. jejuni does not normally
affect or even come into contact with the respiratory system. The inspiration for this model came from
the effective use of an intranasal challenge using Shigella, which proved effective in evaluating potential
vaccine candidates.96 In the case of C. jejuni, an intranasal challenge not only results in exposure of the
respiratory system, but also quickly leads to C. jejuni colonization at systemic sites around the body
within a few days, including persistent colonization of the intestine, mesenteric lymph nodes, liver, and
spleen.96 In the years since the first study utilized this technique, it has been repeated on several occa-
sions, including being used to assess the colonization potential of the cell-adhesion-deficient Peb1A
mutant and it was used to study cytokine production at colonized systemic sites.91,97
Some of the most promising results using mice to study Campylobacter infection have come from
the use of knockout mice, particularly those deficient in key aspects of their immune systems. An early
example of this was a study that assessed C. jejuni infection in both SCID- and RAG2-deficient mice,
both of which are deficient in cellular and humoral immunity.88,98 In these cases, pathogen burdens were
significantly enhanced compared to immunocompetent BALB/c mice; however, in neither case did the
infection trigger diarrhea or significant pathology in a reproducible fashion.
Campylobacter 297
A more successful and widely used approach utilized IL-10 deficient (IL-10−/−) mice to mimic the
intestinal inflammation caused by a C. jejuni infection, in order to assess C. jejuni virulence, rather
than the simple colonization potential.99 IL-10 is one of the key cytokines that regulates inflammation.100
Typically, once inflammation has been induced by infection or injury, the upregulation of IL-10 plays a
critical role in its subsequent resolution. High levels of IL-10 can also prevent inflammation from begin-
ning in the first place, making it an important cytokine for the control of unwanted inflammation and
the collateral damage it can cause. As expected, mice deficient in this cytokine are very susceptible to
developing intestinal inflammation, either from injury, a chemical trigger-like dextran sodium sulfate
(DSS),101 or in response to pathogenic bacteria such as C. jejuni. Several studies have used this infection
model to assess the ability of different C. jejuni isolates from human, animal, and environmental sources
to induce inflammation and pathology.99,102–104 A number of C. jejuni mutants, including those lacking
Campylobacter invasion antigen (Cia) proteins,105 showed a reduced potential to trigger inflammation
in these mice. Additionally, mice doubly deficient in IL-10 as well as one of the several innate immune
receptors such as NOD2, TLR2, and TLR4 were recently used to link signaling by these receptors to the
development of inflammation in this model.106,107
The increased susceptibility of the IL-10−/− mice has proven very useful as a means of overcoming the
seemingly high immune tolerance of mice to C. jejuni; however, it comes with a number of complica-
tions. With IL-10 being one of the linchpins for controlling and resolving inflammation, IL-10−/− mice
are often unable to recover from infection, making C. jejuni infection a chronic and ultimately terminal
infection, rather than the acute, self-limiting infection typically observed in humans. Furthermore, the
sensitivity of these mice to almost any inflammatory trigger often results in the spontaneous develop-
ment of inflammation, likely in response to their own intestinal microbiota. To limit the development of
spontaneous inflammation, IL-10−/− mice are typically kept under germfree conditions,108 adding to the
cost and difficulty of maintaining colonies.
Modifications to the innate immune system have also been investigated as a means of generating an
improved mouse infection model for C. jejuni, as well as for studying the interactions between C. jejuni
and innate immune receptors. When Watson et al. infected single and double knockouts of Nramp1 and
MyD88 with C. jejuni,109 they found that all of them were highly susceptible to C. jejuni colonization,
with these mouse strains carrying a much higher pathogen burden than wild-type mice, including higher
numbers recovered from systemic sites following oral inoculation. However, once again, neither mouse
strain developed any particular signs of inflammation in response to this higher pathogen burden.
With a MyD88 knockout being susceptible to asymptomatic colonization, we took the opposite approach,
and infected mice lacking single IgG IL-1 related receptor (SIGIRR), a protein highly expressed by the
intestinal epithelium, and known to act as a repressor of MyD88-dependent signaling.26 The resulting
mouse exhibits enhanced signaling via MyD88-dependent receptors such as TLRs and IL-1R. Previous
work infecting these mice with Salmonella Typhimurium and Citrobacter rodentium found that their
increased innate signaling impaired microbiota-dependent colonization resistance, resulting in increased
susceptibility to infection along with increased pathogen burdens and the development of more severe
intestinal inflammation in response to infection.110 Unlike IL-10−/− mice, SIGIRR−/− mice do not develop
spontaneous colitis, although they do maintain a slightly higher intestinal inflammatory “tone,” reflect-
ing increased basal levels of several key cytokines.110 Despite being prone to developing more significant
pathology in response to infection or DSS colitis, they are still capable of resolving inflammation after a
bacterial infection has been cleared. This makes them more relevant for studying the type of acute infec-
tion that is normally associated with C. jejuni.
When orally inoculated with C. jejuni, colonized SIGIRR−/− mice developed noticeable signs of
inflammation, primarily within the cecum and proximal colon.26 This corresponded with the primary
site of colonization for C. jejuni, with relatively few microbes being recovered from the small bowel
or at systemic sites. Inflammation was typically characterized by inflammatory and immune cell
infiltration into both the mucosa and submucosa of infected tissues, along with crypt hyperplasia,
increased sloughing of epithelial cells, submucosal edema, and in more severe cases the develop-
ment of ulcers and the loss of crypt structure as the epithelium was damaged/destroyed. Mortality
or severe morbidity was not observed, likely due to C. jejuni’s inability to reach systemic sites in
significant numbers. The development of diarrhea was not typical, but the appearance of mucoid or
298 Laboratory Models for Foodborne Infections
soft stools was common, which differs from typical human infection, in which diarrhea is one of the
most prominent sequelae.
Further investigation into C. jejuni infection of SIGIRR−/− mice linked most of the inflammatory sig-
naling being triggered by C. jejuni to TLR4.26 TLR4−/− and TLR4/SIGIRR double knockouts were found
to display only minimal inflammation and pathology in response to the presence of C. jejuni. This was
also fairly consistent with other results published with mice lacking both IL-10 and TLR4, where sub-
stantially decreased pathology was observed in the absence of TLR4.106 Conversely, although TLR2 had
been previously linked to C. jejuni-induced colitis, in SIGIRR−/− mice, TLR2 appeared to have a protec-
tive role, with TLR2−/− mice and TLR2/SIGIRR double knockouts both developing more severe colitis
following C. jejuni infection.26
A common refrain among those who use mouse models of C. jejuni infection has been the diffi-
culty of establishing consistent infections and colonization numbers, even in knockout mice with more
susceptible immune systems. This is believed to reflect the colonization resistance provided by the
mouse intestinal microbiota. While most birds, ferrets, and humans can become readily infected by
C. jejuni without any manipulation to their intestinal microbiota, the piglet model requires a neonatal
piglet, lacking a fully developed microbiota. Most other animal species, including mice, can harbor
Campylobacter within their intestines transiently, but persistent colonization is largely limited by com-
petition from the intestinal microbiota. In order to allow for reliable and consistent mouse colonization,
some form of disruption to their intestinal microbiota is necessary. Germfree mice, or mice with a
limited microflora are well-known to be very susceptible to C. jejuni colonization, even if they typi-
cally do not develop significant pathology.88 The downside of germfree mice, aside from the difficulty
of maintaining germfree conditions, is the key role microbiota play in the proper development of the
immune system, often leaving germfree mice with an underdeveloped immune system. Fortunately,
a temporary disruption of the microbiota through a single antibiotic treatment was sufficient to allow
for consistent colonization. We treated mice with a single gavage dose of vancomycin 4 h prior to
inoculation with C. jejuni and achieved reliable colonization and consistent pathogen burdens follow-
ing infection with a relatively modest 107 CFU inoculating dose.26 Other studies have used a variety of
antibiotic cocktails to achieve similar results.86 In one study, Bereswill et al. “humanized” mice with
a representative human microbiota, by clearing out the existing mouse microbiota with a cocktail of
antibiotics, followed by inoculation with human fecal samples.111 The newly “humanized” mice became
easily susceptible to C. jejuni colonization by several different strains. The authors characterized a
number of differences between the mouse and human microbial composition; however, precisely how
shifts between mouse and human microbiota opened intestinal niches for C. jejuni colonization remains
unknown. Stahl et al. also carried out a basic characterization of microbiota shifts following vancomy-
cin pretreatment.26 Aside from a dramatic drop in overall bacterial numbers, there were dramatic shifts
in the relative proportions of Firmicutes and Bacteroidetes, however, how those shifts may influence
C. jejuni colonization is presently unknown.
With better knowledge regarding the mechanisms underlying the colonization resistance of the murine
microbiota and new means for increasing the sensitivity of mice to C. jejuni infection, mice may finally
become a relevant infection model for C. jejuni. What is needed now is new research describing the inter-
actions between C. jejuni and the mouse microbiota and immune system to better describe how infection
occurs in mice. Once this is accomplished, we can start drawing more parallels between C. jejuni infec-
tion in mice and humans and employ mouse models to their full potential in understanding the human
disease better.
19.3 Conclusions
Campylobacter jejuni is a common foodborne pathogen that can trigger serious diarrhea in infected
patients. Proper laboratory models for infection have been difficult to establish due to C. jejuni being a
commensal or transient colonizer in most potential animal models of infection. Cellular infection mod-
els, such as the Caco-2 and HT-29 cell lines have proven to be an effective model for studying cellular
interactions. Chicks and germfree mice are useful models for commensal colonization, whereas ferrets,
Campylobacter 299
piglets, and certain knockout mouse strains can be used as effective models for human disease. Herein,
we have discussed how each of these models has been used, the advantages and disadvantages, and some
of what has been learned about Campylobacter from each model.
Many of the mechanisms of pathogenesis for C. jejuni remain either completely unknown or, at best,
poorly characterized. When the first C. jejuni genome sequences failed to reveal identifiable pathogenic-
ity factors akin to those already identified in pathogenic E. coli and Salmonella, a renewed effort was
placed on using known animal models to try and elucidate how C. jejuni colonizes its hosts and how it in
fact causes disease. The lack of animal models that fully replicate the human disease has hindered this
process; however, as a better understanding of the interactions between C. jejuni and the immune system
has started to unfold, better and more relevant animal models are being explored.
REFERENCES
1. Young, K.T., Davis, L.M. & Dirita, V.J. Campylobacter jejuni: molecular biology and pathogenesis. Nat
Rev Microbiol 5, 665–679 (2007).
2. Thabane, M. & Marshall, J.K. Post-infectious irritable bowel syndrome. World J Gastroenterol 15,
3591–3596 (2009).
3. Wieczorek, K. & Osek, J. Antimicrobial resistance mechanisms among Campylobacter. Biomed Res Int
2013, 340605 (2013).
4. van Spreeuwel, J.P. et al. Campylobacter colitis: histological immunohistochemical and ultrastructural
findings. Gut 26, 945–951 (1985).
5. Allos, B.M. Campylobacter jejuni infections: update on emerging issues and trends. Clin Infect Dis 32,
1201–1206 (2001).
6. Newell, D.G. & Fearnley, C. Sources of Campylobacter colonization in broiler chickens. Appl Environ
Microbiol 69, 4343–4351 (2003).
7. Van Dyke, M.I., Morton, V.K., McLellan, N.L. & Huck, P.M. The occurrence of Campylobacter in river
water and waterfowl within a watershed in southern Ontario, Canada. J Appl Microbiol 109, 1053–1066
(2010).
8. Waldenstrom, J. et al. Campylobacter jejuni colonization in wild birds: results from an infection experi-
ment. PLoS One 5, e9082 (2010).
9. Stern, N.J. et al. Campylobacter spp. in Icelandic poultry operations and human disease. Epidemiol
Infect 130, 23–32 (2003).
10. Agunos, A., Waddell, L., Leger, D. & Taboada, E. A systematic review characterizing on-farm sources
of Campylobacter spp. for broiler chickens. PLoS One 9, e104905 (2014).
11. Nichols, G.L. Fly transmission of Campylobacter. Emerg Infect Dis 11, 361–364 (2005).
12. Stanley, K. & Jones, K. Cattle and sheep farms as reservoirs of Campylobacter. J Appl Microbiol
94(Suppl), 104S–113S (2003).
13. Line, J., Hiett, K. & Conlan, A. Comparison of challenge models for determining the colonization dose
of Campylobacter jejuni in broiler chicks. Poult Sci 87, 1700–1706 (2008).
14. Bui, X.T. et al. Survival of Campylobacter jejuni in co-culture with Acanthamoeba castellanii: role of
amoeba-mediated depletion of dissolved oxygen. Environ Microbiol 14, 2034–2047 (2012).
15. Olofsson, J., Axelsson-Olsson, D., Brudin, L., Olsen, B. & Ellstrom, P. Campylobacter jejuni actively
invades the amoeba Acanthamoeba polyphaga and survives within non digestive vacuoles. PLoS One 8,
e78873 (2013).
16. Ikeda, N. & Karlyshev, A.V. Putative mechanisms and biological role of coccoid form formation in
Campylobacter jejuni. Eur J Microbiol Immunol (Bp) 2, 41–49 (2012).
17. Newell, D.G. Animal models of Campylobacter jejuni colonization and disease and the lessons to be
learned from similar Helicobacter pylori models. Symp Ser Soc Appl Microbiol, 57S–67S (2001).
18. Hedstrom, O.R. et al. Pathology of Campylobacter jejuni abortion in sheep. Vet Pathol 24, 419–426
(1987).
19. de Vries, J.J., Arents, N.L. & Manson, W.L. Campylobacter species isolated from extra-oro-intestinal
abscesses: a report of four cases and literature review. Eur J Clin Microbiol Infect Dis 27, 1119–1123 (2008).
20. Blaser, M.J. & Atherton, J.C. Helicobacter pylori persistence: biology and disease. J Clin Invest 113,
321–333 (2004).
300 Laboratory Models for Foodborne Infections
21. Beery, J.T., Hugdahl, M.B. & Doyle, M.P. Colonization of gastrointestinal tracts of chicks by
Campylobacter jejuni. Appl Environ Microbiol 54, 2365–2370 (1988).
22. Fox, J.G., Ackerman, J.I., Taylor, N., Claps, M. & Murphy, J.C. Campylobacter jejuni infection in the
ferret: an animal model of human campylobacteriosis. Am J Vet Res 48, 85–90 (1987).
23. Black, R.E., Levine, M.M., Clements, M.L., Hughes, T.P. & Blaser, M.J. Experimental Campylobacter
jejuni infection in humans. J Infect Dis 157, 472–479 (1988).
24. Lee, A., O’Rourke, J.L., Barrington, P.J. & Trust, T.J. Mucus colonization as a determinant of pathoge-
nicity in intestinal infection by Campylobacter jejuni: a mouse cecal model. Infect Immun 51, 536–546
(1986).
25. Alemka, A., Corcionivoschi, N. & Bourke, B. Defense and adaptation: the complex inter-relationship
between Campylobacter jejuni and mucus. Front Cell Infect Microbiol 2, 15 (2012).
26. Stahl, M. et al. A novel mouse model of Campylobacter jejuni gastroenteritis reveals key pro-
inflammatory and tissue protective roles for Toll-like receptor signaling during infection. PLoS Pathog
10, e1004264 (2014).
27. Sellars, M.J., Hall, S.J. & Kelly, D.J. Growth of Campylobacter jejuni supported by respiration of fuma-
rate, nitrate, nitrite, trimethylamine-N-oxide, or dimethyl sulfoxide requires oxygen. J Bacteriol 184,
4187–4196 (2002).
28. Kendall, J.J., Barrero-Tobon, A.M., Hendrixson, D.R. & Kelly, D.J. Hemerythrins in the microaero-
philic bacterium Campylobacter jejuni help protect key iron-sulphur cluster enzymes from oxidative
damage. Environ Microbiol 16, 1105–1021 (2014).
29. Hugdahl, M.B., Beery, J.T. & Doyle, M.P. Chemotactic behavior of Campylobacter jejuni. Infect Immun
56, 1560–1156 (1988).
30. Lertsethtakarn, P., Ottemann, K.M. & Hendrixson, D.R. Motility and chemotaxis in Campylobacter
and Helicobacter. Annu Rev Microbiol 65, 389–410 (2011).
31. Szymanski, C.M., King, M., Haardt, M. & Armstrong, G.D. Campylobacter jejuni motility and invasion
of Caco-2 cells. Infect Immun 63, 4295–4300 (1995).
32. Hendrixson, D.R. & DiRita, V.J. Identification of Campylobacter jejuni genes involved in commensal
colonization of the chick gastrointestinal tract. Mol Microbiol 52, 471–484 (2004).
33. Kanungpean, D., Kakuda, T. & Takai, S. Participation of CheR and CheB in the chemosensory response
of Campylobacter jejuni. Microbiology 157, 1279–1289 (2011).
34. Lin, J., Sahin, O., Michel, L.O. & Zhang, Q. Critical role of multidrug efflux pump CmeABC in bile
resistance and in vivo colonization of Campylobacter jejuni. Infect Immun 71, 4250–4259 (2003).
35. Flint, A. et al. Phenotypic screening of a targeted mutant library reveals Campylobacter jejuni defenses
against oxidative stress. Infect Immun 82, 2266–2275 (2014).
36. Palyada, K. et al. Characterization of the oxidative stress stimulon and PerR regulon of Campylobacter
jejuni. BMC Genomics 10, 481 (2009).
37. Stahl, M. et al. l-Fucose utilization provides Campylobacter jejuni with a competitive advantage. Proc
Natl Acad Sci USA 108, 7194–7199 (2011).
38. Stahl, M., Butcher, J. & Stintzi, A. Nutrient acquisition and metabolism by Campylobacter jejuni. Front
Cell Infect Microbiol 2, 5 (2012).
39. Hofreuter, D. Defining the metabolic requirements for the growth and colonization capacity of
Campylobacter jejuni. Front Cell Infect Microbiol 4, 137 (2014).
40. Hofreuter, D. et al. Contribution of amino acid catabolism to the tissue specific persistence of
Campylobacter jejuni in a murine colonization model. PLoS One 7, e50699 (2012).
41. Guccione, E. et al. Amino acid-dependent growth of Campylobacter jejuni: key roles for aspartase
(AspA) under microaerobic and oxygen-limited conditions and identification of AspB (Cj0762), essen-
tial for growth on glutamate. Mol Microbiol 69, 77–93 (2008).
42. Naikare, H., Palyada, K., Panciera, R., Marlow, D. & Stintzi, A. Major role for FeoB in Campylobacter
jejuni ferrous iron acquisition, gut colonization, and intracellular survival. Infect Immun 74, 5433–5444
(2006).
43. Palyada, K., Threadgill, D. & Stintzi, A. Iron acquisition and regulation in Campylobacter jejuni. J
Bacteriol 186, 4714–4729 (2004).
44. Everest, P.H. et al. Differentiated Caco-2 cells as a model for enteric invasion by Campylobacter jejuni
and C. coli. J Med Microbiol 37, 319–325 (1992).
Campylobacter 301
45. Monteville, M.R., Yoon, J.E. & Konkel, M.E. Maximal adherence and invasion of INT 407 cells by
Campylobacter jejuni requires the CadF outer-membrane protein and microfilament reorganization.
Microbiology 149, 153–165 (2003).
46. MacCallum, A.J., Harris, D., Haddock, G. & Everest, P.H. Campylobacter jejuni-infected human epi-
thelial cell lines vary in their ability to secrete interleukin-8 compared to in vitro-infected primary
human intestinal tissue. Microbiology 152, 3661–3665 (2006).
47. Van Deun, K. et al. Colonization strategy of Campylobacter jejuni results in persistent infection of the
chicken gut. Vet Microbiol 130, 285–297 (2008).
48. Watson, R.O. & Galan, J.E. Campylobacter jejuni survives within epithelial cells by avoiding delivery
to lysosomes. PLoS Pathog 4, e14 (2008).
49. Davis, L. & DiRita, V. Experimental chick colonization by Campylobacter jejuni. Curr Protoc Microbiol
Chapter 8, Unit 8A.3 (2008).
50. Ruiz-Palacios, G.M., Escamilla, E. & Torres, N. Experimental Campylobacter diarrhea in chickens.
Infect Immun 34, 250–255 (1981).
51. Welkos, S.L. Experimental gastroenteritis in newly-hatched chicks infected with Campylobacter jejuni.
J Med Microbiol 18, 233–248 (1984).
52. Manninen, K.I., Prescott, J.F. & Dohoo, I.R. Pathogenicity of Campylobacter jejuni isolates from ani-
mals and humans. Infect Immun 38, 46–52 (1982).
53. Hermans, D. et al. A tolerogenic mucosal immune response leads to persistent Campylobacter jejuni
colonization in the chicken gut. Crit Rev Microbiol 38, 17–29 (2012).
54. Shanker, S., Lee, A. & Sorrell, T.C. Experimental colonization of broiler chicks with Campylobacter
jejuni. Epidemiol Infect 100, 27–34 (1988).
55. Flanagan, R.C., Neal-McKinney, J.M., Dhillon, A.S., Miller, W.G. & Konkel, M.E. Examination of
Campylobacter jejuni putative adhesins leads to the identification of a new protein, designated FlpA,
required for chicken colonization. Infect Immun 77, 2399–2407 (2009).
56. Hermans, D. et al. Colonization factors of Campylobacter jejuni in the chicken gut. Vet Res 42, 82
(2011).
57. Byrne, C.M., Clyne, M. & Bourke, B. Campylobacter jejuni adhere to and invade chicken intestinal
epithelial cells in vitro. Microbiology 153, 561–569 (2007).
58. Alemka, A. et al. Purified chicken intestinal mucin attenuates Campylobacter jejuni pathogenicity in
vitro. J Med Microbiol 59, 898–903 (2010).
59. de Zoete, M.R., van Putten, J.P. & Wagenaar, J.A. Vaccination of chickens against Campylobacter.
Vaccine 25, 5548–5557 (2007).
60. Shaughnessy, R.G., Meade, K.G., McGivney, B.A., Allan, B. & O’Farrelly, C. Global gene expression
analysis of chicken caecal response to Campylobacter jejuni. Vet Immunol Immunopathol 142, 64–71
(2011).
61. Meade, K.G. et al. Comparative in vivo infection models yield insights on early host immune response
to Campylobacter in chickens. Immunogenetics 61, 101–110 (2009).
62. Shaughnessy, R.G. et al. Innate immune gene expression differentiates the early avian intestinal response
between Salmonella and Campylobacter. Vet Immunol Immunopathol 132, 191–198 (2009).
63. Smith, C.K. et al. Campylobacter colonization of the chicken induces a proinflammatory response in
mucosal tissues. FEMS Immunol Med Microbiol 54, 114–121 (2008).
64. Babakhani, F.K., Bradley, G.A. & Joens, L.A. Newborn piglet model for campylobacteriosis. Infect
Immun 61, 3466–3475 (1993).
65. Munroe, D.L., Prescott, J.F. & Penner, J.L. Campylobacter jejuni and Campylobacter coli serotypes
isolated from chickens, cattle, and pigs. J Clin Microbiol 18, 877–881 (1983).
66. Tzipori, S. et al. Studies with enteroaggregative Escherichia coli in the gnotobiotic piglet gastroenteritis
model. Infect Immun 60, 5302–5306 (1992).
67. Boyen, F. et al. Porcine in vitro and in vivo models to assess the virulence of Salmonella enterica
serovar Typhimurium for pigs. Lab Anim 43, 46–52 (2009).
68. Reid, A.N., Pandey, R., Palyada, K., Naikare, H. & Stintzi, A. Identification of Campylobacter jejuni genes
involved in the response to acidic pH and stomach transit. Appl Environ Microbiol 74, 1583–1597 (2008).
69. Dufour, V. et al. Inactivation of the LysR regulator Cj1000 of Campylobacter jejuni affects host coloni-
zation and respiration. Microbiology 159, 1165–1178 (2013).
302 Laboratory Models for Foodborne Infections
70. van Alphen, L.B. et al. Biological roles of the O-methyl phosphoramidate capsule modification in
Campylobacter jejuni. PLoS One 9, e87051 (2014).
71. Flint, A., Sun, Y.Q. & Stintzi, A. Cj1386 is an ankyrin-containing protein involved in heme trafficking
to catalase in Campylobacter jejuni. J Bacteriol 194, 334–345 (2012).
72. Bell, J.A. & Manning, D.D. A domestic ferret model of immunity to Campylobacter jejuni-induced
enteric disease. Infect Immun 58, 1848–1852 (1990).
73. Fox, J.G. et al. Campylobacter-like organisms isolated from gastric mucosa of ferrets. Am J Vet Res 47,
236–239 (1986).
74. Fox, J.G., Otto, G., Murphy, J.C., Taylor, N.S. & Lee, A. Gastric colonization of the ferret with
Helicobacter species: natural and experimental infections. Rev Infect Dis 13(Suppl 8), S671–S680
(1991).
75. Nemelka, K.W. et al. Immune response to and histopathology of Campylobacter jejuni infection in fer-
rets (Mustela putorius furo). Comp Med 59, 363–371 (2009).
76. Bacon, D.J. et al. A phase-variable capsule is involved in virulence of Campylobacter jejuni 81-176. Mol
Microbiol 40, 769–777 (2001).
77. Goon, S. et al. A σ28-regulated nonflagella gene contributes to virulence of Campylobacter jejuni 81-176.
Infect Immun 74, 769–772 (2006).
78. Yao, R., Burr, D.H. & Guerry, P. CheY-mediated modulation of Campylobacter jejuni virulence. Mol
Microbiol 23, 1021–1031 (1997).
79. Glavis-Bloom, J., Muhammed, M. & Mylonakis, E. Of model hosts and man: using Caenorhabditis ele-
gans, Drosophila melanogaster and Galleria mellonella as model hosts for infectious disease research.
Adv Exp Med Biol 710, 11–17 (2012).
80. Senior, N.J. et al. Galleria mellonella as an infection model for Campylobacter jejuni virulence. J Med
Microbiol 60, 661–669 (2011).
81. Champion, O.L. et al. Galleria mellonella as an alternative infection model for Yersinia pseudotubercu-
losis. Microbiology 155, 1516–1522 (2009).
82. Champion, O.L. et al. Insect infection model for Campylobacter jejuni reveals that O-methyl phos-
phoramidate has insecticidal activity. J Infect Dis 201, 776–782 (2010).
83. Gundogdu, O. et al. The Campylobacter jejuni transcriptional regulator Cj1556 plays a role in the
oxidative and aerobic stress response and is important for bacterial survival in vivo. J Bacteriol 193,
4238–4249 (2011).
84. Fernie, D.S. & Park, R.W. The isolation and nature of campylobacters (microaerophilic vibrios) from
laboratory and wild rodents. J Med Microbiol 10, 325–329 (1977).
85. Field, L.H., Underwood, J.L., Pope, L.M. & Berry, L.J. Intestinal colonization of neonatal animals by
Campylobacter fetus subsp. jejuni. Infect Immun 33, 884–892 (1981).
86. Field, L.H., Underwood, J.L. & Berry, L.J. The role of gut flora and animal passage in the colonisation
of adult mice with Campylobacter jejuni. J Med Microbiol 17, 59–66 (1984).
87. Blaser, M.J., Duncan, D.J., Warren, G.H. & Wang, W.L. Experimental Campylobacter jejuni infection
of adult mice. Infect Immun 39, 908–916 (1983).
88. Chang, C. & Miller, J.F. Campylobacter jejuni colonization of mice with limited enteric flora. Infect
Immun 74, 5261–5271 (2006).
89. Jesudason, M.V., Hentges, D.J. & Pongpech, P. Colonization of mice by Campylobacter jejuni. Infect
Immun 57, 2279–2282 (1989).
90. Stanfield, J.T., McCardell, B.A. & Madden, J.M. Campylobacter diarrhea in an adult mouse model.
Microb Pathog 3, 155–165 (1987).
91. Pei, Z. et al. Mutation in the peb1A locus of Campylobacter jejuni reduces interactions with epithelial
cells and intestinal colonization of mice. Infect Immun 66, 938–943 (1998).
92. Pei, Z. & Blaser, M.J. Pathogenesis of Campylobacter fetus infections. Role of surface array proteins in
virulence in a mouse model. J Clin Invest 85, 1036–1043 (1990).
93. Fernandez, H., Vivanco, T. & Eller, G. Expression of invasiveness of Campylobacter jejuni ssp. jejuni
after serial intraperitoneal passages in mice. J Vet Med B Infect Dis Vet Public Health 47, 635–639
(2000).
94. Bar, W., Becker, K. & Hewel, C. Systemic spread of Campylobacter jejuni after intravenous infections.
FEMS Microbiol Immunol 1, 263–270 (1989).
Campylobacter 303
95. Blaser, M.J. et al. Extraintestinal Campylobacter jejuni and Campylobacter coli infections: host factors
and strain characteristics. J Infect Dis 153, 552–559 (1986).
96. Baqar, S. et al. Murine intranasal challenge model for the study of Campylobacter pathogenesis and
immunity. Infect Immun 64, 4933–4939 (1996).
97. Al-Banna, N.A., Raghupathy, R. & Albert, M.J. Induction of cytokines in different organs after intrana-
sal inoculation of Campylobacter jejuni in mice. Gut Pathog 4, 23 (2012).
98. Hodgson, A.E., McBride, B.W., Hudson, M.J., Hall, G. & Leach, S.A. Experimental Campylobacter
infection and diarrhoea in immunodeficient mice. J Med Microbiol 47, 799–809 (1998).
99. Mansfield, L.S. et al. C57BL/6 and congenic interleukin-10-deficient mice can serve as models of
Campylobacter jejuni colonization and enteritis. Infect Immun 75, 1099–1115 (2007).
100. Kole, A. & Maloy, K.J. Control of intestinal inflammation by interleukin-10. Curr Top Microbiol
Immunol 380, 19–38 (2014).
101. Hansen, J.J., Holt, L. & Sartor, R.B. Gene expression patterns in experimental colitis in IL-10-deficient
mice. Inflamm Bowel Dis 15, 890–899 (2009).
102. Lippert, E. et al. Gnotobiotic IL-10; NF-κB mice develop rapid and severe colitis following
Campylobacter jejuni infection. PLoS One 4, e7413 (2009).
103. Wilson, D.L. et al. Genetic diversity in Campylobacter jejuni is associated with differential colonization
of broiler chickens and C57BL/6J IL10-deficient mice. Microbiology 156, 2046–2057 (2010).
104. Sun, X., Threadgill, D. & Jobin, C. Campylobacter jejuni induces colitis through activation of mam-
malian target of rapamycin signaling. Gastroenterology 142, 86–95.e5 (2012).
105. Samuelson, D.R. et al. The Campylobacter jejuni CiaD effector protein activates MAP kinase signaling
pathways and is required for the development of disease. Cell Commun Signal 11, 79 (2013).
106. Haag, L.M. et al. Campylobacter jejuni induces acute enterocolitis in gnotobiotic IL-10−/− mice via Toll-
like-receptor-2 and -4 signaling. PLoS One 7, e40761 (2012).
107. Sun, X. & Jobin, C. Nucleotide-binding oligomerization domain-containing protein 2 controls host
response to Campylobacter jejuni in Il10−/− mice. J Infect Dis 210, 1145–1154 (2014).
108. Karrasch, T., Kim, J.S., Muhlbauer, M., Magness, S.T. & Jobin, C. Gnotobiotic IL-10−/−; NF-κB(EGFP)
mice reveal the critical role of TLR/NF-κB signaling in commensal bacteria-induced colitis. J Immunol
178, 6522–6532 (2007).
109. Watson, R.O., Novik, V., Hofreuter, D., Lara-Tejero, M. & Galan, J.E. A MyD88-deficient mouse model
reveals a role for Nramp1 in Campylobacter jejuni infection. Infect Immun 75, 1994–2003 (2007).
110. Sham, H.P. et al. SIGIRR, a negative regulator of TLR/IL-1R signalling promotes microbiota dependent
resistance to colonization by enteric bacterial pathogens. PLoS Pathog 9, e1003539 (2013).
111. Bereswill, S. et al. Novel murine infection models provide deep insights into the “menage a trois” of
Campylobacter jejuni, microbiota and host innate immunity. PLoS One 6, e20953 (2011).
20
Cronobacter: Virulence and Pathogenesis
Nemani V. Prasadarao
CONTENTS
20.1 Introduction................................................................................................................................... 305
20.2 Necrotizing Enterocolitis.............................................................................................................. 306
20.2.1 A Rat Model of C. muytjensii-Induced Necrotizing Enterocolitis.................................. 306
20.2.2 C. muytjensii Interaction with Epithelial Cells in Culture............................................... 307
20.2.3 C. muytjensii Interaction with Dendritic Cells................................................................ 308
20.2.4 The Role of Neutrophils and Macrophages in Cronobacter Infection............................ 309
20.2.5 Prevention of Cronobacter-Induced NEC by Lactobacillus........................................... 309
20.3 Septicemia and Meningitis.............................................................................................................310
20.3.1 Serum Tolerance of Cronobacter spp...............................................................................310
20.3.2 A Neonatal Rat Model of Meningitis................................................................................310
20.3.3 Cronobacter Interaction with Brain Endothelial Cells.....................................................311
20.4 C. sakazakii: Motility and Biofilm Formation...............................................................................312
20.5 C. sakazakii Interaction with Caenorhabditis elegans.................................................................313
20.6 Conclusions....................................................................................................................................313
References................................................................................................................................................314
20.1 Introduction
Originally referred to as yellow-pigmented Enterobacter cloacae, it was later classified as a new species
Enterobacter sakazakii. Subsequent characterization enabled the reclassification of these bacteria into a
new genus called Cronobacter [1]. Cronobacter is composed of a diverse group of Gram-negative bacilli,
which includes Cronobacter sakazakii, Cronobacter muytjensii, Cronobacter malonaticus, Cronobacter
dublinensis, Cronobacter turicensis, Cronobacter universalis, and Cronobacter condimenti [2]. Except
Cronobacter condimenti, all other Cronobacter spp. are associated with human infections. They cause
life-threatening infections in neonates due to the consumption of powdered infant formula contami-
nated with Cronobacter [3,4]. Outbreaks of Cronobacter infections in neonatal intensive care units have
resulted in several CDC warnings, and so efforts are in place to improve health care. Considered as an
opportunistic pathogen, Cronobacter causes severe illness in neonates, such as necrotizing enterocolitis,
bacteremia, and meningitis, often in low-birth-weight preterm infants [5]. Infections due to Cronobacter
in normal and immunocompromised adults have also been noted, but these are less severe. Based on
partial 16S rRNA and hsp60 sequencing, four cluster groups of C. sakazakii have been identified among
this diverse group of pathogens [6]. The bacterium can be found in a variety of foods, including dairy-
based foods (cheese), dried meats, and rice. Furthermore, it was also detected in environmental sources
such as soil, livestock facilities, and food preparation units. Compared to other Enterobacteriaceae fam-
ily members, Cronobacter is highly resistant to heat, dryness, and acidic conditions [7]. It also forms
biofilms that function as a protective barrier to withstand environmental stress and obviate immune
surveillance of the host.
Several selective media for detecting Cronobacter have been developed [8,9]. However, these
media are insufficient to support the growth of all strains of Cronobacter [10]. To detect Cronobacter
305
306 Laboratory Models for Foodborne Infections
spp. in powdered infant formula, a one-step enrichment protocol using a chromogenic medium has been
designed [11]. More useful molecular-based detection techniques for understanding the epidemiology
of Cronobacter have also been developed by targeting a number of genes such as 16S rRNA, 16S-23S
rRNA intergenic regions, ompA, zinc-containing metalloprotease, dnaG and gluA genes by real-time
polymerase chain reaction (PCR) [12–15]. Genes that are unique to different species of Cronobacter are
particularly useful as candidate markers in molecular detection protocols [16–18]. Due to limitations of
individual gene-based techniques, whole-genome sequencing may help identify Cronobacter species
and assist in comparing the genotypic and phenotypic features of the pathogen being studied.
Although powdered infant formula has been found to be the main source of Cronobacter for infecting
newborns, transmission from mother to child during the delivery cannot be excluded. It was also reported
that plant material may be a natural source of Cronobacter spp. [19]. In a study performed to characterize
genotypic and phenotypic features of C. sakazakii strains obtained in an outbreak in intensive care units
in France in 1994 [20], a total of 31 stains were collected, and on the basis of 16S rRNA analysis, 30
strains were confirmed as C. sakazakii. Pulsed-field gel electrophoresis (PFGE) typing recognized four
different pulsotypes and clearly demonstrated that C. sakazakii strains exhibit different genotypic and
phenotypic characteristics. This reinforces the necessity for rigorous and careful testing to identify the
strains. Genomic sequencing of C. sakazakii strain ATCC BAA-894 isolated from contaminated infant
formula revealed a single 4.4 Mb chromosome and two plasmids, pESA2 (31 kb) and pESA3 (131 kb) [21].
ATCC BAA-894 strain contains 4392 genes in the core genome, and 21 genes are found to be unique in
5 other C. sakazakii strains. Similar to ATCC BAA-894 strain, C. turicensis contains a similar genome
and 2 plasmids, which also encodes open reading frames (ORFs) for 223 virulence-associated genes [22].
Comparison of pESA3 and pCTU1 (C. turicensis plasmid) by in silico analysis revealed the presence of
two iron-acquisition systems, which may be essential for pathogenesis.
For the pathogenesis of Cronobacter spp., the bacteria must attach to intestinal epithelial cells of
infants fed with contaminated formula, followed by invasion and hematogenous spread. The adherence
capacities of 50 C. sakazakii strains have been studied using HEp2 and Caco-2 cell lines and brain
microvascular endothelial cells, and two distinctive patterns of binding were observed, diffuse adhesion
and localized cluster formation [23]. The adherence of these Cronobacter strains appears to be non-
fimbrial mediated. Of note, some fimbrial clusters have been identified in the genomes of Cronobacter,
although C. sakazakii is the only bacterium that expresses β-fimbriae, but its role in the pathogenesis
is still to be evaluated. The putative virulence factors produced, such as enterotoxin-like compounds,
by Cronobacter were first assessed by the suckling mouse assay [24]. Analysis of various Cronobacter
strains from different brands of infant formula revealed that they contain high levels (500-fold) of heat-
stable endotoxin (lipopolysaccharide or LPS) [20]. Since these formulae contain a variety of bacterial
species, it is not clear what the source of LPS is. However, its presence enhances the translocation of
C. sakazakii across the gut and blood–brain barrier. Surveillance of microbial burden in food products,
growth conditions, and epidemiology would provide clues to developing methods that reduce health
risks in vulnerable individuals. Furthermore, understanding the pathophysiology of the diseases caused
by Cronobacter spp. is also desirable to identify long-term risks in infected neonates and infants. To
gain insights into the pathophysiology of Cronobacter-induced diseases, a careful selection and usage
of animal models is clearly required. Therefore, the purpose of this chapter is to present the animal and
tissue culture models used thus far to identify bacterial and host factors that contribute to Cronobacter
pathogenesis.
20.2 Necrotizing Enterocolitis
20.2.1 A Rat Model of C. muytjensii-Induced Necrotizing Enterocolitis
Premature infants are prone to a variety of diseases including necrotizing enterocolitis (NEC), which
is a serious gastrointestinal disease and occurs in ∼1 in 1000 live births [25]. The etiology of NEC is
multifactorial, and the risk factors include prematurity, formula feeding, and abnormal bacterial coloni-
zation of intestinal tract. To replicate histopathological manifestations of NEC that mimic human NEC,
Cronobacter: Virulence and Pathogenesis 307
investigators have used several animal models to study the disease. The pathological features of NEC
show the presence of mucosal edema, pneumatosis intestinalis (i.e., the presence of gas within the wall
of the intestine), epithelial sloughing/villous atrophy, enterocyte apoptosis, vascular thrombosis, and dis-
continuous necrotic segments of intestine. A variety of bacterial species are thought to cause NEC such
as Enterobacter, Clostridium, and Staphylococcus spp. Since milk-based infant formulas contaminated
with C. sakazakii have been linked to many NEC outbreaks, a newborn rat model of Cronobacter-
induced NEC using the combination of hypoxia and formula feeding has been developed [26]. The strain
used in these studies, obtained from ATCC (strain 51329), was later classified as C. muytjensii. Newborn
rats fed with infant formula mixed with 105 CFU of C. muytjensii produced clinical symptoms similar
to Grade 3 NEC by day 4 postinfection (Figure 20.1). The pathological analysis of intestinal sections
obtained from infected rats showed macroscopic and microscopic features analogous to that seen in
human NEC [27]. The formula-fed and hypoxic group of rats only showed lower pathological symptoms
and had a mortality rate of 40%, whereas the mortality rate increased to 70% with Cronobacter infec-
tion. Scanning electron microscopy of intestines of newborn rats revealed that specimens from formula-
fed and hypoxic rats exhibited intact villi. C. muytjensii infection increased the binding of the bacteria
to villi tips and showed enterocyte blebbing and gap formation in the epithelium.
(C)
FIGURE 20.1 Neonatal rat model of Cronobacter muytjensii-induced necrotizing enterocolitis: Newborn rat pups were
fed with Esbilac formula two times and three times daily and were placed under hypoxic conditions with 5% carbon dioxide
(FF+H) with (A, left) or without (A, right) C. muytjensii infection. FF+H treated rats showed a normal abdominal wall
with a milk spot (B). Abdominal discoloration and evidence of clinical peritonitis were observed in FF+H+C. muytjensii
rats (C). In contrast, control rat pups showed normal intestine (D). (Reprinted with permission from Hunter, C.J., et al., J.
Infect. Dis., 198, 586–593, 2008.)
308 Laboratory Models for Foodborne Infections
C. sakazakii strains induced more apoptosis in Caco-2 cells than the low-binding strains. Approximately
25% of infected Caco-2 cells were apoptotic. Abnormal colonization of bacteria usually results in the
loss of gut barrier integrity, which is an important pathophysiological feature in the development of
NEC. Similar to that in human epithelial cells, high-binding C. sakazakii infection of Caco-2 cells
caused an increase in permeability of the monolayers as assessed by horseradish peroxidase leakage.
C. sakazakii-induced leakage of the tight junctions in Caco-2 cells were due to the disruption of ZO1 at
the periphery of the cells. Moreover, these permeability changes of Caco-2 cells require the activation of
PKC, which in turn activates NO production upon C. sakazakii infection.
Outer membrane protein A (OmpA) is a 35-kDa surface protein of Gram-negative bacteria whose
structure is highly conserved throughout the evolution [30]. OmpA contains eight transmembrane
domains and four extracellular loops. However, some minor changes are present in the extracellular
loops that help differentiate whether the bacterium is pathogenic or nonpathogenic [31]. Comparison
of OmpA sequences among several Gram-negative strains revealed that the extracellular loops of
C. sakazakii are significantly different from other sequences [30]. OmpA of C. sakazakii has been shown
to play a critical role in the invasion of INT407 cells with an invasion frequency of 0.08 + 0.002 at a mul-
tiplicity of infection of 100 [32,33]. This invasion frequency in epithelial cells is considerably negligible
compared to the invasion frequencies of other gut pathogens such as Salmonella or Shigella [34,35]. The
invasion into epithelial cells depends on both microfilament and microtubules [36]. Fibronectin appears
to be critical for the binding of C. sakazakii to epithelial cells [37]. However, deletion of OmpA in this
strain reduced the invasion by 80% but did not reduce the attachment to the cells. Subsequently, a mouse
model of infection has been developed in which oral feeding of 103 CFU C. muytjensii to 3-day-old mice
induces NEC-like pathology such as intestinal dilation and bowel discoloration by 48 h postinfection [38].
In contrast, OmpA− C. muytjensii does not cause such injury and the mice survived. In this model, wild-
type C. muytjensii binds to the intestine efficiently, whereas OmpA− C. sakazakii could not associate
with epithelial cells. Of note, prebiotic galacto-oligosaccharides and polydextrose inhibit the binding of
C. sakazakii 4603 to HEp-2 cells and Caco-2 cells [39].
Furthermore, supernatants of myeloid-derived DCs infected with wild-type C. muytjensii when incu-
bated with Caco-2 cells grown in Transwell inserts increased the permeability of the monolayers [38]. In
contrast, OmpA− C. muytjensii-infected DC supernatants showed only minor permeability changes. The
supernatant of the wild-type strain also caused disruption of tight junctions, which was further increased
by infection. It appears that tight junction disruption is the first step in the loss of epithelial cells in
the pathogenesis of NEC. In support of this concept, incubation of Caco-2 cells with the supernatants
obtained from DCs infected with C. muytjensii enhanced apoptosis of the cells in the presence of the
bacteria, which is prevented by the presence of apoptosis inhibitor, ZVAD. The responsible soluble factor
in C. muytjensii-infected DC supernatants is an anti-inflammatory cytokine, TGFβ. Wild-type strains
produced robust quantities of TGFβ from bone-marrow-derived DCs, while OmpA− C. muytjensii gener-
ated basal levels of the cytokine. Mucosal scrapings obtained from the intestines of infected mice also
showed increased tgfβ transcript levels. Similarly, intestinal homogenates from C. muytjensii-infected
mice also revealed higher concentrations of TGFβ. Anti-TGFβ antibodies, but not anit-IL-10 antibodies,
when added to the supernatants of C. muytjensii-infected DCs prevented the bacteria-induced monolayer
permeability of Caco-2 cells and apoptosis. This inhibitory effect was due to the inhibition of iNOS
expression in the cells. Upon infection, DCs are recruited to the intestinal lamina propria, which extend
their protrusions between enterocytes to sample the pathogen or macromolecules. Using a double-layer
model by culturing monocyte-derived macrophages on the top of a Transwell filter insert and bone-
marrow-derived DCs at the bottom, it was demonstrated that the presence of DCs increased the produc-
tion of TGFβ, thereby increasing the permeability of the Caco-2 monolayers. Corroborating the in vitro
results, depletion of DCs in newborn mice by injecting anti-CD11c antibody at day 1 after birth resulted
in resistance to C. muytjensii-induced NEC and survival as normal, uninfected mice. DC-depleted
mice showed no signs of pathology in intestines. Adoptive transfer of DCs into these mice rendered the
animals susceptible to infection again.
suppressing the iNOS expression [28]. This effect was due to prevention of the binding of C. muytjensii
to IEC-6 cell by L. bulgaricus. Moreover, the probiotic also inhibited C. muytjensii-induced apoptosis
of IEC-6 cells. The observations made in IEC-6 were also reflected in infant rats that were orally fed
with L. bulgaricus and then infected with C. muytjensii as they maintained the structural integrity of
enterocytes by preventing the attachment of Cronobacter under hypoxic conditions. Recent studies have
also shown that the conditioned medium of the probiotic Bifidobacterium infantis protects neonatal mice
from intestinal inflammation when infected with C. sakazakii [45]. The probiotic conditioned medium
reduced enterocyte apoptosis and mucin production and maintained the integrity of the ileal structure
when compared with intestines of mice treated with wild-type bacteria.
Control
X 20 X 40 X 40
(D) (E) (F)
OmpA+ CS
X 20 X 20 X 40
FIGURE 20.2 The presence of OmpA+ C. muytjensii in infected newborn rats: C. muytjensii-fed newborn rat brains were
harvested 48 h postinfection, fixed, sectioned, and stained with the anti-OmpA antibody. Mayer’s hematoxylin was used to
counterstain the sections. Control pups received saline. Arrows indicate the presence of clusters of bacteria in white matter
and the cortex. (Reprinted with permission from Mittal, R., et al., Lab. Invest., 89, 263–277, 2009.)
growth along with microvilli flattening. Furthermore, epithelial degeneration was observed in mucosa
along with focal ulceration, reactive stromal changes, and suppurative inflammation. The binding of
C. muytjensii also caused apoptosis of epithelial cells. Infection of mouse pups with wild-type strain
produced TNF-α, IL-1β, IL-6, and IL-10, and chemokine MIP-2 at highest levels by 48 h postinfection.
Of note, OmpA− C. muytjensii-infected animals showed very low levels of these cytokines and chemo-
kines except for IL-10, the levels of which increased between 96 and 120 h postinfection. Despite a small
number of OmpA− C. muytjensii entering the circulation, further multiplication of the bacteria was not
observed. Follow-up serum bactericidal assays showed that wild-type C. muytjensii was resistant to
killing compared to OmpA− C. muytjensii, which are killed within 1 h postincubation with neonatal rat
serum. Other investigators demonstrated that a plasminogen activator (Cpa) mutant of C. sakazakii was
serum sensitive in comparison with its wild-type strain [51]. Cpa activates plasminogen by inactivat-
ing α2-AP and also proteolytically cleaves complement components C3, C3a, and C4b. Lack of serum
resistance in OmpA− C. muytjensii could be due to alteration of Cpa expression because of downstream
effects of ompA deletion. Taken together, it appears that newborn rats infected with C. muytjensii without
hypoxia also results in NEC-like symptoms followed by penetration of the organism into the blood to
cause meningitis.
(A) (B)
(C) (D)
(E) (F)
FIGURE 20.3 Invasion of C. muytjensii into human brain microvascular endothelial cells (HBMEC): Green-fluorescent-
protein-expressing C. muytjensii were incubated with HBMEC monolayers for 4 h, washed, and fixed with 2% parafor-
maldehyde. The monolayers were imaged, and Z-stacks of confocal images were acquired using a Leica laser scanning
microscope. The images were taken at 63× magnification. (Reprinted with permission from Singamsetty, V.K., et al.,
Microb. Pathog., 45, 181–191, 2008.)
the expression of OmpA and microtubule reorganization in the cells. In this entry process, microtubule
aggregation is observed beneath the bacterial binding sites but not actin accumulation. Although many
bacterial strains utilize microfilament reorganization in eukaryotic cells for internalization, C. muytjensii
entry appears to be a specialized mechanism. PKC-α plays a central role in cytoskeletal reorganization
in the entry of many bacterial pathogens. C. muytjensii invasion of HBMECs also depends on PKC-α
as overexpression of a dominant negative form of PKC-α, PKC-CAT/KR, in HBMECs prevented the
entry of the bacteria. Studies have demonstrated that both PKC-α and PI3-Kinase stabilize microtubules,
and in agreement, PI3-kinase activation is also necessary for C. muytjensii entry into HBMECs. In
contrast, the invasion of C. sakazakii strain (ATCC 29544) into HBMECs was prevented by cytochala-
sin D, an inhibitor of actin microfilaments [53]. However, the invasion also depended on PI3-kinase as
C. sakazakii ATCC 29544 induced activation of Akt, a downstream substrate for PI3-Kinase.
capacity [55]. A mutant strain of C. sakazakii, LWW02, was constructed by deleting a gene encoding
heptosyltransferase I [56]. This mutant exhibited slower growth, higher permeability of the mem-
brane, and surface hydrophobicity. The biofilm formation by this mutant was stronger compared
to wild-type strains. Furthermore, studies have shown that flagella of Cronobacter is responsible
for the formation of biofilms and for binding to Caco-2 cells [57]. To study the virulence genes of
C. sakazakii ATCC 29544, a transposon-mediated random mutant library was generated and screened,
leading to the identification of a mutant deficient in invasion in Caco-2 cells [58]. A novel plasmid
pCSA2 that contains six ORFs and 4938 bp has been identified. Of the six ORFs, one was predicted
to encode methyl-accepting chemotaxis protein (MCP), which contains one MCP domain and two
sensor PAS (Per-Arnt-Sim sensory) domains that show similarity with biofilm dispersion protein
BdlA of Pseudomonas aeruginosa. Deletion of mcp gene significantly reduced the invasion in Caco-2
cells, and complementation with a plasmid-containing mcp gene enabled the mutant to invade the
cells. Lack of invasion into Caco-2 cells is due to reduced adherence to the cells. Corroborating with
the role in adherence and invasion for MCP, 3-day-old rats fed with the mcp-deleted mutant showed
100-fold reduced efficiency to translocate across the gut and deep into liver and spleen. Interestingly,
the deletion of the mcp gene in C. sakazakii resulted in organisms becoming hypermotile (∼31.5 mm
in 8 h on 0.3% TSA agar) compared to the wild-type strain (∼17.3 mm). As flagella are important
for the motility of Cronobacter, analysis of genes involved in flagellar assembly revealed that mcp
deletion enhanced the mRNA levels of fliA and fliC genes. In addition to the role of the mcp gene in
motility, it is also required for biofilm formation. Thus, MCP plays a critical role in the virulence of
C. sakazakii by regulating multiple functions.
20.5 C
. sakazakii Interaction with Caenorhabditis elegans
Besides rat and murine models, several other models have also been used to study bacterial patho-
genesis, such as zebrafish and C. elegans [59,60]. C. elegans is a nonparasitic free-living nematode
that is very useful for studying bacterial interaction due to its well-developed genetic and molecular
tools to manipulate the organism. Studies by Shivamurthy et al. revealed that C. elegans fed with
C. sakazakii died in liquid conditions with LT50 of 134 + 2.8 h, whereas feeding on E. coli OP50
(food source of C. elegans) had no effect [61]. C. sakazakii binding to intestines of C. elegans is
responsible for killing the organism, which increased with time of infection. Only live Cronobacter,
not heat-killed ones, were able to colonize and multiply in the intestines, thereby killing the organ-
ism. The death of C. elegans by Cronobacter was due to the apoptosis of the cells of the intestinal
lumen, a mechanism that appears to be similar to that of human NEC pathogenesis. Subsequent stud-
ies by this group revealed that exposure to C. sakazakii LPS led to paralysis, and eventually death,
of C. elegans [61]. Interestingly, the bacterium modifies the structure of LPS upon interaction with
the host to avoid immune response. Map Kinase pathway plays an important role in the immunity of
C. elegans against C. sakazakii LPS.
20.6 Conclusions
Application of suitable animal models to study bacterial infections in such a way that replicates the
pathogenesis in humans is critical to developing therapeutic strategies. To date, investigators have used
a variety of animal species to understand the pathogenic mechanisms. Commonly present in various
food sources and soil, Cronobacter spp. have been linked to the onset of NEC. Both newborn rat mod-
els under hypoxic conditions and mouse models used to explore the pathogenesis of NEC revealed that
C. sakazakii and C. muytjensii can induce NEC-like symptoms. Further studies are clearly required to
fully elucidate the molecular mechanisms involved in the onset of NEC. C. sakazakii-induced neonatal
meningitis is infrequent but often devastating. The neonatal rat model, which mimics human disease, is
extremely useful to identify the bacterial virulence factors and host response to infection, thereby help-
ing to create appropriate treatment strategies.
314 Laboratory Models for Foodborne Infections
REFERENCES
1. Iversen C, et al. Identification of “Cronobacter” spp. (Enterobacter sakazakii). J Clin Microbiol. 2007;
45:3814–3816.
2. Healy B, et al. Cronobacter (Enterobacter sakazakii): an opportunistic foodborne pathogen. Foodborne
Pathog Dis. 2010; 7:339–350.
3. Drudy D, Mullane NR, Quinn T, Wall PG, Fanning S. Enterobacter sakazakii: an emerging pathogen in
powdered infant formula. Clin Infect Dis. 2006; 42:996–1002.
4. Jaradat ZW, Al Mousa W, Elbetieha A, Al Nabulsi A, Tall BD. Cronobacter spp.—opportunistic food-
borne pathogens. A review of their virulence and environmental-adaptive traits. J Med Microbiol. 2014;
63:1023–1037.
5. Hunter CJ, Bean JF. Cronobacter: an emerging opportunistic pathogen associated with neonatal
meningitis, sepsis and necrotizing enterocolitis. J Perinatol. 2013; 33:581–585.
6. Caubilla-Barron J, et al. Genotypic and phenotypic analysis of Enterobacter sakazakii strains from
an outbreak resulting in fatalities in a neonatal intensive care unit in France. J Clin Microbiol. 2007;
45:3979–3985.
7. Feeney A, Kropp KA, O’Connor R, Sleator RD. Cronobacter sakazakii: stress survival and virulence
potential in an opportunistic foodborne pathogen. Gut Microbes. 2014; 5:711–718.
8. Iversen C, Forsythe SJ. Comparison of media for the isolation of Enterobacter sakazakii. Appl Environ
Microbiol. 2007; 73:48–52.
9. Gičová A, Oriešková M, Oslanecová L, Drahovská H, Kaclíková E. Identification and characterization
of Cronobacter strains isolated from powdered infant foods. Lett Appl Microbiol. 2014; 58:242–247.
10. Yan QQ, Condell O, Power K, Butler F, Tall BD, Fanning S. Cronobacter species (formerly known
as Enterobacter sakazakii) in powdered infant formula: a review of our current understanding of the
biology of this bacterium. J Appl Microbiol. 2012; 113:1–15
11. O’Brien S, Healy B, Negredo C, Fanning S, Iversen C. Evaluation of a new one-step enrichment in
conjunction with a chromogenic medium for the detection of Cronobacter spp. (Enterobacter sakazakii)
in powdered infant formula. J Food Prot. 2009; 72:1472–1475.
12. Hassan AA, Akineden O, Kress C, Estuningsih S, Schneider E, Usleber E. Characterization of the gene
encoding the 16S rRNA of Enterobacter sakazakii and development of a species-specific PCR method.
Int J Food Microbiol. 2007; 116:214–220.
13. Lehner A, Tasara T, Stephan R. 16S rRNA gene based analysis of Enterobacter sakazakii strains from
different sources and development of a PCR assay for identification. BMC Microbiol. 2004; 4:43.
14. Stoop B, Lehner A, Iversen C, Fanning S, Stephan R. Development and evaluation of rpoB based PCR
systems to differentiate the six proposed species within the genus Cronobacter. Int J Food Microbiol.
2009; 136:165–168.
15. Zimmermann J, Schmidt H, Loessner MJ, Weiss A. Development of a rapid detection system for oppor-
tunistic pathogenic Cronobacter spp. in powdered milk products. Food Microbiol. 2014; 42:19–25.
16. Mohan Nair MK, Venkitanarayanan KS. Cloning and sequencing of the ompA gene of Enterobacter
sakazakii and development of an ompA-targeted PCR for rapid detection of Enterobacter sakazakii in
infant formula. Appl Environ Microbiol. 2006; 72:2539–2546.
17. Mullane N, et al. Molecular analysis of the Enterobacter sakazakii O-antigen gene locus. Appl Environ
Microbiol. 2008; 74:3783–3894.
18. Healy B, et al. Microarray-based comparative genomic indexing of the Cronobacter genus (Enterobacter
sakazakii). Int J Food Microbiol. 2009; 136:159–164.
19. Schmid M, et al. Evidence for a plant-associated natural habitat for Cronobacter spp. Res Microbiol.
2009; 160:608–614.
20. Townsend S, Hurrell E, Forsythe S. Virulence studies of Enterobacter sakazakii isolates associated with
a neonatal intensive care unit outbreak. BMC Microbiol. 2008; 18;8:64.
21. Kucerova E, et al. Genome sequence of Cronobacter sakazakii BAA-894 and comparative genomic
hybridization analysis with other Cronobacter species. PLoS One. 2010; 5:e9556.
22. Stephan R, Lehner A, Tischler P, Rattei T. Complete genome sequence of Cronobacter turicensis LMG
23827, a food-borne pathogen causing deaths in neonates. J Bacteriol. 2011; 193:309–310.
23. Mange JP, et al. Adhesive properties of Enterobacter sakazakii to human epithelial and brain microvas-
cular endothelial cells. BMC Microbiol. 2006; 6:58.
Cronobacter: Virulence and Pathogenesis 315
24. Pagotto FJ, Nazarowec-White M, Bidawid S, Farber JM. Enterobacter sakazakii: infectivity and entero-
toxin production in vitro and in vivo. J Food Prot. 2003; 66:370–375.
25. Hunter CJ, Petrosyan M, Ford HR, Prasadarao NV. Enterobacter sakazakii: an emerging pathogen in
infants and neonates. Surg Infect (Larchmt). 2008; 9:533–539.
26. Hunter CJ, et al. Enterobacter sakazakii enhances epithelial cell injury by inducing apoptosis in a rat
model of necrotizing enterocolitis. J Infect Dis. 2008; 198:586–593.
27. Hunter CJ, Chokshi N, Ford HR. Evidence vs experience in the surgical management of necrotizing
enterocolitis and focal intestinal perforation. J Perinatol. 2008; 28(Suppl 1):S14–S17.
28. Hunter CJ, et al. Lactobacillus bulgaricus prevents intestinal epithelial cell injury caused by Enterobacter
sakazakii-induced nitric oxide both in vitro and in the newborn rat model of necrotizing enterocolitis.
Infect Immun. 2009; 77:1031–1043.
29. Liu Q, Mittal R, Emami CN, Iversen C, Ford HR, Prasadarao NV. Human isolates of Cronobacter
sakazakii bind efficiently to intestinal epithelial cells in vitro to induce monolayer permeability and
apoptosis. J Surg Res. 2012; 176:437–447.
30. Krishnan S, Prasadarao NV. Outer membrane protein A and OprF: versatile roles in Gram-negative
bacterial infections. FEBS J. 2012; 279:919–931.
31. Smith SG, Mahon V, Lambert MA, Fagan RP. A molecular Swiss army knife: OmpA structure, function
and expression. FEMS Microbiol Lett. 2007; 273:1–11.
32. Kim K, et al. Outer membrane proteins A (OmpA) and X (OmpX) are essential for basolateral invasion
of Cronobacter sakazakii. Appl Environ Microbiol. 2010; 76:5188–5198.
33. Mohan Nair MK, Venkitanarayanan K. Role of bacterial OmpA and host cytoskeleton in the invasion of
human intestinal epithelial cells by Enterobacter sakazakii. Pediatr Res. 2007; 62:664–669.
34. García-del Portillo F, Finlay BB. Invasion and intracellular proliferation of Salmonella within non-
phagocytic cells. Microbiologia. 1994; 10:229–238.
35. Sansonetti PJ. Genetic and molecular basis of epithelial cell invasion by Shigella species. Rev Infect Dis.
1991; 13(Suppl 4):S285–S292.
36. Kim KP, Loessner MJ. Enterobacter sakazakii invasion in human intestinal Caco-2 cells requires the
host cell cytoskeleton and is enhanced by disruption of tight junction. Infect Immun. 2008; 76:562–570.
37. Nair MK, Venkitanarayanan K, Silbart LK, Kim KS. Outer membrane protein A (OmpA) of Cronobacter
sakazakii binds fibronectin and contributes to invasion of human brain microvascular endothelial cells.
Foodborne Pathog Dis. 2009; 6:495–501.
38. Emami CN, Mittal R, Wang L, Ford HR, Prasadarao NV. Recruitment of dendritic cells is respon-
sible for intestinal epithelial damage in the pathogenesis of necrotizing enterocolitis by Cronobacter
sakazakii. J Immunol. 2011; 186:7067–7079.
39. Quintero M, et al. Adherence inhibition of Cronobacter sakazakii to intestinal epithelial cells by
prebiotic oligosaccharides. Curr Microbiol. 2011; 62:1448–1454.
40. Rossi M, Young JW. Human dendritic cells: potent antigen-presenting cells at the crossroads of innate
and adaptive immunity. J Immunol. 2005; 175:1373–1381.
41. Emami CN, Mittal R, Wang L, Ford HR, Prasadarao NV. Role of neutrophils and macrophages in the
pathogenesis of necrotizing enterocolitis caused by Cronobacter sakazakii. J Surg Res. 2012; 172:18–28.
42. Mittal R, Bulgheresi S, Emami C, Prasadarao NV. Enterobacter sakazakii targets DC-SIGN to
induce immunosuppressive responses in dendritic cells by modulating MAPKs. J Immunol. 2009;
183:6588–6599.
43. Cruz-Córdova A, et al. Flagella from five Cronobacter species induce pro-inflammatory cytokines in
macrophage derivatives from human monocytes. PLoS One. 2012; 7:e52091.
44. Bernardo WM, Aires FT, Carneiro RM, Sá FP, Rullo VE, Burns DA. Effectiveness of probiotics in the
prophylaxis of necrotizing enterocolitis in preterm neonates: a systematic review and meta-analysis.
J Pediatr (Rio J). 2013; 89:18–24.
45. Weng M, Ganguli K, Zhu W, Shi HN, Walker WA. Conditioned medium from Bifidobacteria infantis
protects against Cronobacter sakazakii-induced intestinal inflammation in newborn mice. Am J Physiol
Gastrointest Liver Physiol. 2014; 306:G779–G787.
46. Schwizer S, Tasara T, Zurfluh K, Stephan R, Lehner A. Identification of genes involved in serum
tolerance in the clinical strain Cronobacter sakazakii ES5. BMC Microbiol. 2013; 13:38.
47. Joseph S, Forsythe SJ. Predominance of Cronobacter sakazakii sequence type 4 in neonatal infections.
Emerg Infect Dis. 2011; 17:1713–1715.
316 Laboratory Models for Foodborne Infections
48. Hariri S, Joseph S, Forsythe SJ. Cronobacter sakazakii ST4 strains and neonatal meningitis, United
States. Emerg Infect Dis. 2013; 19:175–177.
49. Stoll BJ, et al. Enterobacter sakazakii invades brain capillary endothelial cells, persists in human
macrophage influencing cytokine secretion and induces severe brain pathology in the neonatal rat.
Microbiology. 2007; 153:3538–3547.
50. Mittal R, Wang Y, Hunter CJ, Gonzalez-Gomez I, Prasadarao NV. Brain damage in newborn rat model
of meningitis by Enterobacter sakazakii: a role for outer membrane protein A. Lab Invest. 2009;
89:263–277.
51. Franco AA, et al. Cpa, the outer membrane protease of Cronobacter sakazakii, activates plasminogen
and mediates resistance to serum bactericidal activity. Infect Immun. 2011; 79:1578–1587.
52. Singamsetty VK, Wang Y, Shimada H, Prasadarao NV. Outer membrane protein A expression in
Enterobacter sakazakii is required to induce microtubule condensation in human brain microvascular
endothelial cells for invasion. Microb Pathog. 2008; 45:181–191.
53. Li Q, et al. PI3K-dependent host cell actin rearrangements are required for Cronobacter sakazakii
invasion of human brain microvascular endothelial cells. Med Microbiol Immunol. 2010; 199:333–340.
54. Hurrell E, Kucerova E, Loughlin M, Caubilla-Barron J, Forsythe SJ. Biofilm formation on enteral
feeding tubes by Cronobacter sakazakii, Salmonella serovars and other Enterobacteriaceae. Int J Food
Microbiol. 2009; 136:227–231.
55. Jung JH, Choi NY, Lee SY. Biofilm formation and exopolysaccharide (EPS) production by Cronobacter
sakazakii depending on environmental conditions. Food Microbiol. 2013; 34:70–80.
56. Wang L, Hu X, Tao G, Wang X. Outer membrane defect and stronger biofilm formation caused by
inactivation of a gene encoding for heptosyltransferase I in Cronobacter sakazakii ATCC BAA-894.
J Appl Microbiol. 2012; 112:985–997.
57. Hartmann I, Carranza P, Lehner A, Stephan R, Eberl L, Riedel K. Genes involved in Cronobacter
sakazakii biofilm formation. Appl Environ Microbiol. 2010; 76:2251–2261.
58. Choi Y, Kim S, Hwang H, Kim KP, Kang DH, Ryu S. Plasmid-encoded MCP is involved in viru-
lence, motility, and biofilm formation of Cronobacter sakazakii ATCC 29544. Infect Immun. 2015;
83:197–204.
59. Fehr A, Eshwar AK, Neuhauss SC, Ruetten M, Lehner A, Vaughan L. Evaluation of zebrafish as a
model to study the pathogenesis of the opportunistic pathogen Cronobacter turicensis. Emerg Microbes
Infect. 2015; 4:e29.
60. Sivamaruthi BS, Ganguli A, Kumar M, Bhaviya S, Pandian SK, Balamurugan K. Caenorhabditis elegans
as a model for studying Cronobacter sakazakii ATCC BAA-894 pathogenesis. J Basic Microbiol. 2011;
51: 540–549.
61. Sivamaruthi BS, Prasanth MI, Balamurugan K. Alterations in Caenorhabditis elegans and Cronobacter
sakazakii lipopolysaccharide during interaction. Arch Microbiol. 2015; 197:327–337.
21
Escherichia
Dongyou Liu
CONTENTS
21.1 Introduction....................................................................................................................................317
21.1.1 Classification, Morphology, and Genomics......................................................................318
21.1.1.1 Classification....................................................................................................318
21.1.1.2 Morphology......................................................................................................318
21.1.1.3 Genomics........................................................................................................ 320
21.1.2 Biology and Epidemiology................................................................................................321
21.1.3 Clinical Features and Pathogenesis...................................................................................321
21.1.3.1 ETEC...............................................................................................................321
21.1.3.2 EPEC............................................................................................................... 322
21.1.3.3 EHEC.............................................................................................................. 322
21.1.3.4 EAEC.............................................................................................................. 322
21.1.3.5 STEAEC......................................................................................................... 323
21.1.3.6 EIEC................................................................................................................ 323
21.1.3.7 DAEC.............................................................................................................. 323
21.1.3.8 AIEC............................................................................................................... 323
21.1.3.9 SCEC............................................................................................................... 323
21.1.3.10 NMEC............................................................................................................. 323
21.1.3.11 UPEC.............................................................................................................. 324
21.1.4 Diagnosis.......................................................................................................................... 324
21.1.5 Treatment and Prevention................................................................................................. 324
21.2 Laboratory Models........................................................................................................................ 325
21.2.1 Animal Models................................................................................................................. 325
21.2.1.1 Rodents........................................................................................................... 325
21.2.1.2 Rabbits............................................................................................................ 325
21.2.1.3 Pigs.................................................................................................................. 326
21.2.1.4 Caenorhabditis elegans Nematode................................................................ 326
21.2.2 In Vitro Models................................................................................................................. 326
21.3 Conclusion..................................................................................................................................... 327
References............................................................................................................................................... 327
21.1 Introduction
The genus Escherichia comprises a small group of Gram-negative, rod-shaped bacterial species that
form a part of gut microbiota in mammalian hosts including humans. As part of the well-known and yet
still incompletely understood Escherichia species, Escherichia coli (represented by O157:H4 strain) is an
important cause of intestinal (enteritis, diarrhea, and dysentery), or extraintestinal diseases (urinary tract
infection, intra-abdominal infection, pneumonia, neonatal meningitis, and sepsis). By first presenting a
brief overview on E. coli classification, biology, epidemiology, clinical features, diagnosis, treatment,
317
318 Laboratory Models for Foodborne Infections
and prevention, this chapter then discusses laboratory models that have been applied to the study of
E. coli, with the goal of unlocking the secrets of its pathogenic mechanisms and aiding in the develop-
ment of improved anti-infection strategies.
21.1.1.2 Morphology
E. coli is a rod-shaped bacterium of about 0.6 μm in diameter and 2 μm in length. The bacterium forms
nonspreading black colonies with a characteristic greenish-black metallic sheen on eosin methylene blue
(EMB) agar, and deep red colonies on MacConkey agar. Other morphological features of note include:
Escherichia
TABLE 21.1
Characteristics of Major Diarrheagenic E. coli Pathotypes
Pathotype Serogroupa Human Disease Other Features
Enterotoxigenic (ETEC) O6, O8, O15, O20, O25, O27, O63, Watery diarrhea (without Colonizing the small intestine, ETEC employs fimbrial adhesins to bind
O78, O80, O85, O115, O128ac, fever) in children enterocytes and produces two proteinaceous enterotoxins: heat-labile LT
O139, O148, O153, O159, O167 <5 years old, and enterotoxin (with similarity to cholera toxin in structure and function) and
travelers’ diarrhea heat-stable ST enterotoxin (inducing cGMP accumulation in the target cells
and a subsequent secretion of fluid and electrolytes into the intestinal lumen).
ETEC strains are noninvasive and do not leave the small intestinal lumen.
Enteropathogenic (EPEC) O26, O55, O86, O111, O114, O119, Profuse watery diarrhea Colonizing the small intestine, EPEC uses an adhesin known as intimin to
O125, O126, O127, O128, O142, O158 in infants bind host intestinal cells, leading to rearrangement of actin, A/E lesion, and
subsequent diarrhea. Being moderately invasive (with the ability to enter
host cells), EPEC produces a number of virulence factors that are similar to
those found in Shigella and that can elicit an inflammatory response.
Enterohemorrhagic (EHEC) O26, O91, O111, O157 among others Watery diarrhea, Colonizing the distal ileum and colon, EHEC induces A/E lesion and employs
hemorrhagic colitis/ fimbriae for attachment (E. coli common pilus, ECP). EHEC produces a
bloody diarrhea, HUS phage-encoded Shiga toxin that elicits an intense inflammatory response.
Being moderately invasive, EHEC (particularly O157:H7 strain) causes
more serious diarrhea (bloody diarrhea without fever) than EPEC, in
addition to HUS and sudden kidney failure.
Enteroaggregative (EAEC) O3, O15, O44, O77, O104, O126 Travelers’ diarrhea, HUS Colonizing the small intestine and/or colon, EAEC (whose fimbriae aggregate
(stx+), and persistent tissue culture cells) produces a hemolysin and an ST enterotoxin similar to
diarrhea (infants) that of ETEC. EAEC is noninvasive and binds to the intestinal mucosa,
leading to watery diarrhea without fever.
Shiga-toxin-producing O104 Food poisoning STEAEC is an EAEC strain (O104:H4) showing typical EHEC phenotypes
enteroaggregative (STEAEC) (Stx production and strong cell adherence) that causes HUS.
Enteroinvasive (EIEC) O28ac, O29, O112ac, O124, O136, Shigellosis/bacillary Colonizing the colon, EIEC causes a syndrome with profuse diarrhea and
O143, O144, O152, O164, O167 dysentery high fever, which is identical to shigellosis.
Diffusely adherent (DAEC) O86, O127, O142, O158 Persistent watery diarrhea Colonizing the intestine, DAEC shows a distinct diffuse adherence to epithelial
(<5 years old) cells (HEp-2 or HeLa). Isolated from children, DAEC harbors Afa/Dr genes.
Some serogroups are associated with more than one diarrheagenic E. coli pathotype. For example, serogroup O86 is mainly considered as EPEC, but it has also been implicated in ETEC,
a
319
320 Laboratory Models for Foodborne Infections
1. Fimbriae (pili), which are of two kinds, common or conjugative. Common fimbriae (about
100–1000 per cell) comprise mainly an acidic hydrophobic protein called fimbrin and may
be divided into seven groups according to the amino acid sequence of their major fimbrin.
Conjugative fimbriae (also known as sex pili, usually number one or a few copies per cell)
facilitate contact between the donor and recipient bacteria, permitting transfer of DNA during
conjugation. Fimbriae are highly antigenic and contain many F antigens.
2. Flagella, which are responsible for bacterial mobility. E. coli flagella (about 5–10 copies per
cell; 5–10 μm in length) are composed of a long filament, a hook, and a basal body and are
arranged randomly around the cell surface (a pattern known as peritrichous flagellation). The
principal component of E. coli flagella is an N-methyl-lysine-rich protein (55 kDa in size)
known as flagellin, and around 20,000 subunits of flagellin are needed to make the flagellar
filament. Flagella are highly antigenic and include a large number of H antigens.
3. Capsule and outer membrane. The outer membrane of E. coli is composed of a lipid bilayer,
which in turn consists of a phospholipid inner leaflet and an LPS outer leaflet, together with
several kinds of membrane proteins in the periplasm (the space between the inner and outer
leaflets/membranes). In the outer membrane, a glycolipid (called the enterobacterial common
antigen or ECA) is found in E. coli. In some E. coli strains, the outer membrane is covered
by a polysaccharide capsule containing K antigens. Under conditions of high osmolarity, low
temperature, and low humidity, other polysaccharides such as M antigens (colanic acids, which
are polymers of glucose, galactose, fucose, and galacturonic acid) are synthesized.
4. Periplasm and cell wall. The periplasm of E. coli is osmotically active and contains over 60
known proteins, including binding proteins for amino acids, sugars, vitamins, and ions; deg-
radative enzymes (phosphatases, proteases, and endonucleases); and antibiotic detoxifying
enzymes (β-lactamases, alkyl sulfodehydrases, and aminoglycoside phosphorylating enzymes).
The cell wall of E. coli is responsible for cell shape and rigidity and is composed of a pepti-
doglycan layer (of one or a few molecules thick) that is anchored to the outer membrane via
covalent links to the major membrane lipoprotein and noncovalent links to porins.
5. Cytoplasmic membrane. The cytoplasmic membrane of E. coli is made up of about 200 distinct
proteins (including those involved in peptidoglycan biosynthesis, cell wall elongation, and cell
division, as well as penicillin-binding proteins that covalently bind β-lactam antibiotics) and
four kinds of phospholipids.
6. Cytoplasm. The cytoplasm of E. coli contains ribosomes and other organelles in which biologi-
cal/biochemical activities necessary for growth are carried out. These activities include meta-
bolic fueling (production of energy, reducing power, and precursor metabolites), biosynthesis
of building blocks, polymerization into macromolecules, and assembly of cell structures [1].
21.1.1.3 Genomics
The genome of E. coli strain MG1655 (a laboratory strain K-12 derivative) consists of a circular DNA
molecule 4.64 Mb in length, with 4288 protein-coding genes (organized into 2584 operons), 7 rRNA
operons, and 86 tRNA genes, together with a number of transposable genetic elements, repeat ele-
ments, cryptic prophages, and bacteriophage remnants. Some pathogenic E. coli strains possess larger
genomes than that of the commensal K-12 strain, such as enterohemorrhagic E. coli strain O157:H7
Sakai (5.50 Mb); enteroaggregative E. coli strain O42 (5.36 Mb); and UPEC isolates CFT073 (5.23 Mb),
536 (4.94 Mb), and UTI89 (5.07 Mb).
While many strains (e.g., strain MG1655) do not have plasmid, others possess one to five plasmids,
which along with chromosomal pathogenicity islands contribute to the plasticity of the E. coli genome.
It is notable that all major diarrheagenic E. coli pathotypes carry at least one virulence-related property
on a plasmid. The plasmid is typically large (>60 megadalton or MDa), of low copy number, of either
conjugative or transmissible incompatibility group, and encodes multiple virulence factors. In addition
to plasmid-encoded clusters of virulence traits and chromosomal pathogenicity islands, transposon-
encoded individual traits (e.g., Shiga toxin) may be also present [7,8].
Escherichia 321
21.1.3.1 ETEC
ETEC is usually acquired from food or water contaminated with human or animal feces. This pathotype
causes watery diarrhea of varied severity (ranging from mild, self-limiting disease to severe cholera-like,
life-threatening illness). With a sudden onset of watery stool (without blood or inflammatory cells) and
vomiting, ETEC infection may lead to dry mouth, rapid pulse, lethargy, decreased skin turgor, decreased
blood pressure, muscle cramps, and shock due to progressive loss of fluids (dehydration) and electrolytes
322 Laboratory Models for Foodborne Infections
(sodium, potassium, chloride, and bicarbonate). The diarrhea is self-limited and generally lasts for only
3–4 days. If hydration is maintained, the patient may recover without any sequelae. ETEC is a major
cause of diarrhea in infants younger than 2 years of age in the developing world, and it is also associated
with travelers’ diarrhea [12].
21.1.3.2 EPEC
EPEC is an attaching and effacing (A/E) pathogen that induces a characteristic A/E lesion (or pedestal-
like structure) on the lumenal surfaces of host small intestine, without the production of Shiga toxins.
In addition to watery diarrhea containing mucus but not blood, other symptoms associated with EPEC
infection include vomiting, fever, malaise, and dehydration. While these symptoms often persist for
several days, chronic EPEC infection is sometimes observed. Occasionally, EPEC may cause bloody
diarrhea after the colonization of the mid-distal small intestine (ileum). EPEC is separated into two
groups according to the presence/absence of EPEC adherence factor plasmid (or pEAF): typical EPEC
(tEPEC) and atypical EPEC (aEPEC). tEPEC possesses pEAF, forms localized adherence pattern,
and includes classical O26, O55, O86, O111, O119, O125, O126, O127, O128ac, O142, O158, whereas
aEPEC lacks pEAF, but harbors other virulence factors and may form diffuse or aggregative adherence
patterns. EPEC is a common cause of watery diarrhea among infants in the developing world and is
also implicated in sporadic diarrheal outbreaks among infants (e.g., day-care facilities) in the developed
world [13].
21.1.3.3 EHEC
EHEC is characterized by the production of Shiga toxins (Stx1 and/or Stx2, leading to its alternative
name of STEC) and the formation of A/E lesion, notably in the cecum and ascending colon [14–16].
EHEC typically causes an afebrile bloody colitis (bloody stools with ulcerations of the bowel) known as
hemorrhagic colitis (HC), which is characterized by the sudden onset of abdominal pain, severe cramps,
and diarrhea within 24 h. In about 10% of patients (e.g., children and the elderly), infection with EHEC
O157 may result in HUS, which is defined by acute renal failure, hemolytic anemia, and thrombocytope-
nia. Colonoscopy examination reveals the presence of edema, erythema (redness), hemorrhage, erosion,
and, occasionally, a long ulcer-like lesion, with a marked narrowing of the luminal space. Histologic
examination indicates destruction of the surface epithelium, neutrophil infiltration of the lamina propria,
and the formation of crypt abscesses. In patients (especially children of <5 years of age and the elderly)
with diarrhea and HUS, renal pathology consists of endothelial swelling and glomerular thrombosis
with congested rather than ischemic glomeruli. The most prevalent EHEC serotype causing outbreaks in
North America and other parts of the world is O157:H7 [17,18]. Besides Shiga toxin, which may account
for the severe complications including HUS, O157:H7 expresses several other virulence factors including
intimin, translocated intimin receptor (Tir), a T3SS, and enterohemolysin [19]. The genes encoding many
of these factors are located on a 44 kb pathogenicity island (also known as the locus of enterocyte efface-
ment or the LEE locus) [20–22]. Cattle act as a primary reservoir for EHEC, although vegetables (let-
tuce, spinach, and sprouts) and fruits may also serve as vehicles for EHEC outbreaks [23].
21.1.3.4 EAEC
EAEC is noted for forming an AA pattern (which is characterized by prominent, “stacked brick” auto-
agglutination of the bacterial cells to each other) during its adhesion to HEp-2 cells [24,25]. EAEC
infection manifests clinically as watery diarrhea with or without blood and mucus, abdominal pain,
nausea, vomiting, and low-grade fever. Since 1980s, EAEC has been recognized as a causative agent of
persistent diarrhea in malnourished children in the developing world, and it also causes both outbreaks
and sporadic diarrhea among travelers and immunocompromised individuals (specifically HIV-infected
patients) in the developed world. Individuals with single-nucleotide polymorphisms (SNPs) in the IL-8
gene promoter and lactoferrin gene may show higher susceptibility to EAEC infection [26]. EAEC is
Escherichia 323
primarily transmitted through contaminated food and water. Food handlers (especially those working in
tourist hotels) are important carriers of EAEC. Interestingly, EAEC-induced travelers’ diarrhea occurs
constantly throughout winter and summer, whereas other diarrheagenic E. coli strains such as ETEC,
EPEC, and EIEC show lower rates of infection in winter [27].
21.1.3.5 STEAEC
STEAEC is an EAEC strain (O104:H4) that has acquired typical EHEC phenotypes such as Stx pro-
duction and strong cell adherence. STEAEC O104:H4 is responsible for HUS in a high percentage of
patients, with a mortality rate of 1%.
21.1.3.6 EIEC
EIEC is pathogenetically, biochemically, and genetically related to Shigella spp. and principally resides in
the large intestine. EIEC causes an invasive inflammatory colitis with a watery diarrhea syndrome that is
indistinguishable from those caused by other E. coli pathotypes and Shigella spp. In severe cases, scanty
dysenteric stools may contain blood and mucus, and this may occur together with fever and severe cramps.
21.1.3.7 DAEC
DAEC is differentiated from other diarrheic E. coli by its diffuse adherence to epithelial cells in the clas-
sical laboratory assay of adherence to HEp-2 or HeLa cells. This diffuse adherence pattern is essentially
linked to the production of adhesins encoded by a family of afa/dra/daa-related operons. DAEC causes
a watery diarrhea in adults and children (with increased risk in those aged 18 months to 5 years) [28,29].
21.1.3.8 AIEC
AIEC adheres to intestinal epithelial cells through type 1 pili (especially FimH adhesin variant) that
interacts with host glycoprotein CEACAM6 in a mannose-associated manner, gains access to the lam-
ina propria, and replicates in macrophages (which represent an environment with acidic pH, oxidative
stress, active proteolytic enzymes, and antimicrobial compounds) [30]. Transient AIEC colonization
elicits intestinal inflammation and alters microbiota composition leading to Crohn’s disease (CD), with
symptoms ranging from fever, fatigue, abdominal pain, cramping, nausea, vomiting, bloody stool, mouth
sores, reduced appetite, weight loss, and perianal disease to diarrhea. Host deficiencies in CD patients
linked with the increased ability of AIEC LF82 to cause infection include the overexpression of the
CEACAM6 and Gp96 receptors in the apical membrane of intestinal epithelial cells (which facilitates
AIEC adhesion and invasion), defects in autophagy related to NOD2, ATG16L1, and IRGM function
and expression (which impair the ability of host cells to resolve infections), altered bile salts metabolism
(which enhances the expression of long polar fimbriae in AIEC, permitting better translocation via
M cells), and decreased levels of protease meprin (which degrades type 1 pili) [31].
21.1.3.9 SCEC
SCEC is associated with sudden high fever with chills, nausea, vomiting, diarrhea, abdominal pain,
hypotension, confusion, anxiety, tachypnea (short of breath), tachycardia (rapid heart rate), and uremia.
21.1.3.10 NMEC
NMEC is responsible for fever, failure to thrive, neurologic signs, jaundice, decreased feeding, periods of
apnea, and listlessness in neonates. Infants of <1 month of age may also show irritability, lethargy, vomiting,
lack of appetite, and seizures, while those >4 months of age display neck rigidity, tense fontanels, and fever.
Older children and adults tend to present with headache, vomiting, confusion, lethargy, seizures, and fever.
324 Laboratory Models for Foodborne Infections
21.1.3.11 UPEC
UPEC utilizes type 1 fimbriae (particularly FimH) to interact with urinary tract host epithelia and is
among the most prevalent extraintestinal bacteria accounting for 90% of all UTIs, including both cystitis
and pyelonephritis. This pathotype has evolved a multitude of virulence factors and strategies to facili-
tate its growth and persistence within the adverse settings of the host urinary tract [32,33].
21.1.4 Diagnosis
E. coli grown on EMB agar produces black colonies with a diagnostic greenish-black metallic sheen. In
addition, being lactose positive, E. coli generates deep red colonies on MacConkey agar, as fermentation
of lactose decreases the medium’s pH and darkens the medium. Other biochemical features of E. coli
include the ability to reduce nitrates to nitrites and to generate succinate, ethanol, acetate, and carbon
dioxide. While most E. coli strains are positive for catalase, they are negative for oxidase, citrate, urease,
and hydrogen sulfide. Further, E. coli is positive for indole production and the methyl red test. Given
that about 98% of E. coli strains are positive in the indole test, it offers a useful approach to differentiate
E. coli from other members of the family Enterobacteriaceae.
Use of epithelial cell lines (e.g., HEp-2 and HeLa) provides another means to characterize E. coli
pathotypes. For instance, HEp-2 cell-adherence assay enables differentiation among EPEC, EAEC,
and DAEC isolates, and HeLa cell monolayers facilitate identification of plaque-forming EIEC isolates.
Further, Y1 adrenal and Chinese hamster ovarian cells may display morphological changes in the pres-
ence of heat-labile enterotoxin (LT)-producing E. coli. Infant mouse physiological assay is applicable
for heat-stable enterotoxin (ST) identification. Guinea pig may be used for detecting keratoconjunctivitis
caused by EIEC isolates. Moreover, Stx-producing E. coli may be verified by mammalian cell cultures,
which are rapidly killed by the Stx.
By targeting the O (lipopolysaccharide), H (flagellar), K (capsular), and F (fimbrial) antigens, sero-
logical procedures (e.g., mannose-resistant agglutination of erythrocytes, passive latex agglutination,
enzyme-linked immunosorbent assay, immunoprecipitation in agar, and Biken test) employing poly-
clonal or monoclonal antibodies offer a more specific way to define E. coli strains and pathotypes.
Recent development of nucleic-acid-based techniques makes rapid, sensitive, and specific identi-
fication and typing of E. coli strains and pathotypes possible. These new-generation methods detect
E. coli genes that encode toxins, pili, and other virulence factors. Indeed, polymerase chain reaction
(PCR) procedures targeting the pap, afa, and sfa genes have proven valuable for UPEC identification.
Application of multiplex and real-time PCR allows for simultaneous detection of several diarrheagenic
E. coli pathotypes [34–38].
vaccines will contribute to the control and prevention of E. coli infection. In this regard, an E. coli
O157:H7 O-specific polysaccharide conjugated to recombinant exotoxin A of Pseudomonas aerugi-
nosa (O157-rEPA) has been shown to elicit a mitigating immune response in both adults and children
2–5 years of age [40].
21.2 Laboratory Models
In order to uncover the virulence mechanisms of pathogenic E. coli, understand bacteria–host cell inter-
actions, determine colonization and attachment factors, investigate host immune response, and evaluate
candidate vaccines, use of laboratory models (both in vivo and in vitro) is critical [41–43].
21.2.1 Animal Models
A variety of animals have been utilized as models for investigating E. coli infection and disease. These
range from mice, rats, rabbits, chickens, pigs, cows, and dogs to baboons and macaques. For example,
gnotobiotic piglets, infant rabbits, calves, chickens, and macaques have proven valuable for assessing
characteristic A/E lesions associated with E. coli O157:H7. Greyhounds and rabbits are useful for exam-
ining naturally occurring HUS-like diseases [i.e., idiopathic cutaneous and renal glomerular vasculopa-
thy of greyhounds (CRVGs) or Alabama rot] [21,44].
21.2.1.1 Rodents
Mice do not show signs of intestinal disease following oral infection with EHEC, although they do
develop kidney damage (acute tubular necrosis instead of prothrombotic condition seen in humans) and
subsequently die [45]. Pretreatment with antibiotics (e.g., streptomycin), use of germfree animals, or
dietary-induced changes (e.g., protein–calorie malnutrition) appears to reduce the natural resistance of
mice to EHEC colonization [46,47]. Another useful mouse model is the intact commensal flora (ICF)
model for which mice harboring normal flora are utilized. ICF mice infected with O157:H7 enable moni-
toring of both colonization and disease (e.g., ruffled fur, lethargy, weight loss, renal tubular damage, and
mortality) [21,48]. A natural mouse pathogen Citrobacter rodentium (which carries a homolog of the
LEE locus of EPEC and EHEC and has the capacity to evoke A/E lesions) may be exploited as a sur-
rogate for E. coli O157:H7 for evaluation of the virulence mechanisms of EHEC. A mouse intoxication
model involving parenteral injection of Stx facilitates in vivo can be used for assessment of the effects of
Shiga toxin (especially Stx2), which are responsible for damage to renal cortical tubule epithelial cells,
paralysis, and mortality (i.e., symptoms associated with HC and HUS) [21].
A neonatal rat model provides a useful method to evaluate systemic infection due to neuropathogenic
E. coli, as colonization of the gastrointestinal tract leads to dissemination of the pathogen along the gut-
lymph-blood-brain course of infection. Similar to human host, neonatal rat shows local inflammation
upon E. coli penetration of the central nervous system [49]. Additionally, use of a rat model of unilateral
E. coli epididymitis reveals that despite the eradication of the pathogen by antimicrobial agents accom-
panied by the reduction of epididymal damage, inflammation in the contralateral epididymis is neverthe-
less present, which may contribute to impaired fertility [50]. On the whole, rodent models are relatively
inexpensive and easy to care for and handle.
21.2.1.2 Rabbits
Suckling New Zealand White (NZW) rabbits are susceptible to oral infection with E. coli O157:H7, lead-
ing to watery diarrhea in young (5–10 days old) but not older (20 days old) rabbits. Histological abnor-
malities (e.g., edema, hemorrhage, the presence of an inflammatory infiltrate, and mucosal epithelial
apoptosis) are observed in the colon. However, suckling rabbits do not develop signs of renal disease [51].
These data suggest that suckling rabbits may be a useful model for studying the intestinal manifestations
of EHEC infection. Nevertheless, older rabbits appear to be recalcitrant to colonization by EHEC [52].
326 Laboratory Models for Foodborne Infections
21.2.1.3 Pigs
The intestine of gnotobiotic piglets [i.e., cesarean-derived colostrum-deprived (CDCD) piglets] is per-
missive for EHEC replication, as oral infection of CDCD piglets with 1010 CFU of E. coli O157:H7
leads to watery diarrhea without blood by 2–4 days postinoculation. Histologic abnormalities include
destruction of the mucosal brush border and inflammation (so called A/E lesions) in the cecum and
colon. Apart from intestinal disease, gnotobiotic piglets also exhibit signs of central nervous system
disease (e.g., vascular damage in the cerebellum, ataxia) and renal lesion upon oral or intraperitoneal
administration of O157:H7. The kidneys of infected gnotobiotic piglets show signs of diffuse glomeru-
lar endothelial swelling and congestion as well as a narrowing of the capillary vessels. In addition,
morphological signs of thrombotic microangiopathy, which is characteristic of HUS in humans, are
also noticeable. Naturally born piglets are found to exhibit more severe neurological disease and more
quickly succumb to EHEC infection than traditional CDCD animals, although no signs of kidney dam-
age are found. Nonetheless, maintenance of gnotobiotic piglets requires considerable veterinary skill,
space, and expense [21].
21.3 Conclusion
Constituting a key member within the genus Escherichia, E. coli is a remarkable bacterium that encom-
passes a large number of strains of varied pathogenic potential. Although many E. coli strains are com-
mon symbiotic organisms in the gut, some demonstrate propensity to cause intestinal and extraintestinal
diseases in humans. To date, 10 pathotypes have been identified among E. coli strains involved in intes-
tinal diseases, including ETEC, EPEC, EHEC, EAEC, STEAEC, EIEC, DAEC, CDEC, NTEC, and
AIEC [53], whereas three pathotypes have been recognized among E. coli strains causing extraintestinal
diseases, i.e., SCEC, NMEC, and UPEC. Extensive past research has uncovered valuable insights in
the biology, epidemiology, and pathogenesis of E. coli infections and contributed to the development
of rapid and precise diagnostic techniques [54]. However, much remains unknown or unclear about
the molecular and immunological basis of E. coli pathogenicity. The application of various laboratory
models is unquestionably necessary in generating new clues to virulence strategies employed by E. coli
organisms [55].
REFERENCES
1. Kaper JB, Nataro JP, Mobley HL. Pathogenic Escherichia coli. Nat Rev Microbiol. 2004;2:123–140.
2. Liu S, et al. Escherichia marmotae sp. nov., isolated from faeces of Marmota himalayana. Int J Syst
Evol Microbiol. 2015;65:2130–2134.
3. Kim KS. Current concepts on the pathogenesis of Escherichia coli meningitis: implications for therapy
and prevention. Curr Opin Infect Dis. 2012;25:273–278.
4. Werber D, et al. Outbreaks of virulent diarrheagenic Escherichia coli—are we in control? BMC Med.
2012;10:11.
5. Makobe CK, Sang WK, Kikuvi G, Kariuki S. Molecular characterization of virulence factors in
diarrhoeagenic Escherichia coli isolates from children in Nairobi, Kenya. J Infect Dev Ctries.
2012;6(8):598–604.
6. Lozer DM, et al. Genotypic and phenotypic analysis of diarrheagenic Escherichia coli strains iso-
lated from Brazilian children living in low socioeconomic level communities. BMC Infect Dis.
2013;13:418.
7. Croxen MA, Finlay BB. Molecular mechanisms of Escherichia coli pathogenicity. Nat Rev Microbiol.
2010;8:26–38.
8. Clements A, Young JC, Constantinou N, Frankel G. Infection strategies of enteric pathogenic Escherichia
coli. Gut Microbes. 2012;3(2):71–87.
9. Keseler IM, et al. EcoCyc: fusing model organism databases with systems biology. Nucleic Acids Res.
2013;41:D605–D612.
10. Jafari A, Aslani MM, Bouzari S. Escherichia coli: a brief review of diarrheagenic pathotypes and their
role in diarrheal diseases in Iran. Iran J Microbiol. 2012;4(3):102–117.
11. Koutsoumpas AL, Smyk DS, Bogdanos DP. E. coli induced experimental model of primary biliary
cirrhosis: at last. Int J Hepatol. 2014;2014. doi: 10.1155/2014/848373.
12. Qadri F, Svennerholm A-M, Faruque ASG, Sack RB. Enterotoxigenic Escherichia coli in developing
countries: epidemiology, microbiology, clinical features, treatment, and prevention. Clin Microbiol Rev.
2005;18(3):465–483.
13. Totsika M, et al. Uropathogenic Escherichia coli mediated urinary tract infection. Curr Drug Targets.
2012;13:1386–1399.
14. Mora A, et al. Characteristics of the Shiga-toxin-producing enteroaggregative Escherichia coli O104:H4
German outbreak strain and of STEC strains isolated in Spain. Int Microbiol. 2011;14(3):121–141.
15. Farfan MJ, Torres AG. Molecular mechanisms that mediate colonization of Shiga toxin-producing
Escherichia coli strains. Infect Immun. 2012;80:903–913.
16. Melton-Celsa A, Mohawk K, Teel L, O’Brien A. Pathogenesis of Shiga-toxin producing Escherichia
coli. Curr Top Microbiol Immunol. 2012;357:67–103.
17. Preussel K, Hohle M, Stark K, Werber D. Shiga toxin-producing Escherichia coli O157 is more likely to
lead to hospitalization and death than non-O157 serogroups—except O104. PLoS One. 2013;8:e78180.
328 Laboratory Models for Foodborne Infections
18. Keithlin J, Sargeant J, Thomas MK, Fazil A. Chronic sequelae of E. coli O157: systematic review and
meta-analysis of the proportion of E. coli O157 cases that develop chronic sequelae. Foodborne Pathog
Dis. 2014;11:79–95.
19. Mayer CL, Leibowitz CS, Kurosawa S, Stearns-Kurosawa DJ. Shiga toxins and the pathophysiology of
hemolytic uremic syndrome in humans and animals. Toxins. 2012;4:1261–1287.
20. Lim JY, Yoon JW, Hovde CJ. A brief overview of Escherichia coli O157:H7 and its plasmid O157.
J Microbiol Biotechnol. 2010;20(1):5–14.
21. Mohawk KL, O’Brien AD. Mouse models of Escherichia coli O157:H7 infection and Shiga toxin injec-
tion. J Biomed Biotechnol. 2011;2011. doi: 10.1155/2011/258185.
22. Barnett Foster D. Modulation of the enterohemorrhagic E. coli virulence program through the human
gastrointestinal tract. Virulence. 2013;4(4):315–323.
23. Ferens WA, Hovde CJ. Escherichia coli O157:H7: animal reservoir and sources of human infection.
Foodborne Pathog Dis. 2011;8:465–87.
24. Kaur P, Chakraborti A, Asea A. Enteroaggregative Escherichia coli: an emerging enteric foodborne
pathogen. Interdiscip Perspect Infect Dis. 2010;2010. doi: 10.1155/2010/254159.
25. Muniesa M, Hammerl JA, Hertwig S, Appel B, Brüssow H. Shiga toxin-producing Escherichia coli
O104:H4: a new challenge for microbiology. Appl Environ Microbiol. 2012;78:4065–4073.
26. Beutin L, Martin A. Outbreak of Shiga toxin-producing Escherichia coli (STEC) O104:H4 infection in
Germany causes a paradigm shift with regard to human pathogenicity of STEC strains. J Food Prot.
2012;75(2):408–418.
27. Estrada-Garcia T, Navarro-Garcia F. Enteroaggregative Escherichia coli pathotype: a genetically het-
erogeneous emerging foodborne enteropathogen. FEMS Immunol Med Microbiol. 2012;66(3):281–298.
28. Servin AL. Pathogenesis of Afa/Dr diffusely adhering Escherichia coli. Clin Microbiol Rev.
2005;18(2):264–292.
29. Mansan-Almeida R, Pereira AL, Giugliano LG. Diffusely adherent Escherichia coli strains isolated
from children and adults constitute two different populations. BMC Microbiol. 2013;13:22.
30. Smith EJ, Thompson AP, O’Driscoll A, Clarke DJ. Pathogenesis of adherent-invasive Escherichia coli.
Future Microbiol. 2013;8:1289–1300.
31. Martinez-Medina M, Garcia-Gil LJ. Escherichia coli in chronic inflammatory bowel diseases: an
update on adherent invasive Escherichia coli pathogenicity. World J Gastrointest Pathophysiol.
2014;5(3):213–227.
32. Agarwal J, Srivastava S, Singh M. Pathogenomics of uropathogenic Escherichia coli. Indian J Med
Microbiol. 2012;30:141–149.
33. Botkin DJ, Galli L, Sankarapani V, Soler M, Rivas M, Torres AG. Development of a multiplex PCR
assay for detection of Shiga toxin-producing Escherichia coli, enterohemorrhagic E. coli, and entero-
pathogenic E. coli strains. Front Cell Infect Microbiol. 2012;2:8.
34. Chandra M, Cheng P, Rondeau G, Porwollik S, McClelland M. A single step multiplex PCR for identifica-
tion of six diarrheagenic E. coli pathotypes and Salmonella. Int J Med Microbiol. 2013;303(4):210–216.
35. Wiles TJ, Mulvey MA. The RTX pore-forming toxin α-hemolysin of uropathogenic Escherichia coli:
progress and perspectives. Future Microbiol. 2013;8:73–84.
36. Fialho OB, et al. Detection of diarrheagenic Escherichia coli using a two-system multiplex-PCR proto-
col. J Clin Lab Anal. 2013;27(2):155–161.
37. Fujioka M, Otomo Y, Ahsan CR. A novel single-step multiplex polymerase chain reaction assay for the
detection of diarrheagenic Escherichia coli. J Microbiol Methods. 2013;92(3):289–292.
38. Souza TB, Lozer DM, Kitagawa SM, Spano LC, Silva NP, Scaletsky IC. Real-time multiplex PCR
assay and melting curve analysis for identifying diarrheagenic Escherichia coli. J Clin Microbiol.
2013;51(3):1031–1033.
39. Bitzan M. Treatment options for HUS secondary to Escherichia coli O157:H7. Kidney Int Suppl.
2009;112:S62–S66.
40. Toledo CC, Arvidsson I, Karpman D. Cross-reactive protection against enterohemorrhagic Escherichia
coli infection by enteropathogenic E. coli in a mouse model. Infect Immun. 2011;79:2224–2233.
41. Garcia-Angulo VA, Kalita A, Torres AG. Advances in the development of enterohemorrhagic
Escherichia coli vaccines using murine models of infection. Vaccine. 2013;31(32):3229–3235.
42. Law RJ, Gur-Arie L, Rosenshine I, Finlay BB. In vitro and in vivo model systems for studying entero-
pathogenic Escherichia coli infections. Cold Spring Harb Perspect Med. 2013;3(3):a009977.
Escherichia 329
43. Kalita A, Hu J, Torres AG. Recent advances in adherence and invasion of pathogenic Escherichia coli.
Curr Opin Infect Dis. 2014;27(5):459–464.
44. Philipson CW, Bassaganya-Riera J, Hontecillas R. Animal models of enteroaggregative Escherichia
coli infection. Gut Microbes. 2013;4(4):281–291.
45. Mallick EM, et al. A novel murine infection model for Shiga toxin-producing Escherichia coli. J Clin
Invest. 2012;122:4012–4024.
46. Ritchie JM. Animal models of enterohemorrhagic Escherichia coli infection. Microbiol Spectr.
2014;2(4). doi: 10.1128/microbiolspec.EHEC-0022-2013.
47. Zotta E, et al. Development of an experimental hemolytic uremic syndrome in rats. Pediatr Nephrol.
2008;23:559–567.
48. Mohawk KL, et al. Pathogenesis of Escherichia coli O157:H7 strain 86-24 following oral infection of
BALB/c mice with an intact commensal flora. Microb Pathog. 2010;48:131–142.
49. Dalgakiran F, Witcomb LA, McCarthy AJ, Birchenough GM, Taylor PW. Non-invasive model of neuro-
pathogenic Escherichia coli infection in the neonatal rat. J Vis Exp. 2014;(92):e52018.
50. Ludwig M, Johannes S, Bergmann M, Failing K, Schiefer HG, Weidner W. Experimental
Escherichia coli epididymitis in rats: a model to assess the outcome of antibiotic treatment. BJU Int.
2002;90(9):933–938.
51. García A, et al. Renal injury is a consistent finding in Dutch belted rabbits experimentally infected with
enterohemorrhagic Escherichia coli. J Infect Dis. 2006;193:1125–1134.
52. Panda A, et al. Escherichia coli O157:H7 infection in Dutch belted and New Zealand white rabbits.
Comp Med. 2010;60:31–37.
53. Croxen MA, et al. Recent advances in understanding enteric pathogenic Escherichia coli. Clin Microbiol
Rev. 2013;26(4):822–880.
54. Parsons BD, Zelyas N, Berenger BM, Chui L. Detection, characterization, and typing of Shiga toxin-
producing Escherichia coli. Front Microbiol. 2016;7:478.
55. Hoogland IC, Houbolt C, van Westerloo DJ, van Gool WA, van de Beek D. Systemic inflammation and
microglial activation: systematic review of animal experiments. J Neuroinflammation. 2015;12:114.
22
Helicobacter
CONTENTS
22.1 Introduction..................................................................................................................................331
22.2 Discovery of Gastric Microorganisms.........................................................................................331
22.3 Gastric Microenvironment of H. pylori.......................................................................................332
22.4 Characteristics and Pathogenesis of H. pylori.............................................................................332
22.5 Infection Route and Childhood Acquisition of H. pylori............................................................333
22.6 Role of H. pylori Infection in Gastric Carcinogenesis................................................................333
22.7 H. pylori Infection Induces No Neoplastic but Hyperplastic Lesions in Mongolian Gerbils...... 334
22.8 Prevention of Gastric Carcinogenesis by Eradication of H. pylori............................................ 336
22.9 Intestinal Metaplasia and Intestinalization of Gastric Cancer................................................... 336
22.10 Exacerbating Factors for Gastric Carcinogenesis: Synergistic Effects of H. pylori
and High-Salt Diet��������������������������������������������������������������������������������������������������������������������� 336
22.11 Chemoprevention of Gastric Carcinogenesis.............................................................................. 337
22.12 Conclusion................................................................................................................................... 337
References............................................................................................................................................... 337
22.1 Introduction
Since Helicobacter pylori was discovered to play a major causative role in chronic gastritis and gastric
carcinogenesis, several attempts have been made to treat and prevent the lesions. In parallel, many exper-
imental studies have confirmed the importance of H. pylori in gastric disease processes. Among animal
models, mice and Mongolian gerbils offer powerful tools for the analysis of H. pylori-associated gastric
lesions.1,2 The Helicobacter genus nowadays contains more than 30 species; this chapter focuses on
H. pylori within the genus and reviews its characteristics, interaction with host environment, and patho-
genesis in gastric disease. As gastric cancer remains a significant contributor to cancer-related death,
prevention of infection and treatment of gastric cancer represent important aspects of cancer control.
331
332 Laboratory Models for Foodborne Infections
H. pylori
(A) (B)
CagA
Type IV Flagella
Src
secretion
system
SHP-2
Urease
VacA
FIGURE 22.1 H. pylori and its schematic view. (A) Immunohistochemical view of H. pylori in the human stomach colo-
nized in the human gastric surface mucin. Top inset: active bacillary form. Bottom inset: coccoid form in the unsatisfactory
condition. The center square is enlarged in (B) as a schematic view. Immunohistochemistry against H. pylori. Original
magnification, 630×. (B) H. pylori utilize flagella to move into gastric surface mucin. CagA is injected through type IV
secretion system and tyrosine phosphorylated with Src family kinase. Phosphorylated CagA binds and activates SHP-2
tyrosine phosphatase. VacA is a vacuolating toxin. Bacterial urease catalyzes urea to ammonia to neutralize hydrogen
chloride in the acidic environment of the stomach.
bacterial genus.8 Here, we concentrate mainly on H. pylori, since most of the laboratory models were
established with this species (Figure 22.1A).
in the former possess EPIYA-A, B, and D motifs and may contribute to the geographical differences
observed in gastric carcinogenesis. Phosphorylated CagA then activates SHP-2 phosphatase, which
then dephosphorylates FAK kinase, resulting in impairment of epithelial cell adhesion and morphol-
ogy.19 Studies in wild-type CagA transgenic mice have confirmed the oncogenicity of this bacterial pro-
tein by demonstrating the development of gastrointestinal epithelial hyperplasia and adenocarcinomas
as well as hematopoietic malignancies, whereas phosphorylation-resistant CagA mice did not develop
disease.20
Broth culture supernatant of H. pylori was known to induce vacuolization of the mammalian cells,
which was characterized as VacA.21,22 VacA binds to receptor protein tyrosine phosphatases (RPTPs) α
and β on the surface of target cells.23 Mice deficient in RPTP type Z (RPTPζ or RPTPβ) revealed toler-
ance to VacA for mucosal damage.24 Oligomerized VacA enters into the cytoplasm through endocytosis
and shows multifunctional toxicity including interference of T cell activation and proliferation, reduction
of mitochondria membrane potential, and formation of vacuolation in endosomal/lysosomal compart-
ments25 (Figure 22.1B).
Incidence of
gastric cancer
(A) S
(B) S
(C) S
(D) S
Early
S
Late
(E) Early S
Late S
Chemopreventive agents S
: Chemical carcinogens
: Inoculation of H. pylori : Eradication S : Sacrifice
FIGURE 22.2 Modifying factors for H. pylori-associated stomach carcinogenesis in the Mongolian gerbil models.
(A) H. pylori infection alone is rarely carcinogenic. (B) Drinking water containing chemical carcinogens including
MNU or MNNG induces stomach cancers. (C) When combined, H. pylori become a strong promoter. (D) Earlier infec-
tion increases the risk compared with a later event. (E) Earlier eradication of H. pylori reduces risk of stomach cancers.
(F) A high-salt diet exacerbates inflammation and increases the incidence of H. pylori-associated cancer. On the other
hand, various natural products and pure chemicals appear to have chemopreventive potential.
Besides the gerbil models, mouse models feature advantages especially for genetic approaches.
Transgenic mice (K19-C2mE transgenic mouse) simultaneously expressing cyclooxygenase-2 (COX-2)
and microsomal prostaglandin E synthase (mPGES)-1 as well as Wnt1 revealed the importance of these
pathways in gastric tumorigenesis with Helicobacter infection.52,53 K19-C2mE mice developed adeno-
carcinomas not only in pyloric mucosa but also in fundic glands with MNU treatment as well as H. pylori
infection, and so are considered as a model for increasing proximal malignancies.54 Interleukin 1β (−/−)
knockout mice showed that IL1β induced by H. pylori infection affected both inflammatory and epithe-
lial cells and enhanced gastric carcinogenesis.55
Long-term infection of H. pylori frequently induced neuroendocrine tumors (NETs) (endocrine cell
hyperplasia/dysplasia and carcinoid tumors) in a Mongolian gerbil model in association with increased
serum gastrin level. Eradication of H. pylori prevents development of NETs in the glandular stomach,
being associated with reduction in serum gastrin levels.56 Proton pump inhibitors (PPIs), routinely used
for treatment of upper gastrointestinal disorders, have raised some concerns regarding the long-term
safety and development of NETs in the stomach. When PPI was administered to H. pylori-infected
Mongolian gerbils, high dose increased NET development with higher serum gastrin in contrast to no
influence at low dose.57
subserosa (several months after inoculation of H. pylori), and these are designated as heterotopic prolif-
erative glands (HPGs).62 HPGs consisted of gastric and/or intestinal metaplastic epithelial cells, the latter
progressed depending on the duration of H. pylori-induced inflammation. HPGs, in turn, dramatically
diminished with the eradication of the bacteria, indicating that HPG is not neoplastic but rather hyper-
plastic. HPGs often resemble differentiated or mucinous adenocarcinomas showing structural abnor-
mality, but lack obvious cellular atypia. Characteristics of HPG include (1) organized polarity of their
component cells; (2) differentiation from gastric phenotypic HPG into intestinal phenotypic HPG some-
times associated with mature Paneth cells; (3) formation of large cystic dilatations containing mucin,
often with calcification; (4) shedding of epithelial cells and necrosis at the tips of lesions; (5) high-grade
inflammation with infiltration of inflammatory cells (neutrophils in acute phase and mononuclear cells in
chronic phase); and (6) organized polarity of proliferating zones. These features are quite different from
those of well-differentiated adenocarcinomas, which are characterized by obvious cellular atypia. HPGs
need to be precisely distinguished from adenocarcinomas (Figure 22.3 and Table 22.1).1,2
(A) (B)
(C) (D)
FIGURE 22.3 Gastric lesions in H. pylori-infected Mongolian gerbils. (A) Submucosal hyperplastic proliferating glands
called HPG after 44-week long inflammation. (B) HPG has drastically disappeared 20 weeks after eradication of the bac-
teria. Well- (C) and poorly (D) differentiated adenocarcinomas developed with carcinogen treatment. (A–D) Hematoxylin
and eosin staining. Original magnification, 25× (A and B); 200× (C); and 400× (D).
TABLE 22.1
Comparison of Submucosal Lesions in H. pylori-Infected Mongolian Gerbils
HPG Tubular Adenocarcinoma
Gland distribution Dispersed Compactly proliferated
Gland shape Large and cystic Relatively small
Intracystic material Eosinophilic, sometimes with calcification Usually transparent
Lining epithelium of cysts Shedding of epithelial cells with necrosis at the tip of Fully lined with atypical cells.
the cyst
Intestinal metaplasia Frequent, sometimes with Paneth cells Relatively infrequent
Stroma Severe inflammatory cell infiltrates. Neutrophils in acute Desmoplastic reaction
phase. Lymphocytes and plasma cells in chronic phase
with lymphoid follicle formation.
Atypia No atypia. Organized polarity. Partly degenerated. Enlarged nuclei with increased
chromatin. Loss of polarity.
336 Laboratory Models for Foodborne Infections
dose-dependent fashion (Figure 22.2). A high-salt diet upregulated the amount of SMCM, suitable for
H. pylori colonization, despite no increment in the corresponding mucin core protein, MUC5AC mRNA.
Although GMCM with its mucin core protein MUC6 was induced against H. pylori infection, high-salt
diet decreased the amount of GMCM, which acts against H. pylori infection by inhibiting the bacterial
cell wall component.12 In an H. pylori-infected MNU-treated mouse model, microarray analysis revealed
overexpression of CD177 and Reg3g in a high-salt diet group, the former being associated with better
prognosis in human cases.78 Reduction of salt intake could thus be one of the most important chemopre-
ventive methods for human gastric carcinogenesis.
22.12 Conclusion
Discovery of H. pylori from the human stomach opened a new avenue to clarify the mechanism of
gastric lesions including chronic atrophic gastritis, intestinal metaplasia, and gastric carcinogenesis.
A large number of epidemiological studies revealed the positive correlation of H. pylori with gastric can-
cer development. In Mongolian gerbil models, H. pylori inoculation caused severe hyperplastic lesions
called HPG and showed strong promoting effects in carcinogen-treated animals; H. pylori infection alone
rarely induced adenocarcinomas. Mouse models are also effective for genetic approach. Eradication of
the bacteria was effective in prevention of gastric carcinogenesis, especially in cases of milder gastritis
without severe atrophy or intestinal metaplasia, both in human and rodent models. Besides eradication
of the bacteria, chemopreventive approaches have been attempted in prevention of gastric neoplasia. It is
necessary to establish effective approaches for application in human trials.
REFERENCES
1.
Tsukamoto, T., Mizoshita, T. & Tatematsu, M. Animal models of stomach carcinogenesis. Toxicol
Pathol 35, 636–648 (2007).
2.
Tsukamoto, T., Toyoda, T., Mizoshita, T. & Tatematsu, M. Helicobacter pylori infection and gastric
carcinogenesis in rodent models. Semin Immunopathol 35, 177–190 (2013).
3.
Warren, J.R. & Marshall, B. Unidentified curved bacilli on gastric epithelium in active chronic gastritis.
Lancet 321, 1273–1275 (1983).
338 Laboratory Models for Foodborne Infections
4. Marshall, B.J. & Warren, J.R. Unidentified curved bacilli in the stomach of patients with gastritis and
peptic ulceration. Lancet 323, 1311–1315 (1984).
5. Goodwin, C.S. et al. Transfer of Campylobacter pylori and Campylobacter mustelae to Helicobacter
gen. nov. as Helicobacter pylori comb. nov. and Helicobacter mustelae comb. nov., respectively. Int
J Syst Bacteriol 39, 397–405 (1989).
6. Hannula, M. & Hanninen, M.L. Phylogenetic analysis of Helicobacter species based on partial gyrB
gene sequences. Int J Syst Evol Microbiol 57, 444–449 (2007).
7. Mikkonen, T.P., Karenlampi, R.I. & Hanninen, M.L. Phylogenetic analysis of gastric and enterohepatic
Helicobacter species based on partial HSP60 gene sequences. Int J Syst Evol Microbiol 54, 753–758
(2004).
8. National Center for Biotechnology Information. The NCBI taxonomy database: Helicobacter. http://www.
ncbi.nlm.nih.gov/taxonomy/?term=helicobacter. U.S. National Library of Medicine, Bethesda, MD.
9. Ota, H. & Katsuyama, T. Alternating laminated array of two types of mucin in the human gastric
surface mucous layer. Histochem J 24, 86–92 (1992).
10. Ota, H. et al. A dual staining method for identifying mucins of different gastric epithelial mucous cells.
Histochem J 23, 22–28 (1991).
11. Kato, S. et al. High salt diets dose-dependently promote gastric chemical carcinogenesis in Helicobacter
pylori-infected Mongolian gerbils associated with a shift in mucin production from glandular to surface
mucous cells. Int J Cancer 119, 1558–1566 (2006).
12. Kawakubo, M. et al. Natural antibiotic function of a human gastric mucin against Helicobacter pylori
infection. Science 305, 1003–1006 (2004).
13. Chan, W.Y. et al. Coccoid forms of Helicobacter pylori in the human stomach. Am J Clin Pathol 102,
503–507 (1994).
14. Sorberg, M., Nilsson, M., Hanberger, H. & Nilsson, L.E. Morphologic conversion of Helicobacter pylori
from bacillary to coccoid form. Eur J Clin Microbiol Infect Dis 15, 216–219 (1996).
15. Tomb, J.F. et al. The complete genome sequence of the gastric pathogen Helicobacter pylori. Nature
388, 539–547 (1997).
16. Sharma, C.M. et al. The primary transcriptome of the major human pathogen Helicobacter pylori.
Nature 464, 250–255 (2010).
17. Backert, S. & Selbach, M. Tyrosine-phosphorylated bacterial effector proteins: the enemies within.
Trends Microbiol 13, 476–484 (2005).
18. Hatakeyama, M. Anthropological and clinical implications for the structural diversity of the Helicobacter
pylori CagA oncoprotein. Cancer Sci 102, 36–43 (2011).
19. Tsutsumi, R., Takahashi, A., Azuma, T., Higashi, H. & Hatakeyama, M. Focal adhesion kinase is a
substrate and downstream effector of SHP-2 complexed with Helicobacter pylori CagA. Mol Cell Biol
26, 261–276 (2006).
20. Ohnishi, N. et al. Transgenic expression of Helicobacter pylori CagA induces gastrointestinal and
hematopoietic neoplasms in mouse. Proc Natl Acad Sci USA 105, 1003–1008 (2008).
21. Cover, T.L. & Blaser, M.J. Purification and characterization of the vacuolating toxin from Helicobacter
pylori. J Biol Chem 267, 10570–10575 (1992).
22. Phadnis, S.H., Ilver, D., Janzon, L., Normark, S. & Westblom, T.U. Pathological significance and
molecular characterization of the vacuolating toxin gene of Helicobacter pylori. Infect Immun 62,
1557–1565 (1994).
23. Wada, A., Yamasaki, E. & Hirayama, T. Helicobacter pylori vacuolating cytotoxin, VacA, is respon-
sible for gastric ulceration. J Biochem 136, 741–746 (2004).
24. Fujikawa, A. et al. Mice deficient in protein tyrosine phosphatase receptor type Z are resistant to gastric
ulcer induction by VacA of Helicobacter pylori. Nat Genet 33, 375–381 (2003).
25. Cover, T.L. & Blanke, S.R. Helicobacter pylori VacA, a paradigm for toxin multifunctionality. Nat Rev
Microbiol 3, 320–332 (2005).
26. Goh, K.L., Chan, W.K., Shiota, S. & Yamaoka, Y. Epidemiology of Helicobacter pylori infection and
public health implications. Helicobacter 16(Suppl 1), 1–9 (2011).
27. Cao, X. et al. Earlier Helicobacter pylori infection increases the risk for the N-methyl-N-nitrosourea-
induced stomach carcinogenesis in Mongolian gerbils. Jpn J Cancer Res 93, 1293–1298 (2002).
28. Kato, S., Kikuchi, S. & Nakajima, S. When does gastric atrophy develop in Japanese children?
Helicobacter 13, 278–281 (2008).
Helicobacter 339
29. Correa, P. et al. Chemoprevention of gastric dysplasia: randomized trial of antioxidant supplements and
anti-Helicobacter pylori therapy. J Natl Cancer Inst 92, 1881–1888 (2000).
30. Malaty, H.M. et al. Age at acquisition of Helicobacter pylori infection: a follow-up study from infancy
to adulthood. Lancet 359, 931–935 (2002).
31. Hu, P.J. et al. Helicobacter pylori associated with a high prevalence of duodenal ulcer disease and a low
prevalence of gastric cancer in a developing nation. Gut 36, 198–202 (1995).
32. Craanen, M.E., Dekker, W., Blok, P., Ferwerda, J. & Tytgat, G.N. Intestinal metaplasia and Helicobacter
pylori: an endoscopic bioptic study of the gastric antrum. Gut 33, 16–20 (1992).
33. Parsonnet, J. et al. Helicobacter pylori infection and the risk of gastric carcinoma. N Engl J Med 325,
1127–1131 (1991).
34. Nomura, A. et al. Helicobacter pylori infection and gastric carcinoma among Japanese Americans in
Hawaii. N Engl J Med 325, 1132–1136 (1991).
35. Forman, D. et al. Association between infection with Helicobacter pylori and risk of gastric cancer:
evidence from a prospective investigation. BMJ 302, 1302–1305 (1991).
36. Graham, D.Y. et al. Effect of treatment of Helicobacter pylori infection on the long-term recurrence of
gastric or duodenal ulcer. A randomized, controlled study. Ann Intern Med 116, 705–708 (1992).
37. Kuipers, E.J. et al. Long-term sequelae of Helicobacter pylori gastritis. Lancet 345, 1525–1528 (1995).
38. Asaka, M. et al. Atrophic changes of gastric mucosa are caused by Helicobacter pylori infection rather
than aging: studies in asymptomatic Japanese adults. Helicobacter 1, 52–56 (1996).
39. Huang, J.Q., Sridhar, S., Chen, Y. & Hunt, R.H. Meta-analysis of the relationship between Helicobacter
pylori seropositivity and gastric cancer. Gastroenterology 114, 1169–1179 (1998).
40. Parsonnet, J. et al. Helicobacter pylori infection and gastric lymphoma. N Engl J Med 330, 1267–
1271 (1994).
41. The EUROGAST Study Group. An international association between Helicobacter pylori infection and
gastric cancer. Lancet 341, 1359–1362 (1993).
42. IARC Working Group on the Evaluation of Carcinogenic Risks to Humans. Infection with Helicobacter
pylori. in Schistosomes, Liver Flukes and Helicobacter pylori, Vol. 61, 177–241, World Health
Organization/International Agency for Research on Cancer, Lyon (1994).
43. Lee, A., Fox, J.G., Otto, G. & Murphy, J. A small animal model of human Helicobacter pylori active
chronic gastritis. Gastroenterology 99, 1315–1323 (1990).
44. Lee, A. et al. A standardized mouse model of Helicobacter pylori infection: introducing the Sydney
strain. Gastroenterology 112, 1386–1397 (1997).
45. Hirayama, F., Takagi, S., Yokoyama, Y., Iwao, E. & Ikeda, Y. Establishment of gastric Helicobacter
pylori infection in Mongolian gerbils. J Gastroenterol 31(Suppl 9), 24–28 (1996).
46. Sugimura, T. & Fujimura, S. Tumour production in glandular stomach of rat by N-methyl-N′-nitro-N-
nitrosoguanidine. Nature 216, 943–944 (1967).
47. Tatematsu, M. et al. Induction of adenocarcinomas in the glandular stomach of BALB/c mice treated
with N-methyl-N-nitrosourea. Jpn J Cancer Res 83, 915–918 (1992).
48. Tatematsu, M. et al. Induction of glandular stomach cancers in Helicobacter pylori-sensitive Mongolian
gerbils treated with N-methyl-N-nitrosourea and N-methyl-N′-nitro-N-nitrosoguanidine in drinking
water. Jpn J Cancer Res 89, 97–104 (1998).
49. Sugiyama, A. et al. Helicobacter pylori infection enhances N-methyl-N-nitrosourea-induced stomach
carcinogenesis in the Mongolian gerbil. Cancer Res 58, 2067–2069 (1998).
50. Shimizu, N. et al. Helicobacter pylori infection enhances glandular stomach carcinogenesis in
Mongolian gerbils treated with chemical carcinogens. Carcinogenesis 20, 669–676 (1999).
51. Shimizu, N. et al. New animal model of glandular stomach carcinogenesis in Mongolian gerbils infected
with Helicobacter pylori and treated with a chemical carcinogen. J Gastroenterol 34(Suppl 11), 61–66
(1999).
52. Oshima, H., Oshima, M., Inaba, K. & Taketo, M.M. Hyperplastic gastric tumors induced by activated
macrophages in COX-2/mPGES-1 transgenic mice. EMBO J 23, 1669–1678 (2004).
53. Oshima, H. et al. Carcinogenesis in mouse stomach by simultaneous activation of the Wnt signaling and
prostaglandin E2 pathway. Gastroenterology 131, 1086–1095 (2006).
54. Takasu, S. et al. Roles of cyclooxygenase-2 and microsomal prostaglandin E synthase-1 expression and
β-catenin activation in gastric carcinogenesis in N-methyl-N-nitrosourea-treated K19-C2mE transgenic
mice. Cancer Sci 99, 2356–2364 (2008).
340 Laboratory Models for Foodborne Infections
55. Shigematsu, Y. et al. Interleukin-1β induced by Helicobacter pylori infection enhances mouse gastric
carcinogenesis. Cancer Lett 340, 141–147 (2013).
56. Cao, L. et al. Development of carcinoid tumors of the glandular stomach and effects of eradication in
Helicobacter pylori-infected Mongolian gerbils. Asian Pac J Cancer Prev 9, 25–30 (2008).
57. Tsukamoto, H. et al. Long-term high-dose proton pump inhibitor administration to Helicobacter pylori-
infected Mongolian gerbils enhances neuroendocrine tumor development in the glandular stomach.
Asian Pac J Cancer Prev 12, 1049–1054 (2011).
58. Honda, S. et al. Development of Helicobacter pylori-induced gastric carcinoma in Mongolian gerbils.
Cancer Res 58, 4255–4259 (1998).
59. Watanabe, T., Tada, M., Nagai, H., Sasaki, S. & Nakao, M. Helicobacter pylori infection induces gastric
cancer in Mongolian gerbils. Gastroenterology 115, 642–648 (1998).
60. Gaddy, J.A. et al. High dietary salt intake exacerbates Helicobacter pylori-induced gastric carcinogen-
esis. Infect Immun 81, 2258–2267 (2013).
61. Shimizu, N. et al. Eradication diminishes enhancing effects of Helicobacter pylori infection on
glandular stomach carcinogenesis in Mongolian gerbils. Cancer Res 60, 1512–1514 (2000).
62. Nozaki, K. et al. Reversibility of heterotopic proliferative glands in glandular stomach of Helicobacter
pylori-infected Mongolian gerbils on eradication. Jpn J Cancer Res 93, 374–381 (2002).
63. Uemura, N. et al. Effect of Helicobacter pylori eradication on subsequent development of cancer after
endoscopic resection of early gastric cancer. Cancer Epidemiol Biomarkers Prev 6, 639–642 (1997).
64. Fukase, K. et al. Effect of eradication of Helicobacter pylori on incidence of metachronous gastric
carcinoma after endoscopic resection of early gastric cancer: an open-label, randomised controlled trial.
Lancet 372, 392–397 (2008).
65. Wong, B.C. et al. Helicobacter pylori eradication to prevent gastric cancer in a high-risk region of
China: a randomized controlled trial. JAMA 291, 187–194 (2004).
66. Yanaoka, K. et al. Eradication of Helicobacter pylori prevents cancer development in subjects with mild
gastric atrophy identified by serum pepsinogen levels. Int J Cancer 125, 2697–2703 (2009).
67. Nozaki, K. et al. Effect of early eradication on Helicobacter pylori-related gastric carcinogenesis in
Mongolian gerbils. Cancer Sci 94, 235–239 (2003).
68. Inada, K. et al. Gastric and intestinal mixed and solely intestinal types of intestinal metaplasia in the
human stomach. Pathol Int 47, 831–841 (1997).
69. Tsukamoto, T. et al. Down regulation of a gastric transcription factor, Sox2, and ectopic expression
of intestinal homeobox genes, Cdx1 and Cdx2: inverse correlation during progression from gastric/
intestinal-mixed to complete intestinal metaplasia. J Cancer Res Clin Oncol 130, 135–145 (2004).
70. Niwa, T. et al. Mixed gastric- and intestinal-type metaplasia is formed by cells with dual intestinal and
gastric differentiation. J Histochem Cytochem 53, 75–85 (2005).
71. Tatematsu, M., Tsukamoto, T. & Inada, K. Stem cells and gastric cancer—role of gastric and intestinal
mixed intestinal metaplasia. Cancer Sci 94, 135–141 (2003).
72. Tajima, K. & Tominaga, S. Dietary habits and gastro-intestinal cancers: a comparative case-control
study of stomach and large intestinal cancers in Nagoya, Japan. Jpn J Cancer Res 76, 705–716 (1985).
73. Tsugane, S., Tsuda, M., Gey, F. & Watanabe, S. Cross-sectional study with multiple measurements of
biological markers for assessing stomach cancer risks at the population level. Environ Health Perspect
98, 207–210 (1992).
74. Joossens, J.V. et al. European Cancer Prevention (ECP) and the INTERSALT Cooperative Research
Group. Dietary salt, nitrate and stomach cancer mortality in 24 countries. Int J Epidemiol 25, 494–504
(1996).
75. Kono, S. & Hirohata, T. Nutrition and stomach cancer. Cancer Causes Control 7, 41–55 (1996).
76. Tatematsu, M., Takahashi, M., Fukushima, S., Hananouchi, M. & Shirai, T. Effects in rats of
sodium chloride on experimental gastric cancers induced by N-methyl-N-nitro-N-nitrosoguani-
dine or 4-nitroquinoline-1-oxide. J Natl Cancer Inst 55, 101–106. (1975).
77. Nozaki, K. et al. Synergistic promoting effects of Helicobacter pylori infection and high-salt diet on
gastric carcinogenesis in Mongolian gerbils. Jpn J Cancer Res 93, 1083–1089 (2002).
78. Toyoda, T. et al. Gene expression analysis of a Helicobacter pylori-infected and high-salt diet-treated
mouse gastric tumor model: identification of CD177 as a novel prognostic factor in patients with gastric
cancer. BMC Gastroenterol 13, 122 (2013).
Helicobacter 341
79. Magari, H. et al. Inhibitory effect of etodolac, a selective cyclooxygenase-2 inhibitor, on stomach
carcinogenesis in Helicobacter pylori-infected Mongolian gerbils. Biochem Biophys Res Commun 334,
606–612 (2005).
80. Yanaoka, K. et al. Preventive effects of etodolac, a selective cyclooxygenase-2 inhibitor, on cancer
development in extensive metaplastic gastritis, a Helicobacter pylori-negative precancerous lesion. Int
J Cancer 126, 1467–1473 (2010).
81. Cao, D. et al. Canolol inhibits gastric tumors initiation and progression through COX-2/PGE2 pathway
in K19-C2mE transgenic mice. PLoS One 10, e0120938 (2015).
23
Klebsiella: Caenorhabditis elegans as a Laboratory
Model for Klebsiella pneumoniae Infection
CONTENTS
23.1 Foodborne Infection: A Global Burden...................................................................................... 344
23.2 K. pneumoniae: A Potent Agent for Foodborne Diseases.......................................................... 344
23.3 Importance of Model Animals in Research............................................................................... 345
23.4 Discovery and Exploration of C. elegans................................................................................... 345
23.5 Other Model Animals to Study K. pneumoniae Infection......................................................... 345
23.6 Characteristic Features of C. elegans......................................................................................... 346
23.7 C. elegans as a Model for Host–Pathogen Interaction Studies................................................... 346
23.8 Key Differences between Innate Immune System of C. elegans and Mammals....................... 347
23.9 p38 MAPK Pathway in C. elegans............................................................................................. 347
23.10 Live K. pneumoniae Was More Pathogenic to C. elegans�������������������������������������������������������� 347
23.11 K. pneumoniae Proliferates inside the Host Intestine and Causes Persistent Infection������������ 347
23.12 Role of p38 MAPK in Immune Defense of Nematode against K. pneumoniae Infection�������� 349
23.13 Role of Toll-Dependent p38 MAPK Pathway in C. elegans against
K. pneumoniae Infection .....������������������������������������������������������������������������������������������������������ 350
23.14 Upregulation of Virulence Gene in K. pneumoniae during Infection........................................ 351
23.15 Conclusion................................................................................................................................... 351
Acknowledgments................................................................................................................................... 351
References............................................................................................................................................... 351
Klebsiella pneumoniae has emerged as one of the most frequent and potent pathogens causing foodborne
infection in humans since 1980. Despite its clinical significance, the absence of appropriate animal mod-
els has hampered efforts in identifying the virulence and host factors that determine the pathogenesis
and susceptibility of host to gastrointestinal infection. For the past few decades, Caenorhabditis elegans
has been recognized as a better laboratory animal model based on its potential to closely recapitulate
human disease. Additionally, owing to its conserved immune pathways with higher animals and avail-
ability of developed transgenic, knockout, and knockdown strains, C. elegans has attracted the atten-
tion of researchers in various fields as a powerful and versatile model system. C. elegans has played a
crucial role in elucidating the pathophysiology and immunology of microbial infections and has thus
provided valuable insights into the pathogenesis and response of host system, from chronic to acute
infection. The information derived from animal model aids researchers to characterize the host response
and helps design chemotherapeutic treatment for the public risk associated with infection caused by
K. pneumoniae-contaminated food commodities. This chapter focuses on C. elegans as a model system
that has been instrumental in uncovering the clues on host–pathogen interactions during K. pneumoniae
infection.
343
344 Laboratory Models for Foodborne Infections
23.2 K
. pneumoniae: A Potent Agent for Foodborne Diseases
Foodborne illness represents an important public health hazard and has a significant economical
impact on developing countries worldwide. Approximately 1.5 million neonatal deaths are caused
by foodborne diarrheal disease alone. Viruses, bacteria, fungi, and parasites are the causative agents
for foodborne infections [5]. Among them, bacteria are implicated in sporadic illness among differ-
ent populations. Prior to 1990s, the species of Salmonella and Campylobacter and Escherichia coli
accounted for most of the outbreaks. Subsequently, K. pneumoniae was shown to cause foodborne
diseases in humans through contaminated vegetables [6] and hamburgers [7]. Given the increased
number and severity of infection, K. pneumoniae has been recognized as one of the most important
foodborne bacterial pathogens.
K. pneumoniae is one of the most versatile and clinically significant pathogens in causing both
nosocomial- and community-acquired infections worldwide. It is a Gram-negative, rod-shaped bac-
terium belonging to the family Enterobacteriaceae [8]. Although it is found as a common component
of normal microflora in the nasopharyngeal and gastrointestinal regions of humans, K. pneumoniae
can cause a wide variety of diseases, ranging from mild infections to life-threatening diseases, in
immunocompromised patients. The diseases caused by K. pneumoniae are bowel infections, pneu-
monia, suppurative infections, bacteremia, septicemia, and inflammation of the urinary tract. In
severe infection cases, K. pneumoniae causes liver abscess along with meningitis and endophthal-
mitis, which accounts for a mortality rate of 20%–55% [8]. Due to its severity in causing infection
in immunocompromised individuals, it is classified as an opportunistic pathogen. As far as hospital-
acquired infection by K. pneumoniae is concerned, the prime significance is its ability to evade the
innate immune response and cause colonization in the intestinal tract of patients and cause symp-
toms. Infection with K. pneumoniae begins with its initial colonization in the gastrointestinal tract,
irrespective of its site of infection [9]. The degree of colonization mainly depends on the potential
of the bacterial strain to form biofilms [10] and emerge resistant to antibiotics. The resistance of K.
pneumoniae toward third-generation cephalosporins and carbapenem were identified in 1983 [11–13].
Since, K. pneumoniae is undergoing multidrug resistance, it is necessary to study its pathogenesis
in a multicellular host system to approach the infection using an alternate therapy. Thus, research-
ers paid a great deal of attention to choose a suitable animal model to decipher the host perspectives
during bacterial infections.
Klebsiella: C. elegans as a Laboratory Model for K. pneumoniae Infection 345
the host immune system. Injection of K. pneumoniae into the murine model alters the level of interleu-
kin-17 (IL-17) [49], tumor necrosis factor alpha, keratinocyte-derived chemokines and bronchoalveolar
fluid, and IL-10 [50] during the course of infection. Furthermore, interaction of K. pneumoniae with
the lung epithelial cells [51] markedly increased the recruitment of neutrophils to the lungs as well as
proinflammatory cytokines and chemokines. A recent study has used the wax moth Galleria mellonella
as a surrogate host model to understand the pathogenesis of K. pneumoniae at the molecular level for
testing new therapies [52]. In that study, it was reported that G. mellonella could distinguish pathogenic
and nonpathogenic Klebsiella strains. Moreover, infection of K. pneumoniae in G. mellonella attenu-
ates the host innate immune response by inhibiting the antimicrobial peptides. Furthermore, the study
by Wu et al. in HepG2 cells of humans suggested that infection with K. pneumoniae attenuates the p38
mitogen-activated protein kinase (p38 MAPK) pathway of host system and that it required lipopolysac-
charide for complete pathogenesis during host–pathogen interaction [53]. A recent study by us has used
C. elegans as a model and reinforced the essential role of Toll-dependent p38 MAPK pathway during
K. pneumoniae infection [54].
(v) (vi)
80
No. of eggs/worm
60
60
40
40
20
20
OP50
K. pneumoniae 0
0
0 10 20 30 40 50 0 4 8 12 24
Time (h) Time of exposure (h)
OP50 control
(D) 60 K. pneumoniae
50
No. of flings/worm/30 s
40
30
20
10
0 4 8 12 24
Time of exposure (h)
FIGURE 23.1 (A) Microscopic images of C. elegans exposed to K. pneumoniae for 12 h at 20°C are shown. Wild-type
C. elegans fed with OP50 showed intact pharynx (i) and (ii) normal eggs. In contrast, nematodes exposed to K. pneumoniae
showed severe physiological defects such as (iii, iv) distended pharynx (indicated by arrowhead), (v) accumulation of eggs
inside the intestine (indicated by arrowhead), (vi) internal hatching, and intestinal colonization (indicated by arrowhead).
Physiological assays showing the impact of K. pneumoniae infection in C. elegans. (B) Survival of C. elegans exposed to
live K. pneumoniae. (C) Cessation of egg laying in nematodes exposed to K. pneumoniae. (D) Inhibition of feeding dur-
ing K. pneumoniae infection. Data are presented as mean ± standard deviation of three biological replicates. Statistical
analysis was performed by one-way ANOVA followed by Duncan’s post hoc analysis.
(Figure 23.2). Measuring the bacterial load inside the intestine of nematodes by colony forming unit (CFU)
assay revealed that the number of bacterial cells in C. elegans exposed for 4 h was 2.7 × 103, and it was
found to be increased to 3.6 × 104, 4.7 × 105, and 5.3 × 106 at 6, 12, and 24 h, respectively (Figure 23.2A).
The results of CFU assay suggested that K. pneumoniae colonized and proliferated inside the host intes-
tine with an increase in residence time.
Klebsiella: C. elegans as a Laboratory Model for K. pneumoniae Infection 349
(A) 5
4
Log CFU/worm
4 6 12 24
Time (h)
(B) 35
30
Fold change in expression
25 pmk-1
tol-1
20
clec-60
15 clec-67
10 lys-1
nlp-29
5
0
KP 6 h KP 12 h KP 24 h KP 36 h
FIGURE 23.2 The histogram showing the CFU of K. pneumoniae inside the intestine of C. elegans in response to time
of infection (A). Changes in the expression profile of N2 nematodes during infection with K. pneumoniae (B). (From
Kamaladevi, A. and Balamurugan, K., Pathog. Dis., 73(5), ftv021, 2015.)
80
Fold change in expression
pmk-1
20
clec-60
60
15 clec-67
40 lys-1
10 clec-87
20 clec-85
5
** ** * nlp-29
* * * ** ** * **
0 0 ** **
0 5 10 15 20 25 30 35 40 45 50 KP 6 h KP 12 h KP 24 h
Time (h) –5
clec-60
Fold change in expression
8 *
clec-67 2 clec-60
**
clec-67
6 lys-1
1.5 * * lys-1
* tol-1 ** * tol-1
** ** *
4 ** clec-87 *
1 * clec-87
* clec-85 clec-85
2 ** nlp-29
**
** ** ** nlp-29
** ** 0.5 **
* *
0 * **
KP 6 h KP 12 h KP 24 h ** ** ** ** **
0
–2 KP 6 h KP 12 h KP 24 h
(E) 7
**
6 **
Fold change in expression
5
4
3 *
2
1
0
rmpA oxyR uge
FIGURE 23.3 The increased susceptibility of p38 MAPK pathway mutants against K. pneumoniae infection (A).
Changes in the expression profile of the tol-1 mutant IG10 (B), sek-1 mutant AU37 (C), pmk-1 mutant KU25 (D), and
regulation of selective virulence factors in K. pneumoniae during interaction with host C. elegans (E). (Figure 23.3A–D
were adapted from Kamaladevi, A. and Balamurugan, K., Pathog. Dis., 73(5), ftv021, 2015; Figure 23.3E unpublished
data.)
Klebsiella: C. elegans as a Laboratory Model for K. pneumoniae Infection 351
genes, such as nlp-29 and lys-1 (Figure 23.3B), that are primarily regulated by p38 MAPK also strongly
suggests the role of toll-dependent p38 MAPK pathway against K. pneumoniae infection. The antimi-
crobial genes clec-60, clec-67, clec-85, and clec-87 were significantly (p < 0.005) upregulated in the
early hours and downregulated at the later hours in the AU37 (sek-1) and KU25 (pmk-1) mutants during
infection, suggesting that K. pneumoniae thwarts the host defense and causes a persistent lethal infection
(Figure 23.3C and D).
23.15 Conclusion
C. elegans has been considered as an excellent animal model to study the host immune factors and patho-
genesis of K. pneumoniae under in vivo conditions. Concerning the pathogenicity of K. pneumoniae to
C. elegans, the killing of C. elegans by K. pneumoniae is mediated by the colonization and prolifera-
tion inside the host intestine. While C. elegans utilizes the Toll-dependent p38 MAPK pathways against
K. pneumoniae infection, K. pneumoniae increases the expression of rmpA, oxyR, and uge genes during
infection. Overall, these interactions underscore the host–pathogen relationship between C. elegans and
K. pneumoniae and will form the basis for identification of potential therapeutic targets against K. pneu-
moniae and for formulation of effective immune therapy in hosts.
Acknowledgments
We thank Caenorhabditis Genetics Center, which is funded by the National Institutes of Health, National
Center for Research Resources, for providing C. elegans N2 WT, mutant strains, and E. coli OP50.
Dr. K. Balamurugan thankfully acknowledges the DBT (BT/PR14932/MED/29/233/2010), ICMR (Sanction
No: 5/3/3/13/2010-ECD-I), DST-SERC Fast Track Young Scientist Scheme [No. SR/FT/LS-83/2009 (G)],
DST-SERB (No. SR/SO/AS-80/2010), UGC Major Research Project [No. 42-222/2013 (SR)], CSIR [No.
37(1460)/11/EMR-II], DST-PURSE [Grant No. SR/S9Z-23/2010/42(G)], FIST (Grant No. SR-FST/LSI-
087/2008), and UGC-SAP-DRS-I [Grant No. F.3-28/2011 (SAP-II)], New Delhi, India, for financial assis-
tances. Financial support to AK by DBT in the form of SRF is thankfully acknowledged. The authors
gratefully acknowledge the computational and bioinformatics help provided by the Alagappa University
Bioinformatics Infrastructure Facility (funded by DBT, GOI; Grant No. BT/BI/25/001/2006).
REFERENCES
1.
Hanson, L. A. et al., Estimating global mortality from potentially foodborne diseases: an analysis using
vital registration data, Popul. Health Metrics, 10, 7, 2012.
2.
Scallan, E. et al., Foodborne illness acquired in the United States—major pathogens, Emerg. Infect.
Dis., 17, 7, 2011.
3.
Mead, P. S. et al., Food-related illness and death in the United States, Emerg. Infect. Dis., 5, 607, 1999.
352 Laboratory Models for Foodborne Infections
4. D’Orazio, S. E., Animal models for oral transmission of Listeria monocytogenes, Front. Cell Infect.
Microbiol., 4, 15, 2014.
5. Atreya, C. D., Major foodborne illness causing viruses and current status of vaccines against the dis-
eases, Foodborne Pathog. Dis., 1, 89, 2004.
6. Puspanadan, S. et al., Detection of Klebsiella pneumoniae in raw vegetables using most probable
number-polymerase chain reaction (MPN-PCR), Int. Food Res. J., 19, 1757, 2012.
7. Sabota, J. M. et al., A new variant of food poisoning: enteroinvasive Klebsiella pneumoniae and
Escherichia coli sepsis from a contaminated hamburger, Am. J. Gastroenterol., 93, 118, 1998.
8. Podschun, R. and Ullmann, U., Klebsiella spp. as nosocomial pathogens: epidemiology, taxonomy, typ-
ing methods, and pathogenicity factors, Clin. Microbiol. Rev., 11, 589, 1998.
9. Won, S. Y. et al., Emergence and rapid regional spread of Klebsiella pneumoniae carbapenemase-
producing Enterobacteriaceae, Clin. Infect. Dis., 53, 532, 2011.
10. Begun, J. et al., Staphylococcal biofilm exopolysaccharide protects against Caenorhabditis elegans
immune defenses, PLoS Pathog., 3, e57, 2007.
11. White, D. G. et al., Antimicrobial resistance among Gram-negative foodborne bacterial pathogens asso-
ciated with foods of animal origin, Foodborne Pathog. Dis., 1, 137, 2004.
12. Cao, X. et al., Molecular characterization of clinical multidrug-resistant Klebsiella pneumoniae iso-
lates, Ann. Clin. Microbiol. Antimicrob., 13, 16, 2014.
13. Yigit, H. et al., Novel carbapenem-hydrolyzing β-lactamase, KPC-1, from a carbapenem-resistant strain
of Klebsiella pneumoniae, Antimicrob. Agents Chemother., 45, 1151, 2001.
14. Müller, B. and Grossniklaus, U., Model organisms—a historical perspective, J. Proteomics, 73, 2054, 2010.
15. Rahme, L. G. et al., Common virulence factors for bacterial pathogenicity in plants and animals,
Science, 268, 1899, 1995.
16. Rahme, L. G. et al., Plants and animals share functionally common bacterial virulence factors, Proc.
Natl. Acad. Sci. USA, 97, 8815, 2000.
17. Maupas, E., Modes et formes de reproduction des nematodes, Arch. Zool. Exp. Gén., 8, 463, 1901.
18. Osche, G., Systematik und phylogenie der gattung Rhabditis (Nematoda), Zool. Jahrb. Abt. Allg. Zool.
Physiol. Tiere, 81, 190, 1952.
19. Fodor, A. et al., Comparison of a new wild-type Caenorhabditis briggsae with laboratory strains of
C. briggsae and C. elegans, Nematologica, 29, 203, 1983.
20. Lai, C. H. et al., Identification of novel human genes evolutionarily conserved in Caenorhabditis ele-
gans by comparative proteomics, Genome Res., 10, 703, 2000.
21. Brenner, S., The genetics of behaviour, Br. Med. Bull., 29, 269, 1973.
22. Hodgkin, J. et al., A novel bacterial pathogen, Microbacterium nematophilum, induces morphological
change in the nematode C. elegans, Curr. Biol., 10, 1615, 2000.
23. Nicholas, H. R. and Hodgkin, J., Innate immunity: the worm fights back, Curr. Biol., 12, R731, 2002.
24. Gravato‐Nobre, M. J. and Hodgkin, J., Caenorhabditis elegans as a model for innate immunity to patho-
gens, Cell. Microbiol., 7, 741, 2005.
25. O’Rourke, D. et al., Genomic clusters, putative pathogen recognition molecules, and antimicrobial genes
are induced by infection of C. elegans with M. nematophilum, Genome Res., 16, 1005, 2006.
26. Jansson, H. B., Adhesion of conidia of Drechmeria coniospora to Caenorhabditis elegans wild type and
mutants, J. Nematol., 26, 430, 1994.
27. Mahajan-Miklos, S. et al., Molecular mechanisms of bacterial virulence elucidated using a Pseudomonas
aeruginosa–Caenorhabditis elegans pathogenesis model, Cell, 96, 47, 1999.
28. Tan, M. W. et al., Killing of Caenorhabditis elegans by Pseudomonas aeruginosa used to model
mammalian bacterial pathogenesis, Proc. Natl. Acad. Sci. USA, 96, 715, 1999.
29. Irazoqui, J. E. et al., Distinct pathogenesis and host responses during infection of C. elegans by
P. aeruginosa and S. aureus, PLoS Pathog., 6, e1000982, 2010.
30. Mallo, G. et al., Inducible antibacterial defense system in C. elegans, Curr. Biol., 12, 1209, 2002.
31. Mellies, J. et al., The global regulator Ler is necessary for enteropathogenic Escherichia coli colonization
of Caenorhabditis elegans, Infect. Immun., 74, 64, 2006.
32. Crossman, L. C. et al., A commensal gone bad: complete genome sequence of the prototypical
enterotoxigenic Escherichia coli strain H10407, J. Bacteriol., 192, 5822, 2010.
33. Sifri, C. D. et al., Caenorhabditis elegans as a model host for Staphylococcus aureus pathogenesis,
Infect. Immun., 71, 2208, 2003.
Klebsiella: C. elegans as a Laboratory Model for K. pneumoniae Infection 353
34. Van Der Hoeven, R. et al., Ce-Duox1/BLI-3 generated reactive oxygen species trigger protective SKN-1
activity via p38 MAPK signaling during infection in C. elegans, PLoS Pathog., 7, e1002453, 2011.
35. JebaMercy, G. et al., Changes in Caenorhabditis elegans life span and selective innate immune genes
during Staphylococcus aureus infection, Folia Microbiol., 56, 373, 2011.
36. Aballay, A. et al., Salmonella typhimurium proliferates and establishes a persistent infection in the
intestine of Caenorhabditis elegans, Curr. Biol., 10, 1539, 2000.
37. Aballay, A. and Ausubel, F. M., Programmed cell death mediated by ced-3 and ced-4 protects
Caenorhabditis elegans from Salmonella typhimurium-mediated killing, Proc. Natl. Acad. Sci. USA,
98, 2735, 2001.
38. Alegado, R. A. and Tan, M. W., Resistance to antimicrobial peptides contributes to persistence of
Salmonella typhimurium in the C. elegans intestine, Cell. Microbiol., 10, 1259, 2008.
39. Burton, E. A. et al., The Caenorhabditis elegans ABL-1 tyrosine kinase is required for Shigella flexneri
pathogenesis, Appl. Environ. Microbiol., 72, 5043, 2006.
40. Kesika, P. et al., Analysis of Shigella flexneri-mediated infections in model organism Caenorhabditis
elegans, Scand. J. Infect. Dis., 43, 286, 2011.
41. Kesika, P. and Balamurugan, K., Studies on Shigella boydii infection in Caenorhabditis elegans and
bioinformatics analysis of immune regulatory protein interactions, Biochim. Biophys. Acta, 1824, 1449,
2012.
42. Zhu, J. and Mekalanos, J. J., Quorum sensing-dependent biofilms enhance colonization in Vibrio chol-
erae, Dev. Cell, 5, 647, 2003.
43. Vaitkevicius, K. et al., A Vibrio cholerae protease needed for killing of Caenorhabditis elegans has a
role in protection from natural predator grazing, Proc. Natl. Acad. Sci. USA, 103, 9280, 2006.
44. Durai, S. et al., Establishment of a Caenorhabditis elegans infection model for Vibrio alginolyticus,
J. Basic Microbiol., 51, 243, 2011.
45. Durai, S. and Balamurugan, K., Changes in Caenorhabditis elegans exposed to Vibrio parahaemolyti-
cus, J. Microbiol. Biotechnol., 21, 1026, 2011.
46. Yu, W. L. et al., Association between rmpA and magA genes and clinical syndromes caused by Klebsiella
pneumoniae in Taiwan, Clin. Infect. Dis., 42, 1351, 2006.
47. Regué, M. et al., A gene, uge, is essential for Klebsiella pneumoniae virulence, Infect. Immun., 72, 54,
2004.
48. Srinivasan, V. B. et al., Role of oxyRKP, a novel LysR-family transcriptional regulator, in antimicrobial
resistance and virulence in Klebsiella pneumoniae, Microbiology, 159, 1301, 2013.
49. Ye, P. et al., Interleukin-17 and lung host defense against Klebsiella pneumoniae infection, Am.
J. Respir. Cell Mol. Biol., 25, 335, 2001.
50. Soares, A. C. et al., Impaired host defense to Klebsiella pneumoniae infection in mice treated with the
PDE4 inhibitor rolipram, Br. J. Pharmacol., 140, 855, 2003.
51. Cortés, G. et al., Role of lung epithelial cells in defense against Klebsiella pneumoniae pneumonia,
Infect. Immun., 70, 1075, 2002.
52. Insua, J. L. et al., Modeling Klebsiella pneumoniae pathogenesis by infection of the wax moth Galleria
mellonella, Infect. Immun., 81, 3552, 2013.
53. Wu, J. H. et al., Mitogen-activated protein kinase (MAPK) signalling pathways in HepG2 cells infected
with a virulent strain of Klebsiella pneumoniae, Cell. Microbiol., 8, 1467, 2006.
54. Kamaladevi, A. and Balamurugan, K., Role of PMK-1/p38 MAPK defense in Caenorhabditis ele-
gans against Klebsiella pneumoniae infection during host–pathogen interaction, Pathog. Dis., 73(5),
ftv021, 2015.
55. Kurz, C. L. and Ewbank, J. J., Caenorhabditis elegans for the study of host–pathogen interactions,
Trends Microbiol., 8, 142, 2000.
56. Marsh, E. K. and May, R. C., Caenorhabditis elegans, a model organism for investigating immunity,
Appl. Environ. Microbiol., 78, 2075, 2012.
57. Shivers, R. P., Youngman, M. J. and Kim, D. H., Transcriptional responses to pathogens in Caenorhabditis
elegans, Curr. Opin. Microbiol., 11, 251, 2008.
58. Kim, D. H. et al., A conserved p38 MAP kinase pathway in Caenorhabditis elegans innate immunity,
Science, 297, 623, 2002.
59. Shivers, R. P. et al., Tissue-specific activities of an immune signaling module regulate physiological
responses to pathogenic and nutritional bacteria in C. elegans, Cell Host Microbe, 6, 321, 2009.
354 Laboratory Models for Foodborne Infections
60. Troemel, E. R. et al., p38 MAPK regulates expression of immune response genes and contributes to
longevity in C. elegans, PLoS Genet., 2, e183, 2006.
61. Aballay, A. et al., Caenorhabditis elegans innate immune response triggered by Salmonella enterica
requires intact LPS and is mediated by a MAPK signaling pathway, Curr. Biol., 13, 47, 2003.
62. Bolz, D. D. et al., A conserved PMK-1/p38 MAPK is required in Caenorhabditis elegans tissue-specific
immune response to Yersinia pestis infection, J. Biol. Chem., 285, 10832, 2010.
63. Shivers, R. P. et al., Phosphorylation of the conserved transcription factor ATF-7 by PMK-1 p38 MAPK
regulates innate immunity in Caenorhabditis elegans, PLoS Genet., 6, e1000892, 2010.
64. Pukkila-Worley, R. et al., Candida albicans hyphal formation and virulence assessed using a
Caenorhabditis elegans infection model, Eukaryot. Cell, 8, 1750, 2009.
65. Pukkila-Worley, R. et al., Candida albicans infection of Caenorhabditis elegans induces antifungal
immune defenses, PLoS Pathog., 7, e1002074, 2011.
24
Proteus
CONTENTS
24.1 Introduction....................................................................................................................................355
24.2 Proteus mirabilis............................................................................................................................355
24.3 Virulence Factors and Biofilm...................................................................................................... 356
24.3.1 Virulence Factors............................................................................................................. 356
24.3.2 Techniques for the Study of Virulence Factors................................................................ 358
24.3.3 Biofilm Formation and Cell-to-Cell Communication...................................................... 359
24.3.4 Techniques for Biofilm Evaluation....................................................................................361
24.4 Host–Pathogen Interaction.............................................................................................................361
24.5 Immunity and Vaccines................................................................................................................ 364
24.6 Conclusion..................................................................................................................................... 365
References............................................................................................................................................... 365
24.1 Introduction
The genus Proteus was named in 1885 by Hauser, inspired by the character of Proteus in Homer’s
Odyssey as he had the power to change his shape to escape different threats. Members of the genus
Proteus are ubiquitously distributed in the environment, and many of them are human pathogens. In
particular, P. mirabilis is an important etiological agent of opportunistic and nosocomial infections.
It causes urinary tract infections (UTIs), which can be severe in patients under catheterization or with
abnormalities in the urinary tract. Pyelonephritis, bacteremia, renal and bladder stones, and catheter
obstruction are among the most common complications in P. mirabilis infection. Occasionally, it can
cause foodborne illness as a result of mishandling during food processing.
In order to uncover its pathogenicity, several virulence factors were expressed in vitro and different
strategies have been developed to assess the contributions of these factors. For example, application of
laboratory models, from pioneer work with cell culture to animal model, has revealed valuable insights
into various aspects of P. mirabilis infection. Indeed, several conserved antigens have been evaluated as
vaccine components using a mouse model of infection. There is no doubt that a thorough understanding
of the fundamental aspects of pathogenicity and host–pathogen interaction of P. mirabilis infection will
be crucial for the formulation of new treatments and preventive strategies.
24.2 P
roteus mirabilis
Proteus mirabilis is a Gram-negative bacterium, a dimorphic and motile member of the Enterobacteriaceae
family. It has the ability to differentiate from short rods into elongated, multinucleate swarm cells that
overexpress flagella [1]. It is a commensal bacterium found in the intestinal microbiota and in the envi-
ronment, including in soil and contaminated water. Generally associated with complicated UTIs, Proteus
355
356 Laboratory Models for Foodborne Infections
may also cause a wide variety of nosocomial infections, including respiratory infections, affecting dif-
ferent organs (eyes, ears, nose, and skin, among others) [2,3].
Contamination of food by enteric pathogens can occur from the farm if human or animal sewage is
used to fertilize soil or if sewage water is used to irrigate crops. Such risks are further increased if the
food is mishandled during processing and preparation, a stage where pathogens could multiply exponen-
tially under favorable conditions [4].
Members of the family Enterobacteriaceae have been considered a potent cause of foodborne out-
breaks [5]. P. mirabilis has been frequently implicated in food deterioration and spoilage. Besides oppor-
tunistic infections, it can also cause food poisoning when consumed in contaminated food such as meat,
vegetables, and seafood [6]. Indeed, P. mirabilis is regularly detected in food. For example, the bacte-
rium was found in 7% of the beef, 10% of the chicken, and 17% of the rice from a cafeteria in Alice Town,
South Africa [7]. In China, P. mirabilis isolated from chicken products showed a high level of antibiotic
resistance, posing a potential threat to public health [8]. Moreover, the presence of blaVEB was confirmed
in a nonhuman P. mirabilis strain isolated from poultry meat in Bern, Switzerland, in addition to the
detection of blaCMY-2 in a sample imported from Austria [9]. CMY-2-positive P. mirabilis isolates are
commonly found in humans, while VEB-6 isolates are less common. Several reports showed that VEB-6
P. mirabilis is responsible for infection in other countries [9]. It is worth noting that P. mirabilis strains
harboring blaCTXM-2 were recently identified in chicken meat sold in markets in southeast Brazil [10].
them repress motility, inhibiting swarm cell differentiation [24]. Bacteria adopt a filamentous morphol-
ogy as a result of the sensor activities of flagella in contact, for example, with a urinary catheter. Contact
to solid surfaces creates a torsional change in the outer membrane, and this is sensed by an upregulator
of the flagellar master operon (Umo) protein, which induces the expression of flagella to produce the
highly flagellated cells that are required for swarming [11]. This motility is related with migration across
different abiotic surfaces including catheters.
Iron acquisition systems are an important virulence determinant that enhances bacterial colonization
of the host cells and survival in the environment [25]. Iron is an essential micronutrient for all liv-
ing organisms, and its acquisition is vital for bacteria considering that only a minor fraction is avail-
able (10 −18 M). In order to capture iron, bacteria have evolved high-affinity iron-scavenging and uptake
systems [25]. The main strategies used by bacteria are the production and uptake of siderophores and
the direct utilization of host iron compounds such as transferrin, lactoferrin, or heme-containing mol-
ecules [25]. Gram-negative Fe(III) acquisition systems usually consist of an outer membrane receptor,
with transport across the outer membrane by TonB/ExbB/ExbD complex, a periplasmic binding protein,
and an inner membrane ABC transporter. In P. mirabilis, there are at least two gene clusters related to
siderophore biosynthesis and ABC transport, three outer membrane proteins induced by iron starvation
involved in heme uptake, and a heme receptor [26,27]. One of the clusters related to siderophore biosyn-
thesis is a novel nonribosomal peptide syntheses (NRPS)-independent siderophore (NIS) named proteo-
bactin, as this was first described in a bacterium [28]. The other one contains the nrp operon, which has
been previously described to be upregulated during iron limitation [29]. This operon is encoded within
the high pathogenicity island (HPI) in P. mirabilis HI4320 that shows homology compared to the HPI of
Yersinia spp. [30]. Infection challenges with mutant strains in different genes involved in yersiniabactin-
related siderophore showed that it contributes to P. mirabilis fitness in vivo [31].
Quorum sensing describes the communication between bacterial cells, whereby a coordinated popula-
tion response is controlled by diffusible molecules produced by individuals, often known as autoinduc-
ers. The influence of quorum-sensing molecules in Proteus strains is much less known compared to
Pseudomonas or Escherichia. N-acyl homoserine lactone (AHL) signaling molecules are utilized by
several Gram-negative species to sense population density and coordinate gene expression [32]. P. mira-
bilis lacks a clear AHL synthase (LuxI) homologue and does not seem to produce this type of signaling
molecule [12,33]. However, P. mirabilis encodes a LuxR family transcriptional regulator and seems to
produce compounds with AHL-like activity. Previous studies found that the addition of exogenous AHL
to a P. mirabilis population had a strain-specific impact on virulence factor expression, swarming, and
biofilm formation [34,35]. The quorum-sensing molecule autoinducer 2 (AI-2), encoded by luxS, can
mediate both intra- and interspecies interactions. P. mirabilis possesses a luxS homologue and produces
AI-2 [36]. However, mutation of luxS in P. mirabilis str. BB2000 does not significantly affect swarming,
virulence factor production, or survival in a mouse model, suggesting that AI-2 does not contribute to
pathogenicity [36]. This lack of phenotype might indicate that P. mirabilis uses LuxS strictly as part of
the activated methyl cycle, particularly as P. mirabilis str. HI4320 contains no clear homologue of the
Lsr system for sensing and responding to AI-2. However, AI-2 produced by P. mirabilis might influence
gene expression in other species that use this signaling molecule. Other studies have reported that the
presence of a quorum-sensing molecule (N-butanoyl homoserine lactone, BHL) increased P. mirabilis
biofilm thickness and ureolytic activity. Laser interferometric determination of diffusion showed that
urea easily diffuses through P. mirabilis biofilm, while acetohydroxamic acid (AHA) is blocked. This
may suggest that the use of urease inhibitors in catheter-associated UTIs (CAUTIs) may be less effective
than in other urease-associated infections [37].
Urease is produced by P. mirabilis and is responsible for the hydrolysis of urea to carbon dioxide and
ammonia [38]. Urease production is induced by urea and is expressed during growth in urine where it
is highly active [39]. In urine, this activity elevates environmental pH and induces crystal formation
(calcium crystals and magnesium ammonium phosphate) [39]. These crystals become trapped within
the polysaccharides produced by attached bacteria, forming the characteristic crystalline biofilms on
catheters [40]. Formation of these crystalline biofilms by P. mirabilis could be considered as a protective
structure as the host immune system and antibiotics cannot penetrate the biofilm [40]. The activity of
this enzyme also causes indirect tissue damage as ammonia becomes toxic for the uroepithelial cells [41].
358 Laboratory Models for Foodborne Infections
P. mirabilis produces two toxins that are implicated in tissue damage and dissemination to the kid-
neys. Hemolysin (HpmA) is a Ca+ -dependent pore-forming cytolytic agent that destabilizes the host cell
by inserting itself into the cell membrane and causing a Na+ efflux [42]. The other is a surface-associated
cytotoxic protease proteus toxic agglutinin (Pta) that is functional only in an alkaline pH; it punctures
the host cell membrane, causing leakage of the cytosol, osmotic stress, and depolymerization of actin
filaments, affecting the integrity of the cell and inducing bladder and kidney damage in the urinary
tract [23,43].
One important virulence factor that protects bacteria against immune response is the metalloprotein-
ase ZapA, which cleaves serum and secretory immunoglobulins A1 (IgA1), IgA2, and IgG [44]. ZapA
might also cleave complement components C1q and C3; cell matrix components such as collagen, fibro-
nectin, and laminin; and cytoskeletal proteins such as actin and tubulin [45].
TABLE 24.1
Identification of P. mirabilis Microbial Factors Using Different Molecular Tools
Gene Genetic Tool References
pmfA Allelic replacement [20]
Flagellin genes Allelic replacement [46]
Protease STM [47]
Mutants with increased sensitivity to Mini-Tn5 mutagenesis [48]
antimicrobial peptides
Swarming-deficient mutants Random transposon mutagenesis [49]
Genes that contribute to pathogenesis of UTI STM [50]
64-kDa iron-regulated OMP; heme receptor Transposon mutagenesis [27]
Fimbrial mutants Allelic replacement [51]
mrpJ Site-directed mutagenesis [28]
Virulence genes STM [52]
qnrC and qnrA1 Site-directed mutagenesis [53]
fliL Null mutations [54]
mrpI; glnE; fimbrial subunit; putative MuA-like Random transposon mutagenesis [55]
DNA-binding protein; rsbA; bcr; putative
transmembrane protein; gltS; lrp; nirB; putative
lipoprotein
Proteus 359
virulence determinants required for colonization of the urinary tract (Table 24.1). They could iden-
tify genes that affected motility, iron acquisition, transcriptional regulation, phosphate transport, urease
activity, cell surface structure, and key metabolic pathways [50].
A traditional tool that allows for the identification of gene functions is random transposon mutagen-
esis. The basis of this technique is in the generation of mutant libraries that harbor a transposon insert,
which abolishes the function of the affected gene. Transposons are mobile genetic elements that can
move within the genome and can affect the function of gene expression. One of the limitations in iden-
tification and separation of nonvirulent mutants from a pool of mutants is that it is time consuming. In
P. mirabilis, there are many studies that report the use of this technique. Recently, Holling et al. [55]
used random transposon mutagenesis to identify genes involved in biofilm formation. They used a mini-
Tn5Km2 transposon that was introduced into the wild-type P. mirabilis strain B4 by conjugal transfer
from the donor organism, Escherichia coli S17.1λpir (Table 24.1). The mutants were screened by the
crystal violet microtiter plate assay to identify bacterial impairment in catheter blockage. The end adja-
cent to the transposon was sequenced in order to identify the mutated gene.
The use of null mutations is also an important strategy as it consists of the insertion of a reprogrammed
intron carried into a plasmid that finally results in a lack of function of a particular gene. Lee et al. [54]
used a fliL knockout mutant to gain further insight into the function of FliL (Table 24.1). The mutant
was generated by a reprogrammed intron, the intron was inserted between nucleotides (nt) 30 and 31 in
fliL. The loss of FliL results in cells that cannot swarm on agar surfaces but can swim in broth cultures.
and dispersion (day 7) [69]. Our group has observed that different P. mirabilis mutant strains express fla-
gella in different stages of biofilm formation generally associated with the dispersion step (Figure 24.1).
In other works, the structure of biofilms in artificial urine (AU) and LB broth, a standard laboratory
medium, has been studied. Different authors have shown that P. mirabilis biofilm formation occurs
rapidly in both LB broth and AU. When grown in LB broth, P. mirabilis produces large and structurally
complex biofilms. In AU, biofilms were less well structurally organized, contained crystalline precipi-
tates that are associated with the urease activity definitive of P. mirabilis, and contained significantly
higher numbers of the swarmer cell form of P. mirabilis [70].
Biofilm development and quorum sensing are closely interconnected processes. Signal molecules
(quorum sensing) are important factors in biofilm formation and development [71]. Different studies
have demonstrated that quorum-sensing molecules, normally associated with the regulation of virulence
factors, could also regulate the development of complex mushroom structures in a P. aeruginosa PAO1
biofilm [72]. Disruption of QS systems has been reported to affect the dynamics of biofilm formation
in some cases, but the specific mechanisms downstream of the QS regulators are, for the most part,
poorly understood [73]. Also, QS antagonists, p-nitrophenyl glycerol and tannic acid, have been shown
to inhibit the quorum-sensing system and subsequently inhibit P. mirabilis biofilm formation in artificial
urine [74].
On the other hand, Stankowska et al. [75] identified the luxS gene coding S-ribosylhomocysteine
lyase responsible for AI-2 synthesis, proving that P. mirabilis uses a quorum-sensing communication
system.
As mentioned before, P. mirabilis can live as single cells (planktonic) or as members of organized
microbial communities called biofilms, which are composed of microorganisms and the extracellular
matrix-forming polymers they produce [73]. Proteus has been shown to produce biofilms in diverse
environments, including industrial and clinical settings [76]. Urinary catheters are the medical devices
that are most frequently colonized by Proteus biofilms. Due to the deposition of crystals within these
biofilms, blockage of the urinary catheter can occur. These episodes of bacteriuria can lead to septicemia
and shock [77]. In addition, the encrustation of urinary catheters can cause trauma to the bladder mucosa
and urethra upon removal of the catheter [77]. Biofilm-based P. mirabilis lifestyle provides a protected
environment against stresses, such as desiccation, attack by the immune system, protozoa ingestion, and
antimicrobials. In the host, biofilm bacteria are protected from the host immune system and treatment
by antibiotics [77]. Other works have demonstrated the biofilm ability to induce host hyperinflammation,
as shown by elevated levels of proinflammatory cytokines [78] and matrix metalloproteases [79], and
excessive numbers of neutrophils [80]. Taking into account that several infections are associated with
biofilm formation, we can consider biofilm as another virulence factor.
FIGURE 24.1 3D reconstruction of P. mirabilis mutant strain that overexpress flagella during biofilm development. Left
panel represents 3D model of bacteria, and right panel shows flagella and extracellular matrix. Images were taken in a
FV300 Olympus Confocal Microscope.
Proteus 361
24.4 Host–Pathogen Interaction
In vivo approaches are still very useful for the study of bacterial pathogens. Although in the last years
efforts have been successful in reducing the use of animals and in refining in vivo-based experimental
methods, the host environment cannot be easily replaced by in vitro techniques.
362 Laboratory Models for Foodborne Infections
(A) (B)
×600 20µm
(C) (D)
FIGURE 24.2 Scanning electron microscopy of P. mirabilis biofilm over catheter material. (Panel A) Latex catheter
without bacteria. The round balls were over all the latex surface and correspond to zinc depositions due to manufacturing.
(Panel B) Silicon catheter without bacteria. (Panel C and D) P. mirabilis growth over latex (C) and silicon (D). In order to
allow bacteria to colonize the surface, the catheter bridge technique was employed. Briefly, small sections of the catheter
were cut and used as bridges in an agar plate (small pictures in Panel C and D). Images were presented in the 115th General
Meeting of the American Society of Microbiology, New Orleans, LA.
Although P. mirabilis is a very ubiquitous bacterium, its capacity to induce uropathogenesis is remark-
able. Animal models have proven valuable in this regard. A classic ascending UTI described initially by
Hagberg et al. [89] for the study of E. coli is still being used by several researchers. In this case, bacte-
ria are slowly introduced into the mouse bladder through a transurethral soft catheter so that different
aspects of urinary infections can be studied.
This model has been traditionally used to assess the ability of different strains to colonize bladders
and kidneys after ascending infection through the urinary tract, typically measured by bacterial count-
ing [21]. Infection of different groups of mice with the different strains or even coinfection of the same
animals with the wild type and isogenic mutants is frequently performed to assess bacterial colonization
competence.
Different studies have been developed to assess the potential of different P. mirabilis UTI treatment-
or prevention-based strategies, including vaccination, using this model. A wide range of vaccination
strategies to prevent UTI, based on different antigens or immunization routes, have been proposed using
this model [90,91].
This model has been recently refined for modern approaches like STM [52], morphological analysis
of kidneys colonization by laser confocal microscopy [92], or transcriptome analysis to compare in vitro
and in vivo P. mirabilis gene expression [93].
Also, different modifications have been performed to increase this animal model input. For example,
an infection model of permanently catheterized mice has been recently developed, which was initially
proposed for the study of Enterococcus faecalis biofilms associated with urinary catheters [94]. This
new approach will help to elucidate the molecular basis and prevention alternatives of in vivo biofilm
formation.
The adhesion of pathogenic bacteria to host cells is crucial for the establishment of the infec-
tion. Subsequent events include tissue colonization and, in some cases, cellular invasion fol-
lowed by intracellular multiplication and/or persistence [95]. Colonization of the epithelium is the
first step in the pathogenesis of P. mirabilis and has been demonstrated by in vitro and in vivo
approaches [3]. However, the mechanisms by which P. mirabilis adhere to epithelial cells are not
fully elucidated [3,96].
Pioneer work by Chippendale et al. showed that P. mirabilis could survive in the epithelial cells, but
no significant replication was noticed [97]. Oelschlaeger and Tall found that P. mirabilis strains isolated
from different sources had varied internalization efficiencies and that microfilaments were important for
Proteus 363
the process [98]. Bacteria were detected in membrane-bound vacuoles after 3 h of infection using human
renal proximal tubular epithelial cells [97].
Torzewska et al. demonstrated that not only could P. mirabilis produce urease with the concomitant
formation of calcium crystals and magnesium ammonium phosphate precipitates but it also could cause
crystallization of urine components inside the cells [99]. The production of crystals inside the cells could
induce cell damage or death, but this was not evaluated by the authors. However, in another work, it was
demonstrated for the first time that both MR/P fimbriae and flagella mediate genotoxic and cytotoxic
effects on eukaryotic cells (Figure 24.3) [16]. It has also been reported that flagella have a role in P. mira-
bilis adhesion [16] (Figure 24.3), as reported in other pathogens [100].
Considering all the information available on eukaryotic cell–Proteus interaction, we can conclude that
P. mirabilis produces several virulence factors that induce damage in cell. The lack of knowledge on the
intracellular life of P. mirabilis could be explained in part because of the wide range of virulence factors
that the bacteria produce. This should be taken into account when cell culture experiments are going to
be set up. These factors include toxins such as Pta and HpmA (bacterial toxin hemolysin) that cause cell
damage, the expression of MR/P fimbriae that mediate adhesion and cytotoxic/genotoxic damage, and
flagella that mediate adhesion and even cell death, among others.
One of the main techniques that allows for visualization of host–pathogen interaction is immunofluo-
rescence. It involves the staining of different cellular structures with antibodies coupled to fluorescent
molecules followed by observation with a fluorescent or confocal microscope. The development of this
type of technique and the use of time-lapse imaging will contribute to the understanding of infection and
to the increase of knowledge of the mechanisms involved in cell invasion.
(A) 7,00 µm
X (B) 7,00 µm
X
Y Y Y Y
7,00 µm
7,00 µm
7,00 µm
7,00 µm
X X
7,00 µm 7,00 µm
(C) 7,00 µm
X
(D) 7,00 µm
X
Y Y Y Y
7,00 µm
7,00 µm
7,00 µm
7,00 µm
X X
7,00 µm 7,00 µm
FIGURE 24.3 Confocal microscopy image of P. mirabilis wild-type adhesion to Vero cell line (kidney); bacteria is
stained with a polyclonal anti-proteus antibody (A), actin is stained with phalloidin-rhodamine (B), and DNA with DAPI
(C). Panel D shows a superimposition of the three images (A–C).
364 Laboratory Models for Foodborne Infections
be observed. On the other hand, the immune response that develops from entering uropathogens in the
urinary tract is particularly complex. The mechanisms of innate immunity would have an important
role in the UTI. The role of different mechanisms in the adaptive immune response in resolving urinary
infection is currently a matter of debate [11]. During the entrance of uropathogens, an early response
in the host that includes recruitment and activation of effector cells at the site of infection results, lead-
ing to an inflammatory response whose magnitude and location explains many of the clinical mani-
festations of UTIs [115]. This response depends on cell receptors present on the uroepithelium that
recognize pathogen-associated molecular patterns (PAMPs), which are highly conserved in different
organisms, [116] and these can be recognized by the innate immune system that then limits the action
of microbial evasion mechanisms and allows for detection of infectious agents from a limited number of
cellular receptors. It has been determined that the detection of PAMPs by toll-like receptors (TLRs) is
crucial for activation of the innate immune response and control of the adaptive immune response that
occurs later [117]. Several studies have shown that bladder epithelial cells, like macrophages, express
TLR4, which mediates a rapid cytokine response against low concentrations of LPS [116,118]. In mac-
rophages, the recognition of LPS requires a CD14 coreceptor [119], which is not present in epithelial
cells. It has also been demonstrated that TLR4 recognizes fimbriae regardless of the presence of the
CD14 [120] coreceptor. The control of UTIs in the bladder depends on TLR4 expression on urothelial
and stromal cells and is needed to drive inflammatory responses [121]. Studies of TLR4 expression in
humans with UTIs demonstrated that lower TLR4 expression attenuates host responses and promotes
asymptomatic carriage of UPEC [122]. TLR5 is essential to mediate flagellin-induced inflammatory
responses; it plays a role in control of infection in both the bladder and kidneys [123]. In humans, a
polymorphism in TLR5 gene abrogates the flagellin signaling cascade and has been associated with an
increased susceptibility to recurrent UTIs [124].
The last TLR identified and characterized is the TLR11, which is specifically expressed in murine
kidney cells [125]. The PAMP that the TLR recognizes was not identified but was observed in an in vitro
assay as a motif present in E. coli uropathogenic strains [126]. Furthermore, in TLR11 knockout mice,
a massive kidney infection has been observed by challenging the animals with a uropathogenic strain of
E. coli. These authors suggest that TLR11 has a role in the innate immune response specifically against
uropathogenic bacteria [125]. Human TLR11 gene is present but not expressed due to the existence of a
stop codon in the reading frame. This finding opens new directions such as the absence of TLR being
one reason for the particular susceptibility of humans to the ITU.
24.6 Conclusion
P. mirabilis has become an interesting model microorganism since it expresses an impressive variety
of virulence factors. Its relation with infection, particularly with UTIs, has increased the necessity for
vaccines and new therapeutic agents. Antimicrobial resistance and biofilm development over different
surfaces including food make P. mirabilis an excellent model for the study of bacterial behavior in differ-
ent conditions. The study of the basic mechanisms used by this bacterium for survival and multiplication
in different environments could contribute to the prevention of infection and even food contamination.
REFERENCES
1.
Hoeniger, J., Influence of pH on Proteus flagella, J Bacteriol., 90, 275–277, 1965.
2.
Penner J., The genera Proteus, Providencia, and Morganella, In I. Balows, G. Truper, W. Harder, and
K. Schleifer, eds., The Prokaryotes, vol. III, 2849–2853, Springer-Verlag KG, Berlin, 1992.
Rózalski, A., Sidorczyk, Z., and Kotełko, K., Potential virulence factors of Proteus bacilli, Microbiol
3.
Mol Biol Rev., 61, 65–89, 1997.
4.
Ghosh, M., Wahi, S., Kumar, M., and Ganguli, A., Prevalence of enterotoxigenic Staphylococcus aureus
and Shigella spp. in some raw street vended Indian foods, Int J Environ Health Res., 17, 151–156, 2007.
Centinkaya, F., Cibik, G., Soyuteniz, E., Ozkin, C., Kayali, R., and Levent, B., Shigella and Salmonella
5.
contamination in various foodstuffs in Turkey, J Food Control, 19, 1059–1063, 2008.
366 Laboratory Models for Foodborne Infections
6. Braide, W., Oranusi, S., Udegbunam, L., Oguoma, O., Akobondu, C., and Nwaoguikpe, R.,
Microbiological quality of an edible caterpillar of an emperor moth, Bunaea alcinoe, J Food Prot., 3,
176–180, 2011.
7. Nyenje, M., Odjadjare, C., Tanih, N., Green, E., and Ndip, R., Foodborne pathogens recovered from
ready-to-eat foods from roadside cafeterias and retail outlets in Alice, Eastern Cape Province, South
Africa: public health implications, Int J Environ Res Public Health, 9, 2608–2619, 2012.
8. Wong, M., Wan, H., and Chen, S., Characterization of multidrug-resistant Proteus mirabilis isolated
from chicken carcasses, Foodborne Pathog Dis., 10, 177–181, 2013.
9. Seiffert, S., Tinguely, R., Lupo, A., Neuwirth, C., Perreten, V., and Endimiani, A., High prevalence of
extended-spectrum-cephalosporin-resistant Enterobacteriaceae in poultry meat in Switzerland: emer-
gence of CMY-2- and VEB-6-possessing Proteus mirabilis, Antimicrob Agents Chemother., 57, 6406–
6408, 2013.
10. Casella, T., et al., Detection of bla CTX-M-type genes in complex class 1 integrons carried by
Enterobacteriaceae isolated from retail chicken meat in Brazil, Int J Food Microbiol., 197, 88–91, 2015.
11. Nielubowicz, G. R., and Mobley, H. L., Host–pathogen interactions in urinary tract infection, Nat Rev
Urol., 7, 430–441, 2010.
12. Pearson, M. M., et al., Complete genome sequence of uropathogenic Proteus mirabilis, a master of both
adherence and motility, J Bacteriol., 190, 4027–4037, 2008.
13. Zunino, P., Geymonat, L., Allen, A. G., Preston, A., Sosa, V., and Maskell, D. J., New aspects of the
role of MR/P fimbriae in Proteus mirabilis urinary tract infection, FEMS Immunol Med Microbiol., 31,
113–120, 2001.
14. Rocha, S. P., et al., Aggregative adherence of uropathogenic Proteus mirabilis to cultured epithelial
cells, FEMS Immunol Med Microbiol., 51, 319–326, 2007.
15. Jansen, A. M., Lockatell, V., Johnson, D. E., and Mobley, H. L., Mannose-resistant Proteus-like fimbriae
are produced by most Proteus mirabilis strains infecting the urinary tract, dictate the in vivo localiza-
tion of bacteria, and contribute to biofilm formation, Infect Immun., 72, 7294–7305, 2004.
16. Scavone, P., Villar, S., Umpiérrez, A., and Zunino, P., Role of Proteus mirabilis MR/P fimbriae and
flagella in adhesion, cytotoxicity and genotoxicity induction in T24 and Vero cells, Pathog Dis.,
73, 2015.
17. Li, X., Rasko, D. A., Lockatell, C. V., Johnson, D. E., and Mobley, H. L., Repression of bacterial motility
by a novel fimbrial gene product, EMBO J., 20, 4854–4862, 2011.
18. Pellegrino, R., Scavone, P., Umpiérrez, A., Maskell, D. J., and Zunino, P., Proteus mirabilis uroepithelial
cell adhesin (UCA) fimbria plays a role in the colonization of the urinary tract, Pathog Dis., 67, 104–107,
2013.
19. Zunino, P., Sosa, V., Allen, A. G., Preston, A., Schlapp, G., and Maskell, D. J., Proteus mirabilis fimbriae
(PMF) are important for both bladder and kidney colonization in mice, Microbiology, 149, 3231–3237,
2003.
20. Massad, G., and Mobley, H. L., Genetic organization and complete sequence of the Proteus mirabilis
pmf fimbrial operon, Gene, 150, 101–104, 1994.
21. Zunino, P., Geymonat, L., Allen, A. G., Legnani-Fajardo, C., and Maskell, D. J., Virulence of a Proteus
mirabilis ATF isogenic mutant is not impaired in a mouse model of ascending urinary tract infection,
FEMS Immunol Med Microbiol., 29, 137–143, 2000.
22. Bode, N. J., Debnath, I., Kuan, L., Schulfer, A., Ty, M., and Pearson, M. M., Transcriptional analysis
of the MrpJ network: modulation of diverse virulence-associated genes and direct regulation of mrp
fimbrial and flhDC flagellar operons in Proteus mirabilis, Infect Immun., 83, 2542–2556, 2015.
23. Armbruster, C. E., and Mobley, H. L. T., Merging mythology and morphology: the multifaceted lifestyle
of Proteus mirabilis, Nat Rev Microbiol., 10, 743–754, 2012.
24. Hartstein, A. I., Garber, S. B., Ward, T. T., Jones, S. R., and Morthland, V. H., Nosocomial urinary tract
infection: a prospective evaluation of 108 catheterized patients, Infect Control, 2, 380–386, 1981.
25. Wooldridge, K. G., and Williams, P. H., Iron uptake mechanisms of pathogenic bacteria, FEMS
Microbiol Rev., 12, 325–348, 1993.
26. Piccini, C. D., Barbe, F. M., and Legnani-Fajardo, C. L., Identification of iron-regulated outer membrane
proteins in uropathogenic Proteus mirabilis and its relationship with heme uptake, FEMS Microbiol
Lett., 166, 243–248, 1998.
Proteus 367
27. Lima, A., Zunino, P., D’Alessandro, B., and Piccini, C., An iron-regulated outer-membrane protein of
Proteus mirabilis is a haem receptor that plays an important role in urinary tract infection and in in vivo
growth, J Med Microbiol., 56, 1600–1607, 2007.
28. Pearson, M. M., and Mobley, H. L. T., Repression of motility during fimbrial expression: identification
of fourteen mrpJ gene paralogs in Proteus mirabilis, Mol Microbiol., 69, 548–558, 2008.
29. Gaisser, S., and Hughes, C., A locus coding for putative non-ribosomal peptide/polyketide synthase
functions is mutated in a swarming-defective Proteus mirabilis strain, Mol Gen Genet., 253, 415–427,
1997.
30. Flannery, E. L., Mody, L., and Mobley, H. L. T., Identification of a modular pathogenicity island that is
widespread among urease-producing uropathogens and shares features with a diverse group of mobile
elements, Infect Immun., 77, 4887–4894, 2009.
31. Himpsl, S. D., Pearson, M. M., Arewång, C. J., Nusca, T. D., Sherman, D. H., and Mobley, H. L. T.,
Proteobactin and a yersiniabactin-related siderophore mediate iron acquisition in Proteus mirabilis, Mol
Microbiol., 78, 138–157, 2010.
32. Boyer, M., and Wisniewski-Dyé, F., Cell–cell signalling in bacteria: not simply a matter of quorum,
FEMS Microbiol Ecol., 70, 1–19, 2009.
33. Belas, R., Schneider, R., and Melch, M., Characterization of Proteus mirabilis precocious swarming
mutants: identification of rsbA, encoding a regulator of swarming behavior, J Bacteriol., 180, 6126–
6139, 1998.
34. Bainton, N. J., et al., A general role for the lux autoinducer in bacterial cell signalling: control of antibi-
otic biosynthesis in Erwinia, Gene, 116, 87–91, 1992.
35. Stankowska, D., Kwinkowski, M., and Kaca, W., Quantification of Proteus mirabilis virulence factors
and modulation by acylated homoserine lactones, J Microbiol Immunol Infect., 41, 243–253, 2008.
36. Schneider, R., Lockatell, C. V., Johnson, D., and Belas, R., Detection and mutation of a luxS-encoded
autoinducer in Proteus mirabilis, Microbiology, 148, 773–782, 2002.
37. Czerwonka, G., Arabski, M., Wąsik, S., Jabłońska-Wawrzycka, A., Rogala, P., and Kaca, W.,
Morphological changes in Proteus mirabilis O18 biofilm under the influence of a urease inhibitor and a
homoserine lactone derivative, Arch Microbiol., 196, 169–177, 2014.
38. Griffith, D. P., Musher, D. Á., and Itin, C., Urease: the primary cause of infection-induced urinary
stones, Invest Urol., 13, 346–350, 1976.
39. Jones, B. D., and Mobley, H. L., Proteus mirabilis urease: nucleotide sequence determination and com-
parison with jack bean urease, J Bacteriol., 171, 6414–6422, 1989.
40. Flores-Mireles, A. L., Walker, J. N., Caparon, M., and Hultgren, S. J., Urinary tract infections: epidemi-
ology, mechanisms of infection and treatment options, Nat Rev Microbiol., 13, 269–284, 2015.
41. Coker, C., Poore, C. A., Li, X., and Mobley, H. L., Pathogenesis of Proteus mirabilis urinary tract infec-
tion, Microbes Infect., 2, 1497–1505, 2000.
42. Jacobsen, S. M., Stickler, D. J., Mobley, H. L. T., and Shirtliff, M. E., Complicated catheter-associated
urinary tract infections due to Escherichia coli and Proteus mirabilis, Clin Microbiol Rev., 21, 26–59,
2008.
43. Alamuri, P., and Mobley, H. L., A novel autotransporter of uropathogenic Proteus mirabilis is both a
cytotoxin and an agglutinin, Mol Microbiol., 68, 997–1017, 2008.
44. Walker, K. E., Moghaddame-Jafari, S., Lockatell, C., Johnson, D., and Belas, R., ZapA, the IgA degrad-
ing metalloprotease of Proteus mirabilis, is a virulence factor expressed specifically in swarmer cells,
Mol Microbiol., 32, 825–836, 1999.
45. Belas, R., Manos, J., and Suvanasuthi, R., Proteus mirabilis ZapA metalloprotease degrades a broad
spectrum of substrates, including antimicrobial peptides, Infect Immun., 72, 5159–5167, 2004.
46. Legnani-Fajardo, C., Zunino, P., Piccini, C., Allen, A., and Maskell, D., Defined mutants of Proteus
mirabilis lacking flagella cause ascending urinary tract infection in mice. Microb Pathog., 21, 395–405,
1996.
47. Zhao, H., Li, X., Johnson, D. E., and Mobley, H. L., Identification of protease and rpoN-associated genes
of uropathogenic Proteus mirabilis by negative selection in a mouse model of ascending urinary tract
infection, Microbiology, 145, 185–195, 1999.
48. McCoy, A. J., Liu, H., Falla, T. J., and Gunn, J. S., Identification of Proteus mirabilis mutants with
increased sensitivity to antimicrobial peptides, Antimicrob Agents Chemother., 45, 2030–2037, 2001.
368 Laboratory Models for Foodborne Infections
49. Jones, B. V., Young, R., Mahenthiralingam, E., and Stickler, D. J., Ultrastructure of Proteus mirabilis
swarmer cell rafts and role of swarming in catheter-associated urinary tract infection, Infect Immun.,
72, 3941–3950, 2004.
50. Burall, L. S., et al., Proteus mirabilis genes that contribute to pathogenesis of urinary tract infection:
identification of 25 signature-tagged mutants attenuated at least 100-fold, Infect Immun., 72, 2922–
2938, 2004.
51. Zunino, P., Sosa, V., Schlapp, G., Allen, A. G., Preston, A., and Maskell, D. J., Mannose-resistant
Proteus like and P. mirabilis fimbriae have specific and additive roles in P. mirabilis urinary tract infec-
tions, FEMS Immunol Med Microbiol., 51, 125–133, 2007.
52. Himpsl, S. D., Lockatell, C. V., Hebel, J. R., Johnson, D. E., and Mobley, H. L., Identification of vir-
ulence determinants in uropathogenic Proteus mirabilis using signature-tagged mutagenesis, J Med
Microbiol., 57, 1068–1078, 2008.
53. Guo, Q., et al., A mutational analysis and molecular dynamics simulation of quinolone resistance pro-
teins QnrA1 and QnrC from Proteus mirabilis, BMC Struct Biol., 10, 33, 2010.
54. Lee, Y.-Y., Patellis, J., and Belas, R., Activity of Proteus mirabilis FliL is viscosity dependent and
requires extragenic DNA, J Bacteriol., 195, 823–832, 2013.
55. Holling, N., et al., Elucidating the genetic basis of crystalline biofilm formation in Proteus mirabilis,
Infect Immun., 82, 1616–1626, 2014.
56. Hensel, M., Shea, J. E., Gleeson, C., Jones, M. D., Dalton, E., and Holden, D. W., Simultaneous identifi-
cation of bacterial virulence genes by negative selection, Science, 269, 400–403, 1995.
57. Mazurkiewicz, P., Tang, C. M., Boone, C., and Holden, D. W., Signature-tagged mutagenesis: barcoding
mutants for genome-wide screens, Nat Rev Genet., 7, 929–939, 2006.
58. Winzeler, E. A., et al., Functional characterization of the S. cerevisiae genome by gene deletion and
parallel analysis, Science, 285, 901–906, 1999.
59. Hall-Stoodley, L., Costerton, J. W., and Stoodley, P., Bacterial biofilms: from the natural environment to
infectious diseases, Nat Rev Microbiol., 2, 95–108, 2004.
60. Costerton, J. W., Stewart, P. S., and Greenberg, E. P., Bacterial biofilms: a common cause of persistent
infections, Science, 284, 1318–1322, 1999.
61. O’Toole, G., Kaplan, H. B., and Kolter, R., Biofilm formation as microbial development, Annu Rev
Microbiol., 54, 49–79, 2000.
62. Simões, L. C., Simões, M., and Vieira, M. J., Adhesion and biofilm formation on polystyrene by drink-
ing water-isolated bacteria, Antonie Van Leeuwenhoek, 98, 317–329, 2010.
63. Lindsay, D., and Von Holy, A., Bacterial biofilms within the clinical setting: what healthcare profession-
als should know, J Hosp Infect., 64, 313–325, 2006.
64. Kiskó, G., and Szabó-Szabó, O., Biofilm removal of Pseudomonas strains using hot water sanitation,
Acta Univ Sapientiae Alimentaria, 4, 69–79, 2011.
65. Belas, R., Biofilms, flagella, and mechanosensing of surfaces by bacteria, Trends Microbiol., 22, 517–
527, 2014.
66. Belas, R., When the swimming gets tough, the tough form a biofilm, Mol Microbiol., 90, 1–5, 2013.
67. Belas, R., Goldman, M., and Ashliman, K., Genetic analysis of Proteus mirabilis mutants defective in
swarmer cell elongation, J Bacteriol., 177, 823–828, 1995.
68. Morgenstein, R. M., Clemmer, K. M., and Rather, P. N., Loss of the WaaL O-antigen ligase prevents
surface activation of the flagellar gene cascade in Proteus mirabilis, J Bacteriol., 192, 3213–3221, 2010.
69. Schlapp, G., Scavone, P., Zunino, P., and Härtel, S., Development of 3D architecture of uropathogenic
Proteus mirabilis batch culture biofilms—a quantitative confocal microscopy approach, J Microbiol
Methods, 87, 234–240, 2011.
70. Jones, S. M., Yerly, J., Hu, Y., Ceri, H., and Martinuzzi, R., Structure of Proteus mirabilis biofilms
grown in artificial urine and standard laboratory media, FEMS Microbiol Lett., 268, 16–21, 2007.
71. Viana E. S, Campos M. E, Ponce A. R, Mantovani H. C, and Vanetti M. C., Biofilm formation and acyl
homoserine lactone production in Hafnia alvei isolated from raw milk, Biol Res., 42, 427–436, 2009.
72. Davies, D. G., Parsek, M. R., Pearson, J. P., Iglewski, B. H., Costerton, J. W., and Greenberg, E. P., The
involvement of cell-to-cell signals in the development of a bacterial biofilm, Science, 280, 295–298,
1998.
73. Monds, R. D., and O’Toole, G. A., The developmental model of microbial biofilms: ten years of a para-
digm up for review, Trends Microbiol., 17, 73–87, 2009.
Proteus 369
74. Jones, S. M., Dang, T. T., and Martinuzzi, R., Use of quorum sensing antagonists to deter the formation
of crystalline Proteus mirabilis biofilms, Int J Antimicrob Agents, 34, 360–364, 2009.
75. Stankowska, D., Czerwonka, G., Rozalska, S., Grosicka, M., Dziadek, J., and Kaca, W., Influence of
quorum sensing signal molecules on biofilm formation in Proteus mirabilis O18, Folia Microbiol., 57,
53–60, 2012.
76. Costerton, J. W., et al., Bacterial biofilms in nature and disease, Annu Rev Microbiol., 41, 435–464,
1987.
77. Stickler, D. J., and Zimakoff, J., Complications of urinary tract infections associated with devices used
for long-term bladder management, J Hosp Infect., 28, 177–194, 1994.
78. Trengove, N. J., Langton, S. R., and Stacey, M. C., Biochemical analysis of wound fluid from nonhealing
and healing chronic leg ulcers, Wound Repair Regen., 4, 234–239, 1996.
79. Trengove, N. J., et al., Analysis of the acute and chronic wound environments: the role of proteases and
their inhibitors, Wound Repair Regen., 7, 442–452, 1999.
80. Diegelmann, R. F., Excessive neutrophils characterize chronic pressure ulcers, Wound Repair Regen.,
11, 490–495, 2003.
81. O’Toole, G. A., and Kolter, R., Initiation of biofilm formation in Pseudomonas fluorescens WCS365
proceeds via multiple, convergent signalling pathways: a genetic analysis, Mol Microbiol., 28, 449–461,
1998.
82. Kanamaru, S., et al., Increased biofilm formation in Escherichia coli isolated from acute prostatitis, Int
J Antimicrob Agents, 28, 21–25, 2006.
83. Klausen, M., et al., Biofilm formation by Pseudomonas aeruginosa wild type, flagella and type IV pili
mutants, Mol Microbiol., 48, 1511–1524, 2003.
84. Pratt, L. A., and Kolter, R., Genetic analyses of bacterial biofilm formation. Curr Opin Microbiol., 2,
598–603, 1999.
85. Watnick, P. I., and Kolter, R., Steps in the development of a Vibrio cholerae El Tor biofilm, Mol
Microbiol., 34, 586–595, 1999.
86. Neu, T. R., and Lawrence, J. R., Investigation of microbial biofilm structure by laser scanning micros-
copy, In K. Muffler, and R. Ulber, eds., Productive Biofilms, 1–51, Springer International Publishing,
Berlin, Heidelberg, 2014.
87. Ustione, A., and Piston, D. W., A simple introduction to multiphoton microscopy, J Microsc., 243,
221–226, 2011.
88. Cox, A. J., and Hukins, D. W., Morphology of mineral deposits on encrusted urinary catheters investi-
gated by scanning electron microscopy, J Urol., 142, 1347–1350, 1989.
89. Hagberg, L., Engberg, I., Freter, R., Lam, J., Olling, S., and Eden, C. S., Ascending, unobstructed uri-
nary tract infection in mice caused by pyelonephritogenic Escherichia coli of human origin, Infect
Immun., 40, 273–283, 1983.
90. Scavone, P., Umpiérrez, A., Maskell, D. J., and Zunino, P., Nasal immunization with attenuated
Salmonella Typhimurium expressing an MrpA–TetC fusion protein significantly reduces Proteus mira-
bilis colonization in the mouse urinary tract, J Med Microbiol., 60, 899–904, 2011.
91. Habibi, M., Karam, M. R. A., Shokrgozar, M. A., Oloomi, M., Jafari, A., and Bouzari, S., Intranasal
immunization with fusion protein MrpH·FimH and MPL adjuvant confers protection against urinary
tract infections caused by uropathogenic Escherichia coli and Proteus mirabilis, Mol Immunol., 64,
285–294, 2015.
92. Jansen, A. M., Lockatell, C. V., Johnson, D. E., and Mobley, H. L., Visualization of Proteus mirabilis
morphotypes in the urinary tract: the elongated swarmer cell is rarely observed in ascending urinary
tract infection, Infect Immun., 71, 3607–3613, 2003.
93. Pearson, M. M., Yep, A., Smith, S. N., and Mobley, H. L., Transcriptome of Proteus mirabilis in the
murine urinary tract: virulence and nitrogen assimilation gene expression, Infect Immun., 79, 2619–
2631, 2011.
94. Guiton, P. S., Hung, C. S., Hancock, L. E., Caparon, M. G., and Hultgren, S. J., Enterococcal biofilm
formation and virulence in an optimized murine model of foreign body-associated urinary tract infec-
tions, Infect Immun., 78, 4166–4175, 2010.
95. Alamuri, P., Löwer, M., Hiss, J. A., Himpsl, S. D., Schneider, G., and Mobley, H. L. T., Adhesion, inva-
sion, and agglutination mediated by two trimeric autotransporters in the human uropathogen Proteus
mirabilis, Infect Immun., 78, 4882–4894, 2010.
370 Laboratory Models for Foodborne Infections
96. Bahrani, F. K., and Mobley, H. L., Proteus mirabilis MR/P fimbrial operon: genetic organization, nucle-
otide sequence, and conditions for expression, J Bacteriol., 176, 3412–3419, 1994.
97. Chippendale, G. R., Warren, J. W., Trifillis, A. L., and Mobley, H. L., Internalization of Proteus mirabi-
lis by human renal epithelial cells, Infect Immun., 62, 3115–3121, 1994.
98. Oelschlaeger, T. A., and Tall, B. D., Uptake pathways of clinical isolates of Proteus mirabilis into human
epithelial cell lines, Microb Pathog., 21, 1–16, 1996.
99. Torzewska, A., Budzyńska, A., Białczak-Kokot, M., and Różalski, A., In vitro studies of epithelium-
associated crystallization caused by uropathogens during urinary calculi development, Microb Pathog.,
71, 25–31, 2014.
100. Duan Q, Zhou M, and Zhu L., Flagella and bacterial pathogenicity, J Basic Microbiol., 52, 1–8, 2012.
101. Foxman, B., The epidemiology of urinary tract infection, Nat Rev Urol., 7, 653–660, 2010.
102. Johnson, J. R., and Brown, J. J., Defining inoculation conditions for the mouse model of ascending uri-
nary tract infection that avoid immediate vesicoureteral reflux yet produce renal and bladder infection,
J Infect Dis., 173, 746–749, 1996.
103. Russo, T. A., Stapleton, A., Wenderoth, S., Hooton, T. M., and Stamm, W. E., Chromosomal restric-
tion fragment length polymorphism analysis of Escherichia coli strains causing recurrent urinary tract
infections in young women, J Infect Dis., 172, 440–445, 1995.
104. Mobley, H. L., and Chippendale, G. R., Hemagglutinin, urease, and hemolysin production by Proteus
mirabilis from clinical sources, J Infect Dis., 161, 525–530, 1990.
105. Mobley, H. L., Vaccines against Escherichia coli and Proteus urinary infections, In M. M. Levine, G. C.
Woodrow, J. B. Kaper, and G. S. Cobon, eds., New Generation Vaccines, 2nd edition, 931–944, Marcel
Dekker, Inc., New York, 1990.
106. O’Hanley, P., Prospects for urinary tract infection vaccines. In H. L. Mobley, and J. W. Warren, eds.,
Urinary Tract Infections: Molecular Pathogenesis and Clinical Management, 405–425, ASM Press,
Washington, DC, 1996.
107. Pazin, G. J., and Braude, A. I., Immobilizing antibodies in pyelonephritis, J Immunol., 102, 1454–1465,
1969.
108. Moayeri, N., Collins, C. M., and O’Hanley, P., Efficacy of a Proteus mirabilis outer membrane protein
vaccine in preventing experimental Proteus pyelonephritis in a BALB/c mouse model, Infect Immun.,
59, 3778–3786, 1991.
109. Legnani-Fajardo, C., Zunino, P., Algorta, G., and Laborde, H. F., Antigenic and immunogenic activ-
ity of flagella and fimbriae preparations from uropathogenic Proteus mirabilis, Can J Microbiol., 37,
325–328, 1991.
110. Pellegrino, R., Galvalisi, U., Scavone, P., Sosa, V., and Zunino, P., Evaluation of Proteus mirabilis struc-
tural fimbrial proteins as antigens against urinary tract infections, FEMS Immunol Med Microbiol., 36,
103–110, 2003.
111. Li, X., Lockatell, C. V., Johnson, D. E., Lane, M. C., Warren, J. W., and Mobley, H. L., Development of
an intranasal vaccine to prevent urinary tract infection by Proteus mirabilis, Infect Immun., 72, 66–75,
2004.
112. Li, X., et al., Use of translational fusion of the MrpH fimbrial adhesin-binding domain with the cholera
toxin A2 domain, coexpressed with the cholera toxin B subunit, as an intranasal vaccine to prevent
experimental urinary tract infection by Proteus mirabilis, Infect Immun., 72, 7306–7310, 2004.
113. Otczyk, D. C., and Cripps, A. W., Mucosal immunization: a realistic alternative, Hum Vaccines, 6,
978–1006, 2010.
114. Scavone, P., Sosa, V., Pellegrino, R., Galvalisi, U., and Zunino, P., Mucosal vaccination of mice with
recombinant Proteus mirabilis structural fimbrial proteins, Microbes Infect., 6, 853–860, 2004.
115. Kunin, C. M., Detection, Prevention, and Management of Urinary Tract Infections, Lea & Febiger,
Philadelphia, 1987.
116. Schilling, J. D., Mulvey, M. A., and Hultgren, S. J., Dynamic interactions between host and pathogen
during acute urinary tract infections, Urology, 57, 56–61, 2001.
117. Bowie, A. G., and Haga, I. R., The role of Toll-like receptors in the host response to viruses, Mol
Immunol., 42, 859–867, 2005.
118. Bäckhed, F., Söderhäll, M., Ekman, P., Normark, S., and Richter-Dahlfors, A., Induction of innate immune
responses by Escherichia coli and purified lipopolysaccharide correlate with organ-and cell-specific
expression of Toll-like receptors within the human urinary tract, Cell Microbiol., 3, 153–158, 2001.
Proteus 371
119. Wright, S. D., Ramos, R. A., Tobias, P. S., Ulevitch, R. J., and Mathison, J. C., CD14, a receptor for
complexes of lipopolysaccharide (LPS) and LPS binding protein, Science, 249, 1431–1433, 1990.
120. Frendéus, B., et al., Escherichia coli P fimbriae utilize the Toll-like receptor 4 pathway for cell activa-
tion, Mol Microbiol., 40, 37–51, 2001.
121. Schilling, J. D., Martin, S. M., Hung, C. S., Lorenz, R. G., and Hultgren, S. J., Toll-like receptor 4 on
stromal and hematopoietic cells mediates innate resistance to uropathogenic Escherichia coli, Proc Natl
Acad Sci USA, 100, 4203–4208, 2003.
122. Hernández, J. G., Sunden, F., Connolly, J., Svanborg, C., and Wullt, B., Genetic control of the variable
innate immune response to asymptomatic bacteriuria, PLoS One, 6, e28289, 2011.
123. Andersen-Nissen, E., Smith, K. D., Bonneau, R., Strong, R. K., and Aderem, A., A conserved surface on
Toll-like receptor 5 recognizes bacterial flagellin, J Exp Med., 204, 393–403, 2007.
124. Hawn, T. R., et al., Toll-like receptor polymorphisms and susceptibility to urinary tract infections in
adult women, PLoS One, 4, e5990, 2009.
125. Zhang, D., et al., A Toll-like receptor that prevents infection by uropathogenic bacteria, Science, 303,
1522–1526, 2004.
126. Lauw, F. N., Caffrey, D. R., and Golenbock, D. T., Of mice and man: TLR11 (finally) finds profiling,
Trends Immunol., 26, 509–511, 2005.
25
Pseudomonas aeruginosa
CONTENTS
25.1 Introduction................................................................................................................................... 373
25.2 Vertebrate Models..........................................................................................................................374
25.2.1 Cystic Fibrosis Mouse Models of Chronic Lung Infection...............................................374
25.2.2 Acute Lung Infection Mouse Models............................................................................... 377
25.2.3 P. aeruginosa Induced Gut-Derived Sepsis Mouse Models............................................. 377
25.2.4 Burn- and Open-Wound Infection Mouse Models........................................................... 378
25.2.5 P. aeruginosa Keratitis Models (Mice, Rabbits, and Guinea Pigs).................................. 378
25.2.6 Otitis Media Models (Mice, Rats, Guinea Pigs, and Chinchilla)..................................... 379
25.2.7 Zebrafish (Danio rerio)..................................................................................................... 379
25.3 Invertebrate Models....................................................................................................................... 379
25.3.1 Drosophila melanogaster................................................................................................. 379
25.3.2 Caenorhabditis elegans.................................................................................................... 380
25.3.3 Dictyostelium discoideum..................................................................................................381
25.3.4 Galleria mellonella (Wax Moth).......................................................................................381
25.3.5 Bombyx mori (Silkworm)................................................................................................. 382
25.4 Plants............................................................................................................................................. 382
25.5 Conclusions................................................................................................................................... 383
References............................................................................................................................................... 383
25.1 Introduction
Classified in the family Pseudomonadaceae, order Pseudomonadales, class Gammaproteobacteria,
the genus Pseudomonas comprises some of the most ubiquitous and diverse Gram-negative bacterial
species in nature that are capable of utilizing a wide range of organic compounds and colonizing a vari-
ety of ecological niches. Among the members of this genus, Pseudomonas aeruginosa is remarkable
for its capacity to inhabit diverse environments, including soil and water, and infect multiple organisms,
such as insects, plants, and animals.1–6 P. aeruginosa is an important opportunistic human pathogen
inflicting predominantly burn, cystic fibrosis (CF), and otherwise immunocompromised patients. It is
a frequent cause of nosocomial infections, being the most common pathogen isolated from patients
hospitalized for longer than 1 week. One reason for its high prevalence is that it is foodborne—found,
for example, in hospital water, food, and feeding tubes—and an efficient intestinal colonizer, especially
upon antibiotic treatment and surgical stress.7 Another reason is its high virulence repertoire, which
includes biofilm formation and quorum-sensing controlled factors.8,9 A third reason is its resistance to
antibiotics.
The multifaceted pathogenicity of P. aeruginosa in humans necessitates the use of various models
of infection and alternative model organisms. Due to ethical considerations and high cost of experi-
menting with vertebrate animals, invertebrates are widely used as alternative model hosts. In the
following sections of this chapter, we describe the mammalian models that recapitulate pivotal aspects
373
374 Laboratory Models for Foodborne Infections
Mouse
Caenorhabditis elegans
Rat
Ear
- Mouse/rat
- Guinea pig
Drosophila melanogaster
- Chinchilla
Eye
- Mouse/rat
- Rabbit Lung
Rabbit - Mouse/rat
- Guigea pig
- Zebrafish
FIGURE 25.1 Model organisms recapitulating aspects of human blood, wound, lung, gut, eye, or ear infection with
P. aeruginosa.
of severe P. aeruginosa pathogenicity, namely, burn and open wound, acute and persistent lung infec-
tion, and bacteremia, as well as less severe but potentially dangerous infections of the ear, eye, and
intestinal tract. Zebrafish and invertebrate models also recapitulate aspects of wound, systemic, or
intestinal/epithelial barrier infection. All established models, including those based on plants, are use-
ful for assessing virulence, the efficacy of various treatments, and the role of host defense to infection
(Figure 25.1 and Table 25.1).
25.2 Vertebrate Models
25.2.1 Cystic Fibrosis Mouse Models of Chronic Lung Infection
CF is an inherited disease of the secretory glands that is caused by mutations in the cystic fibrosis trans-
membrane conductance regulator (CFTR) gene.10 CFTR gene mutations prevent the ion channels of the
lung and other tissues from moving salt and water into and out of cells. As a result, mucus accumulates in
the lungs, trapping bacteria that cause chronic infections.10,11 P. aeruginosa is a common cause of chronic
CF lung infection that may persist for decades12 and leads to mortality in the majority of the cases, due
to progressive lung damage. In 1992, only 3 years after the identification of the CFTR gene, Snouwaert
et al. generated the first CF mouse model.10,13 Since then, several mouse models of CF and the P. aerugi-
nosa lung infection have been developed.14 Although none of them are ideal, mouse models may provide
significant information about the CF pathogenesis and are essential for the preclinical assessment of
new therapeutics.14 A method that mimics very well the human chronic lung infection of CF disease is
based on the introduction of P. aeruginosa-laden agar/agarose or alginate beads in the mouse lung by
transtracheal injection. This model was first described by Cash et al. in 1979 using rats.15 In this model,
agarose beads act as artificial biofilms and protect bacteria from a direct neutrophil attack, facilitating
Pseudomonas aeruginosa 375
TABLE 25.1
P. aeruginosa Models of Infection and Indicative Findings Using Each Model
P. aeruginosa Infection Models Indicative Findings Using the Model
Vertebrate Models
Cystic fibrosis mouse models of • Suitable model for testing anti-inflammatory compounds e.g., BIIL 284.
chronic lung infection • P. aeruginosa and B. cenocepacia interaction in the mouse lung induces
bacterial virulence and concomitant inflammatory response.
Acute lung infection mouse • Increased expression of IL-6 is associated with edema formation and
models decreased lung function.
• IL-17 facilitates neutrophil recruitment in the infected lung areas of
P. aeruginosa-infected mice.
• Immunosenescence leads to impaired neutrophil response in the lungs.
• IL-27 is implicated in sepsis-induced immunosuppression.
• A leukopenic mouse model was developed for testing novel drugs.
P. aeruginosa gut-derived sepsis • The probiotic bacterium B. longum strain BB536 suppresses the intestinal
mouse models colonization of P. aeruginosa.
• IL-1 is critical during gut-derived sepsis.
• The bacteriophage strain KPP10 decreases P. aeruginosa burden and
inflammatory response in the infected mice.
• Surgical stress induces P. aeruginosa PA-I lectin in the mouse intestine,
causing lethal sepsis.
• PA-I lectin and cytotoxic exoproducts compromise the intestinal barrier.
• PA-I lectin is highly expressed during intestinal ischemia/reperfusion injury
in mice and contributes to lethal sepsis.
• Surgical hepatectomy leads to low phosphate levels in the mouse intestine,
which is sensed by P. aeruginosa, which then, enhances its virulence causing
gut-derived sepsis.
Burn-wound and open-wound • P. aeruginosa strain PAO1 quorum-sensing mutants exhibit reduced virulence
sepsis mouse models in this infection model.
• Suitable model for testing new antibacterial agents and treatments.
• Targeting of fragellin protein can be a promising approach for the treatment
of P. aeruginosa-infected burns.
• Negative-pressure wound therapy can prevent sepsis.
• Low GstA4 expression in the muscle causes susceptibility to infection.
P. aeruginosa keratitis models Studies in rabbits:
(mice, rabbits, guinea pigs) • Numerous antimicrobial treatments against P. aeruginosa keratitis have been
assessed.
• The P. aeruginosa proteases, elastase B, and PASP contribute significantly to
the pathogenesis of keratitis, whereas alkaline protease has a lesser contribution.
Studies in mice:
• LiCl and β-catenin promote host resistance against P. aeruginosa keratitis.
• Mouse TREM-2 suppresses corneal inflammation and promotes resistance to
P. aeruginosa infection.
• MRP8/MRP14 signaling amplify the inflammatory responses and increase
corneal susceptibility.
• Extracellular matrix protein Lumican, surfactant protein SP-D, and chemokine
CXCL10 have protective roles against P. aeruginosa keratitis.
• P. aeruginosa proteases, PASP, and MucD contribute significantly to keratitis
pathogenesis.
Studies in guinea pigs:
• Evaluation of P. aeruginosa virulence factors, antimicrobial drugs,
treatments, and host defense in the course of infection.
Otitis media models (mice, rats, • Evaluation of P. aeruginosa virulence factors, inflammatory responses, and
guinea pigs, and chinchilla) treatments.
Zebrafish (Danio rerio) (injection • P. aeruginosa strains PA14 and PAO1 are pathogenic to zebrafish embryos.
or feeding) • Infection is influenced by the developmental stage of the host.
• CFTR mediates resistance against P. aeruginosa infection.
(Continued)
376 Laboratory Models for Foodborne Infections
Invertebrate Models
Drosophila melanogaster (feeding, • Humoral and cellular innate immunity is important against infection.
wounding, or injection) • PA14 escapes from the host defenses by suppressing the expression of
antimicrobial peptides and muscle genes at the wound site.
• Selection of virulence-attenuated mutants, e.g., KerV, which is a conserved
virulence factor in Proteobacteria.
• Expression of the human lactonase PON1 protects flies from P. aeruginosa
infection.
• CHD1 is important for fly intestinal resistance against P. aeruginosa.
• JNK signaling pathway synergizes with Ras1 oncogene to induce stem-cell-
mediated tumorigenesis and invasion/dissemination in the fly midgut and
hindgut, respectively.
• The RhlIR and LasIR quorum-sensing systems are important for the full
virulence in orally infected flies.
Dictyostelium discoideum (feeding) • The rhl quorum-sensing system is required for the full virulence of
P. aeruginosa strain PAO1.
• P. aeruginosa strain PA14 is more virulent than PAO1.
• trpD, pchH, and pchI mutants are attenuated in virulence not only in
D. discoideum but also in flies and mice.
Caenorhabditis elegans (feeding) • P. aeruginosa pathogenesis (slow or fast killing) against C. elegans depends
on the bacterial culture media.
• The transcription factor DAF-19, the bZIP transcription factor zip-2, and the
small organic molecule RPW-24 are important for C. elegans immune
response against P. aeruginosa strain PA14.
• HTS assays are developed for identifying novel antimicrobials.
Galleria mellonella (Wax Moth) • Different P. aeruginosa strains exhibit different strategies of evading the
(feeding or injection) immune system of G. mellonella larvae.
• P. aeruginosa metalloproteinase elastase B stimulates the humoral immune
responses in G. mellonella.
• G. mellonella oral infection model can be useful in investigating
P. aeruginosa virulence mechanisms.
Silkworm (Bombyx mori) • Screening for P. aeruginosa virulence factors in the silkworm larvae.
(feeding or injection) • The NO detoxification enzyme NO reductase is important for full virulence
of P. aeruginosa in the silkworm.
• PvdE and ExoS are important virulence factors for P. aeruginosa to cross
epithelial barriers.
Plants
• Arabidopsis thaliana • P. aeruginosa strains PA14 and PAO1 are pathogenic to A. thaliana and sweet
• Sweet basil (injection) basil.
• The antimicrobial compound rosmarinic acid protects the sweet basil root
only from P. aeruginosa quorum-sensing mutants unable to form biofilms.
• The P. aeruginosa virulence factor pyocyanin inhibits the development of
A. thaliana roots though ethylene-dependent signaling.
• PA14 pathogenicity islands, PAPI-1, and PAPI-2 carry many genes, 11 of
which are necessary for full virulence in both A. thaliana and mice.
• Salicylic acid attenuates the virulence of PA14 against A. thaliana and
C. elegans.
• P. aeruginosa-secreted proteases activate a novel A. thaliana-immune
signaling pathway.
• P. aeruginosa alkaline protease AprA cleaves the bacterial fragellin
monomers in order to prevent immune recognition in both plants and
mammals
Pseudomonas aeruginosa 377
the modeling of delayed bacterial clearance. As a consequence, there is a prolonged neutrophil influx
into the lung as well as accumulation of cytokines that resembles the Pseudomonas lung infection seen
in CF patients.16 Neutrophils are the protagonists of the excessive inflammatory response observed due to
bacterial infections in CF patients,17 but their role in pathogenicity is not clear. Accordingly, many stud-
ies are exploring the effect of specific anti-inflammatory compounds, such as BIIL 284, an antagonist of
the leukotriene B4 (LTB4)-receptor.18 LTB4 is a product of activated neutrophils and macrophages, and
once it makes a complex with its receptor, it triggers NF-κB-dependent inflammatory responses.18 BIIL
284 had previously caused adverse pulmonary reactions when given to CF patients. Therefore, Döring
et al. examined the effect of BIIL 284 treatment in mice infected intratracheally by injection with the
P. aeruginosa strain PAO1 embedded in agar beads.18 Interestingly, BIIL 284 treatment led to decreased
numbers of neutrophils, and consequently increased bacterial numbers, in the mouse lungs.18 These
observations were accompanied with strong presence of bacteria in the blood of the treated mice, com-
pared with the untreated animals, indicating the important role of neutrophils in controlling bacterial
lung infection and preventing sepsis.18 The data also show the importance of mouse infection models for
testing anti-inflammatory drugs before further assessment in clinical trials. Although most studies
are focused on a single pathogen each time, CF disease is more complicated and often includes a number
of different pathogens. Accordingly, the agar beads model of chronic lung infection has also been used
to investigate the interaction between strains of two different opportunistic pathogens, P. aeruginosa
and Burkholderia cenocepacia, in the mouse lung.8 This coinfection increases the inflammatory
response, as compared to the single infections, without any increase in the bacterial load, indicating that
P. aeruginosa interacts with other bacterial species to increase bacterial virulence.8
patients.28 Moreover, in IL-1-deficient mice, P. aeruginosa load and the inflammatory response are sig-
nificantly higher in the liver during gut-derived sepsis.29 This effect is reversed when mice are treated
with the bacteriophage strain KPP10, as compared with the phage-untreated mice.27
Another way to induce lethal gut-derived sepsis is by surgical stress (30% hepatectomy).30 Following
surgical hepatectomy in mice, P. aeruginosa expresses PA-I lectin/adhesin in the intestine of the ani-
mals, indicating that pathogens may sense host stress and respond by expressing specific virulence effec-
tors that promote lethal sepsis.30 The PA-I lectin contributes to damaging the intestinal epithelium barrier
by compromising enterocyte tight junctions.31 Moreover, P. aeruginosa senses low phosphate (Pi) levels
in the mouse intestine following surgical hepatectomy, promoting lethal gut-derived sepsis.32 Similarly,
mice subjected to intestinal ischemia/reperfusion injury exhibit P. aeruginosa PA-I lectin-dependent
translocation from the cecum to other organs including liver, lung, and kidney causing lethal sepsis.33
These, and other studies, suggest that P. aeruginosa exhibits enhanced virulence upon stress, surgery,
and trauma, all of which may promote intestinal pathologies and systemic bacterial spreading.7,30–33
pathogenesis of P. aeruginosa keratitis but also host defense.49–51 Rabbits, on the other hand, have large
eyes similar in size to those of humans and can be used to evaluate several parameters of the disease.42
The strain most commonly used to model bacterial keratitis and the efficacy of multiple treatments
against P. aeruginosa is the New Zealand white rabbit.42,52–59 For example, Chen et al. demonstrated
in two separate studies that lithium chloride (LiCl) and β-catenin promote host resistance against
P. aeruginosa keratitis by reducing the inflammatory responses of the host and by decreasing the
bacterial burden.60,61
Moreover, P. aeruginosa elastase B, protease PASP, MucD, and, to a lesser degree, alkaline protease
contribute significantly to the pathogenesis of keratitis.62–64 Furthermore, induction of the triggering
receptor expressed on myeloid cells-2 (TREM-2) upon infection in cornea scrapes triggers PI3K/Akt
signaling to confer resistance against P. aeruginosa infection.65 In contrast, induction of myeloid-related
protein-8 (MPR8) and MRP14 upon infection in cornea scrapes, despite promoting bacterial clearance,
induces inflammation and concomitant susceptibility to infection.66 Three additional proteins, the extra-
cellular matrix protein Lumican, the Surfactant Protein D (SP-D), and the C-X-C motif chemokine 10
(CXCL10) protect against P. aeruginosa keratitis in mice.67–69
25.3 Invertebrate Models
25.3.1 D rosophila melanogaster
Drosophila melanogaster (the fruit fly), despite its small size (∼2 mm in length), is a great invertebrate
model organism that adequately reflects some aspects of the mammalian pathogenesis of infection.79,80
Its short life cycle and easy rearing allows the production of up to ∼50 adult progeny per female fly within
380 Laboratory Models for Foodborne Infections
2 weeks, which facilitates the large-scale in vivo screening of bacterial mutants. Many human bacte-
rial, fungal, and viral infections can be studied in Drosophila.81 Notwithstanding the lack of adaptive
immunity as we know it in mammals, Drosophila has similar innate immunity, disease-related signaling
pathways, and cellular types to those of mammals. Thus, it is a good model for studying the pathogenic-
ity of microbial infections, including those caused by P. aeruginosa.79,80,82
There are three most common methods to infect Drosophila with P. aeruginosa2,82,83: (1) the feeding
method involves mixing of bacteria with the fly food, which causes intestinal colonization and fly lethal-
ity within a few days; (2) the thoracic or abdominal needle pricking infection, that is, an injury being
caused using a tungsten needle dipped into a bacterial suspension. Accordingly, bacteria are introduced
locally at the wound site and later on spread systemically, killing the flies within 2–4 days; and (3) the
injector pumping, which appears similar to the pricking method, but is actually a method of systemic
infection and involves the injection of a controlled dose of bacteria directly into the fly hemocoel with a
thin glass capillary tip.83
The latter two methods have been used to screen D. melanogaster for virulence-related mutants of
the P. aeruginosa strain PA14, for example the virulence-attenuating factor hudA84 and the hypotheti-
cal methyltransferase KerV, which is conserved among Proteobacteria.85 Moreover, NF-κB and JNK
signaling pathways are important for flies to resist P. aeruginosa infection,86 although highly virulent
P. aeruginosa escapes host defenses by suppressing or evading the induction by these pathways that
would normally activate antimicrobial peptides systemically and muscle genes at the wound site.40,87 In
addition, transgenic expression of the human lactonase paraoxonase-1 (PON1) in flies protects them from
P. aeruginosa wound infection by interfering with the bacterial quorum sensing.88 Thus, human innate
immunity factors such as PON1 can be introduced and studied in Drosophila.89
Using the oral infection model, which recapitulates intestinal colonization and systemic dissemination
of P. aeruginosa, new aspects of bacterial quorum sensing and intestinal pathology have been revealed.
For example, the Drosophila chromatin remodeling factor chromo helicase domain protein 1 (CHD1)
contributes to fly intestinal resistance to P. aeruginosa infection90 and the quorum-sensing factor rhlR
contributes to circumvent the fly cellular immune response when bacteria escape the intestine and spread
systemically.91 Actually, both RhlIR and LasIR quorum-sensing systems are required for full virulence
in orally infected flies.92 In addition, intestinal P. aeruginosa senses Gram-positive bacterial peptido-
glycan to enhance its quorum-sensing-mediated virulence.93 Strikingly, intestinal P. aeruginosa and the
quorum-sensing-produced virulence factor pyocyanin induce intestinal stem-cell-mediated regeneration,
which facilitates tumorigenesis in the presence of oncogenes or in the absence of tumor suppressor
genes.94 Moreover, the activation of the JNK innate immune signaling pathway in the adult Drosophila
hindgut cells during P. aeruginosa infection synergizes with Ras1V12 oncogene expression to induce
enterocyte invasion and dissemination to distant sites.95,96
before they die.106 This phenomenon named “red death” occurs due to the activation of three systems:
the phosphate signaling (PhoB), the MvfR-PQS quorum-sensing system, and the pyoverdine iron acqui-
sition system.106 Recently, Kirienko et al. established a liquid-based killing assay to show that pyover-
dine causes hypoxia-related toxicity to C. elegans and that pyoverdine production by the PA14 strain is
necessary for killing the worms.108
Regarding host defense, many factors important for C. elegans innate immunity against P. aeruginosa
were identified within the last decade, including DAF-19, the ortholog of the human RFX, zip-2, a bZIP
transcription factor and a small organic molecule, 2N(3chloro-4methylphenyl)-quinazoline-2,4diamine
(or RPW-24).99–101 Moreover, Conery et al. established a high-throughput screening (HTS) protocol in
C. elegans for the identification of novel anti-infectives against P. aeruginosa.102 Similarly, Zhou et al.
developed an HTS assay for secondary metabolites of endophytic fungi using extracts of medicinal
plants associated with these fungi to identify bioactive molecules that prolong the survival of C. elegans
after P. aeruginosa infection.103 With the caveat that HTS hits may not be validated in other systems and
that extracts do not provide information on specific chemicals, such studies might serve as a starting
point for the discovery of novel therapeutics.
25.4 Plants
Arabidopsis thaliana is a popular model plant due to its small size, its short life cycle of only 6 weeks—
required for each seed to germinate into a mature plant and produce ∼5000 new seeds—and the large number
of mutant lines and genetic tools available.135 It is also a model host for studying various aspects of infection,
including host immune responses and microbial virulence strategies.4 P. aeruginosa pathogenicity against
A. thaliana involves various steps.136 Infection starts with the syringe-mediated application of the bacteria
on the leaf surface. The bacteria then attach and congregate and enter the plant tissues via stomatal open-
ings.136 Next, bacteria proliferate locally in the substomatal cavity and the intercellular space and destroy
the local plant cells before dispersing systemically.136 P. aeruginosa strains PA14 and PAO1 can infect the
roots of both A. thaliana and sweet basil (Ocimum basilium), killing these plants within a week.137 At the
initial stage of infection, the two strains form biofilm structures on the root surfaces of the plants, which is
reminiscent of the condition of the lungs of cystic fibrosis patients.137 Biofilms confer antibiotic resistance
and persistent pathogenicity to the organism. Accordingly, the antimicrobial compound rosmarinic acid,
a multifunctional caffeic acid ester secreted from the sweet basil root, shows some in vitro activity only
against P. aeruginosa strains unable to form biofilms.137 Also, A. thaliana root development is inflicted by
the multihost P. aeruginosa virulence factor pyocyanin, which induces the production of ROS and sub-
sequent A. thaliana ethylene-dependent signaling.138 The disaccharide trehalose of P. aeruginosa strain
PA14 is a virulence factor that promotes pathogenesis only in A. thaliana and not in other hosts including
mice.139 However, He et al., demonstrated that PA14 carries two pathogenicity islands, PAPI-1 and PAPI-2,
that harbor virulence genes, 11 of which are necessary for full virulence in both A. thaliana and in mice.140
The majority of those genes are present in P. aeruginosa clinical isolates,140 indicating that PA14 could
survive in evolutionarily diverse hosts by using conserved functions. Accordingly, Starkey and Rahme
have published infectivity protocols of A. thaliana and lettuce for screening P. aeruginosa bacterial strains
to identify virulence factors potentially conserved for pathogenicity against other hosts.141 A high through
put (HTP) P. aeruginosa-infection system of Arabidopsis seedlings was also proposed by Gopalan and
Ausubel for the discovery of potent anti-infective agents.142 Furthermore, the A. thaliana pathogenicity
model was used for the identification of host defense mechanisms and factors that repress P. aeruginosa
virulence. For example, plant-derived salicylic acid, which is a phenolic metabolite, attenuates P. aerugi-
nosa virulence against A. thaliana and C. elegans by downregulating the production of several virulence
factors, including pyocyanin, protease, and elastase and by reducing biofilm formation.143 Remarkably,
Cheng et al., discovered a novel A. thaliana immune signaling pathway, namely, a mitogen-activated pro-
tein kinase (MAPK) cascade that is activated by proteases secreted by P. aeruginosa.144 Nevertheless,
P. aeruginosa has developed mechanisms to escape from the immune system of both plants and mammals.
For example, P. aeruginosa alkaline protease AprA cleaves the bacterial fragellin monomers, which are
ligands of pattern-recognition receptors in both plants and mammals, preventing the recognition and clear-
ance of the bacteria from the host.145
Pseudomonas aeruginosa 383
25.5 Conclusions
While all described models are useful, none is ideal. The main asset of mammalian models lies in
the close recapitulation of the human disease, especially regarding host response. Nevertheless, zebraf-
ish and invertebrate models also mimic, to a lesser degree, the pathophysiology of systemic, wound,
or intestinal/barrier epithelial infections. All models, including plants, are able to elucidate aspects of
host defense, including adaptive and innate immunity in the case of mammals and innate immunity in
the case of all the rest. Importantly, all models are suitable for screening P. aeruginosa virulence fac-
tors. As different models may reflect different aspects of infections, involve different virulence factors
and require different treatments may be relevant in each case, it is not clear if some models are always
superior to the others. Therefore, it is valuable to use as many different models of infection as possible
in order to generate the strongest evidence for the relevance of a P. aeruginosa gene, and to provide the
soundest rationale for the implementation of an anti-P. aeruginosa measure.
REFERENCES
1. Filion, G. & Charette, S. J. Assessing Pseudomonas aeruginosa virulence using a nonmammalian host:
Dictyostelium discoideum. Methods Mol. Biol. 1149, 671–680 (2014).
2. Haller, S., Limmer, S. & Ferrandon, D. Assessing Pseudomonas virulence with a nonmammalian host:
Drosophila melanogaster. Methods Mol. Biol. 1149, 723–740 (2014).
3. Koch, G., Nadal-Jimenez, P., Cool, R. H. & Quax, W. J. Assessing Pseudomonas virulence with non-
mammalian host: Galleria mellonella. Methods Mol. Biol. 1149, 681–688 (2014).
4. Baldini, R. L., Starkey, M. & Rahme, L. G. Assessing Pseudomonas virulence with the nonmammalian
host model: Arabidopsis thaliana. Methods Mol. Biol. 1149, 689–697 (2014).
5. Llamas, M. A. & van der Sar, A. M. Assessing Pseudomonas virulence with nonmammalian host:
zebrafish. Methods Mol. Biol. 1149, 709–721 (2014).
6. Munder, A. & Tummler, B. Assessing Pseudomonas virulence using mammalian models: acute infection
model. Methods Mol. Biol. 1149, 773–791 (2014).
7. Markou, P. & Apidianakis, Y. Pathogenesis of intestinal Pseudomonas aeruginosa infection in patients
with cancer. Front. Cell. Infect. Microbiol. 3, 115 (2014).
8. Bragonzi, A. et al. Modelling co-infection of the cystic fibrosis lung by Pseudomonas aeruginosa and
Burkholderia cenocepacia reveals influences on biofilm formation and host response. PLoS One 7,
e52330 (2012).
9. Rumbaugh, K. P., Griswold, J. A., Iglewski, B. H. & Hamood, A. N. Contribution of quorum sensing to the
virulence of Pseudomonas aeruginosa in burn wound infections. Infect. Immun. 67, 5854–5862 (1999).
10. Guilbault, C., Saeed, Z., Downey, G. P. & Radzioch, D. Cystic fibrosis mouse models. Am. J. Respir.
Cell Mol. Biol. 36, 1–7 (2007).
11. Schwiebert, E. M., Benos, D. J., Egan, M. E., Stutts, M. J. & Guggino, W. B. CFTR is a conductance
regulator as well as a chloride channel. Physiol. Rev. 79, S145–S166 (1999).
12. Turner, K. H., Wessel, A. K., Palmer, G. C., Murray, J. L. & Whiteley, M. Essential genome of
Pseudomonas aeruginosa in cystic fibrosis sputum. Proc. Natl. Acad. Sci. USA 112, 4110–4115 (2015).
13. Snouwaert, J. N. et al. An animal model for cystic fibrosis made by gene targeting. Science 257,
1083–1088 (1992).
14. Bragonzi, A. Murine models of acute and chronic lung infection with cystic fibrosis pathogens. Int.
J. Med. Microbiol. 300, 584–593 (2010).
15. Cash, H. A., Woods, D. E., McCullough, B., Johanson, W. G., Jr. & Bass, J. A. A rat model of chronic
respiratory infection with Pseudomonas aeruginosa. Am. Rev. Respir. Dis. 119, 453–459 (1979).
16. Kukavica-Ibrulj, I., Facchini, M., Cigana, C., Levesque, R. C. & Bragonzi, A. Assessing Pseudomonas
aeruginosa virulence and the host response using murine models of acute and chronic lung infection.
Methods Mol. Biol. 1149, 757–771 (2014).
17. Heeckeren, A. et al. Excessive inflammatory response of cystic fibrosis mice to bronchopulmonary
infection with Pseudomonas aeruginosa. J. Clin. Invest. 100, 2810–2815 (1997).
18. Döring, G. et al. BIIL 284 reduces neutrophil numbers but increases P. aeruginosa bacteremia and
inflammation in mouse lungs. J. Cyst. Fibros. 13, 156–163 (2014).
384 Laboratory Models for Foodborne Infections
19. Mizgerd, J. P. & Skerrett, S. J. Animal models of human pneumonia. Am. J. Physiol. Lung Cell. Mol.
Physiol. 294, L387–L398 (2008).
20. Munder, A. et al. Acute intratracheal Pseudomonas aeruginosa infection in cystic fibrosis mice is
age-independent. Respir. Res. 12, 148. doi: 10.1186/1465-9921-12-148 (2011).
21. Wolbeling, F. et al. Lung function and inflammation during murine Pseudomonas aeruginosa airway
infection. Immunobiology 216, 901–908 (2011).
22. Xu, X. et al. Role of Interleukin-17 in defense against Pseudomonas aeruginosa infection in lungs. Int.
J. Clin. Exp. Med. 7, 809–816 (2014).
23. Chen, M. M., Palmer, J. L., Plackett, T. P., Deburghgraeve, C. R. & Kovacs, E. J. Age-related differences
in the neutrophil response to pulmonary Pseudomonas infection. Exp. Gerontol. 54, 42–46 (2014).
24. Cao, J. et al. IL-27 controls sepsis-induced impairment of lung antibacterial host defence. Thorax 69,
926–937 (2014).
25. Lawrenz, M. B. et al. Development and evaluation of murine lung-specific disease models for
Pseudomonas aeruginosa applicable to therapeutic testing. Pathog. Dis. 73(5). pii: ftv025. doi: 10.1093/
femspd/ftv025 (2015).
26. Hirakata, Y. et al. Efficacy of erythromycin lactobionate for treating Pseudomonas aeruginosa bacteremia
in mice. Antimicrob. Agents Chemother. 36, 1198–1203 (1992).
27. Watanabe, R. et al. Efficacy of bacteriophage therapy against gut-derived sepsis caused by Pseudomonas
aeruginosa in mice. Antimicrob. Agents Chemother. 51, 446–452 (2007).
28. Matsumoto, T. et al. Oral administration of Bifidobacterium longum prevents gut-derived Pseudomonas
aeruginosa sepsis in mice. J. Appl. Microbiol. 104, 672–680 (2008).
29. Horino, T. et al. Interleukin-1-deficient mice exhibit high sensitivity to gut-derived sepsis caused by
Pseudomonas aeruginosa. Cytokine 30, 339–346 (2005).
30. Alverdy, J. et al. Gut-derived sepsis occurs when the right pathogen with the right virulence genes meets
the right host: evidence for in vivo virulence expression in Pseudomonas aeruginosa. Ann. Surg. 232,
480–489 (2000).
31. Laughlin, R. S. et al. The key role of Pseudomonas aeruginosa PA-I lectin on experimental gut-derived
sepsis. Ann. Surg. 232, 133–142 (2000).
32. Long, J., Zaborina, O., Holbrook, C., Zaborin, A. & Alverdy, J. Depletion of intestinal phosphate after
operative injury activates the virulence of P. aeruginosa causing lethal gut-derived sepsis. Surgery 144,
189–197 (2008).
33. Fink, D. et al. Pseudomonas aeruginosa potentiates the lethal effect of intestinal ischemia-reperfusion
injury: the role of in vivo virulence activation. J. Trauma 71, 1575–1582 (2011).
34. Stieritz, D. D. & Holder, I. A. Experimental studies of the pathogenesis of infections due to Pseudomonas
aeruginosa: description of a burned mouse model. J. Infect. Dis. 131, 688–691 (1975).
35. Li, N. et al. Systemic inflammatory responses and multiple organ dysfunction syndrome following skin
burn wound and Pseudomonas aeruginosa infection in mice. Shock 40, 152–159 (2013).
36. Barnea, Y. et al. The establishment of a Pseudomonas aeruginosa-infected burn-wound sepsis model
and the effect of imipenem treatment. Ann. Plast. Surg. 56, 674–679 (2006).
37. Barnea, Y. et al. Therapy with anti-flagellin A monoclonal antibody limits Pseudomonas aeruginosa
invasiveness in a mouse burn wound sepsis model. Burns 35, 390–396 (2009).
38. Liu, Y. et al. Negative pressure wound therapy decreases mortality in a murine model of burn-wound
sepsis involving Pseudomonas aeruginosa infection. PLoS One 9, e90494 (2014).
39. Hamblin, M. R., Zahra, T., Contag, C. H., McManus, A. T. & Hasan, T. Optical monitoring and treat-
ment of potentially lethal wound infections in vivo. J. Infect. Dis. 187, 1717–1725 (2003).
40. Apidianakis, Y. et al. Involvement of skeletal muscle gene regulatory network in susceptibility to wound
infection following trauma. PLoS One 2, e1356 (2007).
41. Apidianakis, Y. et al. Down-regulation of glutathione S-transferase α4 (hGSTA4) in the muscle
of thermally injured patients is indicative of susceptibility to bacterial infection. FASEB J. 26,
730–737 (2012).
42. Marquart, M. E. Animal models of bacterial keratitis. J. Biomed. Biotechnol. 2011, 680642 (2011).
43. Davis, S. D. & Chandler, J. W. Experimental keratitis due to Pseudomonas aeruginosa: model for evalu-
ation of antimicrobial drugs. Antimicrob. Agents Chemother. 8, 350–355 (1975).
44. Davis, S. D., Sarff, L. D. & Hyndiuk, R. A. Antibiotic therapy of experimental Pseudomonas keratitis
in guinea pigs. Arch. Ophthalmol. 95, 1638–1643 (1977).
Pseudomonas aeruginosa 385
45. Davis, S. D., Sarff, L. D. & Hyndiuk, R. A. Therapeutic effect of topical antibiotic on untreated eye in
experimental keratitis. Can. J. Ophthalmol. 13, 273–276 (1978).
46. Davis, S. D., Sarff, L. D. & Hyndiuk, R. A. Bacteriologic cure of experimental Pseudomonas keratitis.
Invest. Ophthalmol. Vis. Sci. 17, 916–918 (1978).
47. Malet, F., Colin, J., Jauch, A. & Abalain, M. L. Bacterial keratitis therapy in guinea pigs with lomefloxa-
cin by initially high-followed by low-dosage regimen. Ophthalmic Res. 27, 322–329 (1995).
48. Nanbu, P. N. et al. Heat treatment enhances healing process of experimental Pseudomonas corneal
ulcer. Ophthalmic Res. 36, 218–225 (2004).
49. Van Horn, D. L., Davis, S. D., Hyndiuk, R. A. & Alpren, T. V. Pathogenesis of experimental Pseudomonas
keratitis in the guinea pig: bacteriologic, clinical, and microscopic observations. Invest. Ophthalmol.
Vis. Sci. 17, 1076–1086 (1978).
50. Ijiri, Y. et al. The role of Pseudomonas aeruginosa elastase in corneal ring abscess formation in pseu-
domonal keratitis. Graefes Arch. Clin. Exp. Ophthalmol. 231, 521–528 (1993).
51. Chusid, M. J. & Davis, S. D. Experimental bacterial keratitis in neutropenic guinea pigs: polymorpho-
nuclear leukocytes in corneal host defense. Infect. Immun. 24, 948–952 (1979).
52. Bu, P., Riske, P. S., Zaya, N. E., Carey, R. & Bouchard, C. S. A comparison of topical chlorhexidine,
ciprofloxacin, and fortified tobramycin/cefazolin in rabbit models of Staphylococcus and Pseudomonas
keratitis. J. Ocul. Pharmacol. Ther. 23, 213–220 (2007).
53. Mah, F. S., Romanowski, E. G., Kowalski, R. P., Yates, K. A. & Gordon, Y. J. Zymar (Gatifloxacin 0.3%)
shows excellent Gram-negative activity against Serratia marcescens and Pseudomonas aeruginosa in a
New Zealand white rabbit keratitis model. Cornea 26, 585–588 (2007).
54. McCormick, C. et al. Effectiveness of a new tobramycin (0.3%) and dexamethasone (0.05%)
formulation in the treatment of experimental Pseudomonas keratitis. Curr. Med. Res. Opin. 24,
1569–1575 (2008).
55. Sensoy, D. et al. Bioadhesive sulfacetamide sodium microspheres: evaluation of their effectiveness in
the treatment of bacterial keratitis caused by Staphylococcus aureus and Pseudomonas aeruginosa in a
rabbit model. Eur. J. Pharm. Biopharm. 72, 487–495 (2009).
56. Kowalski, R. P., Romanowski, E. G., Mah, F. S., Shanks, R. M. & Gordon, Y. J. Topical levofloxacin
1.5% overcomes in vitro resistance in rabbit keratitis models. Acta Ophthalmol. 88, e120–e125 (2010).
57. Sanders, M. E. et al. Comparison of besifloxacin, gatifloxacin, and moxifloxacin against strains of
Pseudomonas aeruginosa with different quinolone susceptibility patterns in a rabbit model of keratitis.
Cornea 30, 83–90 (2011).
58. Li, S. A. et al. Therapeutic potential of the antimicrobial peptide OH-CATH30 for antibiotic-resistant
Pseudomonas aeruginosa keratitis. Antimicrob. Agents Chemother. 58, 3144–3150 (2014).
59. Tajima, K. et al. In vivo challenging of polymyxins and levofloxacin eye drop against multidrug-
resistant Pseudomonas aeruginosa keratitis. J. Infect. Chemother. 20, 343–349 (2014).
60. Chen, K. et al. Lithium chloride promotes host resistance against Pseudomonas aeruginosa keratitis.
Mol. Vis. 19, 1502–1514 (2013).
61. Chen, K. et al. β-Catenin promotes host resistance against Pseudomonas aeruginosa keratitis. J. Infect.
67, 584–594 (2013).
62. Thibodeaux, B. A., Caballero, A. R., Marquart, M. E., Tommassen, J. & O’Callaghan, R. J. Corneal
virulence of Pseudomonas aeruginosa elastase B and alkaline protease produced by Pseudomonas
putida. Curr. Eye Res. 32, 373–386 (2007).
63. Tang, A., Caballero, A. R., Marquart, M. E. & O’Callaghan, R. J. Pseudomonas aeruginosa small
protease (PASP), a keratitis virulence factor. Invest. Ophthalmol. Vis. Sci. 54, 2821–2828 (2013).
64. Mochizuki, Y. et al. Pseudomonas aeruginosa MucD protease mediates keratitis by inhibiting neutro-
phil recruitment and promoting bacterial survival. Invest. Ophthalmol. Vis. Sci. 55, 240–246 (2014).
65. Sun, M. et al. TREM-2 promotes host resistance against Pseudomonas aeruginosa infection by sup-
pressing corneal inflammation via a PI3K/Akt signaling pathway. Invest. Ophthalmol. Vis. Sci. 54,
3451–3462 (2013).
66. Deng, Q. et al. MRP8/14 enhances corneal susceptibility to Pseudomonas aeruginosa infection by
amplifying inflammatory responses. Invest. Ophthalmol. Vis. Sci. 54, 1227–1234 (2013).
67. Shao, H., Scott, S. G., Nakata, C., Hamad, A. R. & Chakravarti, S. Extracellular matrix protein lumi-
can promotes clearance and resolution of Pseudomonas aeruginosa keratitis in a mouse model. PLoS
One 8, e54765 (2013).
386 Laboratory Models for Foodborne Infections
68. Heimer, S. R., Evans, D. J., Mun, J. J., Stern, M. E. & Fleiszig, S. M. Surfactant protein D contributes
to ocular defense against Pseudomonas aeruginosa in a murine model of dry eye disease. PLoS One 8,
e65797 (2013).
69. Yoon, G. S. et al. Interferon regulatory factor-1 in flagellin-induced reprogramming: potential
protective role of CXCL10 in cornea innate defense against Pseudomonas aeruginosa infection. Invest.
Ophthalmol. Vis. Sci. 54, 7510–7521 (2013).
70. Trinidad, A. et al. Tissular changes induced by Pseudomonas aeruginosa in an otitis media rat model
with tubal obstruction. Acta Otolaryngol. 127, 132–137 (2007).
71. Trune, D. R. & Zheng, Q. Y. Mouse models for human otitis media. Brain Res. 1277, 90–103 (2009).
72. Byrd, M. S. et al. Direct evaluation of Pseudomonas aeruginosa biofilm mediators in a chronic infection
model. Infect. Immun. 79, 3087–3095 (2011).
73. Hou, W., Li, X. & Xiao, H. Formation characteristics and significance of bacteria biofilm in middle ear
mucosa of rats with chronic suppurative otitis. Clin. Ear Nose Esophagus Head Neck Surg. J. (Chinese)
26, 30–33 (2012).
74. Saussereau, E. & Debarbieux, L. Bacteriophages in the experimental treatment of Pseudomonas
aeruginosa infections in mice. Adv. Virus Res. 83, 123–141 (2012).
75. Jang, C. H., Cho, Y. B., Choi, C. H., Lee, J. S. & Kang, S. I. Effect of anti-adhesion barrier solu-
tion containing ciprofloxacin-hydrocortisone on abraded mucosa with otitis media. Int. J. Pediatr.
Otorhinolaryngol. 77, 19–24 (2013).
76. Clatworthy, A. E. et al. Pseudomonas aeruginosa infection of zebrafish involves both host and pathogen
determinants. Infect. Immun. 77, 1293–1303 (2009).
77. Rawls, J. F., Mahowald, M. A., Goodman, A. L., Trent, C. M. & Gordon, J. I. In vivo imaging and
genetic analysis link bacterial motility and symbiosis in the zebrafish gut. Proc. Natl. Acad. Sci. USA
104, 7622–7627 (2007).
78. Phennicie, R. T., Sullivan, M. J., Singer, J. T., Yoder, J. A. & Kim, C. H. Specific resistance to
Pseudomonas aeruginosa infection in zebrafish is mediated by the cystic fibrosis transmembrane con-
ductance regulator. Infect. Immun. 78, 4542–4550 (2010).
79. Papaioannou, E., Utari, P. D. & Quax, W. J. Choosing an appropriate infection model to study quorum
sensing inhibition in Pseudomonas infections. Int. J. Mol. Sci. 14, 19309–19340 (2013).
80. Apidianakis, Y. & Rahme, L. G. Drosophila melanogaster as a model for human intestinal infection and
pathology. Dis. Model. Mech. 4, 21–30 (2011).
81. Panayidou, S., Ioannidou, E. & Apidianakis, Y. Human pathogenic bacteria, fungi, and viruses in
Drosophila: disease modeling, lessons, and shortcomings. Virulence 5, 253–269 (2014).
82. Fauvarque, M. O. Small flies to tackle big questions: assaying complex bacterial virulence mechanisms
using Drosophila melanogaster. Cell. Microbiol. 16, 824–833 (2014).
83. Apidianakis, Y. & Rahme, L. G. Drosophila melanogaster as a model host for studying Pseudomonas
aeruginosa infection. Nat. Protoc. 4, 1285–1294 (2009).
84. Kim, S. H., Park, S. Y., Heo, Y. J. & Cho, Y. H. Drosophila melanogaster-based screening for multihost
virulence factors of Pseudomonas aeruginosa PA14 and identification of a virulence-attenuating factor,
HudA. Infect. Immun. 76, 4152–4162 (2008).
85. An, D. et al. The pathogenic properties of a novel and conserved gene product, KerV, in proteobacteria.
PLoS One 4, e7167 (2009).
86. Lau, G. W. et al. The Drosophila melanogaster Toll pathway participates in resistance to infection by
the Gram-negative human pathogen Pseudomonas aeruginosa. Infect. Immun. 71, 4059–4066 (2003).
87. Apidianakis, Y. et al. Profiling early infection responses: Pseudomonas aeruginosa eludes host defenses by
suppressing antimicrobial peptide gene expression. Proc. Natl. Acad. Sci. USA 102, 2573–2578 (2005).
88. Stoltz, D. A. et al. Drosophila are protected from Pseudomonas aeruginosa lethality by transgenic
expression of paraoxonase-1. J. Clin. Invest. 118, 3123–3131 (2008).
89. Estin, M. L., Stoltz, D. A. & Zabner, J. Paraoxonase 1, quorum sensing, and P. aeruginosa infection: a
novel model. Adv. Exp. Med. Biol. 660, 183–193 (2010).
90. Sebald, J., Morettini, S., Podhraski, V., Lass-Florl, C. & Lusser, A. CHD1 contributes to intestinal resis-
tance against infection by P. aeruginosa in Drosophila melanogaster. PLoS One 7, e43144 (2012).
91. Limmer, S. et al. Pseudomonas aeruginosa RhlR is required to neutralize the cellular immune response in
a Drosophila melanogaster oral infection model. Proc. Natl. Acad. Sci. USA 108, 17378–17383 (2011).
Pseudomonas aeruginosa 387
92. Lutter, E. I., Purighalla, S., Duong, J. & Storey, D. G. Lethality and cooperation of Pseudomonas aeru-
ginosa quorum-sensing mutants in Drosophila melanogaster infection models. Microbiology 158,
2125–2132 (2012).
93. Korgaonkar, A., Trivedi, U., Rumbaugh, K. P. & Whiteley, M. Community surveillance enhances
Pseudomonas aeruginosa virulence during polymicrobial infection. Proc. Natl. Acad. Sci. USA 110,
1059–1064 (2013).
94. Apidianakis, Y., Pitsouli, C., Perrimon, N. & Rahme, L. Synergy between bacterial infection and genetic
predisposition in intestinal dysplasia. Proc. Natl. Acad. Sci. USA 106, 20883–20888 (2009).
95. Bangi, E., Pitsouli, C., Rahme, L. G., Cagan, R. & Apidianakis, Y. Immune response to bacteria induces
dissemination of Ras-activated Drosophila hindgut cells. EMBO Rep. 13, 569–576 (2012).
96. Christofi, T. & Apidianakis, Y. Ras-oncogenic Drosophila hindgut but not midgut cells use an
inflammation-like program to disseminate to distant sites. Gut Microbes 4, 54–59 (2013).
97. Marsh, E. K. & May, R. C. Caenorhabditis elegans, a model organism for investigating immunity. Appl.
Environ. Microbiol. 78, 2075–2081 (2012).
98. Feinbaum, R. L. et al. Genome-wide identification of Pseudomonas aeruginosa virulence-related genes
using a Caenorhabditis elegans infection model. PLoS Pathog. 8, e1002813 (2012).
99. Estes, K. A., Dunbar, T. L., Powell, J. R., Ausubel, F. M. & Troemel, E. R. bZIP transcription factor zip-2
mediates an early response to Pseudomonas aeruginosa infection in Caenorhabditis elegans. Proc.
Natl. Acad. Sci. USA 107, 2153–2158 (2010).
100. Pukkila-Worley, R. et al. Stimulation of host immune defenses by a small molecule protects C. elegans
from bacterial infection. PLoS Genet. 8, e1002733 (2012).
101. Xie, Y., Moussaif, M., Choi, S., Xu, L. & Sze, J. Y. RFX transcription factor DAF-19 regulates 5-HT
and innate immune responses to pathogenic bacteria in Caenorhabditis elegans. PLoS Genet. 9,
e1003324 (2013).
102. Conery, A. L., Larkins-Ford, J., Ausubel, F. M. & Kirienko, N. V. High-throughput screening for novel
anti-infectives using a C. elegans pathogenesis model. Curr. Protoc. Chem. Biol. 6, 25–37 (2014).
103. Zhou, Y. M. et al. An efficient and novel screening model for assessing the bioactivity of extracts against
multidrug-resistant Pseudomonas aeruginosa using Caenorhabditis elegans. Biosci. Biotechnol.
Biochem. 75, 1746–1751 (2011).
104. Tan, M. W., Mahajan-Miklos, S. & Ausubel, F. M. Killing of Caenorhabditis elegans by
Pseudomonas aeruginosa used to model mammalian bacterial pathogenesis. Proc. Natl. Acad. Sci.
USA 96, 715–720 (1999).
105. Mahajan-Miklos, S., Tan, M. W., Rahme, L. G. & Ausubel, F. M. Molecular mechanisms of bacterial
virulence elucidated using a Pseudomonas aeruginosa–Caenorhabditis elegans pathogenesis model.
Cell 96, 47–56 (1999).
106. Zaborin, A. et al. Red death in Caenorhabditis elegans caused by Pseudomonas aeruginosa PAO1.
Proc. Natl. Acad. Sci. USA 106, 6327–6332 (2009).
107. Gallagher, L. A. & Manoil, C. Pseudomonas aeruginosa PAO1 kills Caenorhabditis elegans by cyanide
poisoning. J. Bacteriol. 183, 6207–6214 (2001).
108. Kirienko, N. V. et al. Pseudomonas aeruginosa disrupts Caenorhabditis elegans iron homeostasis,
causing a hypoxic response and death. Cell Host Microbe 13, 406–416 (2013).
109. Annesley, S. J. & Fisher, P. R. Dictyostelium discoideum—a model for many reasons. Mol. Cell.
Biochem. 329, 73–91 (2009).
110. Li, S. I. & Purugganan, M. D. The cooperative amoeba: Dictyostelium as a model for social evolution.
Trends Genet. 27, 48–54 (2011).
111. Bozzaro, S., Buracco, S. & Peracino, B. Iron metabolism and resistance to infection by invasive bacteria
in the social amoeba Dictyostelium discoideum. Front. Cell. Infect. Microbiol. 3, 50 (2013).
112. Filosa, M. F. & Dengler, R. E. Ultrastructure of macrocyst formation in the cellular slime mold,
Dictyostelium mucoroides: extensive phagocytosis of amoebae by a specialized cell. Dev. Biol. 29,
1–16 (1972).
113. Gaudet, P., Williams, J. G., Fey, P. & Chisholm, R. L. An anatomy ontology to represent biological
knowledge in Dictyostelium discoideum. BMC Genomics 9, 130. doi:10.1186/1471-2164-9-130 (2008).
114. Bozzaro, S. & Eichinger, L. The professional phagocyte Dictyostelium discoideum as a model host for
bacterial pathogens. Curr. Drug Targets 12, 942–954 (2011).
388 Laboratory Models for Foodborne Infections
115. Cosson, P. et al. Pseudomonas aeruginosa virulence analyzed in a Dictyostelium discoideum host
system. J. Bacteriol. 184, 3027–3033 (2002).
116. Carilla-Latorre, S. et al. Dictyostelium transcriptional responses to Pseudomonas aeruginosa: com-
mon and specific effects from PAO1 and PA14 strains. BMC Microbiol. 8, 109. doi:10.1186/1471-2180-
8-109 (2008).
117. Alibaud, L. et al. Pseudomonas aeruginosa virulence genes identified in a Dictyostelium host model.
Cell. Microbiol. 10, 729–740 (2008).
118. Ramarao, N., Nielsen-Leroux, C. & Lereclus, D. The insect Galleria mellonella as a powerful infection
model to investigate bacterial pathogenesis. J. Vis. Exp. (70), e4392. doi:10.3791/4392 (2012).
119. Trevijano-Contador, N. & Zaragoza, O. Expanding the use of alternative models to investigate novel
aspects of immunity to microbial pathogens. Virulence 5, 454–456 (2014).
120. Fedhila, S. et al. Comparative analysis of the virulence of invertebrate and mammalian pathogenic
bacteria in the oral insect infection model Galleria mellonella. J. Invertebr. Pathol. 103, 24–29 (2010).
121. Andrejko, M., Zdybicka-Barabas, A. & Cytrynska, M. Diverse effects of Galleria mellonella infec-
tion with entomopathogenic and clinical strains of Pseudomonas aeruginosa. J. Invertebr. Pathol. 115,
14–25 (2014).
122. Mizerska-Dudka, M. & Andrejko, M. Galleria mellonella hemocytes destruction after infection with
Pseudomonas aeruginosa. J. Basic Microbiol. 54, 232–246 (2014).
123. Andrejko, M. & Mizerska-Dudka, M. Elastase B of Pseudomonas aeruginosa stimulates the humoral
immune response in the greater wax moth, Galleria mellonella. J. Invertebr. Pathol. 107, 16–26 (2011).
124. Andrejko, M. & Mizerska-Dudka, M. Effect of Pseudomonas aeruginosa elastase B on level and activ-
ity of immune proteins/peptides of Galleria mellonella hemolymph. J. Insect Sci. 12, 88 (2012).
125. Zhou, L. et al. Developmental changes for the hemolymph metabolome of silkworm (Bombyx mori L.).
J. Proteome Res. 14(5), 2331–2347 (2015).
126. Ishii, K., Hamamoto, H. & Sekimizu, K. Studies of host–pathogen interactions and immune-related
drug development using the silkworm: interdisciplinary immunology, microbiology, and pharmacology
studies. Drug Discov. Ther. 9(4), 238–246 (2015).
127. Chieda, Y. et al. Pathogenicity of gacA mutant of Pseudomonas aeruginosa PA01 in the silkworm,
Bombyx mori. FEMS Microbiol. Lett. 244, 181–186 (2005).
128. Hossain, M. S. et al. Use of silkworm larvae to study pathogenic bacterial toxins. J. Biochem. 140,
439–444 (2006).
129. Iiyama, K. et al. Effect of superoxide dismutase gene inactivation on virulence of Pseudomonas aerugi-
nosa PAO1 toward the silkworm, Bombyx mori. Appl. Environ. Microbiol. 73, 1569–1575 (2007).
130. Chieda, Y. et al. Inactivation of pyocyanin synthesis genes has no effect on the virulence of Pseudomonas
aeruginosa PAO1 toward the silkworm, Bombyx mori. FEMS Microbiol. Lett. 278, 101–107 (2008).
131. Chieda, Y. et al. Virulence of an exotoxin A-deficient strain of Pseudomonas aeruginosa toward the
silkworm, Bombyx mori. Microb. Pathog. 51, 407–414 (2011).
132. Okuda, J. et al. Complementation of the exoS gene in the pvdE pyoverdine synthesis gene-deficient
mutant of Pseudomonas aeruginosa results in recovery of the pvdE gene-mediated penetration through
the intestinal epithelial cell barrier but not the pvdE-mediated virulence in silkworms. J. Infect.
Chemother. 18, 332–340 (2012).
133. Arai, H. & Iiyama, K. Role of nitric oxide-detoxifying enzymes in the virulence of Pseudomonas
aeruginosa against the silkworm, Bombyx mori. Biosci. Biotechnol. Biochem. 77, 198–200 (2013).
134. Rangel, S. M., Diaz, M. H., Knoten, C. A., Zhang, A. & Hauser, A. R. The role of ExoS in dissemination
of Pseudomonas aeruginosa during pneumonia. PLoS Pathog. 11, e1004945 (2015).
135. Hays, J. B. Arabidopsis thaliana, a versatile model system for study of eukaryotic genome-maintenance
functions. DNA Repair (Amst) 1, 579–600 (2002).
136. Plotnikova, J. M., Rahme, L. G. & Ausubel, F. M. Pathogenesis of the human opportunistic pathogen
Pseudomonas aeruginosa PA14 in Arabidopsis. Plant Physiol. 124, 1766–1774 (2000).
137. Walker, T. S. et al. Pseudomonas aeruginosa–plant root interactions. Pathogenicity, biofilm formation,
and root exudation. Plant Physiol. 134, 320–331 (2004).
138. Ortiz-Castro, R. et al. Pyocyanin, a virulence factor produced by Pseudomonas aeruginosa, alters
root development through reactive oxygen species and ethylene signaling in Arabidopsis. Mol. Plant
Microbe Interact. 27, 364–378 (2014).
Pseudomonas aeruginosa 389
139. Djonovic, S. et al. Trehalose biosynthesis promotes Pseudomonas aeruginosa pathogenicity in plants.
PLoS Pathog. 9, e1003217 (2013).
140. He, J. et al. The broad host range pathogen Pseudomonas aeruginosa strain PA14 carries two patho-
genicity islands harboring plant and animal virulence genes. Proc. Natl. Acad. Sci. USA 101, 2530–
2535 (2004).
141. Starkey, M. & Rahme, L. G. Modeling Pseudomonas aeruginosa pathogenesis in plant hosts. Nat.
Protoc. 4, 117–124 (2009).
142. Gopalan, S. & Ausubel, F. M. A high throughput amenable Arabidopsis-P. aeruginosa system reveals a
rewired regulatory module and the utility to identify potent anti-infectives. PLoS One 6, e16381 (2011).
143. Prithiviraj, B. et al. Down regulation of virulence factors of Pseudomonas aeruginosa by salicylic acid
attenuates its virulence on Arabidopsis thaliana and Caenorhabditis elegans. Infect. Immun. 73, 5319–
5328 (2005).
144. Cheng, Z. et al. Pathogen-secreted proteases activate a novel plant immune pathway. Nature 521(7551),
213–216 (2015).
145. Bardoel, B. W. et al. Pseudomonas evades immune recognition of flagellin in both mammals and plants.
PLoS Pathog. 7, e1002206 (2011).
26
Salmonella
Dongyou Liu
CONTENTS
26.1 Introduction....................................................................................................................................391
26.1.1 Classification, Morphology, and Genomics..................................................................... 392
26.1.1.1 Classification..................................................................................................... 392
26.1.1.2 Morphology....................................................................................................... 392
26.1.1.3 Genomics.......................................................................................................... 393
26.1.2 Biology and Epidemiology............................................................................................... 393
26.1.3 Clinical Features and Pathogenesis.................................................................................. 394
26.1.3.1 Clinical Features............................................................................................... 394
26.1.3.2 Pathogenesis...................................................................................................... 395
26.1.4 Diagnosis.......................................................................................................................... 395
26.1.4.1 Serotyping and Biotyping................................................................................. 395
26.1.4.2 Virulence and Resistance Gene Profiling......................................................... 395
26.1.4.3 Plasmid Profiling.............................................................................................. 396
26.1.4.4 Epidemiological Tracking................................................................................. 396
26.1.5 Treatment and Prevention................................................................................................. 396
26.1.5.1 Treatment.......................................................................................................... 396
26.1.5.2 Prevention......................................................................................................... 396
26.2 Laboratory Models........................................................................................................................ 397
26.2.1 Animal Models................................................................................................................. 397
26.2.1.1 Mice.................................................................................................................. 397
26.2.1.2 Guinea Pigs....................................................................................................... 398
26.2.1.3 Rabbits.............................................................................................................. 398
26.2.1.4 Calves................................................................................................................ 398
26.2.1.5 Chicken............................................................................................................. 398
26.2.1.6 Nonhuman Primates......................................................................................... 398
26.2.1.7 Zebrafish........................................................................................................... 398
26.2.1.8 C . elegans Nematode........................................................................................ 398
26.2.2 In Vitro Models................................................................................................................. 398
26.3 Conclusion..................................................................................................................................... 399
References............................................................................................................................................... 399
26.1 Introduction
Salmonella is a Gram-negative bacterium that was first observed in the Peyer’s patches and spleens of
typhoid patients by Karl Eberth in 1880, and the first culture was obtained by Georg Theodor Gaffky in
1884. Following the isolation of an organism (then called “hog-cholera bacillus”) from the intestine of a
pig by Theobald Smith and Daniel Salmon in 1885, the name Salmonella enterica (Choleraesuis) was pro-
posed by Joseph Leon Lignières in 1900 to honor the contribution by the Daniel Salmon’s group. In 1892,
391
392 Laboratory Models for Foodborne Infections
a pathogen (termed Bacillus typhimurium, now known as nontyphoidal Salmonella typhimurium) asso-
ciated with typhoid fever-like disease in mice was isolated by Friedrich Loeffler. Since then, a large
number of Salmonella serovars have been identified and their roles in the causation of human restricted,
invasive, and systemic disease (typhoid fever) and noninvasive gastroenteritis (nontyphoidal Salmonella
infection) have been clearly determined.
26.1.1.2 Morphology
Salmonella is rod-shaped, non-spore-forming, predominantly motile enterobacterium of approximately
0.7–1.5 μm in diameter and 2–5 μm in length, with peritrichous flagella around the entire bacterial sur-
face. Typically, Salmonella forms transparent and colorless colonies of 1–3 mm, sometimes with dark
center, on MacConkey agar (in mixed culture, Salmonella colonies tend to clear areas of precipitated
bile produced by other organisms); red colonies with or without black centers on xylose lysine desoxy-
cholate (XLD) agar; blue-green to blue colonies with or without large, glossy black centers on Hektoen
enteric (HE) agar; and brown, gray, or black colonies on bismuth sulfite (BS) agar, with surrounding
medium being brown at first, and turning black with longer incubation, producing the so-called halo
effect. Sometimes, Salmonella may produce atypical yellow colonies with or without black centers on
HE and XLD agars or green colonies with little or no darkening of the surrounding medium on BS agar.
Interestingly, colonies formed by Proteus may appear similar to those of Salmonella; however, Proteus
colonies may swarm on blood agar (trypticase soy agar + 5% sheep blood), while Salmonella colonies
Salmonella 393
do not. Apart from Gallinarum and Pallorum serovars, most Salmonella isolates produce motile colo-
nies. It is notable that nonmotile Salmonella primary cultures may switch to the motile phase using a
Craigie tube or ditch plate. As a chemoorganotroph, Salmonella generates energy from organic sources
via oxidation and reduction reaction, and has the capacity to survive with or without oxygen (facultative
anaerobe). Salmonella is positive for catalase, nitrate reduction, methyl red, acid production from glu-
cose, mannitol, and d-mannose [1].
26.1.1.3 Genomics
The genome of S. bongori is 4.46 Mb in size, with GC content of 51.3%, and 4010 protein-coding genes.
This species possesses only a basic set of ancestral Salmonella virulence functions and lacks several
S. enterica-defining metabolic pathways. However, the S. bongori genome contains genes encoding novel
effector proteins (sboA-L) that may have originated from enteropathogenic Escherichia coli (EPEC).
Thus, by inheriting the ancestral Salmonella virulence gene set, and incorporating virulence determi-
nants that resemble those in EPEC, S. bongori may represent an evolutionary link between E. coli and
S. enterica [2].
The genome of typhoid-fever-causing S. Typhi strain CT18 consists of a 4.81-Mb chromosome (with
GC content of 52%) and two plasmids [including a 218-kb multiple-drug-resistance (MDR) incH1 plas-
mid (pHCM1), and a 106-kb cryptic plasmid (pHCM2, possibly derived from a virulence plasmid of
Yersinia pestis)]. Within the chromosome, hundreds of insertions and deletions (ranging in size from
single genes to large islands), 4455 coding genes, and 233 pseudogenes (with some corresponding to
those related to virulence in S. Typhimurium) are found [3].
The genome of gastroenteritis and food-poisoning-inducing S. Typhimurium strain LT2 is composed
of a 4.86-Mb chromosome (with GC content of 52.2%) and a 94-kb virulence plasmid (pSLT). Within the
chromosome, 4631 coding genes and 54 pseudogenes are identified. Compared to those in the S. typhi
genome, 11% of the coding genes in the S. Typhimurium LT2 genome are unique [4].
Salmonella serovars harbor large discrete genomic islands that contain prophage elements and spe-
cialized loci termed Salmonella pathogenicity islands (SPIs). Many Salmonella-specific functions [e.g.,
complete type III secretion systems encoded by SPI-1 (T3SS-1) and SPI-2 (T3SS-2)] may be traced to
the genes located in these large discrete genomic islands, and some of these functions may have been
acquired by S. enterica after splitting from S. bongori. For example, S. bongori lacks SPI-2 (essential for
optimal replication within macrophages), SPI-6 (encoding a type VI secretion system), SPI-13 (required
for survival in chicken macrophages), SPI-14 (encoding an electron transport system), and SPI-16
(bacteriophage remnant carrying genes associated with LPS modification), all of which are present in
S. enterica [2].
amphibians may be passed to animal handlers. Salmonella may persist in a contaminated bathroom for
weeks, and tolerates freezing, although it is sensitive to UV light and heating at 55°C for 90 min, or 60°C
for 12 min.
Both typhoid and nontyphoid Salmonella serovars are distributed throughout the world. The etio-
logic agents of typhoid and paratyphoid fever, S. Typhi and S. Paratyphi A and B (and occasionally C),
collectively known as the enteric fever serovars, thrive in places with a limited sanitation infrastructure
that permits transmission between infected humans without involvement of animal hosts. With the abil-
ity to spread to the deeper tissues of humans (liver, spleen, and bone marrow), S. Typhi [serologically O
(lipopolysaccharide) type 09, 012; H (flagellin) type d; and Vi (extracellular capsule) positive] is respon-
sible for a serious invasive bacterial disease of humans, with an annual global burden of approximately
22 × 107 infections and >200,000 deaths [1,6].
Although infection with NTS usually occurs after ingestion of foods that contain a high concentra-
tion of bacteria, infection through inhalation of bacteria-laden dust is possible in infants. Important
nontyphoidal Salmonella serovars include S. Typhimurium and S. Enteritidis, which are associated with
rapid-onset gastroenteritis as well as serious systemic/invasive diseases (e.g., bacteremia, meningitis)
in immune-deficient individuals, young children, HIV-positive individuals, or patients who happen to
be coinfected with malaria worldwide. In industrialized countries, NTS infection in young infants or
the elderly may sometimes lead to severe invasive diseases accompanied by high case fatality rates.
In the sub-Saharan Africa, NTS diseases are associated with severe invasive disease in the absence of
gastroenteritis [7,8].
26.1.3.2 Pathogenesis
Typhoidal Salmonella serovars breach the intestinal epithelial barrier via phagocytosis and elicit intesti-
nal inflammatory infiltrates consisting primarily of mononuclear cells. After entry into macrophages via
macropinocytosis, typhoidal Salmonella serovars disseminate via the mononuclear phagocyte system
(a network of connective tissue containing immune cells) to other parts of the body, including the liver,
spleen, and bone marrow. During the systemic phase of typhoid fever, Salmonella may transfer from the
liver into the gallbladder, resulting in chronic carrier state [1,12].
On the other hand, NTS serovars enter into M cells through bacterial-mediated endocytosis, which is
accompanied by intestinal inflammation (with a massive influx of neutrophils into the terminal ileum
and colon). Gaining a growth advantage in the inflamed gut due to their ability to respire on tetrathionate
and to grow anaerobically on ethanolamine, NTS serovars disrupt tight junctions between the cells of
the intestinal wall, leading to the increased traffic of ions, water, and immune cells into and out of the
intestine. A combination of inflammation and the disruption of tight junctions ultimately contributes to
vomiting and diarrhea [13].
Salmonella genomes contain many virulence and antimicrobial resistance genes that play crucial parts
in host cell invasion (bapA, siiE, sopB), motility (fliC), intracellular survival (sseF, sseG), plasmids
(spvB, spvC), and ion acquisition (corA, mgtA, mgtB). The T3SS (encoded by genes in pathogenicity
islands SPI-1 and SPI-2) aids the invasion of nonphagocytic cells, colonization of the intestine, induc-
tion of intestinal inflammatory responses and diarrhea, survival and proliferation in macrophages, and
establishment of systemic disease [14–16].
26.1.4 Diagnosis
Conventional laboratory identification of Salmonella organisms is based on phenotyping methods that
assess their morphological, biochemical, and serological characteristics. In particular, the serotyping
technique (targeting somatic and flagellar antigens on the bacterial surface) separates Salmonella into
>2600 serovars in the White–Kauffmann–Le Minor scheme. This provides a useful tool to diagnose
and track Salmonella implicated in outbreaks, despite its obvious limitations (e.g., slow turnaround, skill
demand, and result variability, especially in dealing with rough, monophasic, and nonmotile strains). In
addition, on the basis of their ability to ferment certain substrates, Salmonella strains and serovars (e.g.,
Typhi, Paratyphi A, Paratyphi B, Typhimurium) can be biotyped via the phenotypic lead acetate test [17].
With the ongoing development and refinement in molecular detection technologies, a variety of highly
efficient and specific procedures have become available for identification, subtyping, and tracking of
Salmonella strains. These include serotyping and biotyping, virulence and resistance gene profiling,
plasmid profiling, and epidemiological tracking of clonality and genetic relatedness [1,18].
26.1.4.3 Plasmid Profiling
On the basis of the specific banding patterns of plasmid DNA molecules formed in electrophoretic gel,
plasmid profiling reveals details on the mechanisms and modes of transmission of the virulence or anti-
microbial resistance determinants among Salmonella strains.
26.1.4.4 Epidemiological Tracking
Various gel- and sequence-based genotyping methods may be employed for epidemiological tracking
of Salmonella strain clonality and genetic relatedness. These include pulsed-field gel electrophoresis
(PFGE), PCR-based DNA fingerprinting, multilocus variable-number tandem repeat analysis (MLVA),
multilocus sequence typing (MLST), next-generation sequencing (NGS), etc. [18].
PFGE involves digestion of bacterial chromosomal DNA with a rare cutting enzyme (XbaI or AvrII)
and electrophoretic separation of digested DNA fragments using an alternating electric current (pulsed
field), resulting in a distinct pattern for each Salmonella strain. With sufficiently high discriminative
power, PFGE remains the reference method for subtyping Salmonella organisms.
PCR-based DNA fingerprinting methods examine the genomic contents of Salmonella organisms,
with a goal to differentiate among strains and identify the source of the outbreaks. The commonly
used methods include random amplified polymorphic DNA (RAPD), enterobacterial repetitive inter-
genic consensus (ERIC)-PCR, repetitive extragenic palindromic sequences (REP)-PCR, and PCR-based
single-nucleotide polymorphism (SNP) typing.
MLVA assesses the variability (in terms of nucleotide sequence and unit size) of the genetic entity
called variable number tandem repeat (VNTR), which is present at multiple loci in the Salmonella
genome. MLVA subtyping of Salmonella strains demonstrates serovar specificity, in comparison with
PFGE, which is applicable to all serotypes.
MLST exploits the polymorphisms in selected housekeeping genes (aroC, dnaN, hemD, hisD, purE,
sucA, and thrA) among Salmonella strains to determine the sequence types of the organism. MLST sub-
typing represents a robust technique for epidemiological and evolutionary analyses of Salmonella strains.
PCR-based SNP typing is capable of differentiating Salmonella enteritidis strains that are indistin-
guishable by PFGE and phage typing.
NGS is a high-throughput genome sequencing technique that generates whole-genome sequences of
Salmonella strains in a timely and cost-effective fashion. NGS has proven superior to PFGE for subtyp-
ing Salmonella.
26.1.5.2 Prevention
Proper preparation and cooking of food offers an effective means to prevent the spreading of Salmonella
infection. These include washing hands before preparing food; heating food (chicken and beef) to an
Salmonella 397
internal temperature of 75°C for at least 10 min; avoiding drinking raw (unpasteurized) milk; avoiding
cross-contamination; washing hands with soap after handling reptiles, amphibians, or birds, or after
contact with pet feces.
A number of killed Salmonella whole-cell vaccines (through thermal inactivation, or chemical inacti-
vation with acetone, deoxycholate, or formalin) have been tested in mice. In addition, candidate subunit
vaccines [targeting protein (purified porins, bulk outer membrane proteins, flagellin protein), lipopoly-
saccharide, or O-polysaccharide (OPS)] are being developed. Conjugate vaccines that link Salmonella
OPS to a protein carrier have been shown to enhance the immunogenicity of polysaccharide hapten and
increase functional immunological memory [21–25].
26.2 Laboratory Models
Laboratory models offer valuable approaches for elucidating the pathogenesis of human Salmonella dis-
ease, uncovering insights into immunity to infection, and assessing the efficacy of vaccine preparations.
26.2.1 Animal Models
Due to the fact many Salmonella serovars demonstrate broad host range and that the sources of human
Salmonella infections often come from meat, eggs, and related products, warm-blooded animals of any
kind would be suitable models for foodborne Salmonella enteritis and systemic typhoid [26].
26.2.1.1 Mice
Mouse models of Salmonella infections have a number of obvious advantages, such as relatively low
cost, ready availability, and easy access to host genetics and immunological reagents [27].
Mouse colitis model involves streptomycin pretreatment (which reduces the intestinal microbiota in
mice) followed by oral administration of NTS Salmonella infection, leading to inflammatory colitis and
subsequent systemic infection. Interestingly, different mouse strains demonstrate varied susceptibility
to S. Typhimurium infection, with mouse strain 129sv being resistant and mouse strains C57BL6/J and
BALB/c being susceptible (which lack allele of the Slc11a1 or Nramp1 gene). Mouse colitis model allows
for mechanistic study of intestinal inflammation and Salmonella growth in the intestinal lumen. The
use of this model has helped clarify the role of flagella, T3SS-1, and T3SS-2 in intestinal inflammation
during S. Typhimurium infection; highlighted the contribution of intestinal inflammation to luminal
outgrowth of S. Typhimurium; and revealed the importance of luminal outgrowth of S. Typhimurium to
transmission of the pathogen [28,29].
The mouse typhoid model involves oral administration with NTS serovar S. Typhimurium without
streptomycin pretreatment, since oral challenge of mice with the human host-restricted serovars S. Typhi
and S. Paratyphi does not lead to a productive invasive infection [30]. This oral administration leads to
an invasive, systemic infection (characterized by a predominantly mononuclear leukocyte infiltrate, with
follicular hyperplasia, capillary thrombosis, hemorrhage, and ulcerations at areas of Peyer’s patches
in moribund animals) of the gut-associated lymphoid tissue and RES that resembles typhoid fever in
humans (in terms of entry route, tissue tropism, cellular location of bacteria, immune responses, and
profound hepatosplenomegaly) [31]. Nevertheless, without a capsule, S. Typhimurium does not show any
“stealth” incubation period before the clinical signs and does not express the typhoid toxin and the Vi
capsule polysaccharide (ViCPS) that can reduce toll receptor (TLR)-dependent inflammatory responses
and prevent complement deposition during Salmonella infection. This model has helped verify the role
of SPI-1 effector protein GtgE (present in S. Typhimurium, but absent in S. Typhi) in the proteolyti-
cal inactivation of the Rab29 GTPase, permitting intracellular replication. The absence of this effector
results in the rapid killing of S. typhi inside nonhuman cells [25,28].
Creation of humanized mice provides a sensitive and susceptible model to assess S. Typhi infection that
resembles the pathogenesis of typhoid in humans. The most commonly used humanized mice include
398 Laboratory Models for Foodborne Infections
immunodeficient mice (either Rag2−/− γc−/− or nonobese diabetic scid IL2rγnull) and TLR11-deficient
mice that are engrafted with human hematopoietic stem and progenitor cells (from fetal liver or cord
blood) [26].
26.2.1.2 Guinea Pigs
Guinea pigs represent a useful model for studying intestinal inflammation that results from Salmonella infec-
tion, although preconditioning through starvation and opium treatment is necessary prior to experimentation.
26.2.1.3 Rabbits
Based on New Zealand white rabbits, this model is useful for evaluating immunogenicity and reactoge-
nicity after oral administration of Salmonella serovars. It can also be applied for assessing S. Typhi and
S. Paratyphi A vaccine strains.
26.2.1.4 Calves
Calves infected orally or subjected to surgical ligation of ileal loops are useful for examining the intestinal
inflammation (a severe diffuse infiltrate composed predominantly of neutrophils, necrosis of the upper
mucosa, and formation of a pseudomembrane) and fluid secretion that are associated with S. Typhimurium-
induced gastroenteritis. Application of this model confirmed the ability of S. Typhimurium T3SS-1 in
inducing intestinal inflammation and diarrhea, and the contribution of flagella to neutrophil recruitment
in the intestinal mucosa during S. Typhimurium infection [28,32].
26.2.1.5 Chicken
The chick model has been employed to demonstrate the role of tviA in enhancing S. Typhimurium dis-
semination to internal organs.
26.2.1.6 Nonhuman Primates
Nonhuman primates such as rhesus macaque are susceptible to S. Typhimurium infection, which leads to
gastroenteritis resembling that in humans. Chimpanzees may develop typhoid fever after oral infection
with S. Typhi.
26.2.1.7 Zebrafish
Being strongly susceptible to S. Typhimurium, zebrafish embryos undergo pathological changes (bacte-
remia and proinflammatory response or cytokine storm) within hours after intravenous injection, resem-
bling those seen in humans [33].
26.2.1.8
C . elegans Nematode
This animal model may be utilized for investigation in Salmonella pathogenicity and host-pathogen
interactions.
invasion of, proliferation in, and apoptosis of epithelial cells. Resembling M cells (which lack glyco-
calyx) in Peyer’s patches, a combination of Caco-2 cells and Raji B cells helps uncover critical details
to caveolae-mediated endocytosis that is employed by Salmonella in its adhesion and entry into host
reticuloendothelial system before being taken up by macrophages. Macrophages that are found in the
gut-associated lymphoid tissue may be simulated by murine macrophage like cell line RAW 264.7 and
murine macrophages J774-A.1, yielding clues on Salmonella intracellular survival. Primary dendritic
cells (isolated from bone marrow of animal models or healthy humans) may be employed to investigate
the uptake (phagocytosis) and passive dissemination of Salmonella to systemic sites. In addition, use of
monocytes and granulocytes (such as human monocyte cell line THP-1 and primary cells isolated from
animal models) allows study of survival of Salmonella in unconventional targets [25].
26.3 Conclusion
The genus Salmonella contains some important foodborne pathogens that have the capability to cause
both localized infection (gastroenteritis) and systemic infection (typhoid fever) in humans and other ani-
mals. With the application of biochemical, serological, and molecular techniques together with the use of
various laboratory models (including in vivo animal models and in vitro epithelial, phagocytic, and other
cell models) in the past few decades, the molecular mechanisms of Salmonella invasion and intracellular
survival in host cells have been largely elucidated, and many rapid and accurate diagnostic techniques
for identification and subtyping of Salmonella strains have been developed. Nevertheless, much still has
to be learnt about the most efficient way to vaccinate humans against Salmonella infections. For this
reason, laboratory models will continue to play a vital part in helping decipher the host–bacterium inter-
actions, and design improved control strategies against Salmonella infections.
REFERENCES
1. Crump JA, Sjölund-Karlsson M, Gordon MA, Parry CM. Epidemiology, clinical presentation, labora-
tory diagnosis, antimicrobial resistance, and antimicrobial management of invasive Salmonella infec-
tions. Clin Microbiol Rev. 2015;28(4):901–37.
2. Fookes M, et al. Salmonella bongori provides insights into the evolution of the Salmonellae. PLoS
Pathog. 2011;7(8):e1002191.
3. Parkhill J, et al. Complete genome sequence of a multiple drug resistant Salmonella enterica serovar
Typhi CT18. Nature. 2001;413(6858):848–52.
4. McClelland M, et al. Complete genome sequence of Salmonella enterica serovar Typhimurium LT2.
Nature. 2001;413(6858):852–6.
5. Calenge F, Kaiser P, Vignal A, Beaumont C. Genetic control of resistance to salmonellosis and to
Salmonella carrier-state in fowl: a review. Genet Sel Evol. 2010;42:11.
6. de Jong HK, Parry CM, van der Poll T, Wiersinga WJ. Host–pathogen interaction in invasive salmonel-
losis. PLoS Pathog. 2012;8(10):e1002933.
7. Majowicz SE, et al. The global burden of nontyphoidal Salmonella gastroenteritis. Clin Infect Dis.
2010;50:882–9.
8. Garai P, Gnanadhas DP, Chakravortty D. Salmonella enterica serovars Typhimurium and Typhi as
model organisms: revealing paradigm of host–pathogen interactions. Virulence. 2012;3(4):377–88.
9. Bhan MK, Bahl R, Bhatnagar S. Typhoid and paratyphoid fever. Lancet. 2005;366:749–62.
10. Menendez A, et al. Salmonella infection of gallbladder epithelial cells drives local inflammation and
injury in a model of acute typhoid fever. J Infect Dis. 2009;200:1703–13.
11. Gonzalez-Escobedo G, Marshall JM, Gunn JS. Chronic and acute infection of the gall bladder by
Salmonella Typhi: understanding the carrier state. Nat Rev Microbiol. 2011;9(1):9–14.
12. Gunn JS, et al. Salmonella chronic carriage: epidemiology, diagnosis, and gallbladder persistence.
Trends Microbiol. 2014;22(11):648–55.
13. Pilar AV, Reid-Yu SA, Cooper CA, Mulder DT, Coombes BK. Active modification of host inflammation
by Salmonella. Gut Microbes. 2013;4(2):140–5.
400 Laboratory Models for Foodborne Infections
14. Coburn B, Li Y, Owen D, Vallance BA, Finlay BB. Salmonella enterica serovar Typhimurium pathoge-
nicity island 2 is necessary for complete virulence in a mouse model of infectious enterocolitis. Infect
Immun. 2005;73:3219–27.
15. Andrews-Polymenis HL, Bäumler AJ, McCormick BA, Fang FC. Taming the elephant: Salmonella
biology, pathogenesis, and prevention. Infect Immun. 2010;78:2356–69.
16. Broz P, Ohlson MB, Monack DM. Innate immune response to Salmonella typhimurium, a model enteric
pathogen. Gut Microbes. 2012;3(2):62–70.
17. Andrews JR, Ryan ET. Diagnostics for invasive Salmonella infections: current challenges and future
directions. Vaccine. 2015;33(Suppl 3):C8–15.
18. Ngoi ST, Teh CS, Chai LC, Thong KL. Overview of molecular typing tools for the characterization of
Salmonella enterica in Malaysia. Biomed Environ Sci. 2015;28(10):751–64.
19. Shi C, Singh P, Ranieri ML, Wiedmann M, Moreno Switt AI. Molecular methods for serovar determina-
tion of Salmonella. Crit Rev Microbiol. 2015;41(3):309–25.
20. Weening EH, et al. The Salmonella enterica serotype Typhimurium lpf, bcf, stb, stc, std, and sth fim-
brial operons are required for intestinal persistence in mice. Infect Immun. 2005;73:3358–66.
21. Griffin AJ, McSorley SJ. Development of protective immunity to Salmonella, a mucosal pathogen with
a systemic agenda. Mucosal Immunol. 2011;4(4):371–82.
22. Simon R, Tennant SM, Galen JE, Levine MM. Mouse models to assess the efficacy of non-typhoidal
Salmonella vaccines: revisiting the role of host innate susceptibility and routes of challenge. Vaccine.
2011;29(32):5094–106.
23. Simon R, Levine MM. Glycoconjugate vaccine strategies for protection against invasive Salmonella
infections. Hum Vaccines Immunother. 2012;8(4):494–8.
24. McSorley SJ. Immunity to intestinal pathogens: lessons learned from Salmonella. Immunol Rev.
2014;260(1):168–82.
25. Santos RL. Pathobiology of Salmonella, intestinal microbiota, and the host innate immune response.
Front Immunol. 2014;5:252.
26. Tsolis RM, Xavier MN, Santos RL, Bäumler AJ. How to become a top model: impact of animal experi-
mentation on human Salmonella disease research. Infect Immun. 2011;79(5):1806–14.
27. Lawley TD, et al. Genome-wide screen for Salmonella genes required for long-term systemic infection
of the mouse. PLoS Pathog. 2006;2:e11.
28. Santos RL, et al. Animal models of Salmonella infections: enteritis versus typhoid fever. Microbes
Infect. 2001;3:1335–44.
29. Barthel M, et al. Pretreatment of mice with streptomycin provides a Salmonella enterica serovar
Typhimurium colitis model that allows analysis of both pathogen and host. Infect Immun. 2003;
71:2839–58.
30. Song J, et al. A mouse model for the human pathogen Salmonella typhi. Cell Host Microbe. 2010;8:369–76.
31. Brown DE, McCoy MW, Pilonieta MC, Nix RN, Detweiler CS. Chronic murine typhoid fever is a natu-
ral model of secondary hemophagocytic lymphohistiocytosis. PLoS One. 2010;5:e9441.
32. Tsolis RM, et al. Of mice, calves, and men. Comparison of the mouse typhoid model with other
Salmonella infections. Adv Exp Med Biol. 1999;473:261–74.
33. Torraca V, Masud S, Spaink HP, Meijer AH. Macrophage–pathogen interactions in infectious diseases:
new therapeutic insights from the zebrafish host model. Dis Model Mech. 2014;7(7):785–97.
27
Shigella
CONTENTS
27.1 Shigellosis..................................................................................................................................... 401
27.1.1 Incidence........................................................................................................................... 401
27.1.2 Bacteriology...................................................................................................................... 402
27.1.3 Transmission..................................................................................................................... 402
27.1.4 Pathogenesis..................................................................................................................... 402
27.2 Animal Models............................................................................................................................. 402
27.2.1 Guinea Pigs....................................................................................................................... 402
27.2.2 Mice.................................................................................................................................. 403
27.2.3 Rabbits.............................................................................................................................. 404
27.2.4 Pigs and Piglets................................................................................................................. 404
27.2.5 Chicken............................................................................................................................. 405
27.2.6 Caenorhabditis elegans.................................................................................................... 405
27.2.7 Monkeys........................................................................................................................... 405
27.2.8 Zebrafish........................................................................................................................... 405
27.2.9 Human Cell Lines............................................................................................................. 405
27.3 C onclusion..................................................................................................................................... 406
References............................................................................................................................................... 406
27.1 Shigellosis
27.1.1 Incidence
At the end of the 19th century, young microbiologist Kiyoshi Shiga discovered a unique bacillus causing
dysentery during the dysentery epidemic in Japan, and this bacterium, now known as Shigella dysente-
riae 1, was the first acknowledged member of the genus Shigella.1
Shigella species are crucial causes of traveler’s diarrhea in individuals from industrialized countries
visiting developing areas.2,3 Through accidental hygiene or sanitation breaches, endemic outbreaks of
shigellosis may also occur in developed countries.4 Very recently, the global burden of shigellosis has
been reevaluated.5 Shigellosis generally affects poor populations, especially young children, in the
developing world.5–8
By invading the large intestinal mucosa, Shigella spp. induce major inflammatory destruction and gen-
erate dysenteric symptoms in humans and in nonhuman primates.9 In a few cases, only watery diarrhea
is present.10,11 In humans, remarkably low bacterial loads are able to trigger severe shigellosis.12
Suitable antimicrobial therapy of shigellosis reduces the duration of fever and diarrhea.
Azithromycin and fluoroquinolones are the first-line choice for treatment of shigellosis.13 Currently,
the emergence of multidrug-resistant Shigella variants is the greatest concern among global health
problems.14,15 During the last five decades, extensive research has yielded a large number of vaccine
candidates, but no effective vaccines.16–20 The goal of an effective vaccine against Shigella remains
elusive.
401
402 Laboratory Models for Foodborne Infections
27.1.2 Bacteriology
The Shigella genus includes four species: Shigella dysenteriae, Shigella flexneri, Shigella boydii, and
Shigella sonnei. Based on the antigenicity of the O-specific polysaccharide chain of their lipopolysac-
charide (LPS) molecules, Shigella species are further divided into serotypes: 15 for S. dysenteriae, 15
for S. flexneri (serotypes and subserotypes comprising the recently entitled 7a and 7b subtypes), 20 for
S. boydii, and 1 for S. sonnei.21 In most tropical countries, shigellosis is endemic and is dominated by a
few S. flexneri serotypes, as well as S. dysenteriae 1.19 The dominant isolates in Europe and the United
States are S. sonnei and S. flexneri.19
Shigella is nonmotile and nonflagellated in nature.21 Most shigellae are capable of fermenting glucose,
with a few exceptions (S. flexneri 6, S. boydii 13 and 14, and S. dysenteriae 3).22 S. dysenteriae is inca-
pable of fermenting mannitol, and this property is used as a key feature for its identification.21
For daily laboratory screening of shigellae, two different selective media are generally used, namely
Hektoen enteric (HE) agar and xylose lysine desoxycholate (XLD) agar. Shigella colonies on XLD agar
appear translucent, pink, or red and are smooth in nature. S. dysenteriae 1 colonies on XLD agar are
commonly very small, unlike those of other Shigella species.23 Colonies of shigellae on HE agar appear
green. Serological identification by slide agglutination with polyvalent somatic antigen grouping sera
further confirms the serotypes and subserotypes.
27.1.3 Transmission
The most common route of Shigella transmission is via fecal–oral route. The bacteria may also be spread
by direct contact with an infected person, as shigellae have the ability to survive on human skin for up to
1 h.21,24 Shigella infection requires only a very small inoculum,12 making person-to-person transmission
favorable. Shigella can spread rapidly by foodborne transmission and cause large outbreaks.25–28 Flies
may play a role in spreading shigellae from human excrement to foods.29,30 Shigellae are able to survive
for at least 6 months in water at room temperature.21
27.1.4 Pathogenesis
The cellular pathogenesis of shigellosis is the result of the complicated actions of a large number of bac-
terial virulence factors. The essential machinery for bacterial penetration and intracellular endurance is
usually encoded in a large “virulence plasmid.”31,32 Loss of the virulence plasmid gives rise to avirulent
phenotypes. A type III secretion system (T3SS) is another key factor involved in Shigella invasion.33,34
Shigella bacilli invade the distal region of the colon and rectum,35 where they become imprisoned by
specialized M-cells. The M-cells deliver the bacterial antigens, LPS, and invasive plasmid antigen (Ipa)
proteins to antigen-presenting macrophages and dendritic cells.36 Shigella bacilli are phagocytized by
macrophages, but subsequently escape through apoptosis.37 Before death, the macrophages release pro-
inflammatory cytokines interleukin-1b and interleukin-18.38
These cytokines trigger a strong inflammatory response and stimulate the migration of neutrophils,35
which infiltrate the infected site and destabilize the epithelium.39 The death of infected macrophages and
subsequent destabilization allow penetration of more shigellae into the tissue and invasion of epithelial
cells through the basal membrane. Survival and replication in macrophages represent the fundamental
key to extensive colonization of the intestinal epithelium.40
27.2 Animal Models
27.2.1 Guinea Pigs
The oldest animal model for assaying Shigella invasion is the guinea pig keratoconjunctivitis model,
known as the Sereny test.41 In this invasion assay, a suspension of live shigellae is generally inocu-
lated into the keratoconjunctival sac of guinea pigs. Invasive shigellae disrupt the conjunctival wall and
Shigella 403
cause severe conjunctivitis, which is characterized by redness of the eye and corneal opaqueness. Highly
pathogenic clinical isolates usually cause acute purulent inflammation, which leads to closing of the
eyelids. Noninvasive Shigella isolates cannot invade the ocular mucosa. This assay can determine the
invasive phenotypes of Shigella.
The Sereny test was used to assess immunization efficacy, i.e., determination of protection against
ocular infection, by various research groups.42–48 Although the keratoconjunctivitis assay is regarded as
the gold standard for measuring Shigella invasion, it does not allow accurate quantification of the inflam-
matory response.
A lethal enteric infection in the colon was established in 4-day-starved guinea pigs by the combina-
tion of calcium carbonate and opium.49 In a series of experiments, guinea pigs were inoculated orally
with live S. flexneri 2a ranging from 8 × 105 to 1.4 × 107 CFU, which induced a fatal enteric infection
with ulcerative lesions in the colon. However, this model might not be ideal for the purpose of measuring
protective efficacy, because the fatal effects appear at a relatively early stage of infection.20
The guinea pig colitis model usually induces typical bacillary dysentery after rectal inoculation of
Shigella species (S. flexneri 2a and 5a) without any preparatory treatment like starvation or antibiotic
treatment.50 In this model, guinea pigs were inoculated with four different doses (108, 109, 1010, and 1011
CFU) of Shigella through the intrarectal route. Within 24 h of challenge, all doses conferred significant
signs of bacillary dysentery. This colitis model has been proven successful in many protective effi-
cacy studies.50–52 However, recent reports suggested that it has several limitations53 and methodological
complications.54
The recently developed guinea pig luminal model was found to be sensitive as it mimics human bacil-
lary dysentery.54 In this model, the lumen of the colon was separately infected with virulent S. dysente-
riae 1 and S. flexneri 2a (109 CFU) after ligation of the distal cecum. The ligation was placed at 4-cm
distance from the ileocecal junction. The placement of the surgical tie may facilitate the colonization
of shigellae in the distal colon by controlling the flow of the stool from the cecum. Bacillary dysenteric
symptoms were recorded within 24 h of infection. The luminal model can be used to demonstrate protec-
tive efficacy,54 but it has become untenable because of a surfeit of surgical complications.
To understand the adhesion and invasion of Shigella, in vitro studies were performed on colonic
explants from guinea pigs, wherein bacterial suspensions were incubated with luminary cuts of the cecal
mucosa.55–57
27.2.2 Mice
Mice have been the most frequently used small animal species in preclinical studies of shigellosis. The
major advantages of mice are the ease of handling, availability of a broad spectrum of reagents, and fea-
sibility of genetic manipulation. Mice are also cost-effective for protective efficacy studies.
During oral inoculation studies, Shigella was unintentionally inoculated into the respiratory pas-
sage, leading to pulmonary Shigella infection.58,59 Over the last five decades, the murine pulmonary
model was used to demonstrate protective efficacy against Shigella infection.59–65 Typically, after
immunization through the external nares, mice were generally inculcated with a challenge strain
(10 6 –107 CFU) through the intranasal pathway. Protection was usually demonstrated by the degree
of Shigella invasion into the lungs and the induction of pulmonary pneumonia. The major lacuna
of the pulmonary model is the lack of clinical relevance with respect to the infection site of the
pathogen.20,53
Another murine infection model was demonstrated via oral inculcation of invasive shigellae in new-
born mice.66 This murine model is extremely sensitive because the neonatal gut is only susceptible to
Shigella infection by oral inculcation for 3–4 days after birth.66 Usually, 4-day-old newborn mice were
orally inculcated with S. flexneri 5a (5 × 109 CFU) for experimental shigellosis. Effective Shigella inva-
sion was achieved in the intestinal mucosa of 4-day-old newborn mice, while 5-day-old mice were found
to be resistant.
It has been assumed that murine models are inadequate for determination of protective efficacy.20
However, recent studies have helped overcome this shortcoming.67,68 The offspring of immunized dams
were orally inoculated with invasive shigellae and the level of protection measured by the degree of
404 Laboratory Models for Foodborne Infections
colonization in the bowel. The murine model developed in these studies can be used as a tool for deter-
mination of passive immunization.
To study the interactions of Shigella with the human intestine in vivo, one group of researchers placed
human intestinal xenografts into the subscapular region of 6–8-week-old severe combined immunode-
ficient (SCID) mice.69 Inoculation of Shigella into these human intestinal xenografts led to the develop-
ment of severe inflammation and mucosal destruction at 4 h after infection. This xenograft model is
useful for understanding intracellular communications, but is somewhat limited and strenuous for the
purpose of measuring protective efficacy.20
A recent study showed that intraperitoneal inoculation of S. flexneri 2a (5 × 108 CFU) into mice
provoked acute shigellosis and mimicked human dysentery.70 The inoculated mice had diarrhea within
2 h of infection. This intraperitoneal model was also shown to be useful for evaluating Shigella vaccine
candidates.72
27.2.3 Rabbits
Rabbits are not naturally susceptible to shigellosis, but may acquire the infection under preparatory con-
ditions. New Zealand white rabbits are usually preferred in the Shigella research field because they are
commonly available and inexpensive.
For investigations of Shigella pathogenesis at the cellular level, the ileal-ligated loops assay has
proven beneficial.71–74 For this, ligated segments of the rabbit small intestine are generally inoculated
with various serotypes of S. dysenteriae, S. flexneri, S. boydii, and S. sonnei.71 Within 12–18 h of inoc-
ulation (109 –1010 CFU), bacterial invasion and ulceration can be achieved.71,72 Furthermore, immune
responses can be measured in a quantitative manner after infection of the ligated loops.74 Surgical
complications are one of the drawbacks of the ligated loops assay, and it is not useful for protective
efficacy studies.
Several studies indicated that acute necrosis occurred in the rabbit ileum after oral inoculation of
Shigella, under preparatory treatments like starvation, antibiotic treatment, and stomach acid neutraliza-
tion.75,76 However, there has been no prominent follow-up of such orogastric “conditioned” rabbit models
to date.
Adult rabbits exposed to direct colonic inoculation by cecal bypass also progress to shigellosis.77 Cecal
bypass was achieved by blockade of the cecal lumen above the ileocecal junction with a silk thread liga-
ture, while the ileo-ceco-colic communication was properly maintained. After cecal bypass, bacteria
(107 CFU) were inoculated into the proximal colon. Onset of dysentery occurred within 24 h of inocula-
tion. The colonic infection model was shown to be worthwhile in various Shigella vaccine candidate
screenings.62,78,79 A very recent study clearly demonstrated the usefulness of this model in the assessment
of anti-shigella vaccine candidates.80 Despite encouraging results, the surgical obstacle appears to be the
major lacuna of this model.
27.2.5 Chicken
Very recently, a research group in China reported invasion of clinically isolated S. flexneri 2a into spe-
cific pathogen-free (SPF) chicken.83 SPF chickens at 3 days of age were inoculated by intraperitoneal
injection or crop injection. Inoculation of 3 × 109 CFU resulted in death in 100% of the chickens, while
crop injection did not cause any intestinal clinical signs of dysentery. The ligated intestinal loop of
1-day-old SPF chicken also showed severe congestion and edema. This study has clinical significance
regarding human–poultry cross-infection, although it has a shortcoming regarding vaccine candidate
screening.
27.2.7 Monkeys
In 1965, Formal et al.88 successfully demonstrated immunogenicity and protective efficacy of attenu-
ated Shigella strains against S. flexneri 1b, 2a, and 6 infections in rhesus monkeys (Macaca mulatta).
Both immunization and challenge (5 × 1010 CFU) were performed by the oral route. Later studies by the
same group revealed that typical bacillary dysentery could be induced in rhesus monkeys by oral infec-
tion with S. flexneri without starvation and/or pretreatment with antibiotics.89,90 Subsequent protective
efficacy studies for Shigella vaccine candidates also became effective in rhesus monkeys.91–95 The rhesus
monkey model mimics human shigellosis and the human immune response, making it ideal for assem-
bling preclinical data before human trials of vaccine candidates.
Cynomolgus monkeys (Macaca fascicularis)96,97 and Aotus nancymaae monkeys98 were also estab-
lished as tools for screening Shigella vaccine candidates. Despite auspicious results, the cost of using
these animals can be prohibitive, especially in developing countries where shigellosis is endemic.
27.2.8 Zebrafish
The zebrafish model opens a new avenue to investigate the significance of innate immunity. It provides a
framework where no adaptive responses have yet been developed, because early lymphocytes make their
first appearance in 4-day-old larvae and full adaptive immunity requires several weeks to be displayed.99
An in vivo study of S. flexneri interactions with phagocytes and bacterial autophagy was reported very
recently.100 An inoculum of 4 × 103 S. flexneri 5a resulted in the death of most zebrafish larvae within 48 h
postinfection. Intravenously administered bacteria could survive and replicate in both macrophages and
nonimmune cells. The zebrafish larva represents a valuable new host for analysis of Shigella infection,
although this model cannot define the protective efficacy.
The primary step of shigellosis is invasion of the human colonic mucosa. Host–pathogen interactions
can easily be examined in vitro using cell lines before animal trials are carried out. Several inves-
tigations were performed to disclose host–Shigella interactions at the cellular level using HeLa,101–105
T84,106–108 THP-1,109,110 macrophage J774,111–113 HT-29,114,115 monoblastic U937,113,116,117 human embryonic
kidney (HEK) 293,118,119 human colonic Caco-2 epithelial,120–122 and human CD4+ T (Jurkat)123 cells. The
results identified the relative contributions of antigens, which need to be taken into account for vaccine
development.
27.3 Conclusion
The use of various animal models has already improved our understanding of the in vivo consequences
of intracellular defense mechanisms as well as protective measures against Shigella spp. We expect that
further application of these powerful models will continue to unravel the intricacy of the virulence strat-
egies employed by shigellae to mitigate host’s innate and acquired immune defenses.
REFERENCES
1. Shiga, K. The trend of prevention, therapy and epidemiology of dysentery since the discovery of its
causative organism. N Eng J Med. 215, 1205–1211 (1936).
2. Niyogi, S.K. Shigellosis. J Microbiol. 43, 133–143 (2005).
3. Marchou, B. Traveller’s diarrhea: epidemiology, clinical practice guideline for the prevention and treat-
ment. Presse Med. 42, 76–81 (2013).
4. Bancroft, J.E., Keifer, S.B. & Keene, W.E. Shigellosis from an interactive fountain: implications for
regulation. J Environ Health. 73, 16–20 (2010).
5. GBD 2013 Mortality and Causes of Death Collaborators. Global, regional, and national age–sex specific
all-cause and cause-specific mortality for 240 causes of death, 1990–2013: a systematic analysis for the
Global Burden of Disease Study 2013. Lancet. 385, 117–171 (2015).
6. Osorio, M., Bray, M.D. & Walker, R.I. Vaccine potential for inactivated shigellae. Vaccine. 25, 1581–
1592 (2007).
7. O’Ryan, G.M., Ashkenazi-Hoffnung, L., O’Ryan-Soriano, M.A. & Ashkenazi, S. Management of acute
infectious diarrhea for children living in resource-limited settings. Expert Rev Anti Infect Ther. 12,
621–632 (2014).
8. Lamberti, L.M., Bourgeois, A.L., Fischer Walker, C.L., Black, R.E. & Sack, D. Estimating diarrheal
illness and deaths attributable to Shigellae and enterotoxigenic Escherichia coli among older children,
adolescents, and adults in South Asia and Africa. PLoS Negl Trop Dis. 8, e2705 (2014).
9. Sansonetti, P.J. War and peace at mucosal surfaces. Nat Rev Immunol. 4, 953–964 (2004).
10. Tobias, J. et al. Involvement of main diarrheagenic Escherichia coli, with emphasis on enteroaggrega-
tive E. coli, in severe non-epidemic pediatric diarrhea in a high-income country. BMC Infect Dis. 15,
79 (2015).
11. Zaidi, M.B. & Estrada-García, T. Shigella: a highly virulent and elusive pathogen. Curr Trop Med Rep.
1, 81–87 (2014).
12. Ferreccio, C. et al. Epidemiologic patterns of acute diarrhea and endemic Shigella infections in children
in a poor periurban setting in Santiago, Chile. Am J Epidemiol. 134, 614–627 (1991).
13. Legros, D. ed. Guidelines for the Control of Shigellosis, Including Epidemics due to Shigella dysenteriae
Type 1. World Health Organization, Geneva (2005).
14. Omulo, S., Thumbi, S.M., Njenga, M.K. & Call, D.R. A review of 40 years of enteric antimicrobial
resistance research in Eastern Africa: what can be done better? Antimicrob Resist Infect Control. 4,
1 (2015).
15. Magiorakos, A.P. et al. Multidrug-resistant, extensively drug-resistant and pandrug-resistant bacteria:
an international expert proposal for interim standard definitions for acquired resistance. Clin Microbiol
Infect. 18, 268–281 (2012).
Shigella 407
16. Kraehenbuhl, J.P. & Neutra, M.R. Mucosal vaccines: where do we stand? Curr Top Med Chem. 13,
2609–2628 (2013).
17. Böhles, N. et al. Vaccines against human diarrheal pathogens: current status and perspectives. Hum
Vaccines Immunother. 10, 1522–1535 (2014).
18. Barry, E.M. et al. Progress and pitfalls in Shigella vaccine research. Nat Rev Gastroenterol Hepatol. 10,
245–255 (2013).
19. Levine, M.M., Kotloff, K.L., Barry, E.M., Pasetti, M.F. & Sztein, M.B. Clinical trials of Shigella vac-
cines: two steps forward and one step back on a long, hard road. Nat Rev Microbiol. 5, 540–553 (2007).
20. Kweon, M.N. (2008) Shigellosis: the current status of vaccine development. Curr Opin Infect Dis. 21,
313–318 (2008).
21. Germani, Y. & Sansonetti, P.J. The genus Shigella. In: Dworkin, M. et al., eds. The Prokaryotes, Vol. 6,
3rd ed., pp. 99–122. Springer, New York (2006).
22. Coimbra, R.S. et al. Molecular and phenotypic characterization of potentially new Shigella dysenteriae
serotype. J Clin Microbiol. 39, 618–621 (2001).
23. McIntyre, L. ed. Laboratory Methods for the Diagnosis of Epidemic Dysentery and Cholera. WHO/
CDS/CSR/EDC/99.8, pp. 17–36. Centers for Disease Control and Prevention, Atlanta, GA (1999).
24. Islam, M.S. et al. Detection of non-cultivable Shigella dysenteriae type 1 from artificially contaminated
volunteer’s fingers using fluorescent antibody and PCR techniques. J Diarrheal Dis Res. 15, 65–70 (1997).
25. Boyce, J.M. et al. Patterns of Shigella infection in families in rural Bangladesh. Am J Trop Med Hyg. 31,
1015–1020 (1982).
26. Rennels, M.B. & Levine, M.M. Classical bacterial diarrhea: perspective and update: Salmonella,
Shigella, Escherichia coli, Aeromonas and Flesiomonas. Pediatr Infect Dis J. 5, 91–100 (1986).
27. Scallan, E. et al. Foodborne illness acquired in the United States—major pathogens. Emerg Infect Dis.
17, 7–15 (2011).
28. Vally, H. et al. Proportion of illness acquired by foodborne transmission for nine enteric pathogens in
Australia: an expert elicitation. Foodborne Pathog Dis. 11, 727–733 (2014).
29. Hardy, A. & Watts, J. Studies of the acute diarrheal diseases. XVIII: epidemiology. Public Health Rep.
63, 363 (1948).
30. Levine, O.S. & Levine, M.M. Houseflies (Musca domestica) as mechanical vectors of shigellosis. Rev
Infect Dis. 13, 688–696 (1991).
31. Sansonetti, P.J., Kopecko, D.J. & Formal, S.B. Involvement of a plasmid in the invasive ability of
Shigella flexneri. Infect Immun. 35, 852–860 (1982).
32. Sasakawa, C., Makino, S., Kamato, K. & Yoshikawa, M. Isolation, characterization, and mapping of
Tn5 insertions into the 140 megadalton invasion plasmid defective in the mouse Sereny test in Shigella
flexneri 2a. Infect Immun. 54, 32–36 (1986).
33. Blocker, A. et al. The tripartite type III secreton of Shigella flexneri inserts IpaB and IpaC into host
membranes. J Cell Biol. 147, 683–693 (1999).
34. Blocker, A. et al. Structure and composition of the Shigella flexneri “needle complex,” a part of its type
III secreton. Mol Microbiol. 39, 652–663 (2001).
35. Anand, B.S. et al. Rectal histology in acute bacillary dysentery. Gastroenterology. 90, 654–660 (1986).
36. Phalipon, A. & Sansonetti, P.J. Shigellosis: innate mechanisms of inflammatory destruction of the intes-
tinal epithelium, adaptive immune response, and vaccine development. Crit Rev Immunol. 23, 371–401
(2003).
37. Phalipon, A. & Sansonetti, P.J. Shigella’s ways of manipulating the host intestinal innate and adaptive
immune system: a tool box for survival? Immunol Cell Biol. 85, 119–129 (2007).
38. Chen, Y., Smith, M.R., Thirumalai, K. & Zychlinsky, A. A bacterial invasion induces macrophage apop-
tosis by binding directly to ICE. EMBO J. 15, 3853–3860 (1996).
39. Perdomo, J.J., Gounon, P. & Sansonetti, P.J. Polymorphonuclear leukocyte transmigration promotes
invasion of colonic epithelial monolayer by Shigella flexneri. J Clin Invest. 93, 633–643 (1994).
40. Ashida H. et al. Shigella deploy multiple countermeasures against host innate immune responses. Curr
Opin Microbiol. 14, 16–23 (2011).
41. Sereny, B. Experimental Shigella keratoconjunctivitis: a preliminary report. Acta Microbiol Acad Sci
Hung. 2, 293–296 (1955).
408 Laboratory Models for Foodborne Infections
42. Kerekes, L. Immunity of the guinea pig’s eye after vaccination with Shigella sonnei phase I antigen.
Acta Microbiol Acad Sci Hung. 23, 353–358 (1976).
43. Linde, K. et al. Live Shigella flexneri 2a and Shigella sonnei I vaccine candidate strains with two
attenuating markers. I. Construction of vaccine candidate strains with retained invasiveness but reduced
intracellular multiplication. Vaccine. 8, 25–29 (1990).
44. Verma, N.K. & Lindberg, A.A. Construction of aromatic dependent Shigella flexneri 2a live vaccine
candidate strains: deletion mutations in the aroA and the aroD genes. Vaccine. 9, 6–9 (1991).
45. Hartman, A.B., Powell, C.J., Schultz, C.L., Oaks, E.V. & Eckels, K.H. Small-animal model to measure
efficacy and immunogenicity of Shigella vaccine strains. Infect Immun. 59, 4075–4083 (1991).
46. Hartman, A.B. et al. Local immune response and protection in the guinea pig keratoconjunctivitis
model following immunization with Shigella vaccines. Infect Immun. 62, 412–420 (1994).
47. Mukhopadhaya, A., Mahalanabis, D., Khanam, J. & Chakrabarti, M.K. Protective efficacy of oral
immunization with heat-killed Shigella flexneri 2a in animal model: study of cross protection, immune
response and antigenic recognition. Vaccine. 21, 3043–3050 (2003).
48. Li, W. et al. Development of prophylactic recombinant HPV58-attenuated Shigella live vector vaccine
and evaluation of its protective efficacy and immunogenicity in the guinea pig keratoconjunctivitis
model. Acta Biochim Biophys Sin (Shanghai). 41, 137–145 (2009).
49. Formal, S.B., Dammin, G.J., Labrec, E.H. & Schneider, H. Experimental Shigella infections: character-
istics of a fatal infection produced in guinea pigs. J Bacteriol. 75, 604–610 (1958).
50. Shim, D.H. et al. New animal model of shigellosis in the guinea pig: its usefulness for protective efficacy
studies. J Immunol. 178, 2476– 2482 (2007).
51. Barman, S. et al. Live non-invasive Shigella dysenteriae 1 strain induces homologous protective immu-
nity in a guinea pig colitis model. Microbiol Immunol. 55, 683–693 (2011).
52. Barman, S. et al. Protective immunity by oral immunization with heat-killed Shigella strains in a guinea
pig colitis model. Microbiol Immunol. 57, 762–771 (2013).
53. Kim, Y.J., Yeo, S.G., Park, J.H. & Ko, H.J. Shigella vaccine development: prospective animal models
and current status. Curr Pharm Biotechnol. 14, 903–912 (2013).
54. Barman, S., Saha, D.R., Ramamurthy, T. & Koley, H. Development of a new guinea pig model of shigel-
losis. FEMS Immunol Med Microbiol. 62, 304–314 (2011).
55. Izhar, M., Nuchamowitz, Y. & Mirelman, D. Adherence of Shigella flexneri to guinea pig intestinal cells
is mediated by a mucosal adhesion. Infect Immun. 35, 1110–1118 (1982).
56. Raygoza-Anaya, M., González-Robles, A. & Mora-Galindo, J. In vitro model for the analysis of the inter-
action between Shigella flexneri and the intestinal epithelium. Arch Invest Med (Mex). 21, 305–309 (1990).
57. Guhathakurta, B., Sasmal, D. & Datta, A. Adhesion of Shigella dysenteriae type 1 and Shigella flexneri
to guinea-pig colonic epithelial cells in vitro. J Med Microbiol. 36, 403–405 (1992).
58. Voino-Yasenetsky, M.V. & Voino-Yasenetskaya, M.K. Experimental pneumonia caused by bacteria of
the Shigella group. Acta Morphol Acad Sci Hung. 11, 439–454 (1962).
59. Mallett, C.P., Van de Verg, L., Collins, H.H. & Hale, T.L. Evaluation of Shigella vaccine safety and
efficacy in an intranasally challenged mouse model. Vaccine. 11, 190–196 (1993).
60. Mallett, C.P. et al. Intransal or intragastric immunization with proteosome-Shigella lipopolysaccharide
vaccines protects against lethal pneumonia in a murine model of Shigella infection. Infect Immun. 63,
2382–2386 (1995).
61. Van de Verg, LL et al. Antibody and cytokine responses in a mouse pulmonary model of Shigella flex-
neri serotype 2a infection. Infect Immun. 63, 1947–1954 (1995).
62. Koley, H., Barman, S., Roy, N., Saha, D.R. & Kumar, H. The efficacy and immunogenicity of a live
transconjugant hybrid strain of Shigella dysenteriae type 1 in two animal models. World J Microbiol
Biotechnol. 25, 679–686 (2009).
63. Shim, D.H. et al. Immunogenicity and protective efficacy offered by a ribosomal-based vaccine from
Shigella flexneri 2a. Vaccine. 25, 4828–4836 (2007).
64. Camacho, A.I. et al. Mucosal immunization with Shigella flexneri outer membrane vesicles induced
protection in mice. Vaccine. 29, 8222–8229 (2011).
65. Pore, D., Mahata, N., Pal, A. & Chakrabarti, M.K. Outer membrane protein A (OmpA) of Shigella flex-
neri 2a, induces protective immune response in a mouse model. PLoS One. 6, e22663 (2011).
66. Fernandez, M.I. et al. A newborn mouse model for the study of intestinal pathogenesis of shigellosis.
Cell Microbiol. 5, 481–491 (2003).
Shigella 409
67. Mitra, S., Chakrabarti, M.K. & Koley, H. Multi-serotype outer membrane vesicles of Shigellae confer
passive protection to the neonatal mice against shigellosis. Vaccine. 31, 3163–3173 (2013).
68. Barman, S. et al. Passive immunity with multi-serotype heat-killed Shigellae in neonatal mice. Microbiol
Immunol. 58, 463–466 (2014).
69. Zhang, Z., Jin, L., Champion, G., Seydel, K.B. & Stanley, S.L. Jr. Shigella infection in a SCID mouse-
human intestinal xenograft model: role for neutrophils in containing bacterial dissemination in human
intestine. Infect Immun. 69, 3240–3247 (2001)
70. Yang, J.Y. et al. A mouse model of shigellosis by intraperitoneal infection. J Infect Dis. 209, 203–215 (2014).
71. Arm, H.G., Floyd, T.M., Faber, J.E. & Hayes, J.R. Use of ligated segments of rabbit small intestine in
experimental shigellosis. J Bacteriol. 89, 803–809 (1965).
72. Wassef, J.S., Keren, D.F. & Mailloux, J.L. Role of M cells in initial antigen uptake and in ulcer forma-
tion in the rabbit intestinal loop model of shigellosis. Infect Immun. 57, 858–863 (1989).
73. Sansonetti, P.J., Arondel, J., Cantey, J.R., Prévost, M.C. & Huerre, M. Infection of rabbit Peyer’s patches
by Shigella flexneri: effect of adhesive or invasive bacterial phenotypes on follicle-associated epithe-
lium. Infect Immun. 64, 2752–2764 (1996).
74. Schnupf, P. & Sansonetti, P.J. Quantitative RT-PCR profiling of the rabbit immune response: assessment
of acute Shigella flexneri infection. PLoS One. 7, e36446 (2012).
75. Ahmed, Z.U., Sarker, M.R. & Sack, D.A. Protection of adult rabbits and monkeys from lethal shigellosis
by oral immunization with a thymine-requiring and temperature-sensitive mutant of Shigella flexneri Y.
Vaccine. 8, 153–158 (1990).
76. Etheridge, M.E., Hoque, A.T. & Sack, D.A. Pathologic study of a rabbit model for shigellosis. Lab Anim
Sci. 46, 61–66 (1996).
77. Rabbani, G.H. et al. Development of an improved animal model of shigellosis in the adult rabbit by
colonic infection with Shigella flexneri 2a. Infect Immun. 63, 4350–4357 (1995).
78. Chakrabarti, M.K. et al. Killed oral Shigella vaccine made from Shigella flexneri 2a protects against
challenge in the rabbit model of shigellosis. Acta Paediatr. 88, 161–165 (1999).
79. Mukhopadhaya, A., Mahalanabis, D., Khanam, J. & Chakrabarti, M.K. Protective efficacy of oral
immunization with heat-killed Shigella flexneri 2a in animal model: study of cross protection, immune
response and antigenic recognition. Vaccine. 21, 3043–3050 (2003).
80. Nag, D. et al. Heat killed multi-serotype Shigella immunogens induced humoral immunity and protec-
tion against heterologous challenge in rabbit model. Immunobiology. 220(11), 1275–1283. doi:10.1016/j.
imbio.2015.07.002 (2015).
81. Maurelli, A.T. et al. Shigella infection as observed in the experimentally inoculated domestic pig, Sus
scrofa domestica. Microb Pathog. 25, 189–196 (1998).
82. Jeong, K.I., Zhang, Q., Nunnari, J. & Tzipori, S. A piglet model of acute gastroenteritis induced by
Shigella dysenteriae Type 1. J Infect Dis. 201, 903–911 (2010).
83. Shi, R. et al. Pathogenicity of Shigella in chickens. PLoS One. 9, e100264 (2014).
84. Burton, E.A., Pendergast, A.M. & Aballay, A. The Caenorhabditis elegans ABL-1 tyrosine kinase is
required for Shigella flexneri pathogenesis. Appl Environ Microbiol. 72, 5043–5051 (2006).
85. Kesika, P., Karutha Pandian, S. & Balamurugan, K. Analysis of Shigella flexneri-mediated infections in
model organism Caenorhabditis elegans. Scand J Infect Dis. 43, 286–295 (2011).
86. Kesika, P. & Balamurugan, K. Studies on Shigella boydii infection in Caenorhabditis elegans and
bioinformatics analysis of immune regulatory protein interactions. Biochim Biophys Acta. 1824, 1449–
1456 (2012).
87. George, D.T. et al. Shigella flexneri infection in Caenorhabditis elegans: cytopathological examination
and identification of host responses. PLoS One. 9, e106085 (2014).
88. Formal, S.B., Labrec, E.H., Palmer, A. & Falkow, S. Protection of monkeys against experimental shigel-
losis with attenuated vaccines. J Bacteriol. 90, 63–68 (1965).
89. Formal, S.B., Kent, T.H., Austin, S. & Labrec, E.H. Fluorescent-antibody and histological study of vac-
cinated and control monkeys challenged with Shigella flexneri. J Bacteriol. 91, 2368–2376 (1966).
90. Takeuchi, A., Formal, S.B. & Sprinz, H. Experimental acute colitis in the rhesus monkey following
peroral infection with Shigella flexneri. An electron microscope study. Am J Pathol. 52, 503–529 (1968).
91. Rout, W.R., Formal, S.B., Giannella, R.A. & Dammin, G.J. Pathophysiology of Shigella diarrhea in the
rhesus monkey: intestinal transport, morphological, and bacteriological studies. Gastroenterology. 68,
270–278 (1975).
410 Laboratory Models for Foodborne Infections
92. Formal, S.B. et al. Oral vaccination of monkeys with an invasive Escherichia coli K-12 hybrid express-
ing Shigella flexneri 2a somatic antigen. Infect Immun. 46, 465–469 (1984).
93. Kotloff, K.L. et al. Safety, immunogenicity, and efficacy in monkeys and humans of invasive Escherichia
coli K-12 hybrid vaccine candidates expressing Shigella flexneri 2a somatic antigen. Infect Immun. 60,
2218–2224 (1992).
94. Collins, T.A. et al. Safety and colonization of two novel virG(icsA)-based live Shigella sonnei vaccine
strains in rhesus macaques (Macaca mulatta). Comp Med. 58, 88–94 (2008).
95. Islam, D. et al. Evaluation of an intragastric challenge model for Shigella dysenteriae 1 in rhesus mon-
keys (Macaca mulatta) for the pre-clinical assessment of Shigella vaccine formulations. APMIS. 122,
463–475 (2014).
96. Takasaka, M. et al. Isolation and pathogenicity of provisional serovar 1621–54 of “Shigella” from
imported cynomolgus monkeys. Jpn J Med Sci Biol. 36, 27–37 (1983).
97. Shipley, S.T. et al. A challenge model for Shigella dysenteriae 1 in cynomolgus monkeys (Macaca fas-
cicularis). Comp Med. 60, 54–61 (2010).
98. Gregory, M. et al. Development of an Aotus nancymaae model for Shigella vaccine immunogenicity and
efficacy studies. Infect Immun. 82, 2027–2036 (2014).
99. Torraca, V., Masud, S., Spaink, H.P. & Meijer, A.H. Macrophage–pathogen interactions in infectious
diseases: new therapeutic insights from the zebrafish host model. Dis Model Mech. 7, 785–797 (2014).
100. Mostowy, S. et al. The zebrafish as a new model for the in vivo study of Shigella flexneri interaction with
phagocytes and bacterial autophagy. PLoS Pathog. 9, e1003588 (2013).
101. Sansonetti, P.J., Ryter, A., Clerc, P., Maurelli, A.T. & Mounier, J. Multiplication of Shigella flexneri
within HeLa cells: lysis of the phagocytic vacuole and plasmid-mediated contact hemolysis. Infect
Immun. 51, 461–469 (1986).
102. Clerc, P., Baudry, B. & Sansonetti, P.J. Plasmid-mediated contact haemolytic activity in Shigella spe-
cies: correlation with penetration into HeLa cells. Ann Inst Pasteur Microbiol. 137A, 267–278 (1986).
103. Clerc, P. & Sansonetti, P.J. Entry of Shigella flexneri into HeLa cells: evidence for directed phagocytosis
involving actin polymerization and myosin accumulation. Infect Immun. 55, 2681–2688 (1987).
104. Faherty, C.S. et al. Microarray analysis of Shigella flexneri-infected epithelial cells identifies host fac-
tors important for apoptosis inhibition. BMC Genomics. 11, 272 (2010).
105. Lum, M. & Morona, R. Myosin IIA is essential for Shigella flexneri cell-to-cell spread. Pathog Dis. 72,
174–187 (2014).
106. Zurawski, D.V., Mitsuhata, C., Mumy, K.L., McCormick, B.A. & Maurelli, A.T. OspF and OspC1 are
Shigella flexneri type III secretion system effectors that are required for postinvasion aspects of viru-
lence. Infect Immun. 74, 5964–5976 (2006).
107. Zurawski, D.V., Mumy, K.L., Faherty, C.S., McCormick, B.A. & Maurelli, A.T. Shigella flexneri type
III secretion system effectors OspB and OspF target the nucleus to downregulate the host inflammatory
response via interactions with retinoblastoma protein. Mol Microbiol. 71, 350–368 (2009).
108. Nandakumar, N.S., Pugazhendhi, S. & Ramakrishna, B.S. Effects of enteropathogenic bacteria & lac-
tobacilli on chemokine secretion & Toll like receptor gene expression in two human colonic epithelial
cell lines. Indian J Med Res. 130, 170–178 (2009).
109. Harrison, L.M., van Haaften, W.C. & Tesh, V.L. Regulation of proinflammatory cytokine expression
by Shiga toxin 1 and/or lipopolysaccharides in the human monocytic cell line THP-1. Infect Immun. 72,
2618–2627 (2004).
110. Harrison, L.M., van den Hoogen, C., van Haaften, W.C. & Tesh, V.L. Chemokine expression in the
monocytic cell line THP-1 in response to purified Shiga toxin 1 and/or lipopolysaccharides. Infect
Immun. 73, 403–412 (2005).
111. Clerc, P.L., Ryter, A., Mounier, J. & Sansonetti, P.J. Plasmid-mediated early killing of eukaryotic cells
by Shigella flexneri as studied by infection of J774 macrophages. Infect Immun. 55, 521–527 (1987).
112. Fernandez-Prada, C.M. et al. Shigella flexneri IpaH(7.8) facilitates escape of virulent bacteria from the
endocytic vacuoles of mouse and human macrophages. Infect Immun. 68, 3608–3619.
113. Nonaka, T., Kuwabara, T., Mimuro, H., Kuwae, A. & Imajoh-Ohmi, S. Shigella-induced necrosis and
apoptosis of U937 cells and J774 macrophages. Microbiology. 149, 2513–2527 (2003).
114. Dragoi, A.M. & Agaisse, H. The serine/threonine kinase STK11 promotes Shigella flexneri dissemina-
tion through establishment of cell–cell contacts competent for tyrosine kinase signaling. Infect Immun.
82, 4447–4457 (2014).
Shigella 411
115. Kuehl, C.J., Dragoi, A.M. & Agaisse, H. The Shigella flexneri type 3 secretion system is required for
tyrosine kinase-dependent protrusion resolution, and vacuole escape during bacterial dissemination.
PLoS One. 9, e112738 (2014).
116. Nonaka, T., Kuwae, A., Sasakawa, C. & Imajoh-Ohmi, S. Shigella flexneri YSH6000 induces two types
of cell death, apoptosis and oncosis, in the differentiated human monoblastic cell line U937. FEMS
Microbiol Lett. 174, 89–95 (1999).
117. Yu, J., Oragui, E.E., Stephens, A., Kroll, J.S. & Venkatesan, M.M. Inactivation of DsbA alters the behav-
iour of Shigella flexneri towards murine and human-derived macrophage-like cells. FEMS Microbiol
Lett. 204, 81–88 (2001).
118. Paciello, I. et al. Intracellular Shigella remodels its LPS to dampen the innate immune recognition and
evade inflammasome activation. Proc Natl Acad Sci USA. 110, E4345–E4354 (2013).
119. Rossi, O. et al. Modulation of endotoxicity of Shigella generalized modules for membrane antigens
(GMMA) by genetic lipid A modifications: relative activation of TLR4 and TLR2 pathways in different
mutants. J Biol Chem. 289, 24922–24935 (2014).
120. Mounier, J., Vasselon, T., Hellio, R., Lesourd, M. & Sansonetti, P.J. Shigella flexneri enters human
colonic Caco-2 epithelial cells through the basolateral pole. Infect Immun. 60, 237–248 (1992).
121. Pédron, T., Thibault, C. & Sansonetti, P.J. The invasive phenotype of Shigella flexneri directs a distinct
gene expression pattern in the human intestinal epithelial cell line Caco-2. J Biol Chem. 278, 33878–
33886 (2003).
122. Lima, C.B., Santos, S.A. & Andrade Júnior, D.R. Hypoxic stress, hepatocytes and Caco-2 viability and
susceptibility to Shigella flexneri invasion. Rev Inst Med Trop Sao Paulo. 55, 341–346 (2013).
123. Konradt, C. et al. The Shigella flexneri type three secretion system effector IpgD inhibits T cell migra-
tion by manipulating host phosphoinositide metabolism. Cell Host Microbe. 9, 263–272 (2011).
28
Vibrio: Caenorhabditis elegans as a Laboratory
Model for Vibrio Infections
CONTENTS
28.1 V. cholerae....................................................................................................................................413
28.2 Models to Study V. cholerae.........................................................................................................414
28.3 V. alginolyticus..............................................................................................................................414
28.4 Models to Study V. alginolyticus..................................................................................................415
28.5 V. parahaemolyticus.....................................................................................................................415
28.6 Models to Study V. parahaemolyticus..........................................................................................416
28.7 C. elegans as Model for Studying Bacterial Infection..................................................................417
28.8 C. elegans for Host–Pathogen Interaction Studies.......................................................................418
28.9 C. elegans as a Model for Vibrio spp. Infection...........................................................................418
28.10 Conclusion....................................................................................................................................421
Acknowledgements..................................................................................................................................421
References............................................................................................................................................... 422
Vibriosis is a common illness among seafood consumers that is caused by bacteria of the family
Vibrionaceae. Among many species within the Vibrionaceae family, Vibrio cholerae, Vibrio parahae-
molyticus, Vibrio vulnificus, and Vibrio alginolyticus are important foodborne pathogens in humans.
Transmitted through uncooked or partially cooked seafood, vibriosis produces a range of symptoms,
including cholera, dysentery, diarrhea, gastroenteritis, sepsis, and fasciitis. The virulence, mode of infec-
tion, symptoms, and treatment are different for each of these species. Showing variations in virulent
factors as well as differences between the wild-type and clinical isolates, the pathogenicity of Vibrio spe-
cies is complex. The nematode Caenorhabditis elegans has been the preferred model system for many
investigators to study pathogenesis of this disease. The current chapter summarizes the available models
for studying Vibrio spp. infections, with an emphasis on the utility of C. elegans model.
28.1 V
. cholerae
Vibrio cholerae is facultative anaerobic, Gram-negative, non-spore-forming, curved rod-shaped bac-
terium measuring 1.4–2.6 µm with the ability for respiratory and fermentative metabolism, which can
be defined on the basis of biochemical test and DNA sequence homology analysis. The bacterium is
oxidase-positive, reduces nitrate, and moves using a single sheathed polar flagellum. The growth of the
bacterium is regulated by altering sodium chloride (NaCl) concentration.1 V. cholerae moves by means
of its single polar flagellum, which is driven by the sodium-motive force.2 The ability of the bacterium to
grow in nutrient broth without NaCl makes it unique among Vibrio spp. V. cholera is a causative agent
for the deadly disease cholera, which is considered as a life-threatening intestinal infection. Millions
of people are affected with cholera every year, and the disease accounts for thousands of causalities
413
414 Laboratory Models for Foodborne Infections
worldwide, most commonly in developing countries, and the number of cholera cases increases day by
day.3,4 Infections spread through contaminated water and seafood. In the lumen of the small intestine, the
bacterium begins to multiply persistently, leading to colonization. Recent studies reported the ability of
this pathogen to successfully metabolize glucose, thereby facilitating its proliferation in a glucose-rich
environment like the human intestine.5
V. cholerae initiates infection by adhering to the epithelium while swimming, which is the first step
of colonization. Quorum sensing plays an important role in the colonization of the host system by
V. cholerae.6 Evolution, growth, and virulence of V. cholerae is rigidly controlled by the location of
ribosomal protein gene locus and its distance from the origin of replication.7 During antibiotic treatment,
the pathogen modulates its shape from a rod to a sphere, thereby protecting itself from the antibiotics.8
The availability of model organisms and information about the bacterial genetics has enabled the under-
standing of the few aspects of V. cholerae pathogenicity in humans; however, complete information
about the natural habitat of the bacterium and its interaction with host system remains unknown.
28.3 V
. alginolyticus
Vibrio alginolyticus is a common inhabitant of the marine environment in both temperate and tropical
waters.19,20 V. alginolyticus is also present in estuarine environments and is frequently isolated from bath-
ing areas as free-living bacteria and is also associated with biotic and abiotic surfaces.21 V. alginolyticus
is one of the most common and important pathogen that causes vibriosis in human and marine species,
and infection in humans was first recognized in 1973. The complete genome sequence of V. alginolyticus
ATCC 17749 was revealed recently.22 Infection of fish by V. alginolyticus results in biofilm formation
Vibrio: C. elegans as a Laboratory Model for Vibrio Infections 415
in their intestine, which leads to mortality and potentially significant economic losses. V. alginolyticus
has been described to cause human infections including wound infection (71%), gastroenteritis (12%),
ear infection, and septicemia.23 Other clinical syndromes reported in association with V. alginolyticus
infection include chronic diarrhea in a patient with acquired immunodeficiency syndrome (AIDS), con-
junctivitis, and posttraumatic intracranial infection.
V. alginolyticus produces many extracellular proteases responsible for interaction between the bacte-
rium and cell hosts (human and fish) and plays an important role in infections.24,25 The mechanism of
pathogenicity induced by V. alginolyticus infections is complex and related to several factors, including
cytotoxins, enterotoxins, and lytic enzymes.26,27 The adhesive properties of Vibrio spp. are considered
as the key factor of bacterial pathogenicity and have been implicated as a risk factor for infection in
humans and stressed aquatic animals.21,28,29 A set of genes regulated during environmental stress are
known for their key role in regulating the adhesion process of V. alginolyticus.30 Larger numbers of
Vibrio spp. strains were frequently isolated from zooplankton, especially from copepods.21 The ability
to form biofilms on biotic and abiotic surfaces and to activate a survival state called “Viable but not
cultivable” allow V. alginolyticus strains to persist in seawater under environmental stress conditions.31
In V. alginolyticus, genetic analysis reveals that there is no correlation between the virulence of strains
and the presence of general virulent genes such as tlh, trh, toxR, toxRS, ctxA, and VPI, which are present
and contribute to the virulence in other Vibrio spp. pathogens.32,33
Production of multiple virulent extracellular products (ECP), mainly protease, is used to characterize
the virulent strains of V. alginolyticus.34,35 Extracellular alkaline serine proteases (Asp) have been dem-
onstrated to be important exotoxins of V. alginolyticus strains pathogenic to prawn35 and fish,36 and their
expressions are subjected to the regulation of a LuxO–LuxR quorum-sensing regulatory circuit.37 At
present, little is known about the relationship between the secretion systems and ECP production as well
as their contribution to the pathogenesis in V. alginolyticus. A recent study identified and characterized
the Tat translocation system in V. alginolyticus. The Tat system was strongly involved in the swarming
ability and biofilm formation in this bacterium. In addition, the Tat system in V. alginolyticus was also
required for virulence against the fish model and epithelial cells. These findings suggest that Tat secre-
tion is related to the extracellular protease activity as well as virulence in V. alginolyticus.38
illness around the world.49 Clinical manifestations of V. parahaemolyticus infections include gastroen-
teritis, wound infection, and septicemia. Almost all clinical V. parahaemolyticus show β-hemolysis in
a Wagatsuma agar, and this hemolytic activity is termed the Kanagawa phenomenon (KP).50 The KP
is considered as a good marker to differentiate human pathogenic V. parahaemolyticus strains from
the nonpathogenic strains. Almost all clinical isolates carry tdh, trh, or both genes, whereas 1%–2% of
environmental isolates may likely possess these genes.50 Isolates lacking tdh and trh genes are also iso-
lated from patients with V. parahaemolyticus infection symptoms. Both virulent and avirulent strains of
V. parahaemolyticus are known to occur in a host, leading to in vivo serotype diversity.51 The complete
genome sequence of clinical and environmental isolates were reported recently52,53 followed by draft
genome sequence of 14 Canadian V. parahaemolyticus clinical isolates that were serologically identified
as K group II using polyvalent antisera but were not specifically K serogrouped using monovalent anti-
sera.53 The use of whole-genome sequencing coupled with phylogeny and multiplex polymerase chain
reaction (PCR) assay is also under development for identifying sequence of several V. parahaemolyticus
isolates.54 The complete genome sequence followed by pulsed-field electrophoresis showed that some
V. parahaemolyticus environmental isolates exhibit significant genetic similarity to clinical isolates,
which enables them to make the transition from normal aquatic state to a pathogen during infection.55
Most of the studies exploring the pathogenesis of V. parahaemolyticus on the host focus on TDH and
TRH. TDH is responsible for various cytotoxicity effects that include erythrocyte lysis, disruption of
the microtubule cytoskeleton, and ion flux disturbance in cultured cells.56–58 Very less is known about
the targets of TRH; a study on the purified TRH toxin indicates their effect on cultured erythrocytes
and also result in the fluid accumulation in rabbit ileal loop model.59 Infection by V. parahaemolyticus
bacteria results in cell rounding and disruption of epithelial barrier function, which is mediated through
the effect of TDH on the ion flow; this loss of barrier results in the excessive loss of body fluid and the
condition is termed diarrhea.50
The genetic approach for identifying genes regulating the virulent pathway in V. parahaemolyticus
showed a close association between the urease gene and the TRH gene.60 Hence, it is hypothesized that
possession of the gene for TRH coincides with the presence of the urease gene among many clinical
V. parahaemolyticus strains, making urease gene a marker for identifying clinically pathogenic trh posi-
tive strains.61 Urease is an enzyme involved in the hydrolysis of urea into ammonia and carbon dioxide
and is present across kingdoms in plants, fungi, algae, and bacteria.62 Many bacterial ureases have been
characterized, and the organization of this gene is found to be similar among bacteria with regard to the
structural and accessory genes involved in processing nickel ions.63 Comparing with other Vibrio spp.,
only a small portion of clinical V. parahaemolyticus isolates possess the urease gene.61 But the presence
of urease gene is always associated with presence of trh gene. Specialized PCR techniques such as the
long and accurate PCR have revealed that the distance between these two genes is <8.5 kb.64
Type III secretion systems (T3SSs) are considered as one among the major virulent pathways in many
Gram-negative pathogens; bacteria use this system to translocate the produced virulent factors to the
cytosol of the host cells.65 The T3SS apparatus is found to be highly conserved across Gram-negative
bacteria, and the specific property of the effectors brings about symptomatic effects on the host organ-
ism that vary widely.65 Genome sequence of TDH-positive V. parahaemolyticus strains displayed the
presence of gene sequences that are similar to T3SS present in chromosome 1 and 2, which are named
T3SS1 and T3SS2, respectively.66 T3SS1 gene sequence was found to be present in most or all of the
V. parahaemolyticus strains, whereas the T3SS2 was found only in KP-positive strains. Intensive study on
the T3SS of V. parahaemolyticus revealed that these two systems confer specific phenotypes to the bacteria.
Understanding the effector proteins necessary for the activation of T3SS and environmental influence on
the regulation of T3SS will provide deeper insight into V. parahaemolyticus infection in humans.64
induces cytotoxicity in several cell lines, while T3SS2 can induce cytotoxicity in Caco-2 cells and entero-
toxicity in vivo.69,70 The importance of TDH and T3SSs in V. parahaemolyticus virulence was recently
assessed using a mouse model. These results clarified that T3SS2 rather than TDH was the major con-
tributor to enterotoxicity; however, both T3SS1 and TDH contributed to lethality in a mouse model, sug-
gesting that TDH and/or other proteins(s) that are part of T3SS1 are also likely to contribute to virulence.71
Two different animal models were used to understand the contributions of T3SS1 and T3SS2 to V. para-
haemolyticus virulence.72 Orogastric inoculation of V. parahaemolyticus in neonatal pigs demonstrated
that T3SS2, but not T3SS1, was required for gastrointestinal disease. The study also showed that T3SS1
was necessary to cause mortality during intrapulmonary inoculation, which raises questions on role of
unknown factors related to T3SS1 responsible for lethality.72 Host intestine damage during V. parahae-
molyticus infection was studied using infant rabbit model.73 Enteroinvasiveness of the bacterium has been
reported for a rabbit model, in which the organism invaded, colonized, and produced inflammation in the
small intestine. In another study, an adult zebrafish model was investigated to assess the overall virulence
of V. parahaemolyticus strains.74 The model detected differences in the virulence potential of strains when
animals were challenged intraperitoneally, and this was based on survival time. Differences in survival
were noted irrespective of the source of isolation of the strain (environmental or clinical) and regardless
of the presence or absence of the known virulence factors TDH and TRH, suggesting the influence of
additional virulence factors. The model was also effective in comparing differences in virulence between
the wild-type V. parahaemolyticus strain RIMD2210633 and isogenic pilin mutants.
28.7 C
. elegans as Model for Studying Bacterial Infection
Caenorhabditis elegans was first discovered and described by Emile Maupas, a French zoologist and
botanist, in 1900 from soil in Algeria.75 Earlier, Maupas used Rhabditis as genus name rather than
Caenorhabditis. In 1952, the new subgenus Caenorhabditis was coined,76 which was later raised
into generic status by Dougherty in 1955. The definition of genera is somewhat arbitrary. Even now,
Caenorhabditis is a subgenus name, one among many in the Rhabditis genus, and C. elegans are formally
called Rhabditis (Caenorhabditis) elegans. C. elegans is a eukaryotic and free-living soil nematode. It
has a complex developmental process and includes stages of embryogenesis, morphogenesis, and growth
to an adult. C. elegans demonstrates about 35% of homology to human at genome level and 83% similar-
ity in protein sequence.77 C. elegans was introduced to the scientific community by Sydney Bernner as
an experimental model in 1960s in order to explore animal developmental biology when he published
his first report on the behavior of the nematode.78 According to the C. elegans Sequencing Consortium,
its genome has been completely sequenced, and the genome size is around 100.2 Mb. The availability of
the entire genome sequence is useful in genetic studies, allowing researchers to pick a gene of interest
to study. Surprisingly, it has been shown frequently that human genes replace their C. elegans homologs
when introduced into C. elegans, and many genes in C. elegans can function similar to mammalian genes.
In contrast to other invertebrates, immunity in C. elegans solely depends on the innate immune system
as it lacks the sophisticated adaptive immune signaling.79 With this primary innate immune system, the
nematode’s immune response involves the production of numerous antimicrobial proteins that are con-
served in higher vertebrates and humans. Exploring the innate immune signaling pathways in C. elegans
revealed that many pathways important for development in early stages were reused in the adult stage
for immune signaling. The nematode’s defense involves three major mechanisms against any microbial
infection:80 (1) avoidance behavior, (2) physical barrier, and (3) inducible signaling pathways.
The neuronal system in C. elegans is well equipped in differentiating between the food source and
pathogenic bacterium.81 The repelling behavior against Serratia marcescens shed light on the role of sev-
eral neurons, G-protein-coupled receptors, and TOL-1.82,83 The cuticle of C. elegans is made of collagen
and chitin that act as a strong physical barrier that cannot be pierced easily, thereby protecting from the
entry of pathogens via the dermal layer.84,85 The immune system includes pathogen recognition by PRR
and other signaling cascades like MAPK, DAF-2, UPR, TGF-β, apoptosis, and necrosis pathways.86–88
Forward and reverse genetic studies have identified genes that belong to six signaling cascades, which
are activated upon infection with Gram-positive and Gram-negative bacteria, fungi, and bacterial toxins.
418 Laboratory Models for Foodborne Infections
The six pathways include ERK MAP kinase,89 p38 MAP kinase,90 TGF-β,91 programmed cell death,92
DAF-2/DAF-16 insulin-like receptor signaling,93 and JNK-like MAP kinase.94
28.8 C
. elegans for Host–Pathogen Interaction Studies
Studies in C. elegans have shed light on many pathways of the innate immune system that are phyloge-
netically conserved from worms to humans.95 C. elegans is a very good model to study both the host and
the microbe factors that underscore the host–pathogen interaction.96
M. nematophilum is one among many pathogens that infect C. elegans in its natural environment.
It infects C. elegans and leads to an unusual and visible tail swelling or deformed anal region (Dar),
formerly believed to be a spontaneous heritable and morphological mutation that arises during a regular
genetic cross. Later, Hodgkin et al. revealed that M. nematophilum establishes a specific rectal infection
due to its strong extracellular adherence to the cuticle, resulting in the swelling response and the Dar
phenotype.97,98 The tail-swelling phenotype observed during M. nematophilum infection is related to the
enhancement of immune pathways ERK and MAPK in C. elegans as a response of the innate immune
system against the bacterial infection.89
Studies with mutant strains that lack the bacterially unswollen (Bus) phenotype revealed that the
role of several genes responsible for the Dar phenotype have implications on both the host (capabil-
ity to elicit a swelling response) and the bacterium (ability to adhere and colonize).99 The role of genes
responsible for normal cuticle development and prevention of bacterial adherence have been reported in
previous studies.100 Quantitative molecular studies identified nearly 68 host genes that are induced upon
M. nematophilum infection; most of these genes are pathogen receptor molecules including C-type lectin
domains and lysozymes.101
Drechmeria coniospora is an agriculturally important nematode parasite that infects C. elegans
through the mouth and penetrates the body by means of the proteinaceous hyphae.102 C. elegans triggers
an immune response against the infection through the induction of neuropeptide-like proteins (NLPs).103
Nematocida parisii is a microsporidian parasite of C. elegans, and its infection did not induce basic
immune-responsible genes known to be vital for other pathogenic infections but alters the components of
the immune-signaling pathways (p38 MAPK and insulin signaling/DAF-2 pathways), thereby affecting
animal survival.104 Recent studies have reported the interaction between C. elegans and Orsay virus, a
small, positive-sense RNA virus belonging to the family Nodaviridae.105
using RNAi. By means of a machine learning algorithm called FastMEDUSA, the study identified sev-
eral putative VCC-induced immune regulatory transcriptional factors and transcription factor binding
motifs. Results from the experiments suggest that VCC induces a wide variety of immune-response-
related genes during V. cholerae infection in C. elegans.108
A recent study by Durai et al. established C. elegans as a model for studying phenotypic changes and
regulation of immune-responsible genes in the host against bacterial infection. Preference of C. elegans
between nonpathogenic E. coli OP50 and pathogenic V. alginolyticus was well documented in the study.
The infection was localized using green fluorescent protein (GFP)-tagged pathogen, and intestinal colo-
nization was reported as the major reason for mortality in C. elegans.109 Since C. elegans displayed lawn
avoidance behavior to V. alginolyticus, a liquid killing assay was developed that comprised both M9
buffer and pathogen liquid culture (in a 3:1 ratio). Infection with V. alginolyticus in liquid assay reduced
the lifespan of the nematode significantly. However, earlier reports revealed a reduction in lifespan of
C. elegans exposed to V. cholerae, with complete killing of nematode in approximately 5 days,1 while
with V. vulnificus infection, the TD50 (toxic dose) of the nematode was found to be 9 days.110 Microscopic
image analysis of the infected worm showed colonization of GFP-tagged V. alginolyticus in the pharyn-
geal region and in the intestine of the pathogen-exposed animals. V. alginolyticus was able to colonize
the gut of C. elegans within a short period of time, i.e., after 8 h of infection, and the bacterial load from
the colonized animals were found to be ∼3.7 ± 0.3 × 103 CFU. It was also observed that a bacterial load
of at least 6.5 × 104 CFU of V. alginolyticus was required to cause and maintain infection in C. elegans.
It was also found that V. alginolyticus induced a strong inflammatory response in the intestine, which
appeared to be more severe than the infection caused by the V. cholerae. The persistence of V. alginolyti-
cus leads to the damage of the pharyngeal bulb and distention of intestinal lumen, which was confirmed
by confocal laser scanning microcopy (CLSM) image analyses.
C. elegans are attracted toward the smell of bacteria for a short period of time irrespective of the bacte-
rial nature. On sensing the pathogens, in this case V. alginolyticus, the worms learned to avoid the smell
of the bacterium and defended themselves from infection. The lawn-avoidance behavior of C. elegans
observed in the study could be attributed to its chemosensory response to the pathogen (Figure 28.1).
Zone B V.alginolyticus
E. coli OP50 Zone B
(a) (b)
Zone A
E. coli OP50
Zone B V.alginolyticus
(c)
FIGURE 28.1 Chemotaxis behavior: in all cases, animals were tracked on an agar plate. (a) Animals in the presence of
food source, E. coli OP50 marked at zone A and zone B. (b) Animals in presence of V. alginolyticus marked at zone A and
zone B. (c) Animals in presence of food source, E. coli OP50 marked as zone A and pathogen source, V. alginolyticus
marked as zone B. (Adapted from Durai, S. et al., J. Basic Microbiol., 51, 243–52, 2011. With permission.)
420 Laboratory Models for Foodborne Infections
The immune response of C. elegans against V. alginolyticus infection was analyzed using quantitative
PCR (qPCR). Analogous to the previous studies in M. nematophilum, three candidate genes, viz., lys-7,
clec-60, and clec-87, were analyzed for their expression during infection by having animals treated with a
food source as control.101 The level of expression of the immune effector gene(s) was significantly higher
when compared to the control animals. During the initial stage of infection, the C-type lectin genes clec-60
and clec-87 seem to be responsible for the enhancement in adhesion and antibacterial activity.101 The role
of lysozyme gene lys-7 as a presumptive antimicrobial gene and in induction of immune response as well
as its encoded protein in the upregulation to infection with Gram-negative bacterium S. marcescens has
previously been reported.111
Proteome regulation in C. elegans against V. alginolyticus was also explored by Durai et al.112 Proteins
were separated using two-dimensional differential gel electrophoresis (2D-DIGE), and the differentially
regulated proteins were identified using positive matrix factorization (PMF) and matrix-assisted laser
desorption/ionization–time of flight (MALDI–TOF)/TOF analysis. The results thus obtained were vali-
dated using western blotting for candidate proteins. The corresponding transcriptional regulation was
quantified subsequently using real-time PCR. Interaction network for candidate proteins was predicted
using the search tool for the retrieval of interacting genes/proteins (STRING), and functional validation
was performed using respective mutant strains. Out of the 25 proteins identified, 21 proteins appeared to
be upregulated, while 4 were downregulated (Table 28.1). Upregulated proteins included those involved
in stress response (PDI-2, HSP-6), immune response (KIN-18, GST-8), and energy production (ATP-2),
while proteins involved in structural maintenance (IFB-2) and lipid metabolism (SODH-1) were down-
regulated. The roles of these players in the host system during Vibrio infection were analyzed in vivo
using wild-type and mutant C. elegans. Survival assays using mutants lacking pdi-2, ire-1, and xbp-1
displayed enhanced susceptibility to V. alginolyticus. Cellular stress generated by V. alginolyticus was
determined using reactive oxygen species (ROS) assay. This is the first report of proteome changes in
TABLE 28.1
Differentially Expressed Early Responsive Proteins of C. elegans against V. alginolyticus
Infection
S.No. Protein Gene
A. Upregulated C. elegans Protein against V. alginolyticus Infection
1 Alpha integrin Vab-20
2 Serine/threonine protein kinase kin-18
3 Mitochondrial ribosomal protein, small mrp-25
4 Heat shock protein hsp-6
5 Protein BCCIP homolog ZK1127.4
6 COP9/signalosome and eIF3 complex shared subunit K08F11.3
7 H1 histone hil-1
8 Glucose-6-phosphate 1 dehydrogenase gspd-1
9 Protein disulfide isomerase pdi-2
10 DNA-directed RNA polymerase subunit II phi-14
11 ATP synthase subunit-2 atp-2
12 Glutathione S-transferase 8 gst-8
13 cGMP-dependent protein kinase pkg-1
14 Eukaryotic translation initiation factor 3 subunit eif-3.G
15 Putative aminopeptidase lap-2
16 Nuclear hormone receptor family nhr-44
17 Probable signal recognition protein F55C5.8
B. Downregulated C. elegans Protein against V. alginolyticus Infection
1 Intermediate filament protein ifb-2
2 Sorbitol dehydrogenase sodh-1
3 Putative selenium-binding protein R11G10.2
Vibrio: C. elegans as a Laboratory Model for Vibrio Infections 421
50 µm
50 µm
(a) 50 µm (b) (c)
FIGURE 28.2 CLSM images of C. elegans exposed to GFP-tagged (a) E. coli OP50, (b) V. parahaemolyticus CM2, and
(c) V. parahaemolyticus ATCC.
C. elegans against V. alginolyticus challenge and highlights the significance of unfolded protein response
(UPR) pathway during bacterial infection.
Changes in C. elegans against V. parahaemolyticus infection were reported by Durai et al.113 The study
compared the virulence of two V. parahaemolyticus strains and proved that intestinal colonization of the
bacterium was associated with pharyngeal damage, leading to the death of the nematode in 48 h. The
colonization of V. parahaemolyticus in C. elegans intestine was localized using CLSM, and the intensity
of the colonization was measured and compared with respective controls. The 2.5-dimensional topog-
raphy of the colonized animal clearly showed the colonization of V. parahaemolyticus CM2 throughout
the intestinal lumen of C. elegans (Figure 28.2). The study also showed that transferring the nematodes
to a benign food source cleared the intestinal colonization of V. parahaemolyticus, which was similar
to the earlier observations during Staphylococcus aureus infections.114 The virulence of V. parahaemo-
lyticus is associated with the production of two major toxins, TDH and TRH.115–118 In addition, few
strains lacking these virulence genes/factors were also found to be pathogenic, indicating that infection
was independent of toxin production.119 Results from experiments showed that strain CM2 is negative
to hemolysin test performed in the Wagatsuma agar using human erythrocytes. Molecular studies for
the tdh and trh supported the hemolysin test. However, the presence of tlh favored the notion that CM2
could be a pathogenic strain of V. parahaemolyticus. Previous reports show that tlh has been used for the
molecular-level identifications of the Vibrio species.119–121 C. elegans shows a selective response to food
source and pathogenic bacteria with the phenomenon of olfactory learning.
28.10 Conclusion
With a mode of transmission through seafood contaminants, vibriosis in humans represents a challenge
to researchers. Given that many reoccurring Vibrio species containing various virulent factors (including
antibiotic resistance genes) are being identified every year, these bacteria pose a significant public risk
to human populations. Precautions and proper identification of Vibrio contamination in seafood using
advanced tools provide an effective way to control and reduce risk of infection. Further elucidation of the
molecular mechanisms of Vibrio infections and virulent determinants using model organism will help
identify potent anti-infective compounds that can be used for combating vibriosis.
Acknowledgements
This work was supported in part by the Grants from UGC, DBT, and Department of Science and
Technology to Dr. K. Balamurugan. Financial support to Durai by Lady Tata Memorial Trust’s Senior
Scholarship is thankfully acknowledged.
422 Laboratory Models for Foodborne Infections
REFERENCES
1. Vaitkevicius, K. et al. A Vibrio cholerae protease needed for killing of Caenorhabditis elegans has a
role in protection from natural predator grazing. Proc Natl Acad Sci USA 103, 9280–5 (2006).
2. Halang, P., Vorburger, T. & Steuber, J. Serine 26 in the PomB subunit of the flagellar motor is essential
for hypermotility of Vibrio cholerae. PLoS One 10, e0123518 (2015).
3. Cinar, H.N. et al. Vibrio cholerae hemolysin is required for lethality, developmental delay, and intestinal
vacuolation in Caenorhabditis elegans. PLoS One 5, e11558 (2010).
4. Vanden Broeck, D., Horvath, C. & De Wolf, M.J. Vibrio cholerae: Cholera toxin. Int J Biochem Cell
Biol 39, 1771–5 (2007).
5. Oh, Y.T., Lee, K.M., Bari, W., Raskin, D.M. & Yoon, S.S. (p)ppGpp, a small nucleotide regulator,
directs the metabolic fate of glucose in Vibrio cholerae. J Biol Chem 290(21), 13178–90 (2015).
6. Jung, S.A., Chapman, C.A. & Ng, W.L. Quadruple quorum-sensing inputs control Vibrio cholerae viru-
lence and maintain system robustness. PLoS Pathog 11, e1004837 (2015).
7. Soler-Bistue, A. et al. Genomic location of the major ribosomal protein gene locus determines Vibrio
cholerae global growth and infectivity. PLoS Genet 11, e1005156 (2015).
8. Dorr, T., Davis, B.M. & Waldor, M.K. Endopeptidase-mediated beta lactam tolerance. PLoS Pathog 11,
e1004850 (2015).
9. Spira, W.M., Sack, R.B. & Froehlich, J.L. Simple adult rabbit model for Vibrio cholerae and enterotoxi-
genic Escherichia coli diarrhea. Infect Immun 32, 739–47 (1981).
10. Klose, K.E. The suckling mouse model of cholera. Trends Microbiol 8, 189–91 (2000).
11. Sawasvirojwong, S., Srimanote, P., Chatsudthipong, V. & Muanprasat, C. An adult mouse model of
induced diarrhea for studying pathogenesis and potential therapy of cholera. PLoS Negl Trop Dis 7,
e2293 (2013).
12. Runft, D.L. et al. Zebrafish as a natural host model for Vibrio cholerae colonization and transmission.
Appl Environ Microbiol 80, 1710–7 (2014).
13. De, S.N. & Chatterje, D.N. An experimental study of the mechanism of action of Vibrio cholerae on the
intestinal mucous membrane. J Pathol Bacteriol 66, 559–62 (1953).
14. Dutta, N.K. & Habbu, M.K. Experimental cholera in infant rabbits: A method for chemotherapeutic
investigation. Br J Pharmacol Chemother 10, 153–9 (1955).
15. Pierce, N.F., Greenough, W.B., III & Carpenter, C.C., Jr. Vibrio cholerae enterotoxin and its mode of
action. Bacteriol Rev 35, 1–13 (1971).
16. Blachman, U., Graboff, S.R., Haag, G.E., Gottfeld, E. & Pickett, M.J. Experimental cholera in chinchil-
las: The immune response in serum and intestinal secretions to Vibrio cholerae and cholera toxin. Infect
Immun 10, 1098–104 (1974).
17. Finkelstein, R.A. Cholera, Vibrio cholerae O1 and O139, and Other Pathogenic Vibrios. University of
Texas Medical, Galveston (1996).
18. Millet, Y.A. et al. Insights into Vibrio cholerae intestinal colonization from monitoring fluorescently
labeled bacteria. PLoS Pathog 10, e1004405 (2014).
19. Chakraborty, S., Nair, G.B. & Shinoda, S. Pathogenic vibrios in the natural aquatic environment. Rev
Environ Health 12, 63–80 (1997).
20. Zanetti, S. et al. Differentiation of Vibrio alginolyticus strains isolated from Sardinian waters by ribo-
typing and a new rapid PCR fingerprinting method. Appl Environ Microbiol 65, 1871–5 (1999).
21. Baffone, W. et al. Detection of free-living and plankton-bound vibrios in coastal waters of the Adriatic
Sea (Italy) and study of their pathogenicity-associated properties. Environ Microbiol 8, 1299–305 (2006).
22. Liu, X.F., Cao, Y., Zhang, H.L., Chen, Y.J. & Hu, C.J. Complete genome sequence of Vibrio alginolyti-
cus ATCC 17749T. Genome Announc 3(1), e01500-14 (2015).
23. Hlady, W.G. & Klontz, K.C. The epidemiology of Vibrio infections in Florida, 1981–1993. J Infect Dis
173, 1176–83 (1996).
24. Hare, P., Scott-Burden, T. & Woods, D.R. Characterization of extracellular alkaline proteases and
collagenase induction in Vibrio alginolyticus. J Gen Microbiol 129, 1141–7 (1983).
25. Lee, K.K. Pathogenesis studies on Vibrio alginolyticus in the grouper, Epinephelus malabaricus, Bloch
et Schneider. Microb Pathog 19, 39–48 (1995).
26. Iyer, L., Vadivelu, J. & Puthucheary, S.D. Detection of virulence associated genes, haemolysin and
protease amongst Vibrio cholerae isolated in Malaysia. Epidemiol Infect 125, 27–34 (2000).
Vibrio: C. elegans as a Laboratory Model for Vibrio Infections 423
27. Ottaviani, D. et al. Antimicrobial susceptibility of potentially pathogenic halophilic vibrios isolated
from seafood. Int J Antimicrob Agents 18, 135–40 (2001).
28. Balebona, M.C., Morinigo, M.A. & Borrego, J.J. Role of extracellular products in the pathogenicity of
Vibrio strains on cultured gilt-head seabream (Sparus aurata). Microbiologia 11, 439–46 (1995).
29. Zorrilla, I., Morinigo, M.A., Castro, D., Balebona, M.C. & Borrego, J.J. Intraspecific characterization of
Vibrio alginolyticus isolates recovered from cultured fish in Spain. J Appl Microbiol 95, 1106–16 (2003).
30. Kong, W. et al. Investigation of possible molecular mechanisms underlying the regulation of adhe-
sion in Vibrio alginolyticus with comparative transcriptome analysis. Antonie Van Leeuwenhoek 107,
1197–206 (2015).
31. Ben Kahla-Nakbi, A., Besbes, A., Chaieb, K., Rouabhia, M. & Bakhrouf, A. Survival of Vibrio algino-
lyticus in seawater and retention of virulence of its starved cells. Mar Environ Res 64, 469–78 (2007).
32. Deriu, A., Sechi, L.A., Molicotti, P., Spanu, M.L. & Zanetti, S. Virulence genes in halophilic Vibrio spp.
isolated in common mussels. New Microbiol 25, 93–6 (2002).
33. Xie, Z.Y., Hu, C.Q., Chen, C., Zhang, L.P. & Ren, C.H. Investigation of seven Vibrio virulence genes
among Vibrio alginolyticus and Vibrio parahaemolyticus strains from the coastal mariculture systems
in Guangdong, China. Lett Appl Microbiol 41, 202–7 (2005).
34. Lee, K.K., Chen, F.R., Yu, S.R., Yang, T.I. & Liu, P.C. Effects of extracellular products of Vibrio
alginolyticus on penaeid prawn plasma components. Lett Appl Microbiol 25, 98–100 (1997).
35. Lee, K.K., Yu, S.R. & Liu, P.C. Alkaline serine protease is an exotoxin of Vibrio alginolyticus in kuruma
prawn, Penaeus japonicus. Curr Microbiol 34, 110–7 (1997).
36. Cai, S.H., Wu, Z.H., Jian, J.C. & Lu, Y.S. Cloning and expression of the gene encoding an extracellular
alkaline serine protease from Vibrio alginolyticus strain HY9901, the causative agent of vibriosis in
Lutjanus erythopterus (Bloch). J Fish Dis 30, 493–500 (2007).
37. Rui, H., Liu, Q., Ma, Y., Wang, Q. & Zhang, Y. Roles of LuxR in regulating extracellular alkaline serine
protease A, extracellular polysaccharide and mobility of Vibrio alginolyticus. FEMS Microbiol Lett
285, 155–62 (2008).
38. He, H., Wang, Q., Sheng, L., Liu, Q. & Zhang, Y. Functional characterization of Vibrio alginolyticus
twin-arginine translocation system: Its roles in biofilm formation, extracellular protease activity, and
virulence towards fish. Curr Microbiol 62, 1193–9 (2012).
39. George, M.R., John, K.R., Iyappan, T. & Jeyaseelan, M.J. Genetic heterogeneity among Vibrio algino-
lyticus isolated from shrimp farms by PCR fingerprinting. Lett Appl Microbiol 40, 369–72 (2005).
40. Ye, J. et al. Regulation of Vibrio alginolyticus virulence by the LuxS quorum-sensing system. J Fish Dis
31, 161–9 (2008).
41. Chen, Q., Yan, Q., Wang, K., Zhuang, Z. & Wang, X. Portal of entry for pathogenic Vibrio alginolyticus
into large yellow croaker Pseudosciaena crocea, and characteristics of bacterial adhesion to mucus. Dis
Aquat Organ 80, 181–8 (2008).
42. Harikrishnan, R., Balasundaram, C. & Heo, M.S. Poly d,l-lactide-co-glycolic acid-liposome encap-
sulated ODN on innate immunity in Epinephelus bruneus against Vibrio alginolyticus. Vet Immunol
Immunopathol 147, 77–85 (2012).
43. Wu, C.J., Wang, H., Chan, Y.L. & Li, T.L. Passive immune-protection of small abalone against Vibrio
alginolyticus infection by anti-Vibrio IgY-encapsulated feed. Fish Shellfish Immunol 30, 1042–8 (2011).
44. Liang, H., Xia, L., Wu, Z., Jian, J. & Lu, Y. Expression, characterization and immunogenicity of flagellin
FlaC from Vibrio alginolyticus strain HY9901. Fish Shellfish Immunol 29, 343–8 (2010).
45. Cai, S.H. et al. Loop-mediated isothermal amplification method for rapid detection of Vibrio alginolyti-
cus, the causative agent of vibriosis in mariculture fish. Lett Appl Microbiol 50, 480–5 (2010).
46. Liu, X.F. et al. Pathogenic analysis of Vibrio alginolyticus infection in a mouse model. Folia Microbiol
(Praha) 59, 167–71 (2014).
47. Wang, Y.D. et al. Transcriptome analysis of the effect of Vibrio alginolyticus infection on the innate
immunity-related complement pathway in Epinephelus coioides. BMC Genomics 15, 1102 (2014).
48. Nakasone, N., Insisengmay, S. & Iwanaga, M. Characterization of the pili isolated from Vibrio parahae-
molyticus O3:K6. Southeast Asian J Trop Med Public Health 31, 360–5 (2000).
49. Barker W.H., Jr. & Gangarosa, E.J. Food poisoning due to Vibrio parahaemolyticus. Ann Rev Med 25,
75–81 (1974).
50. Park, K.S. et al. Functional characterization of two type III secretion systems of Vibrio parahaemolyti-
cus. Infect Immun 72, 6659–65 (2004).
424 Laboratory Models for Foodborne Infections
51. Bhoopong, P. et al. Variability of properties of Vibrio parahaemolyticus strains isolated from individual
patients. J Clin Microbiol 45, 1544–50 (2007).
52. Ludeke, C.H., Kong, N., Weimer, B.C., Fischer, M. & Jones, J.L. Complete genome sequences of a
clinical isolate and an environmental isolate of Vibrio parahaemolyticus. Genome Announc 3(2),
e00216-15 (2015).
53. Ronholm, J., Petronella, N., Kenwell, R. & Banerjee, S. Draft whole-genome sequences of 14 Vibrio
parahaemolyticus clinical isolates with an ambiguous K serogroup. Genome Announc 3(2), e00217-
15 (2015).
54. Whistler, C.A. et al. Use of whole genome phylogeny and comparisons in the development of a
multiplex-PCR assay to identify sequence type 36 Vibrio parahaemolyticus. J Clin Microbiol 53(6),
1864–72 (2015).
55. Hazen, T.H. et al. Insights into the environmental reservoir of pathogenic Vibrio parahaemolyticus
using comparative genomics. Front Microbiol 6, 204 (2015).
56. Honda, T. & Iida, T. The pathogenicity of Vibrio parahaemolyticus and the role of the thermostable
direct haemolysin and related haemolysins. Rev Med Microbiol 4, 106–13 (1993).
57. Nishibuchi, M., Fasano, A., Russell, R.G. & Kaper, J.B. Enterotoxigenicity of Vibrio parahaemolyticus
with and without genes encoding thermostable direct hemolysin. Infect Immun 60, 3539–45 (1992).
58. Shirai, H. et al. Molecular epidemiologic evidence for association of thermostable direct hemolysin
(TDH) and TDH-related hemolysin of Vibrio parahaemolyticus with gastroenteritis. Infect Immun 58,
3568–73 (1990).
59. Kishishita, M. et al. Sequence variation in the thermostable direct hemolysin-related hemolysin (trh)
gene of Vibrio parahaemolyticus. Appl Environ Microbiol 58, 2449–57 (1992).
60. Park, K.-S. et al. Genetic characterization of DNA region containing the trh and ure genes of Vibrio
parahaemolyticus. Infect Immun 68, 5742–48 (2000).
61. Suthienkul, O. et al. Urease production correlates with possession of the trh gene in Vibrio parahaemo-
lyticus strains isolated in Thailand. J Infect Dis 172, 1405–8 (1995).
62. Mobley, H.L. & Hausinger, R.P. Microbial ureases: Significance, regulation, and molecular character-
ization. Microbiol Rev 53, 85–108 (1989).
63. Mobley, H.L., Island, M.D. & Hausinger, R.P. Molecular biology of microbial ureases. Microbiol Rev
59, 451–80 (1995).
64. Park, K.-S. et al. Close proximity of the tdh, trh and ure genes on the chromosome of Vibrio parahae-
molyticus. Microbiology 144, 2517–23 (1998).
65. Hueck, C.J. Type III protein secretion systems in bacterial pathogens of animals and plants. Microbiol
Mol Biol Rev 62, 379–433 (1998).
66. Makino, K. et al. Genome sequence of Vibrio parahaemolyticus: A pathogenic mechanism distinct
from that of V. cholerae. Lancet 361, 743–9 (2003).
67. Miyamoto, Y. et al. Simplified purification and biophysicochemical characteristics of Kanagawa
phenomenon-associated hemolysin of Vibrio parahaemolyticus. Infect Immun 28, 567–76 (1980).
68. Takeda, Y. Thermostable direct hemolysin of Vibrio parahaemolyticus. Pharmacol Ther 19, 123–46
(1982).
69. Kodama, T. et al. Identification of two translocon proteins of Vibrio parahaemolyticus type III secretion
system 2. Infect Immun 76, 4282–9 (2008).
70. Park, K.S. et al. Cytotoxicity and enterotoxicity of the thermostable direct hemolysin-deletion mutants
of Vibrio parahaemolyticus. Microbiol Immunol 48, 313–8 (2004).
71. Hiyoshi, H., Kodama, T., Iida, T. & Honda, T. Contribution of Vibrio parahaemolyticus virulence
factors to cytotoxicity, enterotoxicity, and lethality in mice. Infect Immun 78, 1772–80 (2010).
72. Pineyro, P. et al. Development of two animal models to study the function of Vibrio parahaemolyticus
type III secretion systems. Infect Immun 78, 4551–9 (2010).
73. Ritchie, J.M. et al. Inflammation and disintegration of intestinal villi in an experimental model for
Vibrio parahaemolyticus-induced diarrhea. PLoS Pathog 8, e1002593 (2012).
74. Paranjpye, R.N., Myers, M.S., Yount, E.C. & Thompson, J.L. Zebrafish as a model for Vibrio parahae-
molyticus virulence. Microbiology 159, 2605–15 (2013).
75. Maupas, E. Modes et formes de reproduction des nematodes. Arch Zool Exp Gen 8, 463–624 (1901).
76. Osche, G. Systematik und phylogenie der gattung Rhabditis (Nematoda). Zool Jahrb Abt Allg Zool
Physiol Tiere 81, 190–280 (1952).
Vibrio: C. elegans as a Laboratory Model for Vibrio Infections 425
77. Lai, C.H., Chou, C.Y., Ch’ang, L.Y., Liu, C.S. & Lin, W. Identification of novel human genes evolution-
arily conserved in Caenorhabditis elegans by comparative proteomics. Genome Res 10, 703–13 (2000).
78. Brenner, S. The genetics of behaviour. Br Med Bull 29, 269–71 (1973).
79. Fares, H. & Greenwald, I. Genetic analysis of endocytosis in Caenorhabditis elegans: Coelomocyte
uptake defective mutants. Genetics 159, 133–45 (2001).
80. Barriare, A. & Felix, M.-A. Isolation of C. elegans and related nematodes. WormBook, 1–19 (2014).
81. Schulenburg, H. & Ewbank, J.J. The genetics of pathogen avoidance in Caenorhabditis elegans. Mol
Microbiol 66, 563–70 (2007).
82. Pradel, E. et al. Detection and avoidance of a natural product from the pathogenic bacterium Serratia
marcescens by Caenorhabditis elegans. Proc Natl Acad Sci USA 104, 2295–300 (2007).
83. Pujol, N. et al. A reverse genetic analysis of components of the Toll signaling pathway. Curr Biol 16,
1477 (2006).
84. Kim, D.H. et al. A conserved p38 MAP kinase pathway in Caenorhabditis elegans innate immunity. Sci
Signal 297, 623 (2002).
85. Labrousse, A., Chauvet, S., Couillault, C., Kurz, C.L. & Ewbank, J.J. Caenorhabditis elegans is a model
host for Salmonella typhimurium. Curr Biol 10, 1543–5 (2000).
86. Aballay, A., Drenkard, E., Hilbun, L.R. & Ausubel, F.M. Caenorhabditis elegans innate immune
response triggered by Salmonella enterica requires intact LPS and is mediated by a MAPK signaling
pathway. Curr Biol 13, 47–52 (2003).
87. Akira, S. Pathogen recognition by innate immunity and its signaling. Proc Jpn Acad Ser B Phys Biol Sci
85, 143–56 (2009).
88. Couillault, C. & Ewbank, J.J. Diverse bacteria are pathogens of Caenorhabditis elegans. Infect Immun
70, 4705–7 (2002).
89. Nicholas, H.R. & Hodgkin, J. The ERK MAP kinase cascade mediates tail swelling and a protective
response to rectal infection in C. elegans. Curr Biol 14, 1256–61 (2004).
90. Kim, D.H. et al. A conserved p38 MAP kinase pathway in Caenorhabditis elegans innate immunity.
Science 297, 623–6 (2002).
91. Mallo, G.V. et al. Inducible antibacterial defense system in C. elegans. Curr Biol 12, 1209–14 (2002).
92. Aballay, A. & Ausubel, F.M. Programmed cell death mediated by ced-3 and ced-4 protects
Caenorhabditis elegans from Salmonella typhimurium-mediated killing. Proc Natl Acad Sci USA 98,
2735–9 (2001).
93. Garsin, D.A. et al. Long-lived C. elegans daf-2 mutants are resistant to bacterial pathogens. Science
300, 1921 (2003).
94. Huffman, D.L. et al. Mitogen-activated protein kinase pathways defend against bacterial pore-forming
toxins. Proc Natl Acad Sci USA 101, 10995–11000 (2004).
95. Kim, D. Studying host–pathogen interactions and innate immunity in Caenorhabditis elegans. Dis
Model Mech 1, 205–8 (2008).
96. Marsh, E.K. & May, R.C. Caenorhabditis elegans, a model organism for investigating immunity. Appl
Environ Microbiol 78, 2075–81 (2012).
97. Hodgkin, J., Kuwabara, P.E. & Corneliussen, B. A novel bacterial pathogen, Microbacterium nema-
tophilum, induces morphological change in the nematode C. elegans. Curr Biol 10, 1615–8 (2000).
98. Nicholas, H.R. & Hodgkin, J. Innate immunity: The worm fights back. Curr Biol 12, R731–2 (2002).
99. Gravato-Nobre, M.J. & Hodgkin, J. Caenorhabditis elegans as a model for innate immunity to patho-
gens. Cell Microbiol 7, 741–51 (2005).
100. Palaima, E. et al. The Caenorhabditis elegans bus-2 mutant reveals a new class of O-glycans affecting
bacterial resistance. J Biol Chem 285, 17662–72 (2010).
101. O’Rourke, D., Baban, D., Demidova, M., Mott, R. & Hodgkin, J. Genomic clusters, putative pathogen
recognition molecules, and antimicrobial genes are induced by infection of C. elegans with M. nema-
tophilum. Genome Res 16, 1005–16 (2006).
102. Jansson, H.B. Adhesion of conidia of Drechmeria coniospora to Caenorhabditis elegans wild type and
mutants. J Nematol 26, 430 (1994).
103. Couillault, C. et al. TLR-independent control of innate immunity in Caenorhabditis elegans by the TIR
domain adaptor protein TIR-1, an ortholog of human SARM. Nat Immunol 5, 488–94 (2004).
104. Troemel, E.R. et al. p38 MAPK regulates expression of immune response genes and contributes to
longevity in C. elegans. PLoS Genet 2, e183 (2006).
426 Laboratory Models for Foodborne Infections
105. Félix, M.A. et al. Natural and experimental infection of Caenorhabditis nematodes by novel viruses
related to nodaviruses. PLoS Biol 9, e1000586 (2011).
106. Félix, M.A. & Braendle, C. The natural history of Caenorhabditis elegans. Curr Biol 20, R965–9 (2010).
107. Joelsson, A., Liu, Z. & Zhu, J. Genetic and phenotypic diversity of quorum-sensing systems in clinical
and environmental isolates of Vibrio cholerae. Infect Immun 74, 1141–7 (2006).
108. Sahu, S.N. et al. Genomic analysis of immune response against Vibrio cholerae hemolysin in
Caenorhabditis elegans. PLoS One 7, e38200 (2012).
109. Durai, S., Pandian, S.K. & Balamurugan, K. Establishment of a Caenorhabditis elegans infection
model for Vibrio alginolyticus. J Basic Microbiol 51, 243–52 (2011).
110. Dhakal, B.K. et al. Caenorhabditis elegans as a simple model host for Vibrio vulnificus infection.
Biochem Biophys Res Commun 346, 751–7 (2006).
111. Kurz, C.L. et al. Virulence factors of the human opportunistic pathogen Serratia marcescens identified
by in vivo screening. EMBO J 22, 1451–60 (2003).
112. Durai, S., Singh, N., Kundu, S. & Balamurugan, K. Proteomic investigation of Vibrio alginolyticus
challenged Caenorhabditis elegans revealed regulation of cellular homeostasis proteins and their role
in supporting innate immune system. Proteomics 14, 1820–32 (2014).
113. Durai, S., Karutha Pandian, S. & Balamurugan, K. Changes in Caenorhabditis elegans exposed to
Vibrio parahaemolyticus. J Microbiol Biotechnol 21, 1026–35 (2011).
114. Sifri, C.D., Begun, J., Ausubel, F.M. & Calderwood, S.B. Caenorhabditis elegans as a model host for
Staphylococcus aureus pathogenesis. Infect Immun 71, 2208–17 (2003).
115. Ansaruzzaman, M. et al. Pandemic serovars (O3:K6 and O4:K68) of Vibrio parahaemolyticus associ-
ated with diarrhea in Mozambique: Spread of the pandemic into the African continent. J Clin Microbiol
43, 2559–62 (2005).
116. Matsumoto, C. et al. Pandemic spread of an O3:K6 clone of Vibrio parahaemolyticus and emergence of
related strains evidenced by arbitrarily primed PCR and toxRS sequence analyses. J Clin Microbiol 38,
578–85 (2000).
117. Nair, G.B. & Hormazabal, J.C. The Vibrio parahaemolyticus pandemic. Rev Chilena Infectol 22,
125–30 (2005).
118. Okuda, J. et al. Emergence of a unique O3:K6 clone of Vibrio parahaemolyticus in Calcutta, India, and
isolation of strains from the same clonal group from Southeast Asian travelers arriving in Japan. J Clin
Microbiol 35, 3150–5 (1997).
119. Lynch, T. et al. Vibrio parahaemolyticus disruption of epithelial cell tight junctions occurs indepen-
dently of toxin production. Infect Immun 73, 1275–83 (2005).
120. Schulenburg, H., Hoeppner, M.P., Weiner, J., III & Bornberg-Bauer, E. Specificity of the innate immune
system and diversity of C-type lectin domain (CTLD) proteins in the nematode Caenorhabditis elegans.
Immunobiology 213, 237–50 (2008).
121. Wong, D., Bazopoulou, D., Pujol, N., Tavernarakis, N. & Ewbank, J.J. Genome-wide investigation
reveals pathogen-specific and shared signatures in the response of Caenorhabditis elegans to infection.
Genome Biol 8, R194 (2007).
29
Yersinia
Xin Wang, Ran Duan, Junrong Liang, Wenpeng Gu, and Huaiqi Jing
CONTENTS
29.1 Mouse Model................................................................................................................................. 428
29.1.1 Lifestyle and Interaction with Host.................................................................................. 428
29.1.2 Virulence of Pathogen...................................................................................................... 428
29.2 Pig Model...................................................................................................................................... 430
29.3 Caenorhabditis elegans Model......................................................................................................431
29.4 Rabbit Model..................................................................................................................................431
29.5 Cell Models....................................................................................................................................431
29.5.1 Adhesion and Invasion of Mammalian Cells....................................................................432
29.5.2 Modulation of Host Immune Defense Mechanisms..........................................................432
29.6 Conclusion......................................................................................................................................433
References................................................................................................................................................433
427
428 Laboratory Models for Foodborne Infections
highly pathogenic biotype 1B strains.14 Many studies have reported the role of biotype 1A strains in
foodborne outbreaks, in which the pYV plasmid is absent.15
29.1 Mouse Model
29.1.1 Lifestyle and Interaction with Host
After orogastric inoculation of mice, Y. enterocolitica selectively invades the Peyer’s patches (PP) via M
cells.16–18 This invasion leads to recruitment of PMNs, formation of microabscesses comprising extra-
cellular Yersinia, and, finally, complete destruction of the cytoarchitecture of the PP. Subsequently yer-
siniae disseminate to lymph nodes, spleen, and liver where they form microcolonies/microabscesses,
suggesting that Y. enterocolitica disseminates via the lymphatic vessels.16 Similar to in vivo observa-
tions, Yersinia manifests in vitro resistance to phagocytosis by macrophages19,20 and by PMNs.21–24 Once
phagocytosed, Y. pseudotuberculosis and Y. enterocolitica generally do not survive long. These findings
suggest that Y. enterocolitica is an extracellular pathogen and that its survival strategy is based on avoid-
ing the nonspecific immune response.
Recent studies showed that in the oral mouse infection model using RFP- (red fluorescent protein) and
GFP- (green fluorescent protein) expressing yersiniae, the oral Y. enterocolitica infection of mice leads
to monoclonal microabscess formation in PP, spleen, and liver. Furthermore, experiments with red and
green fluorescing yersiniae revealed that only very few yersiniae were able to invade PP from the gut
lumen and that both Yersinia and the host contribute to this phenomenon. In contrast to other enteric
pathogens such as Salmonella and Shigella, Yersinia is predominantly an extracellular pathogen.18,25,26
Initially, innate host defenses such as PMNs, macrophages, and natural killer (NK) cells are involved
in controlling Yersinia infection. But subsequently, a robust adaptive immune response is required to
overcome Yersinia infection. Specific Abs27,28 as well as IFN-γ-producing CD4 and CD8 T cells play
an essential role in clearing Yersinia infection and have been shown to mediate protection in adoptive
transfer experiments.29
Wang et al. investigated the lethality, cytokine alterations, and histopathological changes of patho-
genic Y. enterocolitica bioserotypes 1B/O:8 and 2/O:9 in susceptible BALB/c and resistant C57BL/6
mice. They found that the 50% lethal dose (LD50) for the pathogenic Y. enterocolitica bioserotype 1B/O:8
was 103 CFU in both BALB/c and C57BL/6 mice; while the LD50 for the bioserotype 2/O:9 was 108
CFU in BALB/c mice and 109 CFU in C57BL/6 mice. In BALB/c mice, the bioserotype 2/O:9 induced
a higher level of GM-CSF, IL-6, and TNF-α than the bioserotype 1B/O:8, but the status was reversed
subsequently. Levels of IFN-γ, IL-1β, IL-2, IL-4, IL-5, IL-10, and IL-12 following infection with the
bioserotype 1B/O:8 were always higher than those with the bioserotype 2/O:9 (Figure 29.1). The histo-
pathological changes in the liver and spleen in BALB/c mice infected with the two strains were similar at
different times and doses. These observations indicate the different immunological effects and changes
in the mouse model due to pathogenic Y. enterocolitica 1B/O:8 and 2/O:9 infections.30
29.1.2 Virulence of Pathogen
In an oral mouse model, a phospholipase-negative strain was constructed to ascertain whether phospho-
lipase A plays a role in pathogenesis. At a dose of 108 CFU/mouse, fewer mutant strains were recovered
and less flamed tissue was found from the mesenteric lymph nodes and PP. When given extremely high
dose (109 CFU/mouse), the numbers of foci and the extent of inflammation were noticeably less as well,
confirming the role of phospholipase A in pathogenesis.31
The T3SS is a complex system used to deliver bacterial proteins into the cytosol of host cells.32 The
proteins secreted by T3SSs are known as effectors working together to establish an extracellular infec-
tion that disrupts the actin cytoskeleton and deregulates signal pathways.33
YopM was originally described as Yop48 in Y. enterocolitica.34 According to the LD50 test,
the yopM mutant has a strongly reduced virulence in mice. Bacterial counts in liver and spleen showed the
yopM mutant had a reduced ability to multiply in the host.35 When three challenge routes were done for YopE
mutant, it was avirulent after oral or intraperitoneal infection but virulent after intravenous injection.36
Yersinia 429
80 3000
1000
70 900
Cytokine GM-CSF(pg/ml)
2500
Cytokine IFN-γ(pg/ml)
800
Cytokine IL-1β(pg/ml)
60
700 2000
50 high O:8 high O:8 high O:8
600
high O:9 high O:9 high O:9
40 500 1500
medium O:8 medium O:8 medium O:8
30 medium O:9 400 medium O:9 medium O:9
1000 low O:8
low O:8 300 low O:8
20 low O:9
low O:9 200 low O:9
500
10 100
0 0 0
3 6 9 12 15 24 48 72 h 3 6 9 12 15 24 48 72 h 3 6 9 12 15 24 48 72 h
200
180 1000 1800
Cytokine IFN-4(pg/ml)
900
160 1600
Cytokine IL-2(pg/ml)
Cytokine IL-5(pg/ml)
800
140 high O:8 high O:8 1400 high O:8
high O:9 700 high O:9 high O:9
120 1200
medium O:8 600 medium O:8 medium O:8
100 medium O:9 1000
medium O:9 500 medium O:9
80 low O:8 400 low O:8 800 low O:8
60 low O:9 low O:9 600 low O:9
300
40 200 400
20 100 200
0 0 0
3 6 9 12 15 24 48 72 h 3 6 9 12 15 24 48 72 h 3 6 9 12 15 24 48 72 h
120000
(D) (E) (F)
3000
250
100000 2500
Cytokine IL-10(pg/ml)
Cytokine IL-12(pg/ml)
Cytokine IL-6(pg/ml)
200
80000 high O:8 2000 high O:8 high O:8
high O:9 high O:9 high O:9
150
60000 medium O:8 1500 medium O:8 medium O:8
medium O:9 medium O:9 medium O:9
40000 100 low O:8
low O:8 1000 low O:8
low O:9 low O:9 low O:9
20000 500 50
0 0 0
3 6 9 12 15 24 48 72 h 3 6 9 12 15 24 48 72 h 3 6 9 12 15 24 48 72 h
–500 –50
700
(G) (H) (I)
600
Cytokine TNF-α(pg/ml)
high O:8
500
high O:9
400 medium O:8
medium O:9
300 low O:8
low O:9
200
100
0
3 6 9 12 15 24 48 72 h
–100
(J)
FIGURE 29.1 Comparison of cytokines between two strains in BALB/C mice. (A) GM-CSF, (B) IFN-γ, (C) IL-1β,
(D) IL-2, (E) IL-4, (F) IL-5, (G) IL-6, (H) IL-10, (I) IL-12, and (J) TNF-α. *, The cytokine values were statistically signifi-
cant comparing the two strains in different groups (p < 0.05). ▲, All of the cytokine values were statistically significant
comparing the two strains in every group (p < 0.05). This figure demonstrates the comparison for each cytokine changes
between two strains. (From Wang, X. et al., Mol. Immunol., 55, 365–371, 2013. With permission from Elsevier.)
Compared to extensive study of YopP effects in vitro, the role of YopP in vivo is inadequately inves-
tigated. Monack et al. have shown that Y. pseudotuberculosis induces apoptosis of Mac-1 cells in mice,
which is probably responsible for the attenuated phenotype of a yopJ mutant.37 In 2015, a study using
Listeria model shows for the first time that YopP inhibits the development of an effective CD8 T cell
response in mice.38 In vitro, this is due to the rapid induction of dendritic cell (DC) apoptosis and matura-
tion,39 whereas in vivo a possible mechanism for this could be the inhibition of Ag presentation by DC.38
By using mouse model infected with Y. pseudotuberculosis, Rosqvist et al. showed that a strain unable to
express YopH has a reduced ability to resist phagocytosis,20 while the ability to resist phagocytosis could be
complemented by introduction of a plasmid carrying only the yopH gene, demonstrating that YopH is indeed
involved in the antiphagocytic effect. However, mutation of yopH did not completely abolish the resistance to
phagocytosis, suggesting another virulence factor was involved in this phenomenon.
A systematic study of Yops pathogenicity of the highly virulent mouse O:8 strain administered by oral
and intravenous routes in mice showed the yopH, yopO, yopP, yopE, yopM, and yopQ mutants had only
modest defects in colonization of the small intestine (SI) and PP,40 consistent with that of Y. pseudotuber-
culosis,41 indicating that no single effector is absolutely necessary for the colonizing. YopH, YopQ, and
YopM are important for inducing systemic disease, whereas YopO, YopP, and YopE are dispensable to
reach the spleen and liver. On the other hand, YopT seems to slightly decrease the virulence in both oral
and intravenous model.40
430 Laboratory Models for Foodborne Infections
With the help of the mouse model, the yersinia T3SS has been studied in detail using molecular, cellular,
genetic, and biochemical techniques, yielding insight into the efficiency and sophistication of the complex.
Progression of Y. enterocolitica infection in mouse model closely mirrors that in the human. In natural
infection, Y. enterocolitica enters by the oral route.42 Ruiz-Bravo et al. conducted a comparative experiment
with three challenge routes (oral, intraperitoneal, and intravenous) using moderate–virulent O:9 strains with
or without pYV and noted considerable differences between oral and intraperitoneal infections by the two
strains.43 This suggests the virulence potential of Y. enterocolitica may be influenced by the route of infec-
tion. Therefore, at the beginning of an experiment, the route of infection should be chosen prudently.
29.2 Pig Model
In swine, fecal shedding may stop soon after the ingestion of Y. enterocolitica, while tonsils may carry
the bacterium from hours to months,44 indicating that tonsils represent a more reliable tissue for the
indication of an infection/colonization by Y. enterocolitica. As is known, clinical response to Y. entero-
colitica infection in piglets is related to inoculum size.45 A dose of 4 × 1010 CFU could cause death46;
in contrast, a dose of 3 × 108 CFU would provoke a subclinical response as near as possible to a natural
infection.44 The early infection showed a transient bacteremia within the first 3 h postinfection.45 So if
pigs are infected within hours prior to slaughter, carcass of the animal might not be completely cleared
of the microorganism and thus result in widespread contamination.44
The high prevalence of O:3 strains in fattening pigs indicates serotype- and host-specific colonization
strategies. Comparison of O:3 patient isolate and O:8 8081v strain in minipigs and mice models showed
that small genetic variations contribute to profound changes of the virulence gene expression pattern.47 In
the O:3 strain, IS1667 insertion (an additional promoter) as well as the more stable variant of RovA (invA
transcriptional activator protein) toward Lon protease resulted in high expression of invasion at 37°C,
indicating a fine adjustment of pathogenicity to pig, with a higher body temperature (Figure 29.2).47 They
25°C 37°C
InvA InvA
InvA
RovA RovA
RovA*
RovA*
** ** **
O:3 Transposase invA
PTn PinvA
IS1667
FIGURE 29.2 Different expression of the invasin gene invA in Y. enterocolitica O:3 and O:8. Whole-cell extracts prepared
from overnight cultures of Y. enterocolitica O:3 and O:8 grown at 25°C and 37°C were separated on SDS-polyacrylamide
gels and analyzed by western blotting using polyclonal antibodies directed against InvA. Overview of the invA promoter
region of Y. enterocolitica O:8 (upper panel) and O:3 (lower panel). The transcriptional start site of the invA and the IS1667-
specific promoter are indicated by broken arrows. The blue boxes represent the RovA binding sites. The light blue box
illustrates the RovA binding site in the IS1667 sequence. The RovA dimer is given in red; the stars illustrate the amino acid
substitution that renders the RovA protein less susceptible to the Lon protease. (From Valentin-Weigand, P., et al., Int. J.
Med. Microbiol, 304, 824–834, 2014. With permission from Elsevier.)
Yersinia 431
further uncovered why the cell adhesion and invasion properties were restricted to pig-associated O:3
strains. Coinfection studies with O:3 wild and mutant strains demonstrated that small variations within
the O:3 genome improved colonization/survival in swine but had only a minor effect on the colonization
in mice. In addition, a deletion of the invA abolished long-term colonization in the pigs.48
As the most important reservoir and asymptomatic carriers of Y. enterocolitica prevalent serotype,49,50
the pig (model) is useful for studies ranging from colonization tropisms to research on the virulence
properties of O:3 strains.
29.4 Rabbit Model
Rabbits have been a suitable and reproducible animal model to examine the course of intestinal Yersinia
infection.58–62 In mouse model, diarrhea is not a major symptom.63,64 But the infection of the young rabbit
resembles that in children: the oral inoculation induces both a clear diarrhea and a systemic invasion.59,60,65–67
Thus, Delor and Cornells constructed one for establishment of the virulent role of Yst toxin.68
The pathogenic and immunogenic potential of parental and mutant Y. enterocolitica strains have been
successfully evaluated in oral rabbit model, showing O-antigen is required for full virulence. The strain
that lacks Wzz protein (the O-antigen chain length determinant) is more attenuated than one that lacks
Wzy protein (expressing only one repeat unit of lipopolyasccharide [LPS]).69
On the basis of sodA finding in mouse model,70 Najdenski et al.71 established a rabbit model to show
that the sodA mutant strain not only attenuated in dissemination into various organs but also showed
an improved antibody response. In contrast to previous LPS mutant strains, this mutant was noticeably
more sensitive to leukocytes. Besides, they first observed yersiniae dissemination to brain in experimen-
tal infection. More surprising were the pathomorphological changes not established in tonsils with the
virulent strain but with sodA mutant strain. Together, these showed the potential of sodA mutant to be a
candidate for immunization.
Overall, the susceptibility of rabbits to Y. enterocolitica appeared to be more variable in this study
focused on diarrhea manifestation and immunomorphology.
29.5 Cell Models
In cell models, innate host defenses such as PMNs, macrophages, and NK cells are involved in con-
trolling Yersinia infection initially, but a robust adaptive immune response is required to overcome
Yersinia infection subsequently. Specific Abs27,28 as well as IFN-γ-producing CD4 and CD8 T cells play
432 Laboratory Models for Foodborne Infections
an essential role in clearing Yersinia infection and have been shown to mediate protection in adoptive
transfer experiments.29 Some of the Yops (including YopE, YopH, YopO, and YopM) are delivered into
the host cell cytosol where they damage the cytoskeleton and disrupt the signaling network. Infected
macrophages displayed general features of apoptosis such as membrane blebbing (apoptotic body forma-
tion) and nuclear and cellular shrinkage.
29.6 Conclusion
Y. enterocolitica and Y. pseudotuberculosis are foodborne bacteria associated with gastrointestinal
syndromes (from acute enteritis to mesenteric lymphadenitis), or mesenteric adenitis, and septice-
mia. These pathogens generate a number of proteins that help evade host innate immune response and
replicate inside the host. Use of mouse models revealed the interaction between the host and the extracel-
lular lifestyle of the pathogen. Application of molecular, cellular, genetic, and biochemical techniques
in combination with in vitro models yielded valuable insights into efficiency and sophistication of the
Yersinia T3SS. Comparative studies employing minipig and mouse models uncovered that O:3 strains
utilize serotype- and host-specific colonization strategies in fattening pigs. While C. elegans infection
model is not feasible for pathogenicity study due to inappropriate temperature, rabbit model is suscep-
tible to diarrhea manifestation. Cell models are useful for investigating different stages of yersinia infec-
tion: adhesion and invasion, and evasion of host innate and adaptive immune mechanisms.
REFERENCES
1. Bottone, E.J. Yersinia enterocolitica: The charisma continues. Clin Microbiol Rev 10, 257–76 (1997).
2. Bleves, S. & Cornelis, G.R. How to survive in the host: The Yersinia lesson. Microbes Infect 2,
1451–60 (2000).
3. Spinner, J.L. et al. Neutrophils are resistant to Yersinia YopJ/P-induced apoptosis and are protected
from ROS-mediated cell death by the type III secretion system. PLoS One 5, e9279 (2010).
4. Cover, T.L. & Aber, R.C. Yersinia enterocolitica. N Engl J Med 321, 16–24 (1989).
5. Fukushima, H. & Gomyoda, M. Intestinal carriage of Yersinia pseudotuberculosis by wild birds and
mammals in Japan. Appl Environ Microbiol 57, 1152–5 (1991).
6. Liang, J. et al. Ecology and geographic distribution of Yersinia enterocolitica among livestock and
wildlife in China. Vet Microbiol 178, 125–31 (2015).
7. Wang, X. et al. Canis lupus familiaris involved in the transmission of pathogenic Yersinia spp. in China.
Vet Microbiol 172, 339–44 (2014).
8. Liang, J. et al. Prevalence of Yersinia enterocolitica in pigs slaughtered in Chinese abattoirs. Appl
Environ Microbiol 78, 2949–56 (2012).
9. Wang, X. et al. Distribution of pathogenic Yersinia enterocolitica in China. Eur J Clin Microbiol Infect
Dis 28, 1237–44 (2009).
10. Perry, R.D. & Fetherston, J.D. Yersinia pestis—Etiologic agent of plague. Clin Microbiol Rev 10,
35–66 (1997).
11. Cornelis, G.R. et al. The virulence plasmid of Yersinia, an antihost genome. Microbiol Mol Biol Rev 62,
1315–52 (1998).
12. Cornelis, G.R. Yersinia type III secretion: Send in the effectors. J Cell Biol 158, 401–8 (2002).
13. Cornelis, G.R. Molecular and cell biology aspects of plague. Proc Natl Acad Sci USA 97, 8778–83 (2000).
14. Dhar, M.S. & Virdi, J.S. Strategies used by Yersinia enterocolitica to evade killing by the host: Thinking
beyond Yops. Microbes Infect 16, 87–95 (2014).
15. Bhagat, N. & Virdi, J.S. The enigma of Yersinia enterocolitica biovar 1A. Crit Rev Microbiol 37,
25–39 (2011).
434 Laboratory Models for Foodborne Infections
16. Autenrieth, I.B. & Firsching, R. Penetration of M cells and destruction of Peyer’s patches by Yersinia
enterocolitica: An ultrastructural and histological study. J Med Microbiol 44, 285–94 (1996).
17. Grutzkau, A., Hanski, C., Hahn, H. & Riecken, E.O. Involvement of M cells in the bacterial invasion
of Peyer’s patches: A common mechanism shared by Yersinia enterocolitica and other enteroinvasive
bacteria. Gut 31, 1011–5 (1990).
18. Hanski, C. et al. Immunohistochemical and electron microscopic study of interaction of Yersinia
enterocolitica serotype O8 with intestinal mucosa during experimental enteritis. Infect Immun 57,
673–8 (1989).
19. Fallman, M. et al. Yersinia pseudotuberculosis inhibits Fc receptor-mediated phagocytosis in J774 cells.
Infect Immun 63, 3117–24 (1995).
20. Rosqvist, R., Bolin, I. & Wolf-Watz, H. Inhibition of phagocytosis in Yersinia pseudotuberculosis:
A virulence plasmid-encoded ability involving the Yop2b protein. Infect Immun 56, 2139–43 (1988).
21. Bacon, G.A. & Burrows, T.W. The basis of virulence in Pasteurella pestis: An antigen determining
virulence. Br J Exp Pathol 37, 481–93 (1956).
22. China, B., N’Guyen, B.T., de Bruyere, M. & Cornelis, G.R. Role of YadA in resistance of Yersinia
enterocolitica to phagocytosis by human polymorphonuclear leukocytes. Infect Immun 62, 1275–81
(1994).
23. Ruckdeschel, K., Roggenkamp, A., Schubert, S. & Heesemann, J. Differential contribution of Yersinia
enterocolitica virulence factors to evasion of microbicidal action of neutrophils. Infect Immun 64, 724–
33 (1996).
24. Visser, L.G., Annema, A. & van Furth, R. Role of Yops in inhibition of phagocytosis and killing of
opsonized Yersinia enterocolitica by human granulocytes. Infect Immun 63, 2570–5 (1995).
25. Monack, D.M., Mecsas, J., Ghori, N. & Falkow, S. Yersinia signals macrophages to undergo apoptosis
and YopJ is necessary for this cell death. Proc Natl Acad Sci USA 94, 10385–90 (1997).
26. Lian, C.J., Hwang, W.S. & Pai, C.H. Plasmid-mediated resistance to phagocytosis in Yersinia enteroco-
litica. Infect Immun 55, 1176–83 (1987).
27. Vogel, U., Autenrieth, I.B., Berner, R. & Heesemann, J. Role of plasmid-encoded antigens of Yersinia
enterocolitica in humoral immunity against secondary Y. enterocolitica infection in mice. Microb
Pathog 15, 23–36 (1993).
28. Igwe, E.I. et al. Rational live oral carrier vaccine design by mutating virulence-associated genes of
Yersinia enterocolitica. Infect Immun 67, 5500–7 (1999).
29. Trulzsch, K. et al. Yersinia outer protein P inhibits CD8 T cell priming in the mouse infection model.
J Immunol 174, 4244–51 (2005).
30. Wang, X. et al. Comparison of the cytokine immune response to pathogenic Yersinia enterocolitica
bioserotype 1B/O:8 and 2/O:9 in susceptible BALB/C and resistant C57BL/6 mice. Mol Immunol
55(3–4), 365–71 (2013).
31. Schmiel, D., Wagar, E., Karamanou, L., Weeks, D. & Miller, V. Phospholipase A of Yersinia enteroco-
litica contributes to pathogenesis in a mouse model. Infect Immun 66, 3941–51 (1998).
32. Ghosh, P. Process of protein transport by the type III secretion system. Microbiol Mol Biol Rev 68,
771–95 (2004).
33. Navarro, L., Alto, N.M. & Dixon, J.E. Functions of the Yersinia effector proteins in inhibiting host
immune responses. Curr Opin Microbiol 8, 21–7 (2005).
34. Cornelis, G., Vanootegem, J.-C. & Sluiters, C. Transcription of the yop regulon from Y. enterocolitica
requires trans acting pYV and chromosomal genes. Microb Pathog 2, 367–79 (1987).
35. Mulder, B., Michiels, T., Simonet, M., Sory, M.-P. & Cornelis, G. Identification of additional virulence
determinants on the pYV plasmid of Yersinia enterocolitica W227. Infect Immun 57, 2534–41 (1989).
36. Rosqvist, R., Forsberg, Å., Rimpiläinen, M., Bergman, T. & Wolf‐Watz, H. The cytotoxic protein YopE
of Yersinia obstructs the primary host defence. Mol Microbiol 4, 657–67 (1990).
37. Monack, D.M., Mecsas, J., Bouley, D. & Falkow, S. Yersinia-induced apoptosis in vivo aids in the estab-
lishment of a systemic infection of mice. J Exp Med 188, 2127–37 (1998).
38. Trülzsch, K. et al. Yersinia outer protein P inhibits CD8 T cell priming in the mouse infection model.
J Immunol 174, 4244–51 (2005).
39. Ruckdeschel, K., Richter, K., Mannel, O. & Heesemann, J. Arginine-143 of Yersinia enterocolitica
YopP crucially determines isotype-related NF-κB suppression and apoptosis induction in macrophages.
Infect Immun 69, 7652–62 (2001).
Yersinia 435
40. Trülzsch, K., Sporleder, T., Igwe, E.I., Rüssmann, H. & Heesemann, J. Contribution of the major secreted
yops of Yersinia enterocolitica O:8 to pathogenicity in the mouse infection model. Infect Immun 72,
5227–34 (2004).
41. Logsdon, L.K. & Mecsas, J. Requirement of the Yersinia pseudotuberculosis effectors YopH and YopE
in colonization and persistence in intestinal and lymph tissues. Infect Immun 71, 4595–607 (2003).
42. Ruiz-Bravo, A., Jimenez-Valera, M., Sampedro, A. & Moreno, E. Chromosome-mediated resistance of
Yersinia enterocolitica serotype O9 to intracellular killing by mouse peritoneal macrophages. FEMS
Immunol Med Microbiol 9, 317–24 (1994).
43. Ruiz-Bravo, A., Moreno, E., Sampedro, A. & Jimenez-Valera, M. Experimental infection of mice with
Yersinia enterocolitica serotype O9 by oral and parenteral routes: spreading and enterotropism of viru-
lent yersiniae. Curr Microbiol 38, 257–63 (1999).
44. Thibodeau, V., Frost, E.H., Chenier, S. & Quessy, S. Presence of Yersinia enterocolitica in tissues of
orally-inoculated pigs and the tonsils and feces of pigs at slaughter. Can J Vet Res 63, 96–100 (1999).
45. Shu, D. et al. Experimental infection of newborn piglets with Yersinia enterocolitica: An animal model
of enteritis. N Z Vet J 43, 50–6 (1995).
46. Robins-Browne, R. et al. The pathogenesis of Yersinia enterocolitica infection in gnotobiotic piglets.
J Med Microbiol 19, 297–308 (1985).
47. Valentin-Weigand, P., Heesemann, J. & Dersch, P. Unique virulence properties of Yersinia enteroco-
litica O:3—An emerging zoonotic pathogen using pigs as preferred reservoir host. Int J Med Microbiol
304, 824–34 (2014).
48. Schaake, J. et al. Essential role of invasin for colonization and persistence of Yersinia enterocolitica in
its natural reservoir host, the pig. Infect Immun 82, 960–9 (2014).
49. Bottone, E.J. Yersinia enterocolitica: Overview and epidemiologic correlates. Microbes Infect 1,
323–33 (1999).
50. Fredriksson-Ahomaa, M. Isolation of enteropathogenic Yersinia from non-human sources. in Advances
in Yersinia Research 97–105 (Springer, New York, 2012).
51. Waterfield, N.R. & Wren, B.W. Invertebrates as a source of emerging human pathogens. Nat Rev
Microbiol 2, 833–41 (2004).
52. Spanier, B., Starke, M., Higel, F., Scherer, S. & Fuchs, T.M. Yersinia enterocolitica infection and tcaA-
dependent killing of Caenorhabditis elegans. Appl Environ Microbiol 76, 6277–85 (2010).
53. Darby, C., Hsu, J.W., Ghori, N. & Falkow, S. Caenorhabditis elegans: Plague bacteria biofilm blocks
food intake. Nature 417, 243–4 (2002).
54. Marceau, M. Transcriptional regulation in Yersinia: An update. Curr Issues Mol Biol 7, 151–78 (2005).
55. Straley, S.C. & Perry, R.D. Environmental modulation of gene expression and pathogenesis in Yersinia.
Trends Microbiol 3, 310–7 (1995).
56. Bresolin, G., Neuhaus, K., Scherer, S. & Fuchs, T.M. Transcriptional analysis of long-term adaptation of
Yersinia enterocolitica to low-temperature growth. J Bacteriol 188, 2945–58 (2006).
57. Heermann, R. & Fuchs, T.M. Comparative analysis of the Photorhabdus luminescens and the
Yersinia enterocolitica genomes: Uncovering candidate genes involved in insect pathogenicity. BMC
Genomics 9, 40 (2008).
58. Une, T. Studies on the pathogenicity of Yersinia enterocolitica. I. Experimental infection in rabbits.
Microbiol Immunol 21, 341–63 (1977).
59. Pai, C.H., Mors, V. & Seemayer, T.A. Experimental Yersinia enterocolitica enteritis in rabbits. Infect
Immun 28, 238–44 (1980).
60. Heesemann, J., Schröder, J. & Ulrich, M. Analysis of the class-specific immune response to Yersinia
enterocolitica virulence-associated antigens in oro-gastrically infected rabbits. Microb Pathog 5,
437–47 (1988).
61. Heesemann, J., Gaede, K. & Autenrieth, I. Experimental Yersinia enterocolitica infection in rodents:
A model for human yersiniosis. APMIS 101, 417–29 (1993).
62. Vesselinova, A., Najdenski, H., Nikolova, S. & Wesselinova, D. Arthritis after experimental infection
with Yersinia enterocolitica O:3 in rabbits. J Vet Med Ser B 48, 43–53 (2001).
63. Laird, W. & Cavanaugh, D. Correlation of autoagglutination and virulence of yersiniae. J Clin Microbiol
11, 430 (1980).
64. Schiemann, D. An enterotoxin-negative strain of Yersinia enterocolitica serotype O:3 is capable of
producing diarrhea in mice. Infect Immun 32, 571–4 (1981).
436 Laboratory Models for Foodborne Infections
65. Lian, C.-J., Hwang, W., Kelly, J. & Pai, C. Invasiveness of Yersinia enterocolitica lacking the virulence
plasmid: An in-vivo study. J Med Microbiol 24, 219–26 (1987).
66. O’loughlin, E. et al. Clinical, morphological, and biochemical alterations in acute intestinal yersiniosis.
Pediatr Res 20, 602–8 (1986).
67. O’Loughlin, E., Pai, C. & Gall, D. Effect of acute Yersinia enterocolitica infection on in vivo and in
vitro small intestinal solute and fluid absorption in the rabbit. Gastroenterology 94, 664–72 (1988).
68. Delor, I. & Cornelis, G.R. Role of Yersinia enterocolitica Yst toxin in experimental infection of young
rabbits. Infect Immun 60, 4269–77 (1992).
69. Najdenski, H., Golkocheva, E., Vesselinova, A., Bengoechea, J. & Skurnik, M. Proper expression of the
O-antigen of lipopolysaccharide is essential for the virulence of Yersinia enterocolitica O:8 in experi-
mental oral infection of rabbits. FEMS Immunol Med Microbiol 38, 97–106 (2003).
70. Roggenkamp, A., Bittner, T., Leitritz, L., Sing, A. & Heesemann, J. Contribution of the Mn-cofactored
superoxide dismutase (SodA) to the virulence of Yersinia enterocolitica serotype O8. Infect Immun 65,
4705–10 (1997).
71. Najdenski, H.M., Golkocheva, E.N., Vesselinova, A.M. & Rüssmann, H. Comparison of the course of
infection of virulent Yersinia enterocolitica serotype O:8 with an isogenic sodA mutant in the peroral
rabbit model. Int J Med Microbiol 294, 383–93 (2004).
72. Miller, V.L. & Falkow, S. Evidence for two genetic loci in Yersinia enterocolitica that can promote
invasion of epithelial cells. Infect Immun 56, 1242–8 (1988).
73. Pepe, J.C. & Miller, V.L. The Yersinia enterocolitica inv gene product is an outer membrane protein that
shares epitopes with Yersinia pseudotuberculosis invasin. J Bacteriol 172, 3780–9 (1990).
74. Pierson, D.E. & Falkow, S. The ail gene of Yersinia enterocolitica has a role in the ability of the
organism to survive serum killing. Infect Immun 61, 1846–52 (1993).
75. Goverde, R.L., Jansen, W.H., Brunings, H.A., Huis in ’t Veld, J.H. & Mooi, F.R. Digoxigenin-labelled
inv- and ail-probes for the detection and identification of pathogenic Yersinia enterocolitica in clinical
specimens and naturally contaminated pig samples. J Appl Bacteriol 74, 301–13 (1993).
76. Leo, J.C. & Skurnik, M. Adhesins of human pathogens from the genus Yersinia. Adv Exp Med Biol 715,
1–15 (2011).
77. Paczosa, M.K., Fisher, M.L., Maldonado-Arocho, F.J. & Mecsas, J. Yersinia pseudotuberculosis uses
Ail and YadA to circumvent neutrophils by directing Yop translocation during lung infection. Cell
Microbiol 16, 247–68 (2014).
78. Liang, J. et al. Two novel ail-positive biotype 1A strains of Yersinia enterocolitica isolated in China with
unequal adhesion and invasion properties. Infect Genet Evol 27, 83–8 (2014).
79. Sulakvelidze, A. Yersiniae other than Y. enterocolitica, Y. pseudotuberculosis, and Y. pestis: The
ignored species. Microbes Infect 2, 497–513 (2000).
80. Peters, K.N. & Anderson, D.M. Modulation of host cell death pathways by Yersinia species and the type
III effector YopK. Adv Exp Med Biol 954, 229–36 (2012).
81. Heesemann, J., Sing, A. & Trulzsch, K. Yersinia’s stratagem: Targeting innate and adaptive immune
defense. Curr Opin Microbiol 9, 55–61 (2006).
82. Gebus, C., Faudry, E., Bohn, Y.S., Elsen, S. & Attree, I. Oligomerization of PcrV and LcrV, protective
antigens of Pseudomonas aeruginosa and Yersinia pestis. J Biol Chem 283, 23940–9 (2008).
83. Sodhi, A., Sharma, R.K., Batra, H.V. & Tuteja, U. Recombinant fraction 1 protein of Yersinia pestis
activates murine peritoneal macrophages in vitro. Cell Immunol 229, 52–61 (2004).
84. Orth, K. et al. Inhibition of the mitogen-activated protein kinase kinase superfamily by a Yersinia
effector. Science 285, 1920–3 (1999).
85. Ruckdeschel, K., Richter, K., Mannel, O. & Heesemann, J. Arginine-143 of Yersinia enterocolitica
YopP crucially determines isotype-related NF-κB suppression and apoptosis induction in macrophages.
Infect Immun 69, 7652–62 (2001).
86. Black, D.S. & Bliska, J.B. Identification of p130Cas as a substrate of Yersinia YopH (Yop51), a bacterial
protein tyrosine phosphatase that translocates into mammalian cells and targets focal adhesions. EMBO J
16, 2730–44 (1997).
87. Persson, C., Carballeira, N., Wolf-Watz, H. & Fallman, M. The PTPase YopH inhibits uptake of
Yersinia, tyrosine phosphorylation of p130Cas and FAK, and the associated accumulation of these
proteins in peripheral focal adhesions. EMBO J 16, 2307–18 (1997).
Yersinia 437
88. Andersson, K. et al. YopH of Yersinia pseudotuberculosis interrupts early phosphotyrosine signalling
associated with phagocytosis. Mol Microbiol 20, 1057–69 (1996).
89. Mills, S.D. et al. Yersinia enterocolitica induces apoptosis in macrophages by a process requiring
functional type III secretion and translocation mechanisms and involving YopP, presumably acting as
an effector protein. Proc Natl Acad Sci USA 94, 12638–43 (1997).
90. Schesser, K. et al. The yopJ locus is required for Yersinia-mediated inhibition of NF-κB activation and
cytokine expression: YopJ contains a eukaryotic SH2-like domain that is essential for its repressive
activity. Mol Microbiol 28, 1067–79 (1998).
91. Ruckdeschel, K. et al. Yersinia enterocolitica impairs activation of transcription factor NF-κB:
Involvement in the induction of programmed cell death and in the suppression of the macrophage tumor
necrosis factor α production. J Exp Med 187, 1069–79 (1998).
Section IV
CONTENTS
30.1 Introduction................................................................................................................................... 441
30.2 Foodborne Diseases due to Alternaria Mycotoxins..................................................................... 442
30.3 Investigation and Analysis of Toxigenic Alternaria Strains......................................................... 443
30.4 Laboratory Models for Foodborne Alternaria Analysis............................................................... 445
30.4.1 Animal Laboratory Models.............................................................................................. 447
30.4.2 In Vitro Laboratory Models.............................................................................................. 447
30.4.2.1 Cell Line Systems............................................................................................. 447
30.4.2.2 Other In Vitro Models....................................................................................... 449
30.5 Conclusions................................................................................................................................... 450
Acknowledgments....................................................................................................................................451
References................................................................................................................................................451
30.1 Introduction
Alternaria is a ubiquitous fungal genus covering 275 species,1 and it has been typified as saprobic,
endophytic, and pathogenic. Currently, 24 Alternaria sections, 16 of which are newly described,
and 6 monotypic lineages are recognized.2 Alternaria is associated with a variety of foods. Species
of Alternaria are serious plant pathogens that damage crops in the field and cause postharvest
decays. In addition, with the ability to grow at low temperatures and low water activity (aw),3,4
Alternaria spp. may contaminate and infect fruit and vegetables kept refrigerated.5,6 The most com-
monly reported species include Alternaria alternata, Alternaria tenuissima, Alternaria arbores-
cens, Alternaria radicina, Alternaria brassicae, Alternaria brassicicola, and Alternaria infectoria.
Depending on geographical areas, a specific Alternaria species can predominate over others. For
instance, A. tenuissima is a more frequently isolated species in Argentinean wheat7 than A. alternata
and A. infectoria, which have been reported as the predominant species in cereals in several stud-
ies worldwide.8–12 Alternaria species colonize a range of plants including cereals,13 tomatoes,14 oil-
seeds,15 apples,16 berries, grapes, citrus fruits,17 carrots,18 and others. Some of these moulds may have
a negative impact on foods due to spoilage, causing economic losses and infections.19 It is well known
that Alternaria colonization of ripening ears can result in black pointed grain and impact directly on
flour color of bread-making wheats,20 its spores can be allergenic and cause asthma,21 and some spe-
cies may synthesize mycotoxins whose high toxicity is detrimental to both human health and animal
welfare.
441
442 Laboratory Models for Foodborne Infections
TABLE 30.1
Toxic Secondary Metabolites Produced by the Most Important Alternaria Species and Foods Involved
Species Toxic Secondary Metabolites Foods References
A. alternata AOH, AME, ALT, TeA, ATX-I, ATX-II, Tomato, red pepper, fresh 37,87,88
ATX-III fruit, vegetables, peanut,
wheat
A. tenuissima AOH, AME, ATX-I, -III, TeA Strawberries 28,88
A. arborescens AOH, AME, TEN Fruits, tomato, barley grain 87,89
A. radicina ATX-I, -II, -III, TeA Umbelliferous plants 88
A. brassicae AOH, AME Mustard, opium 90,91
A. brassicicola Small phytotoxic metabolites (causal Rape 92–94
agent of black spot disease)
A. infectoria 4Z-infectopyrone, novae-zelandin A, Wheat 87
novae-zelandin B, phomapyrone A, B,
E, G, F, D
Source: Ostry, V., World Mycotoxin J., 1, 175, 2008.
AOH, alternariol; AME, alternariol methyl ether; ALT, altenuene; TeA, tenuazonic acid; ATX-I, -II, -III,
altertoxins-I, -II, and -III.
Alternaria 443
optimal conditions for ALTX-II production were 0.98 aw and 30°C, although A. tenuissima strains also
accumulate significant amounts of this toxin at 34°C.
Taking into account the ecological conditions for mycotoxin production by Alternaria species, it is
not surprising that these toxins can contaminate a wide range of food and foodstuffs including, e.g.,
citrus, cereals, olives, tomatoes, apples, seeds, berries, and juices (reviewed by Scott,33 see Table 30.1).
The maximum levels of Alternaria mycotoxins reported in marketed food products are in the range of
1–103 μg/kg.24
Alternaria toxins have recently received increasing attention. The European Commission asked the
European Food Safety Authority (EFSA) to review the safety of Alternaria toxins in food and feed.34
EFSA concluded that Alternaria toxins are of high concern for public health. In fact, exposure to
Alternaria toxins has been linked to esophageal cancer in Linxian Province, China, as well as in areas of
South Africa where high levels of A. alternata contamination have been found in grains.23,35,36 However,
due to the possible co-occurrence of mycotoxins in contaminated grains, this correlation may not have
been exclusively ascribed to Alternaria toxins. For this reason, it is important to have adequate methods
first for analyzing and identifying Alternaria species and their toxins and second to examine the toxicity
for foodborne mycotoxin-producing Alternaria.
FOOD
Isolation of Alternaria
Traditional Molecular
methods methods
Microscopy
(Morphology)
Chromatographic
techniques for
mycotoxin analysis
Mycotoxin
purification
-Animal models
-Cell lines
-Other tests
For the evaluation of mycotoxin production by Alternaria, analytical methods based on chromatogra-
phy are often used for their extraction and detection from the food matrices (Figure 30.1). Usually, a sol-
vent extraction from solid foods with organic solvents, such as dichloromethane, methanol, acetonitrile,
or ethyl acetate, is required, although an acidic extraction or a further acidification step is preferable to
increase the recovery for TeA.33 Cleanup procedures are necessary when a complex matrix is involved.
Successive steps of solvent partitioning, solid phase extraction (SPE) columns, or solid phase microex-
traction are common cleanup techniques used in most food matrices.24
Mycotoxin determination has been traditionally carried out by thin-layer chromatography (TLC),
gas chromatography (GC), and mainly high-performance liquid chromatography (HPLC), usually with
ultraviolet (UV) detection, although fluorescence and electrochemical detection have also been used
for certain toxins. Atmospheric pressure chemical ionization (APCI), LC-mass spectroscopy (MS), and
LC-MS/MS have been applied to the determination and confirmation of AOH and AME identity at
sub ng/mL levels.33,38 Several multimycotoxin methods have been recently developed, many of which
include the detection of Alternaria toxins, most of them based on LC-MS/MS systems. A multimethod
for detection of 33 mycotoxins (including AOH and AME) in various food matrices has been developed
by Spanjer et al.,39 based on LC-MS/MS using an electrospray ionization interface (ESI) for detec-
tion and MS/MS with multiple reaction monitoring (MRM). Rasmussen et al.40 developed a method for
simultaneous detection of 27 mycotoxins (including AOH, AME, TeA, and altersetin) in maize silage.
A simple pH-buffered sample extraction has been developed based on the QuEChERS method, and
the detection is made by LC-MS/MS without further cleanup. Wang et al.41 developed an LC-MS/MS
method for detecting 17 mycotoxins (including AOH and AME) with application in traditional Chinese
medicine products, using a MultiPurification Column for cleanup.
Alternaria 445
Even though several techniques are effective for Alternaria mycotoxin quantification in different
food matrices, there are a number of difficulties related to the performance of these methods, such
as the efficiency of sample cleanup, low or unequal recoveries for some of the toxins, availability of
standards, lack of reference materials for food and feedstuffs, etc. In addition, these methods are time
consuming.
As an alternative to traditional methods, nucleic-acid-based techniques are being increasingly
applied to examine the taxonomic relationships among Alternaria species. Most of them have been
focused on small-spored catenulate Alternaria, which show little resolution in their molecular phy-
logeny. However, cladistics analyses of “housekeeping genes” commonly used for other genera,
such as the mitochondrial large subunit (mtLSU) ribosomal DNA, internal transcribed spacer (ITS),
β-tubulin, translation elongation factor α, calmodulin, actin, chitin synthetase, etc., failed to discrimi-
nate among the small-spored species, except for the A. infectoria species group.42–45 There are also
genomic techniques to detect, identify, and quantify toxigenic moulds in foodstuffs. So far, there are
no molecular methods based on genes involved in the Alternaria mycotoxin biosynthesis pathways;
however, some methods, which have used unique conserved genes that distinguish toxigenic and non-
toxigenic Alternaria spp., have been developed successfully. Several molecular methods have been
developed to detect the presence of Alternaria spores and biomass in foods, such as the one reported
by Zur et al.,46 a polymerase chain reaction (PCR)-based method with primers specific to the ITS1
and ITS2 of the 5.8S rRNA gene of Alternaria to detect its presence in commercial tomato products.
The main inconvenience of these methods is that viable and nonviable cells cannot be distinguished,
thus resulting in an overestimation of the amount of spores that can actually produce mycotoxin in a
food product. More recently, Crespo-Sempere et al.47 developed a method including a pretreatment of
samples with nucleic-acid-intercalating dyes (propidium monoazide, PMA) prior to quantitative PCR.
PMA selectively penetrates cells with a damaged membrane, inhibiting DNA amplification during
PCR. The method, based on a primer pair (Alt4 and Alt5) specific to Alternaria spp., allowed quan-
tifying a detection limit of 102 spores/g on tomatoes. Even though the tomato matrix had a protective
effect on the cells against PMA toxicity, reducing the efficiency to distinguish between viable and non-
viable cells, the method is still a suitable tool for quantifying viable Alternaria cells, which could be
useful for estimating potential risks of mycotoxin contamination. The main drawback of these nucleic-
acid-based methods as well as of the traditional identification of Alternaria followed by evaluation of
mycotoxin production is that they only give information about the negative potential effect derived
of the Alternaria presence in foods. Although the latter techniques allow taking corrective actions
to avoid presence of mycotoxin-producing Alternaria on foods, for a more appropriate investigation
of foodborne Alternaria it is necessary to use laboratory animals or cell system models in which the
effect of Alternaria extracts or Alternaria mycotoxin contaminated foods can be evaluated. This could
allow estimating the real risk of the presence of Alternaria in foods.
TABLE 30.2
Animal Models for Evaluating the Toxic Effects of Alternaria Extracts and
Produced Mycotoxins
Animals Alternaria Extracts/Mycotoxin References
Chickens Alternaria extract 55
TeA 10
Rats Alternaria extract 55
Chicken embryos Alternaria extract 53
Mice Alternaria extract 53,55
AOH 50
TeA 48
Dogs TeA 10
TeA, tenuazonic acid; AOH, alternariol.
TABLE 30.3
Cell Line Systems and Other In Vitro Tests Used for Studying the Toxic Effects of Alternaria Mycotoxins
Cell Lines/In Vitro Mycotoxins/Alternaria
Assay Extracts Studied Toxic Effects References
Caco-2 AOH/AME Absorption, metabolism, oxidative stress 59–61
HT-29 AOH/AME DNA-damaging properties 62–65
TeA, ATX-I, -II, -III DNA-damaging properties 66
V79 AOH/AME Cell proliferation, clastogenic potential, 63,67,69
mutagenicity
ATX-I, -II Mutagenicity 68,70
H4IIE ATX-I Mutagenicity 70
MLC AOH Mutagenicity 69
CHL/3T3/L-O2 TeA Cytotoxicity 71
RAW 264.7 AOH DNA-damaging properties, autophagy, 72–74
senescence
HCT116 AOH Cell death mode 75
AOH/AME Cytotoxicity 76
Ishikawa AOH Estrogenic potential 67
H295R AOH Alteration in steroid hormone production 77
RGA AOH Alterations in androgen, progestogen, 77
glucocorticoid, estrogen nuclear receptor
Porcine endometrial AOH Oestrogenic effect 78
Hepa-1c1c7/Hepa- AOH/AME Cytotoxicity 79,80
1c1c4/Hepa-1c1c12
HepG2 AOH Cytotoxicity 81
A431 AOH/AME/ALT Viability and cytotoxicity potential 62
MCF-7 AOH DNA-damaging properties 62
Artemia salina test AOH, AME, ALT, ATX-I, Toxicity 82,83
TeA and Alternaria extracts
Ames Salmonella test Alternaria extracts and Mutagenicity 56,84
ATX-I, -II, and -III
ALT, AOH, AME, ATX-I, Mutagenicity 70,85
TEN, TeA
Human digestive tract Alternaria extracts and AOH Bioaccessibility from food 48
simulation and ALT
AOH, alternariol; AME, alternariol methyl ether; TeA, tenuazonic acid; ATX-I, -II, -III, altertoxins-I, -II and -III;
ALT, altenuene.
Alternaria 447
a main role in the genotoxic properties. In a toxicity-guided fractionation assay, aimed to identify DNA
strand breaking impact compounds in extracts obtained from rice heavily infested with A. alternata, the
HT-29 cell line was used.66 TeA, AOH, and AME do not cause significant DNA-damaging effects, while
ATX-II contributes to the genotoxic effects of the extracts, showing potent DNA-damaging properties in
HT-29 cells. ATX-II does not provoke oxidative stress, while it does influence the cell cycle distribution
of HT-29 cells. The inhibition of proliferation of HT-29 cells (seen in a sulforhodamine B assay) matched
this interference with the cell cycle, thus arguing for effects on inhibition of cell proliferation rather than
cytotoxicity.66
Chinese hamster cell line V79 has been developed from lung tissue of a young male Chinese ham-
ster. This cell line is used to investigate DNA damage, the effect on cell proliferation, and the clasto-
genic potential of AOH. Pfeiffer et al.63 showed that AOH and AME induce DNA strand breaks in a
concentration-dependent manner in the cell line V79. Lehmann et al.67 demonstrated that AOH inhibits
cell proliferation by interference with the cell cycle and induces kinetochore-negative micronucleus
(MN) in cultured V79 cells. ATX II is a potent mutagen in the cell line V79, inducing a concentration-
dependent increase of mutations at the hypoxanthine guanine phosphoribosyltransferase gene locus at
concentrations similar to that of the established mutagen 4-quinoline-N-oxide.68 However, the muta-
genic potency of AOH is at least 50 times lower than that of ATX II.69 In contrast to AOH and AME,
ATX II does not affect the cell cycle of V79 cells. ATX II also causes DNA strand breaks in V79
cells, being more potent than AOH and AME. Schrader et al.70 demonstrated that nitrosylated ATX-I
is mutagenic to V79 cells too. They also used rat hepatoma H4IIE cells to evaluate the mutagenicity of
ATX-I, since they retain more metabolic activities and have been used as a model for assessing chemi-
cal effects on the induction of cytochrome P450, aryl hydrocarbon hydroxylase, and epoxide hydrolase
activities.70 This study concludes that if nitrosylated ATX-I is similarly toxic to other cell types, in par-
ticular esophageal cells, carcinogenesis would be promoted through cell death and the proliferation of
neighboring cells to generate replacements. The sensitivity of H4IIE cells also suggested that exposure
to nitrosylated ATX-I could lead to liver damage/carcinogenesis.
The mutagenicity of AOH at the thymidine kinase (TK) gene locus in mouse lymphoma L5178 tk+/−
(MLC) cells has also been investigated.69 Concentrations higher than or equal to 10 μM AOH give rise
to a significant and concentration-dependent induction of TK mutations in MLC cells. Discrimination
between small and large colonies in the TK assay reveals the predominant induction of small colonies,
which are indicative for extensive chromosomal deletions and correlate with the induction of micronuclei
in MLC cells.
Chinese hamster lung (CHL) cells are also used to evaluate TeA toxicity to mammalian cells. Cell
proliferation inhibition increases with extension of toxin exposure time.71 Total protein content in culture
of cells was measured and showed that TeA decreases the total protein content after 72 h toxin exposure
with half maximal effective concentration (EC50) value of 56.28 μg/mL. In the same study, 3T3 mouse
fibroblasts (3T3 cells) and human hepatocytes (L-O2 cells) showed a similar response. It was concluded
that TeA is the most cytotoxic to 3T3 cells, followed by CHL cells and L-O2 cells. At lower concentra-
tions, TeA had lower cytotoxicity to the human hepatocytes, which suggests that such cells may tolerate
TeA at low concentrations.
The murine macrophage cell line RAW 264.7 was used to test AOH toxicity as a response to DNA dam-
age and to test if AOH induces autophagy, senescence, abnormal morphology, and cell cycle arrest.72–74
High AOH concentrations block cell proliferation and increase the level of reactive oxygen species
(ROS). However they are not directly linked to each other. It seems that AOH-induced DNA damage and
resulting transcriptional changes in the p21, MDM2, and Cyclin B genes contribute to the reduced cell
proliferation, while the expression of Sestrin 2 gene would contribute to the oxidant defense.74 Besides,
it seems that the AOH-induced cell cycle arrest, most probably due to DNA damage and incomplete
decatenation, is followed by very specific morphological changes.72 Furthermore, Solhaug et al.73 found
that autophagy and senescence using the RAW264.7 macrophage model are related to stress responses
caused by the DNA-damaging AOH.
Human colon carcinoma cells HCT116 are used to evaluate the cell death mode and pathways trig-
gered by AOH 75 and the cytotoxic potential of AOH and AME mixtures.76 Cells treated with AOH show
a loss of cell viability by inducing apoptosis.75 Bensassi et al.76 demonstrated that the exposure of HTC116
Alternaria 449
cells to low cytotoxic AOH doses causes a moderate cytotoxicity. However, when AOH and AME are
combined, they exert a significant increase in their toxic potential.
The cell line Ishikawa allows examining the estrogenic potential of AOH.67 In this study, the estroge-
nicity of AOH was about 10,000-fold weaker than of the endogenous hormone E2. To identify if AOH
may alter steroidogenesis, an in vitro screening assay based on measuring alterations in steroid hormone
production and the expression of several important genes that encode the various enzymes involved
in steroidogenesis using the human adrenocortical carcinoma cell line H295R has been performed by
Frizzel et al.77 They demonstrated that AOH has the ability to interfere with steroidogenesis pathway
since it modifies the expression of important genes in steroidogenesis in H295R cells.
Human breast adenocarcinoma cell line RGA is of great relevance to evaluate the effect of AOH on
the androgen, progestogen, glucocorticoid, and estrogen nuclear receptors present in those cells.77 These
authors concluded that AOH has a weak estrogenic activity when tested in the estrogen-responsive RGA
cells. The sensitivity of AOH to different cell types may be an important factor when considering this
mycotoxin oestrogenic effect. In another study performed by Willemsen et al.,78 the estrogenic influence
of the ER α in porcine endometrial cells was not detected.
The evaluation of AOH and AME toxicity in the mouse hepatoma Hepa-1c1c7 cell line with intact
aryl hydrocarbon receptor (AhR) signaling and the Hepa-1c1c4 and Hepa-1c1c12 cell lines, which are
deficient for the AhR nuclear translocator (ARNT) or the AhR, respectively,79 has been performed. It was
demonstrated that AOH and AME are novel inducers of the AhR/ARNT pathway, which mediates induc-
tion of CYP1A1 and apoptosis and might thus contribute to the toxicity of these mycotoxins. Burkhardt
et al.80 used the same three mouse hepatoma (Hepa-1) cell lines with intact and compromised AhR sig-
naling to compare their activities for hydroxylation, methylation, and glucuronidation.
The liver hepatocellular carcinoma HepG2 cell line is widely used to evaluate the cytotoxic effects
of AOH.81 In this study, HepG2 cells were treated at different concentrations over 24, 48, and 72 h. The
half maximal inhibitory concentration (IC50) values were from 65 to 96 μM for AOH. Pfeiffer et al.63 also
demonstrated that AOH and AME cause a concentration-dependent induction of DNA strand breaks in
HepG2 cells.
The human vulva carcinoma A431 cell line allows testing the viability and cytotoxicity potential of
AOH, AME, and ALT as well as to perform a single-cell gel electrophoresis (comet assay) and inmuno-
band depletion assay.62 They demonstrated that AOH and AME significantly increase the rate of DNA
strand breaks in A431 at micromolar concentrations, whereas ALT does not affect DNA integrity up to
100 μM. The inmunoband depletion assay allowed for the observation that AOH affects topoisomerase,
preferentially the IIα isoform. This latter conclusion was also obtained by the same authors62 when they
utilized the human breast adenocarcinoma MCF-7 cell line to evaluate the effects of AOH on topoisomer-
ase I and II by means of relataxion and cleavage assays and decatenation and cleavage assays, respectively.
also examined the major Alternaria toxins (ALT, AOH, AME, ATX-I, TEN, and TeA) by the Ames
Salmonella test, using strains TA98 and TA100 to detect histidine revertants that result from frame-
shift and base pair mutations, respectively. In the absence of nitrosylation, they found that ATX-1
was positive for mutagenicity in TA98, while the all other toxins were negative. In general, nitro-
sylation increases mutagenic potencies, although no effect was observed on TEN and TeA mutage-
nicities. These findings suggest that the presence of mould contamination that contains ATX-I in
foodstuff could pose a potent carcinogenic health hazard when combined with a diet high in nitrites
and nitrates. In a later work, Schrader et al.85 reexamined mutagenicity of these toxins with and
without nitrosylation, using Ames Salmonella strain TA97, sensitive to frameshift mutations at a run
of C’s, as well as strains TA102 and TA104, reverted by base pair mutations at AT sites and more
sensitive to oxidative damage. The results suggested that ATX-I, AME, and AOH induce mutations at
AT sites, possibly through oxidative damage. In addition, nitrosylation enhances ATX-I mutagenicity
at runs of C’s.
Several methods have been developed for simulating the human digestive tract. Dall’Asta et al.48 used a
method proposed by Versantvoort et al.86 for assessing the bioaccessibility of mycotoxins from food, and
another one that involved the use of fecal water obtained as a mixture from healthy volunteers follow-
ing an omnivore diet. Samples from the simulated digestion assay were taken at the end of the duodenal
phase and directly analyzed by LC–MS/MS. Samples from the fecal fermentation assay were taken after
30 min and 24 h of incubation and analyzed by LC–MS/MS after high-speed centrifugation. Their results
showed that both AOH and ALT were totally stable along the simulated human gastrointestinal tract.
Neither digestive conditions nor human gut microbiome seemed to be able to degrade AOH and ALT, as
these were fully recovered in the fecal water after 24 h of incubation.
30.5 Conclusions
Alternaria species are found in a wide range of foods including cereals, fruits, and vegetables due to
their ability to grow at low temperatures and low aw. Although some Alternaria spp. may be allergenic
and cause asthma, the synthesis of mycotoxins when the environmental conditions are adequate is the
most worrying undesirable effect. In spite of the fact that Alternaria species produce a huge number of
secondary metabolites, only a small proportion is considered toxic to humans and animals. The most
important Alternaria mycotoxins are AOH, AME, ALT, TeA, TEN, ATX-I, ATX-II, and ATX-III.
The diagnosis of mycotoxin-producing Alternaria and Alternaria toxins in food could be performed
by morphological, biochemical/analytical, and genetic molecular techniques. However, laboratory mod-
els based on animals or in vitro (cell lines or biological tests) systems are needed to examine the toxic
effects of Alternaria and their toxins on humans and animals, thus completing the investigation in food-
borne Alternaria.
Several animal models based on chickens, rats, dogs, and mice could be used to evaluate toxic effects
of Alternaria culture extracts and some of the main purified Alternaria toxins. They may be adapted for
foodborne analysis.
In addition, cell lines and other models (Ames Salmonella test, A. salina, etc.) are also available
for this purpose. Ethical and scientific reasons due to the use of high number of animals and the toxin
amount required for these experiments are the main limitations on the performance of laboratory toxic-
ity for animal model assays. Cell system models could be a good alternative for foodborne Alternaria
analysis, with human cell lines such as Caco-2 and HT-29, the Chinese hamster V79 cells, rat H4IIE
hepatoma cells, or murine macrophage cell line RAW 264.7 being the most appropriate. Overall, these
assays demonstrated that all Alternaria toxins are cytotoxic and that AOH and AME are also genotoxic
and mutagenic; however, AOH is a weak estrogenic. TeA has been reported to exert cytotoxic properties
as well as being acutely toxic. The altertoxins ATX-I, ATX-II, and ATX-III have been shown to be more
mutagenic and acutely toxic than AOH and AME.
The toxicity of Alternaria mycotoxins still needs a further research. Not enough information is
available on the toxicokinetics, including metabolism, of the most toxicologically relevant foodborne
Alternaria toxins.
Alternaria 451
Acknowledgments
This work has been funded by the Spanish Comisión Interministerial de Ciencia y Tecnología with the
projects AGL2013-45729P and Carnisenusa CSD2007-00016, Consolider Ingenio 2010 and GRU09162
of the Junta de Extremadura and FEDER. Dr. Alicia Rodríguez was supported by a postdoctoral research
fellowship (PO12016) from the Gobierno de Extremadura, Consejería de Empleo, Empresa e Innovación,
and the Fondo Social Europeo (FSE).
REFERENCES
1. Simmons, E., Alternaria: An Identification Manual, CBS Biodiversity Series 6, CBS Fungal Biodiversity
Centre, Utrecht, NL, 2007.
2. Woudenberg, J.H.C. et al., Alternaria redefined, Stud. Mycol., 75, 171, 2013.
3. Hasan, H.A.H., Alternaria mycotoxins in black rot lesion of tomato fruit-conditions and regulation of
their production, Mycopathologia, 130, 171, 1995.
4. Magan, N., Cayley, G.R. and Lacey, J., The effect of water activity and temperature on mycotoxin
production by Alternaria alternata in culture and on wheat grain, Appl. Environ. Microbiol., 47, 1113,
1984.
5. Barkai-Golan, R., Postharvest Diseases of Fruits and Vegetables: Development and Control, Elsevier,
Amsterdam, NL, 2001.
6. Müller, M., Alternaria infestation of corn silage and hay, Zbl. Mikrobiol., 146, 481, 1991.
7. Patriarca, A. et al., Mycotoxin production by Alternaria strains isolated from Argentinean wheat, Int. J.
Food Microbiol., 119, 219, 2007.
8. Andersen, B. et al., Associated field mycobiota on malt barley, Can. J. Bot., 74, 854, 1996.
9. Kosiak, B. et al., Alternaria and Fusarium in Norwegian grains of reduced quality a matched pair
sample study, Int. J. Food Microbiol., 93, 51, 2004.
10. Logrieco, A., Moretti, A. and Solfrizzo, M., Alternaria toxins and plant disease: An overview of origin,
occurrence and risks, World Mycotoxin. J., 2, 129, 2009.
11. Medina, A. et al., Survey of the mycobiota of Spanish malting barley and evaluation of the mycotoxin
producing potential of species of Alternaria, Aspergillus and Fusarium, Int. J. Food Microbiol., 108,
196, 2006.
12. Webley, D.J. et al., Alternaria toxins in weather-damaged wheat and sorghum in the 1995–1996
Australian harvest, Aust. J. Agric. Res., 48, 1249, 1997.
13. Li, F.Q. and Yoshizawa, T., Alternaria mycotoxins in weathered wheat from China, J. Agric. Food
Chem., 48, 2920, 2000.
14. Andersen, B. and Frisvad, J.C., Natural occurrence of fungi and fungal metabolites in moldy tomatoes,
J. Agric. Food Chem., 52, 7507, 2004.
15. Vinas, I. et al., Natural occurrence of aflatoxin and Alternaria mycotoxins in oilseed rape from Catalonia
(Spain)—Incidence of toxigenic strains, Mycopathologia, 128, 175, 1994.
16. Robiglio, A.L. and López, S.E., Mycotoxin production by Alternaria alternata strains isolated from
red-delicious apples in Argentina, Int. J. Food Microbiol., 24, 413, 1995.
17. Tournas, V.H. and Katsoudas, E., Mould and yeast flora in fresh berries, grapes and citrus fruits, Int. J.
Food Microbiol., 105, 11, 2005.
18. Solfrizzo, M. et al., Liquid chromatographic determination of Alternaria toxins in carrots, J. AOAC Int.,
87, 101, 2004.
19. Dagnas, S. and Membré, J.M., Predicting and preventing mold spoilage of food products, J. Food
Protect., 76, 538, 2012.
20. Culshaw, F. et al., Blackpoint of Wheat, p. 46. Home Grown Cereals Authority, London, 1988.
21. Kustrzeba-Wójcicka, I., et al., Alternaria alternata and its allergens: A comprehensive review, Clin. Rev.
Allergy Immunol., 47, 354, 2014.
22. Fernández-Cruz, M.L., Mansilla, M.L. and Tadeo, J.L., Mycotoxins in fruits and their processed prod-
ucts: Analysis, occurrence and health implications, J. Adv. Res., 1, 113, 2010.
23. Panigrahi, S., Alternaria toxins, in Handbook of Plant and Fungal Toxicants, p. 319, D’Mello, J.P.F.
(Ed.), CRC Press, Boca Raton, FL, 1997.
452 Laboratory Models for Foodborne Infections
24. Ostry, V., Alternaria mycotoxins: An overview of chemical characterization, producers, toxicity, analy-
sis and occurrence in foodstuffs, World Mycotoxin J., 1, 175, 2008.
25. Bottalico, A. and Logrieco, A., Toxigenic Alternaria species of economic importance, in Mycotoxins in
Agriculture and Food Safety, p. 65, Sinha, K.K. and Bhatnagar, D. (Eds.), Marcel Dekker, New York,
1998.
26. Magan, N. and Baxter, E.S., Relationship between environmental factors and tenuazonic acid pro-
duction by Alternaria isolates from sorghum, in Occurrence and Significance of Mycotoxins, p. 309,
Skudamore, K.A. (Ed.), Slough Central Science Laboratories, Slough, UK, 1993.
27. Oviedo, M.S. et al., Effect of environmental factors on tenuazonic acid production by Alternaria alter-
nata on soybean-based media, J. Appl. Microbiol., 107, 1186, 2009.
28. Patriarca, A. et al., Temperature and water stress impacts on growth and production of altertoxin-II by
strains of Alternaria tenuissima from Argentinean wheat, World Mycotoxin J., 7, 329, 2014.
29. Magan, N. and Lacey, J., The effect of water activity and temperature on mycotoxin production by
Alternaria alternata in culture and on wheat grain, in Trichothecenes and Other Mycotoxins, p. 243,
Lacey, J. (Ed.), John Wiley & Sons, Chichester, UK, 1985.
30. Oviedo, M.S. et al., Impact of water activity and temperature on growth and alternariol and alter-
nariol monomethyl ether production of Alternaria alternata isolated from soybeans, J. Food Protect.,
73, 336, 2010.
31. Oviedo, M.S. et al., Influence of water activity and temperature on growth and mycotoxin production by
Alternaria alternata on irradiated soya beans, Int. J. Food Microbiol., 149, 127, 2011.
32. Pose, G. et al., Water activity and temperature effects on mycotoxin production by Alternaria alternata
on a synthetic tomato medium, Int. J. Food Microbiol., 142, 348, 2010.
33. Scott, P.M., Analysis of agricultural commodities and foods for Alternaria mycotoxins, J. AOAC Int.,
84, 1809, 2001.
34. EFSA Panel on Contaminants in the Food Chain, Scientific opinion on the risks for animal and public
health related to the presence of Alternaria toxins in feed and food, EFSA J., 9, 2407, 2011.
35. Liu, G.T. et al., Etiological role of Alternaria alternata in human esophageal cancer, Chin. Med. J., 105,
394, 1992.
36. Liu, G.T. et al., Relationships between Alternaria alternata and oesophageal cancer, in Relevance to
Human Cancer of N-Nitroso Compounds, Tobacco Smoke and Mycotoxins, p. 258, O’Neill, I.K., Chen,
J. and Bartsch, H. (Eds.), International Agency for Research on Cancer, Lyon, FR, 1991.
37. Greco, M. et al., Toxigenic Alternaria species from Argentinean blueberries, Int. J. Food Microbiol.,
154, 187, 2012.
38. Scussel, V.M. et al., Multi-toxins in cork stoppers and fermented beverages by liquid chromatography-
mass spectrometry including Alternaria toxins, in XIIth International IUPAC Symposium on Mycotoxins
and Phycotoxins, poster 1356, Istanbul, TR, 2007.
39. Spanjer, M.C., Rensen, P.M. and Scholten, J.M., LC-MS/MS multi-method for mycotoxins after single
extraction, with validation data for peanut, pistachio, wheat, maize, cornflakes, raisins and figs, Food
Addit. Contam., 25, 472, 2008.
40. Rasmussen, R.R. et al., Multi-mycotoxin analysis of maize silage by LC-MS/MS, Anal. Bioanal. Chem.,
397, 765, 2010.
41. Wang, S. et al., Simultaneous determination of seventeen mycotoxins residues in Puerariae lobatae radix
by liquid chromatography-tandem mass spectrometry, J. Pharm. Biomed. Anal., 98, 201, 2014.
42. Andrew, M., Peever, T.L. and Pryor, B.M., An expanded multilocus phylogeny does not resolve morpho-
logical species within the small-spored Alternaria species complex, Mycologia, 101, 95, 2009.
43. Peever, T.L. et al., Molecular systematics of citrus-associated Alternaria species, Mycologia, 96, 119, 2004.
44. Pryor, B.M. and Gilbertson, R.L., Molecular phylogenetic relationships amongst Alternaria species and
related fungi based upon analysis of nuclear ITS and mt SSU rDNA sequences, Mycol. Res., 104, 1312,
2000.
45. Roberts, R.G., Reymond, S.T. and Andersen, B., RAPD fragment pattern analysis and morphological
segregation of small-spored Alternaria species and species groups, Mycol. Res., 104, 151, 2000.
46. Zur, G. et al., Development of a PCR-based assay for the detection of Alternaria fungal contamination
in food products, J. Food Protect., 62, 1191, 1999.
47. Crespo-Sempere, A. et al., Propidium monoazide combined with real-time quantitative PCR to quantifiy
viable Alternaria spp. contamination in tomato products, Int. J. Food Microbiol., 165, 214, 2013.
Alternaria 453
48. Dall’Asta, C., Cirlini, M. and Falavigna, C., Mycotoxins from Alternaria: Toxicological implications,
Adv. Mol. Toxicol., 8, 107, 2014.
49. Juan-García, A. et al., Efectos tóxicos de alternariol por ensayos in vitro: Revisión, Rev. Toxicol., 31,
196, 2014.
50. Schuchardt, S., Ziemann, C. and Hansen, T., Combined toxicokinetic and in vivo genotoxicity study on
Alternaria toxins, EFSA supporting publication, EN-679, 2014.
51. Altemöller, M., Podlech, J. and Fenske, D., Total synthesis of altenuene and isoaltenuene, Eur. J. Org.
Chem., 7, 1678, 2006.
52. Koch, K. et al., Total synthesis of alternariol, J. Org. Chem., 70, 3275, 2005.
53. Griffin, G.F. and Chu, F.S., Toxicity of the Alternaria metabolites alternariol, alternariol monomethyl
ether, altenuene and tenuazonic acid in the chicken embryo assay, Appl. Environ. Microbiol., 46, 1420,
1983.
54. Pero, R.W. et al., Toxicity of metabolites produced by the “Alternaria”, Environ. Health Perspect., 4, 87,
1973.
55. Sauer, D.B. et al., Toxicity of Alternaria metabolites found in weathered sorghum grain at harvest,
J. Agric. Food Chem., 26, 1380, 1978.
56. Scott, P.M. and Stoltz, D.R., Mutagens produced by Alternaria alternata, Mutat. Res., 78, 33, 1980.
57. Cencič, A. and Langerholc, T., Functional cell models of the gust and their applications in food
microbiology—A review, Int. J. Food Microbiol., 141, S4, 2010.
58. Rousset, M., The human colon carcinoma cell lines HT-29 and Caco-2: Two in vitro models for the study
of intestinal differentiation, Biochimie, 68, 1035, 1986.
59. Fernández-Blanco, C., Font, G. and Ruiz, M., Oxidative stress of alternariol in Caco-2 cells, Toxicol.
Lett., 229, 458, 2014.
60. Fernández-Blanco, C., Font, G. and Ruiz, M., Oxidative DNA damage and disturbance of antioxidant
capacity by alternariol in Caco-2 cells, Toxicol. Lett., 235, 61, 2015.
61. Burkhardt, B., Pfeiffer, E. and Metzler, M., Absorption and metabolism of the mycotoxins alternariol
and alternariol-9-methyl ether in Caco-2 cells in vitro, Mycotoxin Res., 25, 149, 2009.
62. Fehr, M. et al., Alternariol acts as a topoisomerase poison, preferentially affecting the IIα isoform, Mol.
Nutr. Food Res., 53, 441, 2009.
63. Pfeiffer, E., Eschbach, S. and Metzler, M., Alternaria toxins: DNA strand-breaking activity in mam-
malian cells in vitro, Mycotoxin Res., 23, 152, 2007.
64. Tiessen, C. et al., Modulation of the cellular redox status by the Alternaria toxins alternariol and alter-
nariol monomethyl ether, Toxicol. Lett., 216, 23, 2013.
65. Schwarz, C., Kreutzer, M. and Marko, D., Minor contribution of alternariol, alternariol monomethyl
ether and tenuazonic acid to the genotoxic properties of extracts from Alternaria alternate infested rice,
Toxicol. Lett., 214, 46, 2012.
66. Schwarz, C. et al., Characterization of a genotoxic impact compound in Alternaria alternata infested
rice as altertoxin II, Arch. Toxicol., 86, 1911, 2012.
67. Lehmann, L., Wagner, J. and Metzler, M., Estrogenic and clastogenic potential of the mycotoxin alter-
nariol in cultured mammalian cells, Food Chem. Toxicol., 44, 398, 2006.
68. Fleck, S.C. et al., Alternaria toxins: Altertoxin II is a much stronger mutagen and DNA strand breaking
mycotoxin than alternariol and its methyl ether in cultured mammalian cells, Toxicol. Lett., 214, 27,
2012.
69. Brugger, E.M. et al., Mutagenicity of the mycotoxin alternariol in cultured mammalian cells, Toxicol.
Lett., 164, 221, 2006.
70. Schrader, T.J. et al., Examination of Alternaria alternata mutagenicity and effects of nitrosylation using
the Ames Salmonella test, Teratog. Carcinog. Mutagen., 21, 261, 2001.
71. Zhou, B. and Qiang, S., Environmental, genetic and cellular toxicity of tenuazonic acid isolated from
Alternaria alternata, Afr. J. Biotechnol., 7, 1151, 2008.
72. Solhaug, A. et al., Alternariol induces abnormal nuclear morphology and cell cycle arrest in murine
RAW 264.7 macrophages, Toxicol. Lett., 219, 8, 2013.
73. Solhaug, A. et al., Autophagy and senescence, stress responses induces by the DNA-damaging myco-
toxin alternariol, Toxicology, 326, 119, 2014.
74. Solhaug, A. et al., Mechanisms involved in alternariol-induced cell cycle arrest, Mutat. Res., 738 1,
2012.
454 Laboratory Models for Foodborne Infections
75. Bensassi, F. et al., Cell death induced by Alternaria mycotoxin alternariol, Toxicol. In Vitro, 26, 915,
2012.
76. Bensassi, F. et al., Combined effects of alternariols mixture on human colon carcinoma cells, Toxicol.
Mech. Methods, 25, 56, 2015.
77. Frizzel, C. et al., An in vitro investigation of endocrine disrupting effects of the mycotoxin alternariol,
Toxicol. Appl. Pharm., 271, 64, 2013.
78. Willemsen, P. et al., Use of reporter cell lines for detection of endocrine-disrupter activity, Anal.
Bioanal. Chem., 378, 655, 2004.
79. Schreck, I. et al., The Alternaria mycotoxins alternariol and alternariol methyl ether induce cytochrome
P450 1A1 and apoptosis in murine hepatoma cells dependent on the aryl hydrocarbon receptor, Arch.
Toxicol., 86, 625, 2012.
80. Burkhardt, B. et al., Mouse hepatoma cell lines differing in aryl hydrocarbon receptor-mediated signal-
ing have different activities for glucuronidation, Arch. Toxicol., 86, 643, 2012.
81. Juan-García, A. et al., Cytotoxic effects and degradation products of three mycotoxins: Alternariol,
3-acetyl-deoxynivalenol and 15-acetyl-deoxynivalenol in liver hepatocellular carninoma cells, Toxicol.
Lett., 235, 8, 2015.
82. Zajwoski, P., Grabarkiewicz-Szcesna, J. and Schmidt, R., Toxicity of mycotoxins produced by four
Alternaria species to Artemia salina larvae, Mycotoxin Res., 7, 11, 1991.
83. Panigrahi, S. and Dallin, S., Toxicity of the Alternaria spp. metabolites, tenuazonic acid, alternariol,
altertoxin-I, and alternariol monomethyl ether to brine shrimp—(Artemia salina L) larvae, J. Sci. Food
Agric., 66, 493, 1994.
84. Stack, M.E. and Prival, M.J., Mutagenicity of the Alternaria metabolites altertoxins I, II, and III, Appl.
Environ. Microbiol., 52, 718, 1986.
85. Schrader, T.J. et al., Further examination of the effects of nitrosylation on Alternaria alternata myco-
toxin mutagenicity in vitro, Mutat. Res., 606, 61, 2006.
86. Versantvoort, C.H. et al., Applicability of an in vitro digestion model in assessing the bioaccessibility of
mycotoxins from food, Food Chem. Toxicol., 43, 31, 2005.
87. Andersen, B. et al., Characterization of Alternaria strains from Argentinean blueberry, tomato, walnut
and wheat, Int. J. Food Microbiol., 196, 1, 2015.
88. Lee, H.B., Patriarca, A. and Magan, N., Alternaria in food: Ecophysiology, mycotoxin production and
toxicology, Mycobiology, 43, 93, 2015.
89. Ntasiou, P. et al., Identification, characterization and mycotoxigenic ability of Alternaria spp. causing
core rot of apple fruit in Greece, Int. J. Food Microbiol., 197, 22, 2015.
90. Sharma, S. and Sohal, B.S., Resistance to Alternaria blight in Brassica juncea (var. RLM619) induced
by combination of elicitors and inoculation with Alternaria brassicae under controlled environment,
Indian J. Agric. Sci., 27, 180, 2014.
91. Pastirčák, M. and Fejér, J., A preliminary survey of fungi on opium poppy in Slovakia, Acta Hort., 1036,
157, 2014.
92. MacDonald, M.V. and Ingram, D.S., Towards the selection in vitro for resistance to Alternaria brassici-
cola (Schw.) Wilts., in Brassica napus ssp. oleifera (Metzg.) Sinsk., winter oilseed rape. New Phytol.,
104, 621, 1986.
93. MacKinnon, S.L., Keifer, P. and Ayer, W.A., Components from the phytotoxic extract of Alternaria
brassicicola, a black spot pathogen of canola, Phytochemistry, 51, 215, 1999.
94. Otani, H. et al., Production of a host-specific toxin by germinating spores of Alternaria brassicicola,
Physiol. Mol. Plant Pathol., 52, 285, 1998.
31
Aspergillus
CONTENTS
31.1 Introduction....................................................................................................................................455
31.2 The Genus Aspergillus...................................................................................................................455
31.3 Mycotoxins of Aspergillus Species and Mycotoxicoses............................................................... 456
31.3.1 Aflatoxins......................................................................................................................... 456
31.3.2 Ochratoxins...................................................................................................................... 457
31.3.3 Fumonisins....................................................................................................................... 458
31.3.4 Patulin............................................................................................................................... 458
31.3.5 Other Mycotoxins Produced by Aspergilli...................................................................... 458
31.4 Laboratory Models for Foodborne Aspergillus Mycotoxicoses................................................... 459
31.4.1 Invertebrate Models.......................................................................................................... 459
31.4.2 Rodent Models.................................................................................................................. 460
31.4.3 Human and Animal Cell Lines........................................................................................ 479
31.5 Conclusions................................................................................................................................... 487
Acknowledgments................................................................................................................................... 487
References............................................................................................................................................... 487
31.1 Introduction
Members of the genus Aspergillus are well-known for their detrimental effects on human and animal
health. Invasive and superficial infections caused by Aspergillus species are more and more frequently
reported, and the genus is on the top of the list of filamentous fungal genera of clinical importance.
However, clinical infections due to Aspergilli are rarely foodborne. On the other hand, the ability of cer-
tain members of the genus to produce various mycotoxins frequently results in serious health problems,
both in animals and humans, that are commonly known as Aspergillus mycotoxicoses.
This chapter aims to give an overview about the laboratory models used to study Aspergillus myco-
toxicoses, with a special focus on the most recent publications applying rodent models (rats and mice)
and mammalian cell lines, the application of which proved especially fruitful to gain more insight into
the toxic and carcinogenic effects of AFB1, OTA, and other Aspergillus mycotoxins.
455
456 Laboratory Models for Foodborne Infections
characteristic of the genus reminded him an aspergillum (holy water sprinkler) used in Catholic liturgy.
These fungi were later recognized as active decomposers and also as the causal agents of diseases in
animals and humans. Today, Aspergillus is considered one of the most economically important and most
widely distributed filamentous fungal genera on our planet [1]. The genus Aspergillus is taxonomically
divided into 8 subgenera and more than 20 sections consisting of about 300–350 species [2,3].
Aspergillus species can be both beneficial and harmful for mankind. The greatest economic ben-
efit of the genus is the ability to produce industrial enzymes (e.g., amylases, glycosidases, pectinases,
proteases), and organic acids (e.g., citric acid, gluconic acid, itaconic acid) [1]. Aspergillus oryzae,
A. sojae, and A. tamarii are known as the “koji moulds” and have been used for centuries in oriental
food fermentation processes for the production of soy sauce and sake [1]. Aspergilli can also produce
a series of secondary metabolites with useful pharmaceutical and biotechnological properties (e.g., the
cholesterol-lowering drug lovastatin produced by A. terreus and some other species). However, taxa of
this genus may also have serious negative impacts on animal and human health as they cause different
diseases called aspergilloses. The most common human pathogen from the genus is A. fumigatus, which
is responsible for more than 90% of both invasive and noninvasive human aspergilloses; however, other
species of the genus are also capable of causing infections [4]. Various fungal diseases like keratitis or
onychomycosis, allergic bronchopulmonary aspergillosis (ABPA), and asthma have been reported to be
caused by the growth or spores of Aspergillus species (among others). Additionally, Aspergilli produce a
range of mycotoxins, which can be harmful to animals and humans. Aspergillus species can contaminate
foods and feeds at different pre- and postharvest, as well as processing or handling stages. The most
economically important Aspergillus mycotoxins identified as contaminants in foods and feeds are the
aflatoxins, ochratoxins, patulin, and fumonisins.
AFs are hepatotoxic and hepatocarcinogenic and usually considered as belonging to the most
potent naturally occurring carcinogens [15,16]. The diseases caused by AF consumption are known
as aflatoxicoses. Acute aflatoxicosis is the result of moderate-to-high level AF consumption. Several
deaths were attributed to aflatoxicosis in tropical and subtropical regions of the world, primar-
ily due to the consumption of contaminated corn [17]. Chronic aflatoxicosis results from prolonged
ingestion of low-to-moderate levels of AFs and can be characterized by usually subclinical effects that
are difficult to recognize. Comprehensive studies indicated that AF is a risk factor for hepatocellular
carcinoma in humans, especially in Asia and the sub-Saharan part of Africa [18,19]. The International
Agency for Research on Cancer (IARC) classified AFs as group I category carcinogens [20].
Because of its toxicity, the content of AF is restricted in food and feed products in more than 100
countries [21].
31.3.2 Ochratoxins
Ochratoxins (OTs) were discovered in 1965 during a systematic examination of the metabolites of
moulds, including Aspergillus ochraceus [22]. Ochratoxin A (OTA) is a cyclic pentaketide dihydroiso-
coumarin derivative linked to an l-β-phenylalanine by an amide bond. The most toxic member of the
group is OTA which is a chlorinated pentaketide. Several OTA-derived metabolites have also been iden-
tified: OTB is a dechlorinated analogue of OTA, while OTC is a chlorinated ethyl ester derivative. OTα is
the isocoumarin core of OTA, while OTβ is a dechlorinated analogue of OTα. Other derivatives include
4-hydroxy-OTA, 10-hydroxy-OTA, OTA methyl ester, OTB ethyl and methyl esters, and several amino
acid analogues [23,24].
Several nephropathies affecting animals and humans can be attributed to OTA, including the Danish
porcine nephropathy as well as renal disorders detected in other animals [25]. In the case of humans, OTA
is often mentioned as the possible causative agent of the Balkan Endemic Nephropathy [26], although
recently aristolochic acid was suggested to play a major role in the etiology of this disease [27]. OTA also
proved to exhibit immunosuppressive, teratogenic, hepatotoxic, and carcinogenic properties [28]. The
IARC classified OTA as a possible human carcinogen in the Group 2B category [20].
Under cooler, temperate climates, OTs are mainly produced by Penicillium species (P. verrucosum
and P. nordicum), while in warmer and tropical regions they are produced by a series of Aspergillus
spp. Aspergillus isolates usually produce both OTA and its dechlorinated analogue OTB, while
Penicillium spp. produce OTA only. In Aspergillus section Circumdati, the recently described new
species A. affinis, A. fresenii, A. occultus, A. pulvericola, A. sesamicola [29,30], as well as A. cre-
tensis, some isolates of A. ochraceopetaliformis (formerly described as A. flocculosus), A. muricatus,
A. ochraceus, A. pseudoelegans, A. roseoglobulosus, A. steynii, and A. westerdijkiae are able to pro-
duce consistently high or medium amounts of OTA. Seven further species of the genus produce OTA
inconsistently or in trace amounts only, including A. ostianus, A. melleus, A. persii, A. salwaensis, A.
sclerotiorum, A. subramanianii, and A. westlandensis. The most relevant species for potential OTA
production in rice, coffee, beverages, and other foodstuffs are A. ochraceus, A. steynii, and A. west-
erdijkiae. In Aspergillus section Flavi, A. albertensis and A. alliaceus are capable of OTA production
in synthetic media [31]. Among the representatives of Aspergillus section Nigri, A. carbonarius, A.
niger, A. sclerotioniger, and A. welwitschiae proved to be potential OTA-producing fungi [32,33]. The
majority of A. carbonarius isolates can produce large amounts of OTA; however, different surveys
reported about only 5%–15% of A. niger and A. welwitschiae isolates producing OTA in smaller
quantities [33].
OTs can be detected in several agricultural products including cereals, cocoa, coffee beans, corn,
figs, grapes, peanut, red pepper, rice, soybeans, and spices [34]. A. westerdijkiae proved to be impor-
tant in the OTA contamination of coffee beans in Thailand [35], while A. alliaceus was found respon-
sible for the OTA contamination of figs in California [36]. Members of Aspergillus section Nigri,
primarily A. carbonarius, are responsible for the OTA contamination of grapes and grape-derived
products [37].
458 Laboratory Models for Foodborne Infections
31.3.3 Fumonisins
Several species of the genera Aspergillus and Fusarium (e.g., Fusarium verticillioides) are able to
produce fumonisins (for details, see Chapter 34 on Fusarium). Sequencing the genome of Aspergillus
niger resulted in the discovery of a region homologous with the fumonisin gene cluster of Fusaria
[38]. The production of fumonisins by A. niger was confirmed later [39], while recent studies revealed
that another black Aspergillus species, A. welwitschiae can also produce fumonisins [33]. Black
Aspergilli produce mainly fumonisins B2 and B4, but other analogues have also been identified in
smaller quantities [40].
Black Aspergilli and fusaria are both among the saprotrophic fungi that are widely distributed. Among
the fumonisin-producing Aspergillus species, A. niger is an important opportunistic pathogen of grapes,
causing raisin mould and bunch or berry rot [41]. It is also able to infect apples, corn, mangoes, onions,
peanuts, and dried meat products [42]. In contrast to fusaria, which produces large amounts of fumoni-
sins in plant-extract- (barley, carrots, malt, oat, potatoes, rice)-containing media, Aspergilli produce
fumonisins in high quantities on substrates with low water activities [39].
31.3.4 Patulin
The water-soluble lactone patulin (PAT; 4-hydroxy-4H-furo[3,2-c]pyran-2(6H)-one) is produced by
many fungi via the polyketide pathway. It was first isolated in 1943 from Penicillium species during a
screening effort aimed at finding novel antibiotics from fungi [43]. As it was discovered independently
in multiple laboratories, various names (e.g., clavacin, expansine, claviformin, clavatin, gigantic acid,
myocin C) were given [44]. PAT was the first compound which was examined under the brand name
Tercinin in a controlled clinical trial to treat common cold [45]; however, it proved to be unsuccessful,
furthermore, even toxic to animals and humans [46].
Although PAT has been reported from up to 30 fungal genera [47], recent studies revealed that it is
produced only by a limited number of species from the genera Aspergillus, Byssochlamys, Paecilomyces,
and Penicillium. According to our recent knowledge, within the genus Aspergillus only three represen-
tatives of section Clavati (A. clavatus, A. giganteus, and A. longivesica) are capable of producing PAT
[48]. The other PAT-producing Aspergilli are rare species and cannot be considered as health hazards in
foods and feeds.
PAT provokes congestion and edema of hepatic, pulmonary, and gastrointestinal blood vessels and
tissues. Regarding its acute toxicity, PAT mainly induces gastrointestinal disorders with distension,
ulceration, and bleeding [49]. It was suggested to be partly responsible for a series of animal intoxications
in France, Germany, Hungary, and Japan [50]. Regarding the carcinogenicity of PAT, its subcutaneous
injection produced local sarcomas in rats [49].
as well as Monascus species are also known producers of this nephrotoxic compound [52]. Citrinin is a
common contaminant of grains, food, and feedstuffs (e.g., corn, oats, wheat, wheat bran, fruit juices) [57].
If ingested by animals and humans, citrinin can cause chronic diseases.
Several further Aspergillus mycotoxins could also be detected in foods and feeds [16]. The extensive
genome sequencing efforts in Aspergilli resulted in the recent clarification of the molecular background
behind the production of a series of Aspergillus mycotoxins (e.g., gliotoxin [58], kojic acid [59], naph-
thopyrones, asperfuranone, and orsellinic acid [60]). However, further studies are needed to clarify the
possible health effects of these mycotoxins on animals and humans.
CYP-nicotinamide adenine dinucleotide phosphate reductase gene) proved to be more resistant to the
growth-inhibitory effect of AFB1 exposure than N2. The results indicated that C. elegans can metabolize
AFB1 into DNA-binding metabolites in a CYP-dependent manner. González-Hunt et al. [82] examined
the effects of several known toxins including AFB1 on C. elegans and found more damage caused to the
mitochondrial than to the nuclear DNA, and AFB1 also caused dopaminergic neurodegeneration, which
supports the role for mitochondrial DNA damage in this process.
31.4.2 Rodent Models
As most popular laboratory animal models for the examination of Aspergillus mycotoxins are rodents
(principally rats and mice), a broad search strategy was used to collect full text articles and—in cases
when full text access was not available—abstracts from the PubMed database. The search was performed
in article titles and abstracts using the following keyword combinations in order to find specific litera-
ture: aflatoxin OR ochratoxin OR sterigmatocystin OR patulin AND rat OR mouse OR mice. (Although
certain Aspergillus species are known to produce fumonisins as well, FB1 and related compounds are
discussed in Chapter 34: Fusarium of this book.) The search was restricted to papers published in and
after 2010 in order to focus on recent literature. Articles and abstracts were screened to include studies
with relevant information (exact definition of the rodent model and the mycotoxin studied, as well as
details about the dosage and the way of administration).
Table 31.1 summarizes the studies published since 2010 that have used rat models to study
Aspergillus mycotoxins [83–119]. Among the retrieved records, relevant data were found in 37 publi-
cations. The most popular rat models in these studies were Fisher rats (12 studies), Sprague-Dawley
rats (12 studies), and Wistar rats (11 studies), but other rat lines (Charles Foster rats, Dark Agouti
rats, and hybrids of Sprague-Dawley female rats and a Fisher male rat) were also applied in single
studies. Male rats were used in the majority of the studies, with a few exceptions applying both
male and female, or just female animals. The age of the model animals ranged from 28 days to 15
months. The most commonly studied Aspergillus mycotoxin in rat models was AFB1 (22 studies),
followed by OTA (14 studies), AFG1, and STC (1–1 study). Depending on the aim of the particular
study, the Aspergillus mycotoxins were administered through diet, oral or gastric gavage, intraperi-
toneally, intramuscularly, intratracheally, intravenously, or subcutaneously. The majority of these
studies examined AFB1-induced hepatotoxicity [96,99,102,109,114,115] and hepatocellular carci-
noma development [90,101,104,118], as well as the protective effects of different compounds against
them [83,88,89,92,105,107,108]. In the case of OTA, many studies focused on its renal accumula-
tion, toxicity, and induction of kidney failure [84,86,93,95,96,103,112]. AFG1 was examined for lung
toxicity in male Sprague-Dawley rats [110], while the potential link between STC and esophageal
cancer was studied in Wistar rats [87]. Supriya and Reddy [91] used female Wistar rats to examine
the effects of in utero exposure to graded doses of AFB1 on development, behavior, and reproduction
and found that AFB1 severely compromised postnatal development of neonatal rats, caused a delay in
the descent of testes, and a reduction in steroidogenesis and spermatogenesis that were accomplished
by suppressed reproduction at adulthood. Other studies examined compounds like ginseng extract
[108,116] and lycopene [106] for their protective effects against the genotoxicity of AFB1 and OTA,
respectively.
Table 31.2 summarizes the studies published since 2010 that have used mouse models to study
Aspergillus mycotoxins [120–174]. Among the retrieved records, relevant data were found in 55 pub-
lications. The most popular mouse model for studying Aspergillus mycotoxins is the albino, labora-
tory-bred BALB/c strain (16 studies), followed by the black inbred strain C57BL/6 and its mutant and
hybrid (e.g., B6C3F1) derivatives (8 studies), the largely noninbred Swiss albino mice (7 studies), and
ICR outbred mice (6 studies). Other mouse models used include Kunming, Swiss Webster (CFW),
MF1, CF-1, and A/J mice. Male animals were used in more studies; however, the use of females was
more frequent than in the case of rat models. The age of the treated mice ranged from newborns to
12-month-old animals. Similar to the studies using rat models, the most commonly studied Aspergillus
mycotoxin in mouse models was also AFB1, followed by OTA, PAT, AFM1, AFG1, and STC. The myco-
toxins were administered to the mice through diet, oral gavage, intraperitoneally, intratracheally, or
TABLE 31.1
Aspergillus
Studies Using Rat Models for the Examination of Aspergillus Mycotoxins Published Since 2010 Based on PubMed
Rat Model MT Dose/Administration Aim of the Study Major Findings References
Male Wistar AFB1 1.5 mg/kg intraperitoneally To study the protective effect of herbal ethanolic The plant extracts showed significant protection [83]
albino extracts of Ixora coccinea, Rhinacanthus nasuta, and against AFB1-induced liver damage by lowering
(200–250 g) Spilanthes ciliata against AFB1-induced the activity of serum enzymes and restoring the
hepatotoxicity decrease in GSH levels
Weaning male OTA 289 μg/kg/d given by gavages To demonstrate that OTA cytotoxicity can cause kidney Exposure to OTA induced significant lesions to [84]
Wistar albino through a stomach tube for 28 d failure the renal corpuscles, which can reduce the
(35–40 g) functional capacity of kidney and lead to kidney
failure
Male Wistar AFB1 0.24 mg/kg by gastric gavage for To investigate the power of conventional primary Conventional hepatocyte cultures mimicked the in [85]
Hannover up to 14 d hepatocyte cultures along with their epigenetically vivo induced changes, which may be suitable for
stabilized counterparts in mimicking in vivo genotoxic the identification of genotoxic carcinogens that
insults do not require extrahepatic activation
Male Wistar OTA 3 mg/kg/d gavage feed To analyze the effect of OTA on three human and two The carcinogenic effects in three human cell [86]
rat renal proximal tubular models models were significantly reflected in a rat
in vivo model and partially in two rat in vitro
models, which clearly evidenced the lack of
species specificity between humans and rats in
the biological perturbations caused by OTA
4-w-old male STC 30 μg/kg intraperitoneally injected To investigate the effects of sterigmatocystin exposure STC exposure downregulated TAP1 and LMP2, [87]
Wistar in rats with reflux esophagitis which directly affected the tumor immunity by
(40–50 g) allowing transformed cells to escape the host
immune surveillance, thereby promoting
esophageal cancer
10–12 w old AFB1 3 mg/kg b.w. intraperitoneally To evaluate whether antioxidant CAPE relieves CAPE induced protective effects on the [88]
male Wistar oxidative stress in AFB1-induced liver injury AFB1-induced hepatotoxicity by modulating free
albino (200 ± radical production, biochemical values, and
20 g) histopathological alterations
2-m-old male AFB1 20 μg/d through gavage for 3 w To investigate the effect of Dialium guineense pulp Free and bound phenolic extract of D. guineense [89]
Wistar albino phenolic extract on AFB1-induced oxidative fruit enhanced the detoxification of AFB1
(150 ± 2.54 g) imbalance in rat liver
Wistar AFB1 100 and 200 μg/kg To explore the effect of EGb on expressions of Cox-2 EGb can regulate the expression of GST-Pi, but it [90]
and GST-Pi in the pathogenesis of HCC does not seem to have an effect on Cox-2
expression in the liver of rats with the risk of HCC
461
(Continued)
TABLE 31.1 (Continued)
462
Studies Using Rat Models for the Examination of Aspergillus Mycotoxins Published Since 2010 Based on PubMed
Rat Model MT Dose/Administration Aim of the Study Major Findings References
Female Wistar AFB1 10, 20, or 50 μg/kg b.w. daily from To examine the embryonic exposure to graded doses of In utero exposure to AFB1 severely compromised [91]
(210–220 g) gestation d 12–19 AFB1 on development, behavior, and reproduction postnatal development of neonatal rats and
intramuscularly caused a delay in testes descent and reduction in
steroidogenesis and spermatogenesis that were
accomplished by suppressed reproduction at
adulthood
3-w-old male AFB1 200 μg/kg b.w. intraperitoneally To investigate genotoxic and antigenotoxic effects of PRBE significantly reduced micronucleus [92]
Wistar (approx. PRBE in rats using liver micronucleus assay formation and mitotic index in the liver of
70 g) AFB1-initiated rats
16-w-old male OTA Fed diets with 3 and 5 μg/g b.w. for To measure OTA concentration in whole kidneys of rats Apparent accumulation of OTA in kidney is due [93]
and female females and males, respectively exposed chronically to dietary toxin to binding to plasma proteins and long half-life
Wistar in plasma
Male Fischer OTA Daily dietary intake of 50 mg/kg To compare the carcinogenic response between the Carcinoma incidence (4/34 rats) through dietary [94]
344 b.w. for up to 2 y NTP carcinogenic gavage dose regimen and exposure is considerably less than in the NTP
administration of the same dosage via artificially study (16/51)
contaminated diet
15-m-old male OTA 6.25 mg by oral gavage To evaluate the urinary metabolomic profile through 1H-NMR metabolomics revealed a progressive [95]
Fischer (445, 1H-NMR spectra cyclic shift in principal components data cluster
Aspergillus
Studies Using Rat Models for the Examination of Aspergillus Mycotoxins Published Since 2010 Based on PubMed
Rat Model MT Dose/Administration Aim of the Study Major Findings References
Male F344/N AFB1 NTP 2000 feed 1 ppm To check whether a subset of genes from a previously Evaluation of gene signatures in archival tissues [98]
derived AFB1 gene signature would be observed in can be an important toxicological tool for
archival RNA from fresh frozen liver, be replicated in evaluating critical molecular events associated
FFPE liver RNA, and also be present in other FFPE with chemical exposures
tissues like the kidney and lung as secondary or
alternate targets for carcinoma
Male F344 AFB1 0.25, 0.75, and 1.5 mg/kg b.w. by To identify mechanisms and potential biomarkers for p53 signaling pathway induced by oxidative [99]
(80–120 g) oral gavage predicting the development and progression of damage was the crucial step in AFB1-induced
AFB1-induced acute hepatotoxicity through integrated acute hepatotoxicity, whereas gluconeogenesis
analysis of general toxicity studies, transcriptomics, and lipid metabolism disorder were found to be
and metabolomics profiles the major metabolic effects after acute AFB1
exposure
6–7-w-old male OTA 0, 70 or 210 μg/kg b.w. gavage To explore the relationship between OTA-induced OTA caused apparent kidney damage within 13 w [100]
F344 oxidative damage and carcinogenicity in the liver and but exerted limited effect on oxidative stress
kidney parameters
Male F344 AFB1 1.5 mg/kg b.w. by gavages To elucidate the functional complexity of microRNAs Abnormally expressed cancer-related miRNAs [101]
in AFB1-induced hepatocellular tumorigenesis were identified
3-w-old (newly AFB1 Diets containing 0, 1, 5, 10, or To develop an experimental animal model of dietary AFB1-exposed animals exhibited dose-dependent [102]
weaned) male 20 ppm aflatoxin exposure to investigate the relationship wasting and stunting, liver pathology, and
inbred F344 or between AFB1 toxin exposure and growth disturbance suppression of hepatic targets of growth
outbred and to explore candidate mechanisms responsible for hormone signaling
Sprague- this association
Dawley
6–7 w-old male OTA 0, 21, 70, or 210 μg/kg b.w. by oral To evaluate a range of novel biomarkers of renal The histopathological alterations induced by OTA [103]
F344/N gavage toxicity to test their ability in detecting acute and were best reflected by changes in urinary Kim-1
chronic kidney injury marker
(Continued)
463
TABLE 31.1 (Continued)
464
Studies Using Rat Models for the Examination of Aspergillus Mycotoxins Published Since 2010 Based on PubMed
Rat Model MT Dose/Administration Aim of the Study Major Findings References
Male F344/N AFB1 1 ppm in feed for 90 d To define gene expression changes that might relate to 49 novel, differentially-expressed transcripts [104]
carcinogenesis produced by AFB1 exposure prior to including two unique, unannotated, hepatic
onset of malignancy and to begin a high-resolution AFB1-responsive transcripts overexpressed by
map of the rat liver transcriptome 10- to 25-fold were identified
Male 344/NHsd AFB1 200 μg/kg b.w. via oral gavage To assess and characterize the chemoprotective efficacy CDDO-Im completely protected against [105]
daily for 4 w of synthetic oleanane triterpenoid CDDO-Im against AFB1-induced liver cancer compared to a 96%
AFB1-induced HCC incidence in AFB1 group. With CDDO-Im
treatment, integrated level of urinary AFB1-N7-
guanine was significantly reduced and
aflatoxin-NAC, a detoxification product, was
consistently elevated after the first AFB1 dose.
Also, the hepatic burden of GST-P-positive foci
and the toxicogenomic RNA expression
signature were largely absent with CDDO-Im
intervention
Male Sprague- OTA 2.2 mg/kg b.w. orally by gastric To demonstrate the effects of free radical scavengers OTA administration caused oxidative damage, [106]
Dawley gavage MEL and CoQ10 on cells damaged by OTA and MEL or CoQ10 treatment ameliorated the
(230–250 g) OTA-induced tissue injuries
Aspergillus
Studies Using Rat Models for the Examination of Aspergillus Mycotoxins Published Since 2010 Based on PubMed
Rat Model MT Dose/Administration Aim of the Study Major Findings References
Male Sprague- AFG1 30 μg/kg b.w. intratracheally To identify the acute toxicity of AFG1 in AT-II cells AFG1 induces structural and functional [110]
Dawley instilled impairment in AT-II cells involved in the acute
(110–130 g, toxicity of AFG1 in lung tissues
120.2 ± 5.9 g)
Male Sprague- OTA 0.2 mg/kg b.w. single dose by oral To develop combinatorial platform of LC–MS/MS and LC–MS/MS method determined OTA with a run [111]
Dawley (200 ± gavage LC–TOF-MS to investigate in vivo kinetics and time of 7.0 min. Simultaneously, an LC–
20 g) biotransformation of OTA in rats TOF-MS method unambiguously identified the
metabolites of OTA in a total run time of 14 min
Male Sprague- OTA 0.5 mg/kg b.w. for 14 d by To investigate the possible protective effect of lycopene Lycopene might be partially protective against [112]
Dawley intragastric lavage against the renal toxic effects of OTA OTA-induced nephrotoxicity and oxidative stress
(<200 g) as it increases GPx1 activity and GSH levels and
decreases apoptotic cell death
12-w-old male OTA 0.5 mg/kg/d by intragastric lavage To investigate the protective effects of lycopene against Lycopene provided a protective effect against [113]
Sprague- for 7 and 14 d the genotoxicity of OTA in rat tissues using the OTA-induced DNA damage, which was
Dawley alkaline comet assay evidenced by decreased tail moment and
(250–260 g) intensity in both rat kidney and liver cells
2-m-old AFB1 Fed diets with 4, 10, 25, and 50 μg/ To evaluate the toxicological impacts on rats fed with Various negative impacts on liver and kidney [114]
Sprague- kg b.w. AFB1-contaminated feedstuffs functions were observed in AFB1-contaminated
Dawley groups, which was positively correlated with
AFB1 concentrations
6–8-w-old male AFB1 0.3 mg/kg/b.w. oral or Transcriptome profiling of rat liver samples treated The resulting large toxicogenomics dataset can [115]
Sprague- intraperitoneal, intravenous or with 27 chemicals to evaluate the utility of RNA-Seq serve as a resource to characterize various
Dawley subcutaneous injection in safety assessment and toxicity mechanism aspects of transcriptomic changes (e.g.,
elucidation alternative splicing) that are byproducts of
chemical perturbation
3-m-old female AFB1 80 μg/kg b.w. AFB1 or/and 100 μg/ To evaluate the protective role of PGE against the PGE induced potential protective effects against [116]
Sprague- and kg b.w. FB1 orally synergistic effect of subchronic administration of the oxidative stress and genotoxicity of these
Dawley FB1 AFB1 and FB1 on DNA and gene expression in rat mycotoxins
(100–120 g)
(Continued)
465
TABLE 31.1 (Continued)
466
Studies Using Rat Models for the Examination of Aspergillus Mycotoxins Published Since 2010 Based on PubMed
Rat Model MT Dose/Administration Aim of the Study Major Findings References
Hybrids of OTA Dietary intake of 5 ppm OTA To explore whether the tumor promoter sodium Female mammary tumorigenesis was advanced [117]
Sprague- barbiturate could shorten the otherwise long latency by 6 ms in all rats given the OTA-barbiturate
Dawley female between exposure to OTA and tumorigenesis
and a F344 male
18- to 20-w-old AFB1 1 mg/kg b.w. of to the 18-h fasted To describe the histopathological progression of AFB1 toxicity activates the oxidative stress [118]
male Charles rats on 2 consecutive d AFB1-induced HCC in the liver of rats proinflammatory pathway, which induces
Foster albino intraperitoneally hepatocarcinogenesis
(200–220 g)
7-w-old dark OTA Fed with 2.5, 7, 40, and 100 μg/kg To examine OTA metabolism in the liver and kidney of The distribution of OTA in male and female rat [119]
Agouti wheat male and female rats using reduced GSH and NAC kidney is not significantly different, but OTA
conjugates along with nontoxic OTα as biomarkers metabolism is greater in male than female rats
d, day; w, week; m, month; AFB1, aflatoxin B1; AT-II, alveolar type II; CAPE, caffeic acid phenethyl ester; CDDO-Im, 1-[2-cyano-3-,12-dioxooleana-1,9(11)-dien-28-oyl]imidazole; CAT,
catalase; CoQ10, coenzyme Q10; Cox-2, cyclooxygenase-2; DNA, desoxyribonucleic acid; EgB, Ginkgo biloba extract; FB1, fumonisin B1; FFPE, formalin-fixed, paraffin-embedded;
GPx1, glutathione peroxidase 1; GSH, glutathione; GST, glutathione S-transferase; HCC, hepatocellular carcinoma; KGE, Korean ginseng extract; KRG, Korean red ginseng; LC–MS/MS,
liquid chromatography–mass spectrometry/mass spectrometry; LC–TOF-MS, liquid chromatography–time of flight-mass spectrometry; LMP2, low molecular weight protein 2; MEL,
melatonin; MT, mycotoxin; NAC, N-acetylcysteine; NMR, nuclear magnetic resonance; NTP, National Toxicology Program; OTA, ochratoxin A; PAT, patulin; PGE, Panas ginseng extract;
PRBE, purple rice bran extract; RNA, ribonucleic acid; SF, sulforaphane; SOD, superoxide dismutase; STC, sterigmatocystin; TAP1, transporter associated with antigen processing 1;
Aspergillus
Studies Using Mouse Models for the Examination of Aspergillus Mycotoxins Published Since 2010 Based on PubMed
Mouse Model MT Dose/Administration Aim of the Study Major Findings References
7-w-old male BALB/c OTA Single dose of 3.5, 7, 35, 70, 289, To test whether the acute exposure to OTA In utero exposure to OTA caused adducts in the [120]
(20 g ± 2 g) 578, 1056 μg/kg/b.w. by oral via food and via exposure in utero causes testicular DNA of male offspring that were
gavage, feed containing increasing adducts in testicular DNA and to test similar to DNA adducts observed in the
amount of OTA (0.5, 1.4, 8, 20 μg/ whether these lesions are identical to those kidney and testis of gavage-fed adults,
kg/b.w.) every day during 4 w, a that can be produced in the kidney and supporting a possible role for OTA in
single intraperitoneal injection of testis by the consumption of OTA testicular cancer
OTA (2.5 mg/kg) at gestation day 17
6- to 8-w-old female PAT 5–20 μg by gastric intubation on days To investigate the adjuvant activity of Patulin increased the allergic immune response [121]
BALB/c ByJ 0, 1, 13, 14, 17, and 18, orally on mycotoxins on allergic airway in mice by modulating the Th1/Th2 balance
days 18, 21, and 22, 100 ng inflammation via direct effects on IL-12 secretion in DCs
intranasally on day 22 and by inducing oxidative stress
4- to 6-w-old BALB/c AFB1 Single dose at 240 μg/kg b.w. once a To evaluate the role of HOC in the Oval cells have produced tumors in liver [122]
athymic, nu/nu, male nude week intraperitoneally and fed in development of hepatocellular carcinoma following transfection with hepatitis B virus
(18.0–22.0 g) the drinking water (400 μg/kg d), x gene (HBx) and treatment with AFB1
continuously for 16 w
Male BALB/c (20–25 g) AFB1 250 μg/kg b.w. intraperitoneally To evaluate the hepatoprotective effect of The treatment of CCE showed a total reduction [123]
CCE on AFB1-induced liver damage in of AFB1-induced oxidative damage markers
mice by measuring MDA level, protein and genotoxicity markers and decreased the
carbonyls generation, and expression of expressions of proapoptotic proteins p53 and
Hsp 70 and Hsp 27 in liver bax
6- to 8-w-old male BALB/c AF 625 μg/kg b.w./d given directly into To investigate the antioxidant effect of AF exhibited adverse effects on most of the [124]
the stomach through a gastric tube pumpkin seed oil against the oxidative- oxidative stress markers, but the
stress-inducing potential of aflatoxin administration of pumpkin seed oil
diminished aflatoxin-induced adverse effects
(Continued)
467
TABLE 31.2 (Continued)
468
Studies Using Mouse Models for the Examination of Aspergillus Mycotoxins Published Since 2010 Based on PubMed
Mouse Model MT Dose/Administration Aim of the Study Major Findings References
3-w-old specific pathogen- ST 3, 30, 300, and 3000 μg/kg To explore the short-term immunotoxic The proportion of CD8+ T cells was decreased [125]
free male BALB/c intraperitoneally effects of ST, specifically on FoxP3+ Tregs in the thymus in ST (3 μg/kg) group, while
and pDCs, by observing changes in that of CD4+ and CD8+ T cells was increased
number/expression of FoxP3+ Tregs, in the spleen in two treatment groups (3 and
pDCs, and CD4+, CD8+ T cells 30 μg/kg); the proportion of FoxP3+ Tregs
and FoxP3 expressions were all significantly
increased in mPBMCs, the thymus, and the
spleen. Importantly, the population of pDCs
significantly decreased in the thymus but
increased in the spleen, which is due to a
temporary immune response triggered by the
ST inhibition
BALB/c ST 3 mg/kg intraperitoneally To evaluate the putative effects of ST on the Downregulation of TNF-α, IL-6, and IL-12 [126]
expression of TNF-α, IL-6, and IL-12 at mRNA expression in mPBMCs and peritoneal
mRNA levels in mPBMCs and peritoneal macrophage cells was observed, and serum
macrophage cells and on the serum TNF-α TNF-α and IL-6 levels were also decreased
and IL-6 levels
4- to 6-w-old inbred female AFB1 0.1 mL at a daily dose of 30, 15, and To focus on the immunosuppressor activity AGE has increased the level of INF-γ and IL-4 [127]
Aspergillus
Studies Using Mouse Models for the Examination of Aspergillus Mycotoxins Published Since 2010 Based on PubMed
Mouse Model MT Dose/Administration Aim of the Study Major Findings References
Nude/BALB/c AFB1 1 × 107 cells injected subcutaneously To investigate the underlying mechanisms ATR mediated the DNA damage repair [129]
of AFB1-induced effects on ATR function response and subsequent neoplastic
and neoplastic transformation of the cells transformation in P40 B-2A13 cells
when B-2A13 cells were exposed to low
levels of AFB1 (0.1–10 nM)
10-w-old female BALB/c AFM1 100 mg/kg b.w orally (by gavage) To evaluate a new AFM1-binding/degrading The isolated Lactobacillus plantarum MON03 [130]
(21 ± 2 g) microorganism for biologic detoxification, (LP) was found to display significant binding
to examine its ability to degrade AFM1 in ability to AFM1 in PBS (93%) and was taken
liquid medium and to evaluate its potential for in vivo study, where administration of LP
for in vivo preventive effects against with AFM1 strongly reduced the cytotoxic/
AFM1-induced immunotoxicity and genotoxic adverse effects of AFM1
genotoxicity
4-w-old female BALB/c AFB1, 2.5, 5.0, and 5.0 mg/kg b.w. of AFB1, To assess the individual and combined toxic AFB1, ZEA, and DON induced liver injury and [131]
ZEA, ZEA, and DON, respectively, effects of AFB1, ZEA, and DON within the were associated with induced oxidative stress
DON administered orally via gavage liver and an upregulation of the apoptotic genes
needle Caspase-3 and Bax, along with a
downregulation of the antiapoptotic gene
Bcl-2; AFB1 + DON displayed synergistic
hepatotoxic effects, while AFB1 + ZEA
displayed antagonistic hepatotoxic effects
10-w-old female BALB/c AFB1 0.25 mg/kg b.w. AFB1 and 0.27 mg/kg To evaluate a new AFB1- and AFM1-binding/ Lactobacillus plantarum MON03 (LP) was [132]
and AFM1 administered by oral gavage degrading microorganism for biological isolated and displayed significant binding
AFM1 detoxification, examine its ability to ability to AFB1 and AFM1 in PBS (i.e., 82%
degrade AFB1 and AFM1 in liquid medium, and 89%, respectively); in vivo study was
and evaluate its potential for in vivo conducted where LP strongly reduced the
preventive effects against AFB1- and adverse effects of each mycotoxin
AFM1-induced immunomodulation
BALB/c AFG1 100 μg/kg body weight orally To understand the phenotypic alterations of Increased MHC-II expression in alveolar [133]
AT-II cells, which may provide new insight epithelium was observed and associated with
into the immune function of AT-II cells enhanced Treg infiltration in mouse lung
involved in AFG1-induced pulmonary tissues and also with AFG1-induced
tumorigenesis inflammation
(Continued)
469
TABLE 31.2 (Continued)
470
Studies Using Mouse Models for the Examination of Aspergillus Mycotoxins Published Since 2010 Based on PubMed
Mouse Model MT Dose/Administration Aim of the Study Major Findings References
Female BALB/c (18–20 g) AFG1 100 μg/kg b.w. orally using the To examine whether oral gavage of AFG1 Inflammatory responses were heightened in the [134]
gavage technique induces chronic lung inflammation and lung alveolar septum 3 and 6 months after
how it contributes to carcinogenesis oral administration of AFG1; 12 months later,
AFG1 induced alveolar epithelial hyperplasia
and adenocarcinoma; upregulation of NF-κB,
p-STAT3, and Cox-2 was also induced in lung
adenocarcinoma
8- to 12-w-old male OTA 0.85 mg/kg b.w. inoculated To investigate the protective effects of The tested polyphenols reduced the toxicity [135]
BALBb/c (20–25 g) intraperitoneally luteolin, ChlA, and CafA against caused by OTA on different target cells with
cyto-genotoxic effects caused by OTA good protective effect, ChlA being the
compound that showed the best effects
5- to 6-w-old female Swiss PAT Topical dose of 400 nmol To investigate the skin carcinogenic Topical application of PAT resulted in cell [136]
albino potential of topically applied PAT proliferation, which is mediated by
ROS-induced MAPKs signaling pathway,
leading to transcriptional activation of
downstream target proteins c-fos, c-jun, and
transcription factor NFκB, showing that PAT
has dermal tumorigenic potential
Aspergillus
Studies Using Mouse Models for the Examination of Aspergillus Mycotoxins Published Since 2010 Based on PubMed
Mouse Model MT Dose/Administration Aim of the Study Major Findings References
6- to 7-w-old female Swiss OTA Topical application of 25, 50, 100, To evaluate the effect of a single topical OTA has cell proliferative and tumor- [139]
albino (20 ± 3 g) and 200 nmol/0.2 mL acetone application of OTA at lower doses on skin promoting potential in mouse skin, which
epidermis and to evaluate the skin-tumor- involves EGFR-mediated MAPKs and Akt
promoting potential of OTA pathways along with NF-κB and AP-1
transcription factors and that cyclin-D1 and
Cox-2 are the target genes responsible for
tumor-promoting activity of OTA
Young male inbred Swiss OTA 50 μg and 100 μg in 0.2 mL olive oil/ To evaluate the ameliorative effects of an Administration of Emblica officinalis aqueous [140]
albino (30–33 g) animal/d for 45 d orally aqueous extract of Emblica officinalis on extract (2 mg/animal/d) and OTA caused a
OTA-induced lipid peroxidation in the significant amelioration in the OTA-induced
kidney and liver lipid peroxidation in liver and kidney
60-d-old male Swiss albino AFB1 66.6 μg/kg b.w./d orally by gavage To investigate the protective effect of Nephrotoxicity induced by AFB1 was [141]
(30–40 g) esculin against pro-oxidant AFB1-induced ameliorated by esculin, which is due to its
nephrotoxicity antioxidant, anticarcinogenic, and ROS-
scavenging properties
Male Swiss albino (30 ± AFB1 2 μg/30 g b.w orally To investigate the ability of the Tinospora T. cordifolia showed protection against [142]
5 g) cordifolia to scavenge free radicals AF-induced nephrotoxicity due to the
generated during aflatoxicosis presence of alkaloids such as a choline,
tinosporin, isocolumbin, palmatine,
tetrahydropalmatine, and magnoflorine
6-w-old male ICR AFB1 0.75 mg/kg b.w. orally To examine the biochemical mechanisms Quercetin does not directly protect against [143]
associated with the effects of quercetin on AFB1-mediated liver damage in vivo but
AFB1-mediated liver damage exerts a partial role in promoting antioxidative
defense systems and inhibiting lipid
peroxidation
6-w-old male ICR AFB1 0.75 mg/kg b.w. orally To investigate the biochemical mechanisms RCMF attenuates AFB1-mediated damage to [144]
of the RCMF effects on AFB1-mediated the liver, which is at least partially related to
liver damage the restoration of antioxidant defense systems
and an increase in AFB1–GSH conjugate
formation
4-w-old female ICR AF, Fed diets with 597 μg/kg AF, 729 μg/ To investigate the regulation of multiple Naturally contained mycotoxins are toxic in [145]
ZEN, kg ZEN, and 3.1 mg/kg DON mycotoxins on the oxidative stress vivo and able to induce oxidative stress
DON
471
(Continued)
TABLE 31.2 (Continued)
472
Studies Using Mouse Models for the Examination of Aspergillus Mycotoxins Published Since 2010 Based on PubMed
Mouse Model MT Dose/Administration Aim of the Study Major Findings References
ICR OTA 1, 5, or 10 μM To explore the cytotoxic effects exerted by In vitro exposure to OTA triggers apoptosis and [146]
OTA on the blastocyst stage of mouse retards early postimplantation development
embryos, on subsequent embryonic after transfer of embryos to host mice; OTA
attachment, on outgrowth in vitro, and induces apoptosis-mediated injury of mouse
following in vivo implantation via embryo blastocysts via ROS generation and promotes
transfer mitochondrion-dependent apoptotic signaling
processes that impair subsequent embryonic
development
4-w-old female ICR DON, Fed diets with 3.875 mg/kg DON, To investigate the effects of a mycotoxin- Mycotoxin-contaminated diet adversely [147]
ZEN, 1897 μg/kg ZEN, and 806 μg/kg AF contaminated diet on oocyte quality affected the developmental competence of
and ovaries and decreased the oocyte quality
AF
7- to 9-w-old male CD1 OTA For acute tests, 1 mg/kg b.w. and To evaluate OTA-degrading and detoxifying OTA was degraded efficiently by Cupriavidus [148]
10 mg/kg b.w. for 72 h; for chronic potential of Cupriavidus basilensis OR16 basilensis OR1; OTα did not display
test, 0.5 mg/kg b.w. for 21 d via oral strain nephrotoxic effects in vivo
gavage
Male Kunming (22 ± 2 g) PAT 1 mg/kg intraperitoneally To investigate PAT-induced hepatotoxicity GTP exerted antioxidative activity in reducing [149]
and genotoxicity and the antioxidant and hepatic ROS and TBARS level and increasing
Aspergillus
Studies Using Mouse Models for the Examination of Aspergillus Mycotoxins Published Since 2010 Based on PubMed
Mouse Model MT Dose/Administration Aim of the Study Major Findings References
Swiss Webster AFB1 — To express the putative proteins tGSTAs in Recombinant tGSTAs detoxified AFBO, [152]
an Escherichia coli heterologous system whereas their hepatic forms did not, which
and to characterize the functional implies that the hepatic forms of these
properties of products of the subunits with enzymes are silenced by one or more
respect to their enzymatic activities toward regulatory mechanisms
prototype GST substrates and toward
enzymatically generated AFBO.
9-w-old male CF-1 (28.5 ± PAT A single dose of 1.0, 2.5, or 3.75 mg/ To investigate the genotoxic effects of PAT induced DNA damage in the brain, liver, [153]
1.27 g) kg b.w. intraperitoneally patulin in multiple organs (brain, kidney, and kidneys and demonstrated a high
liver, and urinary bladder) using an in vivo correlation between decreased GSH content
comet assay and increased lipid peroxidation and DNA
damage, suggesting interrelationship between
the pro-oxidant and genotoxic effects of PAT;
pretreatment administration of N-acetyl-
cysteine reduced PAT-induced DNA damage
Female A/J (16–19 g) AFB1 50 mg/kg To examine the effect of in vivo treatment 2 h posttreatment, AFB1 increased 8-OHdG [154]
with a single tumorigenic dose of AFB1 on levels in mouse lung DNA, but did not alter
the formation and repair of oxidative DNA 8-OHdG levels in liver or 5-OHdC levels in
damage in lung and liver lung or liver; AFB1 treatment also increased
BER activity in mouse lung but did not affect
hepatic BER activity; levels of OGG1
immunoreactive protein were increased in
both lung and liver; the results are consistent
with oxidative DNA damage contributing to
the carcinogenicity of AFB1
Male MF1 (25–30 g) PAT Orally administered with 1 mL of To assess the biochemical and PAT-contaminated apple juice resulted in liver, [155]
apple juice contaminated with histopathological effects of PAT in apple kidney, and neurotoxicological effects
152.5 ppb of PAT for 6 w juice samples collected from different
outlets retailing in Jeddah, Kingdom of
Saudi Arabia
(Continued)
473
TABLE 31.2 (Continued)
474
Studies Using Mouse Models for the Examination of Aspergillus Mycotoxins Published Since 2010 Based on PubMed
Mouse Model MT Dose/Administration Aim of the Study Major Findings References
Sexually mature female OTA Single dose at 3.0 mg/kg b.w. To evaluate the possible regulatory effect Dlx5 is a target for OTA and the inhibition of [156]
C57BL/6 intraperitoneally induced by OTA administration during a its function, directly or indirectly, could be
critical moment of gestation on expression the cause of the observed differentiation
of some homeotic related genes in order to defects
understand the possible mechanism of OTA
in the induction of differentiation defects
16 timed-pregnant AFB1 7 μg/g body b.w. intraperitoneally To test whether specific intestinal bacteria Intestinal colonization by Helicobacter [157]
Helicobacter-free C3H/ promote liver cancer in chemical and viral hepaticus was sufficient to promote AF- and
HeN female, C57BL/6 transgenic mouse models Hepatitis C virus transgene-induced HCC;
FL-N/35 crossed with neither bacterial translocation to the liver nor
C3H/HeN induction of hepatitis was necessary
Gnmt−/− and wild-type AFB1 10 mg/kg of b.w. at 7 d of age and To test whether the Gnmt−/− mice are Liver tumor formation in AFB1-treated [158]
129/B6 (129sv X second treatment of 40 μg AFB1 per susceptible to AFB1 carcinogenesis and to Gnmt−/− mice occurred earlier than in solvent -
C57BL/6) mouse at 9 w of age test whether Gnmt deficiency may treated Gnmt−/− mice and AFB1-treated wild
intraperitoneally accelerate AFB1-induced liver type mice; Gnmt deficiency increased the
tumorigenesis susceptibility to AFB1 related HCC; 5
detoxification pathway-related genes: Cyp1a2,
Cyp3a44, Cyp2d22, Gsta4, and Abca8a were
Aspergillus
Studies Using Mouse Models for the Examination of Aspergillus Mycotoxins Published Since 2010 Based on PubMed
Mouse Model MT Dose/Administration Aim of the Study Major Findings References
AlbCre:Keap1flox/flox and AFB1 0.8 mg/kg b.w. orally To examine whether genetic or The inability to rescue GSTA3 knockout mice [160]
GSTA3 knockout mice pharmacologic activation of Nrf2 signaling from AFB1 genotoxicity through the Nrf2
were crossed to produce could rescue the hypersensitive GSTA3 transcriptional program indicated that Gsta3
compound knockout mouse from the genotoxicity of is unilaterally responsible for the
GSTA3:Alb:Cre:Keap1flox/ AFB1 detoxification of AF in mice
flox DKO
Male C57BL/6JOlaHsd AFB1 1 ppm, ∼0.15 mg/kg b.w./d via feed To characterize the potential genotoxic Supplementation with quercetin at ∼350 mg/kg [161]
for 7 d properties of quercetin in the small bw/d for 12 w in mice showed no
intestine as well as in the liver by upregulation of genotoxicity-related pathways
transcriptome analysis in order to provide in liver and small intestine
new additional information to evaluate the
safety of quercetin
HBsAg transgenic mice, AFB1 Single dose of 6 μg/g b.w. To explore the option of utilizing mouse The utility of these mouse models provided a [162]
C57BL/6J- intraperitoneally models to understand in a systematic and rich resource for the longitudinal analysis of
Tg(Alb1HBV)44Bri/J longitudinal manner the molecular molecular changes and biomarkers associated
were crossed to C57BL/6J pathways that are progressively with HCC
WT to obtain HBsAg and deregulated by various etiological factors
WT in contributing to HCC formation and
report the initial findings in characterizing
their validity
4-d-old B6C3F1 AFB1 Single dose of 6 mg/kg AFB1 on d 4 To assess the capability of AFB1 to generate After 3 w, 10-fold increase in the Spi− mutant [163]
after birth via intraperitoneal DNA sequence changes including those fraction (MF) was observed and after 10 w, a
injection driven by nonhomologous recombination further increase was observed; the MF in the
in vivo by analyzing the Spi− (Sensitive to gpt gene was also increased at 10 w compared
P2 Inhibition) phenotype resulting from to 3 w; no gender-specific differences were
the disruption of red and gam genes in the found in the Spi− or gpt MFs; the Spi−
λEG10 transgene of gpt delta B6C3F1 spectrum was dominated by GC to TA
mice transversions, with one exceptionally strong
hotspot at position 314, which is the dominant
mutation seen in people exposed to AF
(Continued)
475
TABLE 31.2 (Continued)
476
Studies Using Mouse Models for the Examination of Aspergillus Mycotoxins Published Since 2010 Based on PubMed
Mouse Model MT Dose/Administration Aim of the Study Major Findings References
4-w-old male ICR mice AFB1 50 μg/kg b.w. via intraperitoneal To determine changes in testicular gene Differential expression of genes was noted [164]
(N 32) injection daily for 45 d expression due to exposure to AFB1 and to during cell differentiation, and extracellular
investigate which cell types were affected space was observed in the testis; renin was
by treatment with AFB1 commonly represented amongst many
clusters and chosen for further analyses; renin
was upregulated in response to AFB1,
especially in males that maintained fertility
despite AFB1 treatment
7- to 9-w-old male AFB1 Fed diets with 0, 0.2 or 1.0 ppm AFB1 To determine if chronic exposure of mice to Chronic exposure to AFB1 increased NER [165]
heterozygous p53 low levels of AFB1 alters NER in lung and activity in wild-type mice, and this response
knockout mice (p53 (+/−), liver and to determine if such alterations was diminished in heterozygous p53-
B6.129-Trp53tm1BrdN5) are affected in p53-deficient mice knockout mice, indicating that loss of one
and wild type controls allele of p53 limits the ability of NER to be
(p53 (+/+)) upregulated in response to DNA damage
5- to 7-w-old male OTA Fed diets containing 1, 15, or 40 mg To investigate OTA’s mode of action (MOA) OTA induced renal damage, but no tumors [166]
p53+/− (P53N5-T) and OTA/kg (w 1 and 2) and 0.5, 2, or in p53 heterozygous (p53 +/− ) and p53 were observed in either strain, indicating that
p53 +/+ (P53N5-W) 10 mg OTA/kg (w 3–26) homozygous (p53 +/+) mouse p53 heterozygosity conferred little additional
sensitivity to OTA; the lowest observed effect
Aspergillus
Studies Using Mouse Models for the Examination of Aspergillus Mycotoxins Published Since 2010 Based on PubMed
Mouse Model MT Dose/Administration Aim of the Study Major Findings References
10-w-old male p53- OTA 5 mg/5 mL/kg orally To investigate the role of p53 in the OTA induced DSBs at the carcinogenic target [167]
proficient and p53- progression from OTA-induced DNA site in the kidneys; p53 prevented formation
deficient mice and damage to gene mutations of DSBs and following gene mutation, due to
p53-proficient and p53/p21-mediated cell cycle control
p53-deficient gpt delta
mice (acquired by
intercrossing between
p53-deficient mice, gpt
delta transgenic mice and
C57BL/6 mice)
7- to 9-w-old male AFB1 Fed diets with 0, 0.2, or 1.0 ppm To determine if chronic exposure of mice to Chronic exposure to AFB1 did not affect BER [168]
heterozygous p53 AFB1 alters BER in lung and liver and to in lungs or livers of heterozygous p53-
knockouts and their determine if p53 heterozygosity affects knockout mice. BER activity was lower in
wild-type controls BER activity livers from p53 wild-type mice exposed to
(B6.129-Trp53tm1Brd N5) 1.0 ppm AFB1 versus those exposed to 0.2 ppm
AFB1, an effect that was not attributable to
liver cell death or altered levels of OGG1
Male and female FVB AFB1 20 μg/kg b.w. diet To develope transgenic mice specifically Transgenic mice specifically expressing ADTZ [169]
expressing ADTZ in parotid glands by gene in the parotid gland were successfully
transgenic technology toward AFB1 developed, and the ADTZ produced in the
detoxification transgenic mice was shown to reduce the
incidence of AFB1 sepsis
7- to 10-w-old 129/ AFB1 50 mg/kg b.w. intraperitoneally To determine the effect of ogg1 deficiency ogg1 status did not have a significant effect on [170]
C57Bl/6 female on AFB1-induced oxidatively damaged AFB1-induced oxidatively damaged DNA or
wild-type, heterozygous DNA and tumorigenesis tumorigenesis, but deletion of one or both
(+/−) or homozygous alleles of ogg1 did increase susceptibility to
(−/−) ogg1 gene mice other aspects of AFB1 toxicity
Adult male albino mice AFB1 9 mg/kg b.w. orally To investigate the effects of AFB1 and AFB1 and ethanol coexposure induced severe [171]
(19.75 + 2.0 g) ethanol coexposure on biomarkers of oxidative damage to the liver; thus, humans
hepatic damage consuming excessive amount of ethanol and
diets contaminated with AFB1 simultaneously
may be at greater risk of the hepatotoxic
effects of these compounds
477
(Continued)
TABLE 31.2 (Continued)
478
Studies Using Mouse Models for the Examination of Aspergillus Mycotoxins Published Since 2010 Based on PubMed
Mouse Model MT Dose/Administration Aim of the Study Major Findings References
ICR female mice (18–22 g) AF, Fed with low-dose mycotoxin- To identify any epigenetic effects of a Diets containing mycotoxins increased the [172]
ZEA containing diet of DON (581 μg/kg), mycotoxin-containing diet, including levels of DNA methylation, H3K9me3, and
DON ZEA (285 μg/kg), and AF (121 μg/ altered DNA methylation, H3K9 H4K20me3 and decreased the levels of
kg); and fed with high-dose methylation, H3K27 methylation, and H3K27me3 and H4K20me2; these altered
mycotoxin-containing diet of DON H4K20 methylation on reduced oocyte epigenetic modifications may be one of the
(1,163 μg/kg), ZEA (569 μg/kg), and developmental competence reasons for the reduced oocyte developmental
AF (242 μg/kg) competence
Theiler outbred (TO) mice AFB1 20 mg/kg b.w. on GD 13 both To study the toxicokinetics of AF in Large amounts of AFB1 are easily and rapidly [173]
intraperitoneally and orally pregnant mice absorbed both from the gastrointestinal tract
and through the peritoneum
Pdn/Pdn (Polydactyly OTA 2 mg/kg b.w. intraperitoneally on day To investigate gender-dependent differences A high incidence of NTD in male Pdn/Pdn was [174]
Nagoya) mouse (Gli3Pdn) 7.5 of gestation in the incidence of NTD induced by OTA induced by OTA treatment, which might be
in the Pdn/Pdn mouse due to complicated altered gene expressions
among Gli3, Wnt7b, Wnt8b, Fez1, Barx1,
Lim1, Dmrt1, Igf1, Fog2, Dax1, and Sox9,
and in particular, upregulation and gender-
dependent difference in Barx1 and gender-
dependent difference in Sox9 gene
480
Studies Using Human and Animal Cell Lines for the Examination of Aspergillus Mycotoxins Published Since 2010 Based on PubMed
Applied
Applied Cell Line MT Concentration Aim of the Study Major Findings References
Huh7 human AFB1 2.5 and 1–10 μM To develope a stable cell line with constitutive expression A cell line, Huh7–1A2-I-E, with high expression [184]
hepatocellular of human CYP1A2 in Huh7 cells level of CYP1A2 was developed and evaluated for
carcinoma cells identification of CYP1A2 inhibitors and for studies
of cytotoxicity resulting from CYP-mediated drug
metabolism; cellular toxicity of AFB1 in Huh7–1A2-
I-E cells could be prevented by furafylline, a
CYP1A2 inhibitor
HepG2 human AFB1 10 μM To perform a screen of mycotoxins that pose a known AFB1 was able to activate PXR, a known regulator of [185]
hepatocellular AFM1 environmental threat to human and animal health for the liver xenobiotic metabolism, in human hepatocytes,
carcinoma cells AFG1 ability to activate PXR function in a human hepatocyte and it could upregulate the expression of PXR-
cell line dependent genes responsible for AFB1
biotransformation, including CYP3A4
HepG2 and AFB1 2.5 μM To investigate the effects of AFB1 on key elements in AFB1 has stimulated hepatoma cell migration through [186]
SMMC-7721 IGF-IR signaling pathway and the effects of AFB1 on IGF-IR/IRS2 axis
hepatoma cells and hepatoma cell migration
immortalized human
Chang liver cells
Aspergillus
Studies Using Human and Animal Cell Lines for the Examination of Aspergillus Mycotoxins Published Since 2010 Based on PubMed
Applied
Applied Cell Line MT Concentration Aim of the Study Major Findings References
HepaRG cells AFB1 1 and 5 mM To analyze the effects of HBV infection and replication on AFB1 exposure decreased the HBV replication, [190]
DNA damage by AFB1 and vice-versa in HepaRG, a cell whereas DNA damage by AFB1 and subsequent
line with hepatocyte-like morphology that metabolizes p53 induction was not affected by the presence of
AFB1 and supports HBV infection the virus; thus, in HepaRG cell line, AFB1 and
HBV do not cooperate to increase DNA damage
by AFB1
HepaRG cells AFB1 0.05, 0.1, 0.5, and To describe an improved methodology for the in vitro HepaRG hepatocytes exposed to various genotoxic [191]
1.0 μM micronucleus test using an enriched proportion of compounds requiring or not requiring
HepaRG hepatocyte-like cells and based on a complete bioactivation compared favorably with those
in situ assay, thereby avoiding cell detachment after reported in various other cell types; they supported
chemical exposure and allowing cell division after a the view that metabolically competent HepaRG
mitogenic stimulation cells have unique potential benefits for testing
genotoxic compounds using the in vitro
micronucleus assay
Human hepatocytes AFB1 0, 1, 2, and 3 μM To study the application of IdMOC in the definition of the The three model toxicants showed three distinct [192]
and 3T3-L1 mouse role of hepatic metabolism on the cytotoxicity of three cytotoxic profiles: TMX was cytotoxic to 3T3 cells
fibroblast cells model toxicants: CPA, AFB, and TMX via the in the absence of hepatocytes, with slightly lower
coculturing of the metabolically competent human cytotoxicity towards both 3T3 cells and hepatocytes;
hepatocytes with the metabolically incompetent mouse AFB was selectively toxic toward the human
3T3 fibroblasts hepatocytes and relatively noncytotoxic toward 3T3
cells; CPA cytotoxicity to the 3T3 cells was found to
be significantly enhanced by the presence of the
hepatocytes
Hep3B human STC 10−12–10−6 M To investigate the potential cytogenetic effects of STC, STC alone or in combination with OTA and/or CTN [193]
hepatocellular OTA OTA, and CTN alone or in combination, at pM to μM had a cytotoxic and cytogenetic potential even at
carcinoma cells CTN concentrations, on the human hepatocellular cancer cell picomolar concentrations on human hepatoma
line Hep3B cells in vitro
(Continued)
481
TABLE 31.3 (Continued)
482
Studies Using Human and Animal Cell Lines for the Examination of Aspergillus Mycotoxins Published Since 2010 Based on PubMed
Applied
Applied Cell Line MT Concentration Aim of the Study Major Findings References
H4IIE rat hepatoma AFB1 0.31, 0.63, 1.25, To evaluate the effects of mycotoxins alone or combined AFB1 and FB1, either alone or in combination, [194]
cells and spleen FB1 2.50, 5, 10, 20, on activation and expression of Cyp1A and its increased cyp1A transcription and Cyp1A activity,
cells from male 30, 40, and transcription factor Ahr in hepatoma cell line H4IIE and as well as upregulated the Ahr activity, but the
Wistar inbred rats 60 μM AFB1 rat spleen mononuclear cells response potency was significantly higher for the
and 1.56, 3.12, mixture, indicating the existence of an interaction
6.25, 12.50, 25, between both toxins
50, 100, 150,
200, and
300 μM FB1
RPTEC/TERT1 OTA 300 nM To characterize the metabolome of the human RPTEC/ Metabolic profiling of RPTEC/TERT1 cells could [195]
human renal TERT1 cell line and define the response over a 72-hour report the effect of chemical exposure on multiple
proximal tubular period to low-level exposure of several compounds that cellular pathways at low-level exposure and
epithelial cells affect kidney using NMR-based metabolic profiling produced different response profiles for the different
approach compounds, with a greater number of major
metabolic effects observed in the toxin-treated cells
RPTEC/TERT1 OTA 300 nM To investigate the effects of several renal chemical After exposure to two renal carcinogens (OTA and [196]
human renal carcinogens on primary cilium in proximal tubular KBrO3), RPTEC/TERT1 cells lost primary cilium;
Aspergillus
Studies Using Human and Animal Cell Lines for the Examination of Aspergillus Mycotoxins Published Since 2010 Based on PubMed
Applied
Applied Cell Line MT Concentration Aim of the Study Major Findings References
IHKE immortalized OTA 0.01 nM–50 μM To confirm the proposed structure of OTα amide and OTα amide was less cytotoxic compared to OTA; [199]
human kidney cells OTα investigate if this compound can also be formed during thus, the thermal degradation of OTA to OTα
thermal processing, as it is commonly used in food amide might be a detoxification process; complete
technology structure elucidation of OTα amide was performed
via NMR/MS
Vero African green OTA 10, 20, 30, 40, To investigate the cytoprotective effect of quercetin on Quercetin pretreatment prevented the OTA-induced [200]
monkey kidney cells and 50 μM OTA-induced toxicity, with specific reference to oxidative stress and apoptosis in Vero cell line
oxidative stress, intracellular calcium flux, and levels of
protective antioxidant enzymes
J774A.1 murine AFB1 0.01 and To study the effect of AFB1, AFB2, and AFG1 exposure, Exposure of macrophages to low doses of AF [201]
macrophage cells AFB2 0.1 ng/mL alone and in combination, on the secretion of key resulted in a statistically significant change in the
AFG1 pro- and anti-inflammatory cytokines from J774A.1 cells secretion of a number of cytokines; AF exposure
affected expression levels of key cell surface
markers involved in the inflammatory response
Jurkat human OTA 10 μM To gain insights into the molecular mechanisms Diverse modes of action are involved in direct [202]
lymphoblastic underlying direct immunotoxicity immunotoxicity, and a set of pathways or genes,
T-cells rather than one single gene, could be used to
screen compounds for direct immunotoxicity
Jurkat human AFB1 3, 15, 30, and To investigate effects of AFB1 and AFM1 on immune Both AFB1 and AFM1 produced significant [203]
lymphoblastic AFM1 50 μM function decreases in cell proliferation, whereas only minor
T-cells effects were noted on IL-2 and IFN-γ cytokines
mRNA expression; AFB1, but not AFM1, at the
highest concentration induced a marked increase
in IL-8 mRNA expression
(Continued)
483
TABLE 31.3 (Continued)
484
Studies Using Human and Animal Cell Lines for the Examination of Aspergillus Mycotoxins Published Since 2010 Based on PubMed
Applied
Applied Cell Line MT Concentration Aim of the Study Major Findings References
Caco-2 human OTA 1 ng/mL, To improve the assessment of the toxicological impact of Induction of CYP1A1 activity and inhibition of [204]
colorectal 1.6–100 μg/mL, dietary contaminants (OTA , DON, IMA, and BEN) at CYP3A4 activity occurred in Caco-2 cells at a
adenocarcinoma 4, 40, 400, realistic human exposure levels, with a special focus on realistic intestinal concentration of IMA, while
cells 4,000, and the intestinal compartment OTA, DON, and BEN had no effect; some
H4IIE rat hepatoma 40,000 ng/mL bacterial stress genes were induced in a range of
cells, HepG2-Luc realistic concentrations, following exposure to
human hepatoma DON and IMA; BEN clearly provoked an
cells, and MMV-Luc estrogenic receptor agonistic activity in a human
human breast cancer estrogen-sensitive reporter cell line
cells
HCT116, SW620, PAT 1.25, 2.5, 5, 10, To study the effect of PAT on cancer cells and study its A new anticancer mechanism of PAT, suggesting that [205]
and Caco-2 human and 20 μM intracellular mechanism EGR-1 phosphorylation by oxidative stress
colorectal cells following PAT treatment might trigger EGR-1
binding to ATF3 promoter, which leads to the
expression of apoptotic proteins and cell growth
arrest of HCT-116 cells
Caco-2 human AFB1, OTA AFB1: 0.0038 ng/ To evaluate the effects of low levels of each mycotoxin in Individual toxicity showed that OTA was the most [206]
Aspergillus
Studies Using Human and Animal Cell Lines for the Examination of Aspergillus Mycotoxins Published Since 2010 Based on PubMed
Applied
Applied Cell Line MT Concentration Aim of the Study Major Findings References
SAOS-2 human AFB1 5 and 50 ng/mL To describe an unexpected toxicity of AFB1 toward the The expression of vitamin D receptor in [208]
osteosarcoma cells vitamin D receptors osteosarcoma cell line SAOS-2 is significantly
downmodulated by exposure to AFB1
CYP19- AFB1, 0.1% v/v To evaluate the effect of MTs (AFB1, zeranol, and ZEA) ZEA suppressed exon I.3 and II-specific expression [209]
overexpressing zeranol, ZEA on aromatase using 4 cell lines of aromatase and competitively inhibited the
MCF-7aro human enzyme activity; taken together, ZEA was a
breast cancer cells, potential aromatase inhibitor among the three MTs
JEG-3 human
placental cells,
MCF-7 human
breast
adenocarcinoma
cells, T98G human
Caucasian
glioblastoma cells
B-13 and B-13/H rat AFB1 100, 200, and To establish the utility of B-13 cells in toxicity and B-13 cell has generated hepatocyte-like cells with [210]
pancreatic 400 nM genotoxicity screening, CYP induction, susceptibility to functional drug metabolism and transporter
progenitor cells toxins, and transporter gene expression activities that could alone—or in a humanized
form—be used to screen for hepatotoxic and
genotoxic end points in vitro
A549 human lung AFG1 0.5, 2, 4, and To explore whether AFG1 activates the ROS/MAPK/ AFG1 induced the oxidative DNA damage and [211]
carcinoma cells 10 mg/L apoptosis pathway to cause cell damage in human AT-II triggered apoptosis through ROS-mediated JNK and
cells like the cell line A549 p38 signaling pathways in A549 cells, which
resulted in AFG1-induced AT-II cell damage
(Continued)
485
TABLE 31.3 (Continued)
486
Studies Using Human and Animal Cell Lines for the Examination of Aspergillus Mycotoxins Published Since 2010 Based on PubMed
Applied
Applied Cell Line MT Concentration Aim of the Study Major Findings References
INS-1 rat insulinoma PAT 0, 1, 10, 100, To analyze the effects of the microbial secondary Of the three compounds tested, PAT was the most [212]
cells 1,000, and metabolites pyrrolnitrin, phenazine, and PAT on INS-1 toxic; after both 24 and 72 h exposure,
10,000 ng/mL rat pancreatic β-cells concentrations ≥10 ng/mL caused significant cell
death with complete loss of viability at
10,000 ng/mL; PAT at 10 and 100 ng/mL caused a
significant reduction of insulin secretion per live cell
with 20 mM glucose; treatment of INS-1 cells with
PAT had no effect on insulin mRNA levels; PAT
showed no effect on the transcription of the insulin
gene at any concentration tested
NCTC clone L929 AFB1 100, 500, and To detect the antimutagenic and antiproliferative The isolated fractions obtained from octopus contain [213]
murine 1000 ng/plate compounds in octopus (Paraoctopus limaculatus) compounds with chemoprotective properties that
subcutaneous reduce the mutagenicity of AFB1 and proliferation
connective tissue of cancer cell lines
cells, and M12.
C3.F6 murine B cell
lymphoma cells
revealed that OTA causes significant deciliation in a model of the proximal tubule [196], that the
hypoxia inducible factor 2α (but not 1α) plays a crucial role in prevention of diminishment of vascular
endothelial growth factor production evoked by OTA in kidney proximal tubular epithelial cells [197],
and that with its high antioxidant potential, quercetin protects Vero cells from OTA-induced oxida-
tive stress and apoptosis [200]. The immunomodulatory or immunotoxic properties of Aspergillus
mycotoxins were studied on J774A.1 murine macrophages [201] and human T-cell lines [202,203].
Studies on human colon carcinoma cell lines (Caco-2, HCT116, and SW620) revealed important data
about the toxicity of AFB1, OTA, and PAT [204–206]; for the latter, a new anticancer mechanism
was proposed [205]. Further cell lines used since 2010 to study Aspergillus mycotoxins include Sp2/0
mouse myeloma cells and 2C10 and 2E6 mouse hybridoma cells [207]; SAOS-2 human osteosar-
coma cells [208], MCF-7aro human breast cancer cells, JEG-3 human placental cells, MCF-7 human
breast adenocarcinoma cells, and T98G human Caucasian glioblastoma cells [209]; B-13 and B-13/H
rat pancreatic progenitor cells [210]; A549 human lung carcinoma cells [211]; INS-1 rat insulinoma
cells [212]; as well as L929 murine subcutaneous connective tissue cells and M12.C3.F6 murine B cell
lymphoma cells [213].
31.5 Conclusions
Since 2010, the most popular laboratory models to study various aspects of Aspergillus mycotoxins were
animal models—primarily mice and rat lines—and human cell lines—mainly different liver and kid-
ney cells. Studies performed with the application of these laboratory models provided large amount of
important scientific information about the genotoxicity, organotoxicity, carcinogenicity, and metabolism
of Aspergillus mycotoxins, as well as about the protective, antioxidant, and immunostimulatory effects
of plant-derived metabolites, extracts, and oils against mycotoxicoses. The results from the studies dis-
cussed in this chapter clearly indicate that these laboratory models have the potential to further remain
in the frontline of mycotoxin research.
Acknowledgments
This work was supported by the Indian National Science Academy and the Hungarian Academy of
Sciences, as well as by projects NKFI-K115690 (National Research and Innovation Office, Hungary)
and GINOP-2.3.3-15-2016-00006 (Széchenyi 2020 Programme). Csaba Vágvölgyi thanks the visiting
professor program of King Saud University, Riyadh.
REFERENCES
1.
Raper, K.B., Fennell, D.I., The Genus Aspergillus, Williams and Wilkins, Baltimore, MD, 1965.
2.
Peterson, S.W., Phylogenetic analysis of Aspergillus species using DNA sequences from four loci,
Mycologia, 100, 205, 2008.
3.
Samson, R.A., Varga, J., What is a species in Aspergillus? Med. Mycol., 47, S13, 2009.
4.
Hope, W.W., Walsh, T.J., Denning, D.W., The invasive and saprophytic syndromes due to Aspergillus
spp., Med. Mycol., 43, S207, 2005.
5.
Blout, W.P., Turkey “X” disease, Turkeys, 9, 52, 1961.
6.
Van der Zijden, A.S.M., Blanche Koelensmid, W.A.A., Boldingh, J., Barrett, C.B., Ord, W.O., Philp, J.,
Aspergillus flavus and turkey X disease: Isolation in crystalline form of a toxin responsible for turkey X
disease, Nature, 195, 1060, 1962.
7.
Squire, R.A., Ranking animal carcinogens: A proposed regulatory approach, Science, 214, 877, 1981.
8.
Varga, J., Frisvad, J.C., Samson, R.A., A reappraisal of fungi producing aflatoxins, World Mycotoxin J.,
2, 263, 2009.
9.
Varga, J., Frisvad, J.C., Samson, R.A., Two new aflatoxin producing species, and an overview of
Aspergillus section Flavi, Stud. Mycol., 69, 57, 2011.
488 Laboratory Models for Foodborne Infections
10. Goncalves, S.S. et al., Aspergillus novoparasiticus: A new clinical species of the section Flavi, Med.
Mycol., 50, 152, 2012.
11. Rank, C. et al., Distribution of sterigmatocystin in filamentous fungi, Fungal Biol., 115, 406, 2011.
12. Olsen, M. et al., Aspergillus nomius, an important aflatoxin producer in Brazil nuts? World Mycotoxin
J., 1, 123, 2008.
13. Versilovskis, A., De Saeger, S., Sterigmatocystin: Occurrence in foodstuffs and analytical methods—
An overview, Mol. Nutr. Food Res., 54, 136, 2010.
14. Nielsen, K.F., Mycotoxin production by indoor molds, Fungal Genet. Biol., 39, 103, 2003.
15. Chu, F.S., Mycotoxins: Food contamination, mechanisms, carcinogenic potential and preventive mea-
sures, Mutat. Res., 259, 291, 1991.
16. Bennett, J.W., Klich, M., Mycotoxins, Clin. Microbiol. Rev., 16, 497, 2003.
17. Nyikal, J. et al., Outbreak of aflatoxin poisoning—Eastern and Central Provinces, Kenya, January–July
2004, Morbid. Mortal. Week. Rep., 53, 790, 2004.
18. Groopman, J.D., Johnson, D., Kensler, T.W., Aflatoxin and hepatitis B virus biomarkers: A paradigm for
complex environmental exposures and cancer risk, Cancer Biomarkers, 1, 5, 2005.
19. Eaton, D.L., Groopman, J.D., The Toxicology of Aflatoxins: Human Health, Veterinary, and Agricultural
Significance, Academic Press, San Diego, CA, 1994.
20. International Agency for Research on Cancer (IARC), Some Traditional Herbal Medicine, Some
Mycotoxins, Naphthalene and Styrene, IARC Monographs on the Evaluation on the Carcinogenic Risks
to Humans, Vol. 82, IARC Press, Lyon, 2002.
21. van Egmond, H.P., Schothorst, R.C., Jonker, M.A., Regulations relating to mycotoxins in food:
Perspectives in a global and European context, Anal. Bioanal. Chem., 389, 147, 2007.
22. van der Merwe, K.J., Steyn, P.S., Fourie, L., Scott, D.B., Theron, J.J., Ochratoxin A, a toxic metabolite
produced by Aspergillus ochraceus Wilh, Nature, 205, 1112, 1965.
23. Moss, M.O., Mode of formation of ochratoxin A, Food Addit. Contam., 13, S5, 1996.
24. Xiao, H., Marquardt, R.R., Frohlich, A.A., Ling, Y.Z., Synthesis and structural elucidation of analogs of
ochratoxin A, J. Agric. Food Chem., 43, 524, 1995.
25. Smith, J.E., Moss, M.O., Mycotoxins: Formation, Analysis and Significance, John Wiley & Sons,
Chichester, 1985.
26. Krogh, P. et al. 1977. Balkan (endemic) nephropathy and foodborne ochratoxin A: Preliminary results
of a survey of foodstuff, Acta Pathol. Microbiol. Scand. Sect. B, 85, 238, 1977.
27. Grollman, A.P. et al., Aristolochic acid and the etiology of endemic (Balkan) nephropathy, Proc. Natl.
Acad. Sci. USA, 104, 12129, 2007.
28. O’Brien, E., Dietrich, D.R., Ochratoxin A: The continuing enigma, Crit. Rev. Toxicol., 35, 33, 2005.
29. Davolos, D. et al., Aspergillus affinis sp. nov., a novel ochratoxin A-producing Aspergillus species (sec-
tion Circumdati) isolated from decomposing leaves, Int. J. Syst. Evol. Microbiol., 62, 1007, 2012.
30. Visagie, C.M. et al., Ochratoxin production and taxonomy of the yellow aspergilli (Aspergillus section
Circumdati), Stud. Mycol., 78, 1, 2014.
31. Varga, J. et al., Ochratoxin production by Aspergillus species, Appl. Environ. Microbiol., 62, 4461, 1996.
32. Samson, R.A. et al., New ochratoxin or sclerotium producing species in Aspergillus section Nigri, Stud.
Mycol., 50, 45, 2004.
33. Perrone, G. et al., Aspergillus niger contains the cryptic phylogenetic species A. awamori, Fungal Biol.,
115, 1138, 2011.
34. Varga, J. et al., Recent advances in ochratoxin research I. Production, detection and occurrence of och-
ratoxins, Cereal Res. Commun., 29, 85, 2001.
35. Noonim, P. et al., Fumonisin B2 production by Aspergillus niger from Thai coffee beans, Food Addit.
Contam., 26, 94, 2009.
36. Bayman, P., Baker, J.L., Ochratoxins: A global perspective, Mycopathologia, 162, 215, 2006.
37. Varga, J., Kozakiewicz, Z., Ochratoxin A in grapes and grape-derived products, Trends Food Sci.
Technol., 17, 72, 2006.
38. Pel, H.J. et al., Genome sequencing and analysis of the versatile cell factory Aspergillus niger CBS
513.88, Nat. Biotechnol., 25, 221, 2007.
39. Frisvad, J.C. et al., Fumonisin B2 production by Aspergillus niger, J. Agric. Food Chem., 55, 9727, 2007.
40. Varga, J. et al., Fumonisin contamination and fumonisin producing black aspergilli in dried vine fruits
of different origin, Int. J. Food Microbiol., 143, 143, 2010.
Aspergillus 489
41. Varga, J. et al., Molecular diversity of agriculturally important Aspergillus species, Eur. J. Plant Pathol.,
110, 627, 2004.
42. Pitt, J. I., Hocking, A.D., Fungi and Food Spoilage, 2nd ed., Blackie Academic and Professional,
London, UK, 1997.
43. Birkinshaw, J.H. et al., Patulin in the common cold. Collaborative research on a derivative of Penicillium
patulum Bainier. II. Biochemistry and chemistry, Lancet, 245, 625, 1943.
44. Moake, M.M., Padilla-Zakour, O.I., Worobo, R.W., Comprehensive review of patulin control methods in
foods, Compr. Rev. Food Sci. Food Saf., 1, 8, 2005.
45. Chalmers, I., Clarke, M., The 1944 patulin trial: The first properly controlled multicentre trial con-
ducted under the aegis of the British Medical Research Council, Int. J. Epidemiol., 33, 253, 2004.
46. Walker, K., Wiesner, B.P., Patulin and clavicin, Lancet, 246, 294, 1944.
47. Steinman, R., Seigle-Murandi, F., Sage, L., Krivobok, S., Production of patulin by micromycetes,
Mycopathologia, 105, 129, 1989.
48. Varga, J. et al., Taxonomic revision of Aspergillus section Clavati based on molecular, morphological
and physiological data, Stud. Mycol., 59, 89, 2007.
49. Joint FAO/WHO Expert Committee on Food additives (JECFA), Position Paper on Patulin, The Hague,
The Netherlands, 9–13 March, 1998.
50. Scott, P.M., Patulin. In Mycotoxins, ed. I.F.H. Purchase, pp. 383–403. Elsevier, Amsterdam, The
Netherlands, 1974.
51. Holzapfel, C.W., The isolation and structure of cyclopiazonic acid, a toxic metabolite of Penicillium
cyclopium westling, Tetrahedron, 24, 2101, 1968.
52. Frisvad, J.C., Smedsgaard, J., Larsen, T.O., Samson, R.A., Mycotoxins, drugs and other extrolites pro-
duced by species in Penicillium subgenus Penicillium, Stud. Mycol., 49, 201, 2004.
53. Larsen, T.O., Smedsgaard, J., Nielsen, K.F., Hansen, M.A., Samson, R.A., Frisvad, J.C., Production of
mycotoxins by Aspergillus lentulus and other medically important and closely related species in section
Fumigati, Med. Mycol., 45, 225, 2007.
54. Betina, V., Mycotoxins—Chemical, biological and environmental aspects, Bioact. Mol., 9, 27, 1989.
55. Samson, R.A. et al., New species in Aspergillus section Terrei, Stud. Mycol., 69, 39, 2011.
56. Houbraken, J., Frisvad, J.C., Samson, R.A., Taxonomy of Penicillium section Citrina, Stud. Mycol., 70
138, 2011.
57. Li, F.Q., Xu, G.R., Li, Y.W., Chen, Y., Ji, R., Natural occurrence of citrinin in Monascus products. Wei
Sheng Yan Jiu, 34, 451, 2005.
58. Gardiner, D.M., Howlett, B.J., Bioinformatic and expression analysis of the putative gliotoxin biosyn-
thetic gene cluster of Aspergillus fumigatus, FEMS Microbiol. Lett., 248, 241, 2005.
59. Terabayashi, Y. et al., Identification and characterization of genes responsible for biosynthesis of kojic
acid, an industrially important compound from Aspergillus oryzae, Fungal Genet. Biol., 47, 953, 2010.
60. Klejnstrup, M.L. et al., Genetics of polyketide metabolism in Aspergillus nidulans, Metabolites, 2, 100,
2012.
61. Reiss, J., Insecticidal and larvicidal activities of the mycotoxins aflatoxin B1 rubratoxin B, patulin and
diacetoxyscirpenol towards Drosophila melanogaster, Chem. Biol. Interact., 10, 339, 1975.
62. Kirk, H.D., Ewen, A.B., Emson, H.E., Blair, D.G., Effect of aflatoxin B1 on development of Drosophila
melanogaster (Diptera), J. Invertebr. Pathol., 18, 313, 1971.
63. Lamb, M.J., Lilly, L.J., Induction of recessive lethals in Drosophila melanogaster by aflatoxin B1, Mutat.
Res., 11, 430, 1971.
64. Chinnici, J.P., Melone, P.D., Genetic aspects of aflatoxin B1 resistance in Drosophila melanogaster, J.
Hered., 76, 85, 1985.
65. Llewellyn, G.C., Chinnici, J.P., Variation in sensitivity to aflatoxin B1 among several strains of
Drosophila melanogaster (Diptera), J. Invertebr. Pathol., 31, 37, 1978.
66. Chinnici, J.P., Booker, M.A., Llewellyn, G.C., Effect of aflatoxin B1 on viability, growth, fertility, and
crossing over in Drosophila melanogaster (Diptera), J. Invertebr. Pathol., 27, 255, 1976.
67. Fahmy, M.J., Fahmy, O.G., Swenson, D.H., Aflatoxin B1-2,3-dichloride as a model of the active metabo-
lite of aflatoxin B1 in mutagenesis and carcinogenesis, Cancer Res., 38, 2608, 1978.
68. Chinnici, J.P., Erlanger, L., Charnock, M., Jones, M., Stein, J., Sensitivity differences displayed by
Drosophila melanogaster larvae of different ages to the toxic effects of growth on media containing
aflatoxin B1, Chem. Biol. Interact., 24, 373, 1979.
490 Laboratory Models for Foodborne Infections
69. Chinnici, J.P., Genetic resistance to aflatoxin B1 toxicity in Drosophila melanogaster: Chromosome
substitution analysis, J. Hered., 71, 275, 1980.
70. Fahmy, M.J., Fahmy, O.G., Intervening DNA insertions and the alteration of gene expression by carcino-
gens, Cancer Res., 40, 3374, 1980.
71. Chinnici, J.P., Gunst, K., Llewellyn, G.C., Effects of mycotoxin pretreatment on aflatoxin B1 post-
treatment toxicity in Drosophila melanogaster (Diptera), J. Invertebr. Pathol., 41, 321, 1983.
72. Foerster, R.E., Würgler, F.E., In vitro studies on the metabolism of aflatoxin B1 and aldrin in testes of
genetically different strains of Drosophila melanogaster, Arch. Toxicol., 56, 12, 1984.
73. Belitsky, G.A., Khovanova, E.M., Budunova, I.V., Sharuptis, H.G., Mycotoxin induction of somatic mosa-
icism in Drosophila and DNA repair in mammalian liver cell cultures, Cell Biol. Toxicol., 1, 133, 1985.
74. Melone, P.D., Chinnici, J.P., Selection for increased resistance to aflatoxin B1 toxicity in Drosophila
melanogaster, J. Invertebr. Pathol., 48, 60, 1986.
75. Shibahara, T., Ogawa, H.I., Ryo, H., Fujikawa, K., DNA-damaging potency and genotoxicity of afla-
toxin M1 in somatic cells in vivo of Drosophila melanogaster, Mutagenesis, 10, 161, 1995.
76. Karekar, V., Joshi, S., Shinde, S.L., Antimutagenic profile of three antioxidants in the Ames assay and
the Drosophila wing spot test, Mutat. Res., 468, 183, 2000.
77. Sidorov, R.A., Ugnivenko, E.G., Khovanova, E.M., Belitsky, G.A., Induction of tumor clones in D. mela-
nogaster wts/+ heterozygotes with chemical carcinogens, Mutat. Res., 498, 181, 2001.
78. Sişman, T., The protective effect of hydrated sodium calcium aluminosilicate against the adverse effects
of aflatoxin B1 on D. melanogaster, Toxicol. Ind. Health, 22, 173, 2006.
79. Mutlu, A.G., Increase in mitochondrial DNA copy number in response to ochratoxin A and methanol-
induced mitochondrial DNA damage in Drosophila, Bull. Environ. Contam. Toxicol., 89, 1129, 2012.
80. Lee, S.E., Campbell, B.C., In vitro metabolism of aflatoxin B1 by larvae of navel orangeworm, Amyelois
transitella (Walker) (Insecta, Lepidoptera, Pyralidae) and codling moth, Cydia pomonella (L.) (Insecta,
Lepidoptera, Tortricidae), Arch. Insect. Biochem. Physiol., 45, 166, 2000.
81. Leung, M.C., Goldstone, J.V., Boyd, W.A., Freedman, J.H., Meyer, J.N., Caenorhabditis elegans gener-
ates biologically relevant levels of genotoxic metabolites from aflatoxin B1 but not benzo[a]pyrene in
vivo, Toxicol. Sci., 118, 444, 2010.
82. González-Hunt, C.P. et al., Exposure to mitochondrial genotoxins and dopaminergic neurodegeneration
in Caenorhabditis elegans, PLoS One, 9, e114459, 2014.
83. Shyamal, S. et al., Hepatoprotective effect of three herbal extracts on aflatoxin B1-intoxicated rat liver,
Singapore Med. J., 51, 326, 2010.
84. Abdu, S., Ali, A., Ansari, S., Cytotoxic effect of ochratoxin A on the renal corpuscles of rat kidney:
Could ochratoxin A cause kidney failure? Histol. Histopathol., 26, 543, 2011.
85. Doktorova, T. et al., Comparison of genotoxicant-modified transcriptomic responses in conventional and
epigenetically stabilized primary rat hepatocytes with in vivo rat liver data, Arch. Toxicol., 86, 1703, 2012.
86. Jennings, P. et al., Transcriptomic alterations induced by ochratoxin A in rat and human renal proximal
tubular in vitro models and comparison to a rat in vivo model, Arch. Toxicol., 86, 571, 2012.
87. Tong, P.Z., Zhang, G.J., Zhang, X.H., Yan, X., Wang, J.L., Effects of sterigmatocystin on esophageal
epithelium and experimental reflux esophagitis in rats, Mol. Med. Rep., 8, 1043, 2013.
88. Akçam, M. et al., Caffeic acid phenethyl ester modulates aflatoxin B1-induced hepatotoxicity in rats,
Cell Biochem. Funct., 31, 692, 2013.
89. Adeleye, A.O. et al., Phenolic extract of Dialium guineense pulp enhances reactive oxygen species
detoxification in aflatoxin B1 hepatocarcinogenesis, J. Med. Food, 17, 875, 2014.
90. Ou, C., Zheng, H.P., Su, J.J., Cao, J., Li, G.J., Li, L.Q., Effect of Ginkgo biloba extract on the expressions
of Cox-2 and GST-Pi in rats with hepatocellular carcinoma risk, Afr. Health Sci., 14, 37, 2014.
91. Supriya, C., Reddy, P.S., Prenatal exposure to aflatoxin B1: Developmental, behavioral, and reproductive
alterations in male rats, Sci. Nat., 102, 1, 2015.
92. Suwannakul, N., Punvittayagul, C., Jarukamjorn, K., Wongpoomchai, R., Purple rice bran extract
attenuates the aflatoxin B1-induced initiation stage of hepatocarcinogenesis by alteration of xenobiotic
metabolizing enzymes, Asian Pac. J. Cancer Prev., 16, 3371, 2015.
93. Mantle, P., Kilic, M., Mor, F., Ozmen, O., Contribution of organ vasculature in rat renal analysis for
ochratoxin A: Relevance to toxicology of nephrotoxins, Toxins, 7, 1005, 2015.
94. Mantle, P., Kulinskaya, E., Lifetime, low-dose ochratoxin A dietary study on renal carcinogenesis in
male Fischer rats. Food Addit. Contam. A. Chem. Anal. Control Expo. Risk Assess., 27, 1566, 2010.
Aspergillus 491
95. Mantle, P.G., Nicholls, A.W., Shockcor, J.P., 1H NMR spectroscopy-based metabolomic assessment of
uremic toxicity, with toxicological outcomes, in male rats following an acute, mid-life insult from och-
ratoxin A, Toxins, 3, 504, 2011.
96. Corcuera, L.-A., Ibáñez-Vea, M., Vettorazzi, A., González-Peñas, E., Cerain, A.L.D., Validation of a
UHPLC-FLD analytical method for the simultaneous quantification of aflatoxin B1 and ochratoxin A in
rat plasma, liver and kidney, J. Chromatogr. B, 879, 2733, 2011.
97. Koissi, N. et al., Lactone metabolite common to the carcinogens dioxane, diethylene glycol, and
N-nitrosomorpholine: Aqueous chemistry and failure to mediate liver carcinogenesis in the F344 rat,
Chem. Res. Toxicol., 25, 1022, 2012.
98. Merrick, B.A. et al., Testing an aflatoxin B1 gene signature in rat archival tissues, Chem. Res. Toxicol.,
25, 1132, 2012.
99. Lu, X. et al., Integrated analysis of transcriptomics and metabonomics profiles in aflatoxin B1-induced
hepatotoxicity in rat, Food Chem. Toxicol., 55, 444, 2013.
100. Qi, X. et al., Ochratoxin A induces rat renal carcinogenicity with limited induction of oxidative stress
responses, Toxicol. Appl. Pharmacol., 280, 543, 2014.
101. Yang, W. et al., Genome-wide miRNA-profiling of aflatoxin B1-induced hepatic injury using deep
sequencing, Toxicol. Lett., 226, 140, 2014.
102. Knipstein, B. et al., Dietary aflatoxin-induced stunting in a novel rat model: Evidence for toxin-induced
liver injury and hepatic growth hormone resistance, Pediatr. Res., 78(2), 120–127, 2015.
103. Hoffmann, D. et al., Evaluation of a urinary kidney biomarker panel in rat models of acute and sub-
chronic nephrotoxicity, Toxicology, 277, 49, 2010.
104. Merrick, B.A. et al., RNA-seq profiling reveals novel hepatic gene expression pattern in aflatoxin B1
treated rats, PLoS One, 8, e61768, 2013.
105. Johnson, N.M. et al., Complete protection against aflatoxin B1-induced liver cancer with a triterpenoid:
DNA adduct dosimetry, molecular signature, and genotoxicity threshold, Cancer Prev. Res., 7, 658, 2014.
106. Yenilmez, A. et al., Antioxidant effects of melatonin and coenzyme Q10 on oxidative damage caused by
single-dose ochratoxin A in rat kidney, Chin. J. Physiol., 53, 310, 2010.
107. Kim, Y.-S. et al., Protective effect of Korean red ginseng against aflatoxin B(1)-induced hepatotoxicity
in rat, J. Ginseng Res., 35, 243, 2011.
108. Abdel-Aziem, S.H., Hassan, A.M., Abdel-Wahhab, M.A., Dietary supplementation with whey protein
and ginseng extract counteracts oxidative stress and DNA damage in rats fed an aflatoxin-contaminated
diet, Mutat. Res. Genet. Toxicol. Environ. Mutagen., 723, 65, 2011.
109. Fiala, J.L.A. et al., Sulforaphane-mediated reduction of aflatoxin B1-N7-guanine in rat liver DNA:
Impacts of strain and sex, Toxicol. Sci., 121, 57, 2011.
110. Shen, H. et al., Impairment of alveolar type-II cells involved in the toxicity of aflatoxin G1 in rat lung,
Food Chem. Toxicol., 50, 3222, 2012.
111. Han, Z. et al. Combinatorial approach of LC–MS/MS and LC–TOF-MS for uncovering in vivo kinetics
and biotransformation of ochratoxin A in rat, J. Chromatogr. B, 925, 46, 2013.
112. Palabiyik, S.S. et al., Protective effect of lycopene against ochratoxin A induced renal oxidative stress
and apoptosis in rats, Exp. Toxicol. Pathol., 65, 853, 2013.
113. Aydin, S. et al., The carotenoid lycopene protects rats against DNA damage induced by ochratoxin A,
Toxicon, 73, 96, 2013.
114. Wei, W. et al., Evaluation of toxicological impacts on Sprague-Dawley rat by successively low dose of
aflatoxin B1, J. Sci. Food Agric., 94, 3017, 2014.
115. Gong, B. et al., Transcriptomic profiling of rat liver samples in a comprehensive study design by RNA-
Seq, Sci. Data, 1, 140021, 2014.
116. Hassan, A.M., Abdel-Aziem, S.H., El-Nekeety, A.A., Abdel-Wahhab, M.A., Panax ginseng extract
modulates oxidative stress, DNA fragmentation and up-regulate gene expression in rats sub chronically
treated with aflatoxin B1 and fumonisin B1, Cytotechnology, 67, 861–871, 2015.
117. Mantle, P.G., Dobrota, M., Gillett, C.E., Odell, E.W., Pinder, S.E., Oncological outcomes in rats given
nephrocarcinogenic exposure to dietary ochratoxin A, followed by the tumour promoter sodium barbital
for life: A pilot study, Toxins, 2, 552, 2010.
118. Singh, K., Maurya, B., Trigun, S., Activation of oxidative stress and inflammatory factors could account
for histopathological progression of aflatoxin-B1 induced hepatocarcinogenesis in rat, Mol. Cell.
Biochem., 401, 185, 2015.
492 Laboratory Models for Foodborne Infections
119. Tozlovanu, M. et al., Glutathione conjugates of ochratoxin A as biomarkers of exposure, Arh. Hig. Rada
Toksikol., 63, 417, 2012.
120. Jennings-Gee, J.E., Tozlovanu, M., Manderville, R., Miller, M.S., Pfohl-Leszkowicz, A., Schwartz,
G.G., Ochratoxin A: In utero exposure in mice induces adducts in testicular DNA, Toxins, 2, 1428, 2010.
121. Schütze, N., Lehmann, I., Bönisch, U., Simon, J.C., Polte, T., Exposure to mycotoxins increases the
allergic immune response in a murine asthma model, Am. J. Respir. Crit. Care Med., 181, 1188, 2010.
122. Li, C.-H. et al., Hepatic oval cell lines generate hepatocellular carcinoma following transfection with
HBx gene and treatment with aflatoxin B1 in vivo, Cancer Lett., 311, 1, 2011.
123. Brahmi, D., Bouaziz, C., Ayed, Y., Ben Mansour, H., Zourgui, L., Bacha, H., Chemopreventive effect of
cactus Opuntia ficus indica on oxidative stress and genotoxicity of aflatoxin B1, Nutr. Metab. (Lond.), 8,
73, 2011.
124. Eraslan, G., Kanbur, M., Aslan, Ö., Karabacak, M., The antioxidant effects of pumpkin seed oil on
subacute aflatoxin poisoning in mice, Environ. Toxicol., 28, 681, 2013.
125. Liu, Y. et al., Sterigmatocystin alters the number of FoxP3+ regulatory T cells and plasmacytoid den-
dritic cells in BALB/c mice, Food Chem. Toxicol., 50, 1920, 2012.
126. Zhang, Y., Yao, Z.G., Wang, J., Xing, L.X., Xia, Y., Zhang, X.H., Effects of sterigmatocystin on TNF-α,
IL-6 and IL-12 expression in murine peripheral blood mononuclear cells and peritoneal macrophages in
vivo, Mol. Med. Rep., 5, 1318, 2012.
127. Larypoor, M., Bayat, M., Zuhair, M.H., Akhavan Sepahy, A., Amanlou, M., Evaluation of the number
of CD4(+) CD25(+) FoxP3(+) treg cells in normal mice exposed to AFB1 and treated with aged garlic
extract, Cell J., 15, 37, 2013.
128. Abbès, S., Salah-Abbès, J.B., Sharafi, H., Jebali, R., Noghabi, K.A., Oueslati, R., Ability of Lactobacillus
rhamnosus GAF01 to remove AFM1 in vitro and to counteract AFM1 immunotoxicity in vivo.
J. Immunotoxicol., 10, 279, 2013.
129. Zhang, Z. et al., Cytochrome P450 2A13 mediates the neoplastic transformation of human bronchial
epithelial cells at a low concentration of aflatoxin B1, Int. J. Cancer, 134, 1539, 2014.
130. Ben Salah-Abbès, J., Abbès, S., Jebali, R., Haous, Z., Oueslati, R., Potential preventive role of lactic acid
bacteria against aflatoxin M1 immunotoxicity and genotoxicity in mice, J. Immunotoxicol., 12, 107, 2015.
131. Sun, L.H., Lei, M.Y., Zhang, N.Y., Zhao, L., Krumm, C.S., Qi, D.S., Hepatotoxic effects of mycotoxin
combinations in mice, Food Chem. Toxicol., 74, 289, 2014.
132. Jebali, R., Abbès, S., Salah-Abbès, J.B., Younes, R.B., Haous, Z., Oueslati, R., Ability of Lactobacillus
plantarum MON03 to mitigate aflatoxins (B1 and M1) immunotoxicities in mice, J. Immunotoxicol., 12,
290, 2015.
133. Shen, H. et al., Enhanced phenotypic alterations of alveolar type II cells in response to aflatoxin
G1-induced lung inflammation, J. Cell Physiol., 230, 1199, 2015.
134. Liu, C. et al., Oral administration of aflatoxin G1 induces chronic alveolar inflammation associated with
lung tumorigenesis, Toxicol. Lett., 232, 547, 2015.
135. Cariddi, L.N. et al., Polyphenols as possible bioprotectors against cytotoxicity and DNA damage induced
by ochratoxin A, Environ. Toxicol. Pharmacol., 39, 1008, 2015.
136. Saxena, N., Ansari, K.M., Kumar, R., Chaudhari, B.P., Dwivedi, P.D., Das, M., Role of mitogen acti-
vated protein kinases in skin tumorigenicity of patulin, Toxicol. Appl. Pharmacol., 257, 264, 2011.
137. Jha, A., Shah, K., Verma, R.J., Aflatoxin-induced biochemical changes in liver of mice and its mitiga-
tion by black tea extract, Acta Pol. Pharm., 69, 851, 2012.
138. Jha, A., Krithika, R., Manjeet, D., Verma, R.J., Protective effect of black tea infusion on aflatoxin-
induced hepatotoxicity in mice, J. Clin. Exp. Hepatol., 3, 29, 2013.
139. Kumar, R. et al., Ochratoxin A-induced cell proliferation and tumor promotion in mouse skin by acti-
vating the expression of cyclin-D1 and cyclooxygenase-2 through nuclear factor-kappa B and activator
protein-1, Carcinogenesis, 34, 647, 2013.
140. Chakraborty, D., Verma, R., Ameliorative effect of Emblica officinalis aqueous extract on ochratoxin-
induced lipid peroxidation in the kidney and liver of mice, Int. J. Occup. Med. Environ. Health, 23, 63,
2010.
141. Naaz, F., Abdin, M.Z., Javed, S., Protective effect of esculin against prooxidant aflatoxin B1-induced
nephrotoxicity in mice, Mycotoxin Res., 30, 25, 2014.
142. Gupta, R., Sharma, V., Ameliorative effects of Tinospora cordifolia root extract on histopathological
and biochemical changes induced by aflatoxin-B1 in mice kidney, Toxicol. Int., 18, 94, 2011.
Aspergillus 493
143. Choi, K.C. et al., Inhibitory effects of quercetin on aflatoxin B1-induced hepatic damage in mice, Food
Chem. Toxicol., 48, 2747, 2010.
144. Choi, K.C. et al., Chemoprevention of a flavonoid fraction from Rhus verniciflua Stokes on aflatoxin
B1-induced hepatic damage in mice, J. Appl. Toxicol., 31, 150, 2011.
145. Hou, Y.J. et al., Mycotoxin-containing diet causes oxidative stress in the mouse, PLoS One, 8, e60374,
2013.
146. Hsuuw, Y.D., Chan, W.H., Yu, J.S., Ochratoxin A inhibits mouse embryonic development by activating
a mitochondrion-dependent apoptotic signaling pathway, Int. J. Mol. Sci., 14, 935, 2013.
147. Hou, Y.J. et al., Oocyte quality in mice is affected by a mycotoxin-contaminated diet, Environ. Mol.
Mutagen., 55, 354, 2014.
148. Ferenczi, S. et al., A new ochratoxin A biodegradation strategy using Cupriavidus basilensis r16 strain,
PLoS One, 9, e109817, 2014.
149. Song, E. et al., Hepatotoxicity and genotoxicity of patulin in mice, and its modulation by green tea poly-
phenols administration, Food Chem. Toxicol., 71, 122, 2014.
150. Song, E. et al., Selenium supplementation shows protective effects against patulin-induced brain dam-
age in mice via increases in GSH-related enzyme activity and expression, Life Sci., 109, 37, 2014.
151. Miller, J.D., Sun, M., Gilyan, A., Roy, J., Rand, T.G., Inflammation-associated gene transcription and
expression in mouse lungs induced by low molecular weight compounds from fungi from the built envi-
ronment, Chem. Biol. Interact., 183, 113, 2010.
152. Kim, J.E., Bunderson, B.R., Croasdell, A., Coulombe, R.A. Jr., Functional characterization of alpha-
class glutathione S-transferases from the turkey (Meleagris gallopavo), Toxicol. Sci., 124, 45, 2011.
153. de Melo et al., DNA damage in organs of mice treated acutely with patulin, a known mycotoxin, Food
Chem. Toxicol., 50, 3548, 2012.
154. Guindon-Kezis, K.A., Mulder, J.E., Massey, T.E., In vivo treatment with aflatoxin B1 increases DNA
oxidation, base excision repair activity and 8-oxoguanine DNA glycosylase 1 levels in mouse lung,
Toxicology, 321, 21, 2014.
155. Al-Hazmi, M.A., Patulin in apple juice and its risk assessments on albino mice, Toxicol. Ind. Health, 30,
534, 2014.
156. Napoletano, M. et al., Ochratoxin A induces craniofacial malformation in mice acting on Dlx5 gene
expression, Front. Biosci., 2, 133, 2010.
157. Fox, J.G. et al., Gut microbes define liver cancer risk in mice exposed to chemical and viral transgenic
hepatocarcinogens, Gut, 59, 88, 2010.
158. Liu, S.P. et al., Higher susceptibility to aflatoxin B1-related hepatocellular carcinoma in glycine
N-methyltransferase knockout mice, Int. J. Cancer, 128, 511, 2011.
159. Chawanthayatham, S. et al., Prenatal exposure of mice to the human liver carcinogen aflatoxin B1
reveals a critical window of susceptibility to genetic change, Int. J. Cancer, 136, 1254, 2015.
160. Kensler, K.H. et al., Genetic or pharmacologic activation of Nrf2 signaling fails to protect against afla-
toxin genotoxicity in hypersensitive GSTA3 knockout mice, Toxicol. Sci., 139, 293, 2014.
161. Hoek-van den Hil, E.F., van Schothorst, E.M., van der Stelt, I., Hollman, P.C., Keijer, J., Rietjens, I.M.,
Quercetin tests negative for genotoxicity in transcriptome analyses of liver and small intestine of mice,
Food Chem. Toxicol., 81, 34, 2015.
162. Teoh, W.W. et al., Molecular characterization of hepatocarcinogenesis using mouse models, Dis. Model
Mech., 8, 743, 2015.
163. Wattanawaraporn, R. et al., A single neonatal exposure to aflatoxin B1 induces prolonged genetic dam-
age in two loci of mouse liver, Toxicol. Sci., 128, 326, 2012.
164. Austin, K.J., Cockrum, R.R., Jons, A.M., Alexander, B.M., Cammack, K.M., Renin mRNA is upregu-
lated in testes and testicular cells in response to treatment with aflatoxin B1, Theriogenology, 77, 331,
2012.
165. Mulder, J.E., Bondy, G.S., Mehta, R., Massey, T.E., Up-regulation of nucleotide excision repair in mouse
lung and liver following chronic exposure to aflatoxin B1 and its dependence on p53 genotype, Toxicol.
Appl. Pharmacol., 275, 96, 2014.
166. Bondy, G.S. et al., Effects of chronic ochratoxin A exposure on p53 heterozygous and p53 homozygous
mice, Toxicol. Pathol., 43(5), 715–729, 2015.
167. Kuroda, K. et al., Role of p53 in the progression from ochratoxin A-induced DNA damage to gene muta-
tions in the kidneys of mice, Toxicol. Sci., 144, 65, 2015.
494 Laboratory Models for Foodborne Infections
168. Mulder, J.E., Bondy, G.S., Mehta, R., Massey, T.E., The impact of chronic aflatoxin B1 exposure and p53
genotype on base excision repair in mouse lung and liver, Mutat. Res., 773, 63, 2015.
169. Guan, L.Z. et al., The aflatoxin-detoxifizyme specific expression in mouse parotid gland, Transgenic
Res., 24, 489, 2015.
170. Mulder JE, Turner PV, Massey TE. Effect of 8-oxoguanine glycosylase deficiency on aflatoxin B1
tumourigenicity in mice, Mutagenesis, 30, 401, 2015.
171. Adedara, I.A., Owumi, S.E., Uwaifo, A.O., Farombi, E.O., Aflatoxin B1 and ethanol co-exposure induces
hepatic oxidative damage in mice, Toxicol. Ind. Health, 26, 717, 2010.
172. Zhu, C.C. et al., Effect of mycotoxin-containing diets on epigenetic modifications of mouse oocytes by
fluorescence microscopy analysis, Microsc. Microanal., 20, 1158, 2014.
173. Bastaki, S.A., Osman, N., Kochiyil, J., Shafiullah, M., Padmanabhan, R., Abdulrazzaq, Y.M.,
Toxicokinetics of aflatoxin in pregnant mice, Int. J. Toxicol., 29, 425, 2010.
174. Ueta, E. et al., Gender-dependent differences in the incidence of ochratoxin A-induced neural tube
defects in the Pdn/Pdn mouse, Congenit. Anom. (Kyoto), 50, 29, 2010.
175. Koes, M.T., Forrester, L.J., Brown, H.D., Metabolism of aflatoxin B1 by the guinea-pig, Food Cosmet.
Toxicol., 11, 463, 1973.
176. Peden, W.M., Richard, J.L., Thurston, J.R., Sacks, J.L., Effects of pre-treatment with aflatoxin on a
second aflatoxin treatment in guinea pigs, Mycopathologia, 99, 107, 1987.
177. Thurston, J.R., Sacks, J.M., Richard, J.L., Peden, W.M., Driftmier, K., Complement, bacteriostatic, and enzy-
matic activities in sera from guinea pigs given aflatoxin and/or rubratoxin, Am. J. Vet. Res., 50, 356, 1989.
178. Netke, S.P., Roomi, M.W., Tsao, C., Niedzwiecki, A., Ascorbic acid protects guinea pigs from acute
aflatoxin toxicity, Toxicol. Appl. Pharmacol., 143, 429, 1997.
179. Schiller, F., Lippold, U., Heinze, R., Hoffmann, A., Seffner, W., Liver fibrosis in guinea pigs experimen-
tally induced by combined copper and aflatoxin application, Exp. Toxicol. Pathol., 50, 519, 1998.
180. Stĕtinová, V., Grossmann, V., Kvĕtina, J., Changes in the gastrointestinal tract, cardiovascular function
and some drug metabolizing processes in rats and guinea-pigs intoxicated with aflatoxin B1, Pol. J.
Pharmacol., 50, 135, 1998.
181. Miranda, D.D. et al., Analysis of DNA damage induced by aflatoxin B1 in Dunkin–Hartley guinea pigs,
Mycopathologia, 163, 275, 2007.
182. Ungar, H., Sullman, S.F., Zuckerman, A.J., Acute and protracted changes in the liver of Syrian hamsters
induced by a single dose of aflatoxin B1. Observations on pathological effects of the solvent (dimethyl-
formamide), Br. J. Exp. Pathol., 57, 157, 1976.
183. Rajmon, R. et al., Combined effects of repeated low doses of aflatoxin B1 and T-2 toxin on the Chinese
hamster, Vet. Med. Czech, 46, 301, 2001.
184. Chu, C.C., Pan, K.L., Yao, H.T., Hsu, J.T., Development of a whole-cell screening system for evaluation
of the human CYP1A2-mediated metabolism, Biotechnol. Bioeng., 108, 2932, 2011.
185. Ratajewski, M., Walczak-Drzewiecka, A., Sałkowska, A., Dastych, J., Aflatoxin upregulate CYP3A4
mRNA expression in a process that involves the PXR transcription factor, Toxicol. Lett., 205, 146, 2011.
186. Ma, Y., Kong, Q., Hua, H., Luo, T., Jiang, Y., Aflatoxin B1 up-regulates insulin receptor substrate 2 and
stimulates hepatoma cell migration, PLoS One, 7(10), e47961. doi: 10.1371/journal.pone.0047961, 2012.
187. Hanioka, N. et al., Effect of aflatoxin B1 on UDP-glucuronosyltransferase mRNA expression in HepG2
cells, Chemosphere, 89, 526, 2012.
188. Tolosa, L., Donato, M.T., Pérez-Cataldo, G., Castell, J.V., Gómez-Lechón, M.J., Upgrading cytochrome
P450 activity in HepG2 cells co-transfected with adenoviral vectors for drug hepatotoxicity assessment,
Toxicol. In Vitro, 26, 1272, 2012.
189. Chaudhary, H., Jena, P.K., Seshadri, S., Evaluation of hydro-alcoholic extract of Eclipta alba for its
multidrug resistance reversal potential: An in vitro study, Nutr. Cancer, 65, 775, 2013.
190. Lereau, M. et al., Interactions between hepatitis B virus and aflatoxin B1: Effects on p53 induction in
HepaRG cells, J. Gen. Virol., 93, 640, 2012.
191. Jossé, R., Rogue, A., Lorge, E., Guillouzo, A., An adaptation of the human HepaRG cells to the in vitro
micronucleus assay, Mutagenesis, 27, 295, 2012.
192. Li, A.P., Uzgare, A., LaForge, Y.S., Definition of metabolism-dependent xenobiotic toxicity with co-
cultures of human hepatocytes and mouse 3T3 fibroblasts in the novel integrated discrete multiple organ
co-culture (IdMOC) experimental system: Results with model toxicants aflatoxin B1, cyclophosphamide
and tamoxifen, Chem. Biol. Interact., 199, 1, 2012.
Aspergillus 495
193. Anninou, N., Chatzaki, E., Papachristou, F., Pitiakoudis, M., Simopoulos, C., Mycotoxins’ activity at
toxic and sub-toxic concentrations: Differential cytotoxic and genotoxic effects of single and combined
administration of sterigmatocystin, ochratoxin A and citrinin on the hepatocellular cancer cell line
Hep3B, Int. J. Environ. Res. Public Health, 11, 1855, 2014.
194. Mary, V.S., Valdehita, A., Navas, J.M., Rubinstein, H.R., Fernández-Cruz, M.L., Effects of aflatoxin B1,
fumonisin B1 and their mixture on the aryl hydrocarbon receptor and cytochrome P4501A induction,
Food Chem. Toxicol., 75, 104, 2015.
195. Ellis, J.K. et al., Metabolic response to low-level toxicant exposure in a novel renal tubule epithelial cell
system, Mol. Biosyst., 7, 247, 2011.
196. Radford, R. et al., Carcinogens induce loss of the primary cilium in human renal proximal tubular epi-
thelial cells independently of effects on the cell cycle, Am. J. Physiol. Renal Physiol., 302, F905, 2012.
197. Stachurska, A., Kozakowska, M., Jozkowicz, A., Dulak, J., Loboda, A., Aristolochic acid I and ochra-
toxin A differentially regulate VEGF expression in porcine kidney epithelial cells—The involvement of
SP-1 and HIFs transcription factors, Toxicol. Lett., 204, 118, 2011.
198. Lei, M., Zhang, N., Qi, D., In vitro investigation of individual and combined cytotoxic effects of aflatoxin
B1 and other selected mycotoxins on the cell line porcine kidney 15, Exp. Toxicol. Pathol., 65, 1149,
2013.
199. Bittner, A., Cramer, B., Harrer, H., Humpf, H.U., Structure elucidation and in vitro cytotoxicity of och-
ratoxin α amide, a new degradation product of ochratoxin A, Mycotoxin Res., 31, 83, 2015.
200. Ramyaa, P., Padma, V.V., Ochratoxin-induced toxicity, oxidative stress and apoptosis ameliorated by
quercetin–modulation by Nrf2, Food Chem. Toxicol., 62, 205, 2013.
201. Bruneau, J.C., Stack, E., O’Kennedy, R., Loscher, C.E., Aflatoxins B1, B2 and G1 modulate cytokine secre-
tion and cell surface marker expression in J774A.1 murine macrophages, Toxicol. In Vitro, 26, 686, 2012.
202. Shao, J. et al., Toxicogenomics-based identification of mechanisms for direct immunotoxicity, Toxicol.
Sci., 135, 328, 2013.
203. Luongo, D., Russo, R., Balestrieri, A., Marzocco, S., Bergamo, P., Severino, L., In vitro study of AFB1
and AFM1 effects on human lymphoblastoid Jurkat T-cell model, J. Immunotoxicol., 11, 353, 2014.
204. Ribonnet, L. et al., Potential of an in vitro toolbox combined with exposure data as a first step for the
risk assessment of dietary chemical contaminants, Food Addit. Contam. Part A. Chem. Anal. Control
Expo. Risk Assess., 28, 1136, 2011.
205. Kwon, O. et al., Patulin induces colorectal cancer cells apoptosis through EGR-1 dependent ATF3 up-
regulation, Cell Signal., 24, 943, 2012.
206. Clarke, R., Connolly, L., Frizzell, C., Elliott, C.T., Cytotoxic assessment of the regulated, co-existing
mycotoxins aflatoxin B1, fumonisin B1 and ochratoxin, in single, binary and tertiary mixtures, Toxicon,
90, 70, 2014.
207. Liu, A., Ye, Y., Chen, W., Wang, X., Chen, F., Expression of V(H)-linker-V(L) orientation-dependent
single-chain Fv antibody fragment derived from hybridoma 2E6 against aflatoxin B1 in Escherichia
coli, J. Ind. Microbiol. Biotechnol., 42, 255, 2015.
208. Costanzo, P., Santini, A., Fattore, L., Novellino, E., Ritieni, A., Toxicity of aflatoxin B1 towards the
vitamin D receptor (VDR), Food Chem. Toxicol., 76, 77, 2015.
209. Wang, Y., Wong, T.Y., Chan, F.L., Chen, S., Leung, L.K., Assessing the effect of food mycotoxins on
aromatase by using a cell-based system, Toxicol. In Vitro, 28, 640, 2014.
210. Probert, P.M et al., Utility of B-13 progenitor-derived hepatocytes in hepatotoxicity and genotoxicity
studies, Toxicol. Sci., 137, 350, 2014.
211. Shen, H. et al., Aflatoxin G1-induced oxidative stress causes DNA damage and triggers apoptosis through
MAPK signaling pathway in A549 cells, Food Chem. Toxicol., 62, 661, 2013.
212. Nisr, R.B., Russell, M.A., Chrachri, A., Moody, A.J., Gilpin, M.L., Effects of the microbial secondary
metabolites pyrrolnitrin, phenazine and patulin on INS-1 rat pancreatic β-cells, FEMS Immunol. Med.
Microbiol., 63, 217, 2011.
213. Moreno-Félix, C. et al., Bioactive lipidic extracts from octopus (Paraoctopus limaculatus): Antimutagenicity
and antiproliferative studies, Evid. Based Complement. Altern. Med., 2013, 273582, 2013.
32
Candida
María Jesús Andrade, Mar Rodríguez, Alicia Rodríguez, and Juan José Córdoba
CONTENTS
32.1 Introduction................................................................................................................................... 497
32.2 Foodborne Candida Infections..................................................................................................... 498
32.3 Investigation of Foodborne Candida Cases.................................................................................. 498
32.4 Laboratory Models for Analyzing Foodborne Candida Infections.............................................. 501
32.5 Conclusions................................................................................................................................... 506
Acknowledgements................................................................................................................................. 506
References............................................................................................................................................... 506
32.1 Introduction
The Candida genus comprises more than 200 recognized species1,2 of which more than 40 are impli-
cated in human infections.3 Candida is a natural inhabitant of human skin, intestinal and genitourinary
tracts, and mucosal surfaces. Being an opportunistic pathogen, Candida causes disease when the host
defenses are impaired. With recent increase in immunosuppressed patients, Candida infections have
been described with higher frequency.4,5
Nonetheless, the presence of Candida in foods has obvious positive effects, such as in fermented
foods and beverages. This fungus also contributes to the synthesis of some healthy compounds such as
vitamins and antioxidants.6 On the contrary, its negative effects on food products relate to food spoilage,
but not usually to food safety problems. Thus, Candida species are frequently included in daily food
consumption of human beings, as disorders attributable to pathogenic Candida through foods have been
described as improbable.2,7 Consequently, its effect on human health has been scarcely evaluated in the
literature and more attention has been paid to clinical Candida infections.
Candida albicans is the major species of the genus associated with human disease,1 although it is
not involved in foodborne infection. In addition, other Candida species that are frequently referred
to as “non-albicans” Candida species have been isolated as being responsible for infection including
C. glabrata, C. tropicalis, C. parapsilosis, and C. krusei.2,5 In the recent years, the occurrence of infec-
tions caused by such “non-albicans” Candida species has increased. The abovementioned five species
are the causative agents of more than 90% of Candida infections.2,8 In spite of the fact that other Candida
species like C. kefyr, C. rugosa, C. guilliermondii, and C. famata have been reported as causative agents
of clinical infection, they have been rarely isolated from patients.3,5,9
Candida can cause a wide variety of infections in humans1 that range from superficial infection of skin
and mucosal membranes of intestinal and genital tracts,2 including cutaneous and oropharyngeal candi-
diasis and vaginitis,1 to systematic infections. The latter are of significant concern since they encompass
life-threatening diseases such as candidemia, endophthalmitis, endocarditis, osteomyelitis, and dissemi-
nated and central nervous system infections.1,8 Such severe invasive infections cause high morbidity and
mortality rates.8,10
497
498 Laboratory Models for Foodborne Infections
Isolation Identification/detection
Seminested/nested PCR
Microscopy
Chromogenic media
(morphology)
(CHROMagar Candida, NASBA
Candida ID 2)
Caco-2
Mice
IPEC-J2
Diagnosis of Candida infections has traditionally relied on a combination of phenotypic features of the
pathogenic yeast and the clinical signs of the infection.2
When analyzing the clinical presenting features, a combination of tests is necessary to obtain an
adequate diagnosis.2 However, the main problem is that clinical signs are often nonspecific for Candida
disease.
Direct microscopic investigation of appropriate clinical samples and culture is included within the
phenotypic techniques. Microscopic morphology of Candida shows important differences depending on
environmental conditions and yeast species. Thus, Candida can produce round or elongated blastoco-
nidia, pseudohyphae, true hyphae, or chlamydospores.1 However, C. glabrata rarely produces pseudohy-
phae or hyphal structures.2
To study the other phenotypic features, the isolation of the causative agent is necessary. Sabouraud’s
dextrose agar (SDA) is the most commonly used primary isolation medium for Candida. However, it
does not normally allow differentiation between species.34 The colonies of Candida species are cream/
white colored with smooth and wet looking. Nevertheless, C. albicans has the capability to reversibly
change the surface appearance of its colony from smooth to rough as well as from creamy/white to
opaque, a modification that corresponds to an alteration in cell shape from round (oval) to elongate.2
After isolating Candida as the causative agent of an infection, the identification at species level of
the obtained isolates has to be performed. The assimilation and fermentation patterns can be studied
for such purpose35 by using the available commercial kits (API® 20 C AUX, API® Candida, VITEK® 2,
bioMérieux; AuxaColor™ 2, Bio-Rad Laboratories). Such commercial techniques have been used to test
for Candida of both food and clinical origin.
In addition, several chromogenic media for the direct and presumptive identification of Candida on the
basis of the colony color have been developed. Thus, agars such as CHROMagar Candida (CHROMagar
Company) and Candida ID 2 (bioMérieux) allow the differentiation of some of the most significant
pathogenic Candida species.36–38 The use of such media allows reducing the time of analysis in com-
parison with conventional phenotypic and biochemical methods. Despite the fact that the CHROMagar
Candida was originally developed for identifying Candida isolates of clinical origin, it has been used for
differentiation and presumptive identification of the most frequently isolated yeast species from different
ingredients used for salads.39
500 Laboratory Models for Foodborne Infections
Species identification of Candida based on the abovementioned phenotypic methods is not always suf-
ficient in spite of using different tests5 because of their possible ambiguous or inconclusive results. This
fact together with the fact that they are often time consuming cause that infections are often not diag-
nosed or the diagnosis is obtained too late. Consequently, these methods are of limited clinical value, at
least when they are used alone.
Nonculture diagnostic methods are available as alternative and supplementary to the conven-
tional ones.
Thus, serological tests can be performed by detecting antigens and antibodies. Such tests are used
in combination with conventional techniques for improving the diagnosis. The mannan and (1,3)-β-d-
glucan detection is widely used. The mannan constituent of the Candida cell wall is a polysaccharide
used as a target of many serological tests performed in serum or plasma specimens since it induces a
strong antibody response.40 Tests based on the detection of antibodies directed against the mannan anti-
gen are recommended in combination with those based on Candida antigens for improving the sensitiv-
ity and the specificity of the diagnosis.41 The commercial Platelia Candida antigen and antibody tests
(Bio-Rad Laboratories) are the most commonly used serological methods for detecting Candida based
on mannan.41,42 However, since the antibodies could be not produced in immunocompromised patients,
it is very complicated to diagnose Candida infections in them.34,41 (1,3)-β-d-Glucan is also a cell wall
component not only of Candida species but also of most of the other fungi. Several assays have been
developed for quantifying such compounds in blood as a tool in the diagnosis of many pathogenic fungi
including Candida.40,43
The detection of d-arabinitol, a metabolite produced by some of the Candida species with clinical
importance, has also been proposed for diagnosis.40
The serological methods have limitations for the diagnosis of candidal infections due to their sensi-
tivity and specificity. Besides, they cannot be used alone for reliable identification of clinical Candida
isolates as previously described for traditional techniques.
To overcome the limitations of culture and nonculture methods, nucleic-acid-based procedures have
been developed since they are powerful tool because of their rapidity and sensitivity. The literature
contains a lot of data regarding molecular-based technology for clinical diagnosis of Candida. Lewis
et al.2,35 summarized the polymerase chain reaction (PCR) methods published for detecting Candida
species from clinical samples, with most of them being based on rDNA regions as target. These authors
considered that such methods could also be used for screening food products. However, the type of
sample, enrichment processes, and DNA extraction should be optimized for the particular food and for
removal of any interfering compounds within.2
Identification by sequencing of rRNA genes has been widely used for identifying Candida of both
food and clinical origins.28,44,45 However, such technique despite being a valid method for identifying
Candida is time consuming, expensive, tedious, technically demanding, and inappropriate for routine
applications.46,47
Among various molecular-based methods, real-time PCR (qPCR) has advantages of speed, sensitivity,
and specificity.48–50
Multiplex PCR detecting different targets simultaneously and semi-nested/nested PCR incorporating
two round of amplification reactions have also been developed.51–54
Other molecular-based methods for detecting Candida RNA include nucleic-acid-sequence-based
amplification (NASBA),55 etc.
Despite the vast number of suitable published molecular methods for detecting and/or identifying
Candida species, they are still lacking standardization. Thus, it has been suggested that a combination of
methods, including the molecular ones, may be performed to obtain a certain diagnosis.5,43
Novel technologies such as matrix-assisted laser desorption/ionization time-of-flight mass spectrom-
etry (MALDI-TOF MS) have been reported as rapid and accurate methods for routine identification of
Candida associated with human and food.46,47,56,57 However, some of the authors have stated the necessity
of improving the databases of the available systems for such technique.
The former methods can detect Candida involved in foodborne infection or in patients with clinical
symptoms of disease. However, the study of candidal infections has also involved the subsequent inves-
tigation of the yeast in animals and other laboratory model systems.58 They are necessary to provide
Candida 501
new insights for clarifying and fully understanding the mechanisms leading to human Candida disease
independently of whether the yeast is of clinical or food origin. Such powerful systems will be detailed
in the next section.
TABLE 32.1
Laboratory Models Used to Investigate Gastrointestinal Candida Infections
Candida Species
Model Type Detected Main Measured Parameters References
Mouse C. albicans Histopathology, yeast morphology and burden, 75
C. glabrata inflammatory response
C. albicans Yeast burden, histopathology 68
C. tropicalis
C. parapsilosis
C. albicans Yeast burden, role of MAPK pathways 71
C. albicans Yeast burden, colonization and invasion, mortality 70
C. tropicalis
C. albicans Yeast burden and morphology, immune response 74
C. albicans Yeast burden and morphology 72
Mouse, piglet C. albicans Yeast burden and morphology, gene expression 73
Drosophila larvae C. albicans Microscopy, systematic response 84
Caco-2 cells C. albicans LDH assay, genetic analysis, immune response 90
C. tropicalis
C. parapsilosis
C. krusei
C. tropicalis Adhesion, cell damage, gene expression 89
C. albicans Yeast burden and morphology, adhesion, topography of 88
cells, transcriptional regulation during adhesion
C. albicans Adhesion, cell damage, invasion, transmigration, yeast 66
morphology, viability, and hydrolases
Caco-2 and IPEC-J2 cell C. albicans Yeast morphology, transepithelial electrical resistance 27
lines C. rugosa
C. tropicalis
C. krusei
502 Laboratory Models for Foodborne Infections
vaccination strategies. The advantages and limitations of both in vivo models have been summarized by
Naglik et al.58 The main advantage of nonvertebrate animal models consists of lower cost and fewer ethi-
cal concerns than vertebrate ones. Vertebrate animal model systems are more adequate tools for studying
the host–Candida interactions than the nonvertebrate animal ones due to the fact that the used animals
and their environment can be precisely controlled in a manner that is more relevant to humans. A good
in vivo model for investigating Candida infections should be as reproducible as possible regarding the
process of colonization and invasion at a specific portal of entry leading to infection as well as the major
clinical symptoms detected in the human disease, relatively easy to set up, and closely match the specific
immune defects or hormonal conditions associated with infection at that particular site.59,60 The cost
of the model should also be considered in the choice.60 In addition, the host, the tested Candida strain,
and the route of its administration are of critical importance to obtain reliable experimental conclusions
when selecting the in vivo experimental model.61
Although many clinical animal models have been developed, rodents (mainly mice) are the most
commonly used laboratory ones (Table 32.1) because of ethical reasons and their relative anatomic and
immunological similarity to human.60,62 Moreover, they are widely available, easy to handle, and rela-
tively inexpensive.63 However, since C. albicans is an opportunistic pathogen of humans but not a natural
inhabitant of rodents, such models could give responses to infections in a different way than human
beings, and so comparison between such models and human studies could be difficult.58,64,65 Such differ-
ence should be considered when interpreting experimental data.61
In the case of modeling gastrointestinal Candida infections, mice are the animals selected by the
majority of researchers,65 and rats have been occasionally used.58 However, most of them are mainly
focused on studying the later dissemination of Candida because systematic dissemination normally
originates from the gastrointestinal tract.60,66
In gastrointestinal models, it has to be taken into account that the colonization of the gastrointesti-
nal tract by Candida seems to be apparently inhibited by the innate defenses of the host and mucosal
immune response and also by competition of Candida with the native microbial population of the tract.67
Thus, for establishing the gastrointestinal Candida infection the normal mucosal microbial population
must be removed or the yeast must become implanted in the gastrointestinal tract before colonization
by this microbial population.67 Gastrointestinal models are thus commonly based on the use of immu-
nosuppressed adult mice or neonatal mice.68 For neonatal models, no pretreatment is necessary as for
adult ones, which have to be treated for removing their natural gastrointestinal microbial population by
using immunocompromising agents (such as by antibiotic administration) prior to Candida infection.60,67
Germ-free or transgenic mice could also be used for this purpose.58 The performed mice manipulations
have to be considered when interpreting the obtained results.58 Adult mice have been considered as more
suitable since their handling and care are easier and less time consuming and their mortality due to the
inoculation is minimal.68 Adult mice can be inoculated either intragastrically or orally by using contami-
nated food or drinking water,60 the second option being the most adequate for studying gastrointestinal
infections related to foodborne Candida as the inoculum should be administered via the physiologically
relevant portal of entry.61 Since mice are coprophagous, the gastrointestinal mucosa of the animal fed
with Candida contaminated food seems to be exposed to high levels of the yeast.67 The majority of the
clinical models based on gastrointestinal infection have carried out the injection of the yeast directly into
the tract. After Candida inoculation, the maintenance of antibacterial antibiotic administration is neces-
sary for achieving sustained gastrointestinal colonization69 and allowing yeast dissemination to occur.60
The level of Candida inoculum used for gastrointestinal models ranges from about 107 to 108 cells
suspended in a sterile saline solution, mainly phosphate buffered saline (PBS), when performing
intragastric administration.68,70–73 The inoculum level ranges from about 106 to 107 cells/mL via drink
administration.74,75
The behavior of non-albicans Candida species, including C. tropicalis, C. glabrata, and C. parapsilo-
sis, has also been analyzed by using gastrointestinal models, but scarcely. Several authors have compared
the virulence of C. albicans and C. tropicalis in a gastrointestinal model in mice, with C. tropicalis
appearing as more virulent than C. albicans.60,70 When comparing the virulence of C. tropicalis, C. gla-
brata, and C. albicans in a gastrointestinal mouse model, the three species colonized the gastrointestinal
tract, with C. tropicalis showing the least ability and the subsequent dissemination was only observed in
Candida 503
C. albicans and C. tropicalis.68 The virulence of C. glabrata was also less than that shown by C. albi-
cans by using this kind of models.75 It seems that virulence mechanisms differ between Candida species,
but more research using experimental animal models is necessary.
The most usually measured parameters in mice Candida gastrointestinal models during the coloniza-
tion and the later dissemination include yeast burden at various sites, organ histopathology, and mortality
of animals (Table 32.1). Candida burden has been determined in fecal and cecal contents and also in
organs such as esophagus, stomach, intestines, cecum and colon, and internal organs (kidney, spleen,
lungs, liver).68,70,72,74,75 Visualization under microscope of stools and histologic examination of different
organs for identifying yeast or filamentous forms have also been reported for most of gastrointestinal
models. It seems that hyphae, but not yeast, are implied in the inflammatory and immune responses dur-
ing colonization of the gastrointestinal tract,74 having a significant role in the pathogenesis by invading
epithelial cells and causing damage of tissues.76 The ability to change morphology from yeast to hyphal
form (dimorphism) in response to environmental changes is one of the best studied virulence attributes
and characteristics of C. albicans.77 However, the conversion of C. albicans to hyphae has been described
by other authors as important but not essential for pathogenesis and tissue invasion.78
Regarding Candida immunity in gastrointestinal models, the host immune response during Candida
colonization of the gastrointestinal tract has been scarcely studied. Vautier et al.74 performed an immu-
noassay for analyzing cytokine levels after inoculating antibiotic-treated mice with C. albicans via their
drinking water containing 107 yeast cells/mL together with antibiotics. Kidney, stomach, intestines, and
caecum were used for such analysis. They reported that colonization of the gastrointestinal tract by
C. albicans was not influenced by Th17 immunity.
The inflammatory response in gastrointestinal tissues was studied by Westwater et al.75 in an assay
based on phenotyping of GR1 granulocytes. C. glabrata and C. albicans were separately administrated
to germ-free mice via drink. C. glabrata did not evoke a granulocytic inflammatory response in the host,
in contrast to C. albicans.
Few studies have investigated the genes expressed in C. albicans during the gastrointestinal coloniza-
tion in mice and also in piglets. White et al.73 extracted RNA from different tissues of infected piglets and
from cecum and ileum contents from mice, which was used for quantitative real-time reverse transcrip-
tase PCR (qRT-PCR) and microarrays. Their aim was the study of the genes expressed by C. albicans
during its growth in the gastrointestinal tract. In a separate work, Prieto et al.71 used a gastrointestinal
model in antibiotic-treated mouse to determine the role of the mitogen-activated protein kinase (MAPK)
pathways of C. albicans in the colonization of the gastrointestinal tract. Such pathways represent one of
the main mechanisms of adaptation to environmental changes in C. albicans and play an important role
in virulence.71
In the above-described gastrointestinal models based on the host immune and inflammatory responses
and genetic studies, Candida loads were also determined in different sites of the gastrointestinal tract
and in stools.
Laboratory models focused on other vertebrates animals such as monkeys,79 piglets,80 rabbits,81 and
guinea pigs82 have also been proposed.60 In spite of the fact that the larger the animal model, the easier
it is to sample repeatedly,58 the use of those animals larger in size than mice implies higher cost and
husbandry requirements, more intense monitoring during experimental infection, and fewer available
molecular reagents.61 In addition, they are not available as genetically defined strains.61
Due to the fact that research involving mammals is generally expensive, complex, and fraught with
ethical concerns, alternative animal models for studying clinical Candida infection have been used.83
Zebrafish (Danio rerio), a vertebrate minihost, and some nonvertebrate minihosts, including wax moth
(Galleria mellonella) and silk worm (Bombyx mori) larvae, the fruit fly (Drosophila melanogaster), and a
nematode (Caenorhabditis elegans), have been thus reported for studying Candida virulence.60 A limita-
tion of experimental models based on minihosts is that many of them lack an adaptive immune system.60
A Drosophila larvae model has been reported as appropriate to assay the host–pathogen interaction due
to an intestinal infection by Candida and the factors required for disseminated infection.84 The similarity
of mammals’ and flies’ immune response allows analyzing the genes involved.84 However, the differ-
ences between fly and mammalian pathophysiology have to also be considered when using Drosophila
intestinal models,83 as previously mentioned for murine models. Since in the developed D. melanogaster
504 Laboratory Models for Foodborne Infections
model the larvae were fed with Candida contaminated food, such experimental model should be well
suited for analyzing foodborne Candida infections. This characteristic is also attributable to the model
described by Pukkila-Worley et al.,85 who studied C. albicans pathogenesis using C. elegans as host,
which was fed with artificially contaminated food, and described the utility of the assay for screening
the genes involved in Candida morphogenesis and virulence. Since both the genome of D. melanogaster
and C. elegans have been sequenced,62 genetic analyses are available when performing experimental
Candida models, which is not possible for wax moth and silk worm models because of lack of genome
sequencing.62 The use of invertebrate animal models has the disadvantage that they have to be kept at
temperatures below the normal human body temperature, except for Galleria, which could affect the
Candida–host interactions.60
On the other hand, Candida, including foodborne species, commonly grow as a biofilms in medi-
cal devices, thereby providing protection from the host immune system and antimicrobial therapies.86
Scarce works based on experimental models for evaluating the formation of biofilms in invasive devices
are available.86
Although animals offer a good model for analyzing Candida infections, they have some limitations,87
such as they are not appropriate for wide laboratory testing and routine analysis since special facilities
and special trained personnel are needed. Besides, ethical considerations should be taken into account
when animals are used. The absence of Candida as native microorganisms in the gastrointestinal tract of
rodent models should also be considered when searching for a good model for Candida analysis.
Several in vitro alternative methods based on the use of cell lines and epithelial organotypic models
are available for such purpose, which are less expensive and time consuming to obtain results than ani-
mal models87 (Table 32.1). They have been developed for Candida of clinical origin but not for foodborne
Candida, as mentioned for animal models. However, such models could be adapted for such purpose.
When selecting an in vitro model, its availability and easy handling for high-throughput testing as well
as its good human predictive power must be checked.87
Most of the performed investigations based on the use of cell lines for host–Candida interaction stud-
ies have been focused on the ability of Candida (mainly C. albicans) to adhere to different epithelia.
For studying foodborne Candida, the most interesting cell lines are those of intestinal origin. Specific
studies must be carried out in gastrointestinal models since the environment of the different anatomic
sites that could be colonized by Candida differs significantly, particularly with respect to pH, epithelium
metabolism, or surface characteristics.88 Due to the fact that the attachment of Candida to intestinal epi-
thelium is the initial step in the infection, it is an essential parameter to be analyzed in a gastrointestinal
model based on cell lines. The ability of C. albicans to adhere to and invade into intestinal cells for its
translocation across the intestinal barrier and the subsequent dissemination through the body have been
demonstrated by using intestinal cell lines.66 To analyze the adhesion of C. albicans to an intestinal
epithelium in vitro, the Caco-2 cell line has been the most commonly used. Caco-2 is a human epithelial
colon adenocarcinoma cell line.87 Intestinal cell models should be functionally similar to the in vivo situ-
ation,87 and such cells have this characteristic since they resemble morphologically as well as biochemi-
cally primary enterocytes in many characteristics.88
Some examples of the use of Caco-2 cell line will be briefly described below.
Sohn et al.88 infected monolayers of Caco-2 grown to confluency with C. albicans. Polystyrene plates
were used for an adhesion assay. After applying C. albicans to the confluent monolayers, adherent
C. albicans cells counting was performed. Scanning electron microscopy (SEM) was also carried out for
visualizing the surface topographies of cells and the presence of hyphae. The Caco-2 cells had a brush
border surface consisting of microvilli structures, which seemed to be tightly linked to C. albicans
hyphae. The transcriptional regulation during adhesion of C. albicans to Caco-2 cells was analyzed by
genome-wide Candida DNA microarray analysis using total RNA isolated from the adhesion assay in
order to identify how C. albicans responds on the transcriptional level to the environmental stimuli dur-
ing adhesion to intestinal cell. They found that the adherence of C. albicans to Caco-2 cells requires the
expression of adhesive structures on the yeast surface with the ability of interacting very tightly with the
host tissues.
Negri et al.89 also evaluated the attachment of C. tropicalis to intestinal epithelial cells by using Caco-2
cells. Besides analyzing adhesion of C. tropicalis to a confluent layer of intestinal cells, the degree of
Candida 505
the epithelial cell damage was assessed by measuring the lactate dehydrogenase (LDH) activity, and the
inhibition of cellular activity was determined by tetrazolium salt reduction. C. tropicalis was able to
adhere to and to damage Caco-2. However, differences were observed in this ability depending on the
type of used cells. This fact was also strain dependent. Since the secretion of hydrolytic enzymes such
as secreted aspartyl proteinases (SAPs) is considered an important factor for C. tropicalis virulence, the
expression of four genes encoding SAPs (SAPT1, SAPT2, SAPT3, and SAPT4) present in C. tropicalis
was also investigated by qPCR after RT-PCR, which was affected by C. tropicalis adhesion to the cells.
The SAP expression was also strain and human cell line dependent. This fact highlights the necessity to
check specific Candida strains in specific anatomic sites.
Dalle et al.66 also investigated the potential of C. albicans to adhere to and invade into Caco-2 cells.
They performed adherence assay by using epifluorescence after calcofluor white staining, SEM, and
measurement of LDH release, among other techniques. These authors found that adhesion, invasion, and
damage of C. albicans depended not only on yeast morphology and activity, but also on the epithelial
cell type and the differentiation stage of the epithelial cells, indicating that epithelial cells differ in their
susceptibility to C. albicans. Besides, they reported that hyphae seem to have a dominant role in penetra-
tion of monolayers of intestinal cells in relation to yeasts.
Intestinal Caco-2 cells have been recently used for analyzing the host immune response to Candida
since intestinal epithelial cells not only represent a physiologic barrier for the yeasts, but also have a
crucial role in the innate immune system as they, for example, produce cytokines and antimicrobial
peptides.90 The effect of C. albicans as well as C. krusei, C. tropicalis, and C. parapsilosis on the pro-
duction of human β-defensin 2 (HBD-2), a key component of the innate immune system, by Caco-2 cell
line has been studied by RT-q PCR.90 To determine the HBD-2 secretion induction and HBD-2 peptide
expression in Caco-2 cells infected with Candida spp., enzyme linked immunosorbent assay (ELISA)
and Western blot analysis were carried out, respectively. Candida spp. stimulated HBD-2 expression in
and release from Caco-2 cells, with C. albicans inducing the highest levels of HBD-2.
Cell lines have been interestingly used for investigating the probiotic potential of some yeasts from
food including Candida species, but their pathogenic potential was also analyzed.27 C. rugosa, C. tropi-
calis, and C. krusei, which are considered causative agents of infections, were studied. They were previ-
ously isolated from Fura, a spontaneously fermented pearl millet product consumed in West Africa. As
control, the authors included a pathogenic yeast strain of C. albicans. These authors used two intestinal
cell lines: Caco-2 and IPEC-J2. In spite of the fact that IPEC-J2 was isolated from the small intestine
of neonatal pig,87 it has been used for studying pathogen/beneficial interactions of microorganisms due
to its similarity to the human intestinal epithelial cells.27 In such study, after growing both cell lines
in different media, the cells were grown at 37°C in a humidified atmosphere of 5% CO2 and 95% air
until a confluent monolayer was obtained. The culture media were changed every second day, and the
cells were passaged every 5–7 days by trypsination with 1% trypsin–EDTA 10×. Cell passages used
were P55–57 and P26–28 for Caco-2 and IPEC-J2, respectively. The transepithelial electrical resistance
(TEER) assay was used for analyzing the potential pathogenic effect (and also the probiotic one) of used
yeasts by means of evaluating the effect of yeast strains on cell polarity. To obtain a monolayer of polar-
ized intestinal epithelial cells, both cell lines were cultured on polycarbonate membrane inserts. After
reaching confluence, Caco-2 cells were cultured on the membranes for 21 days in order to obtain cell
differentiation, while IPEC-J2 cells were cultured for 16 days in order to obtain a proper mucus layer. An
inoculum of 106 cells/mL of yeasts was added and incubated at 37°C in a humidified atmosphere of 5%
CO2 and 95% air. The TEER of monolayers with Caco-2 or IPEC-J2 cell lines was then measured and
the morphology of yeasts observed by microscopy. Moreover, C. krusei, C. tropicalis, C. rugosa, and
C. albicans were able to survive under conditions simulating passage through the human gastrointestinal
tract [37°C, pH 2.5, and 0.3% (w/v) bile salts]. The ability of potential pathogenic foodborne Candida
to survive the conditions in the gastrointestinal tract should also be evaluated such as the survival and
growth at 37°C and the presence of high bile salt concentration and low pH.
Epithelial organotypic models have been described as effective as an in vitro model for evaluating
the invasion of C. albicans too. Such models allow studying the tissue degradation in Candida infec-
tion as well as conducting experiments to characterize pathogenesis-related genes. The mucosal epithe-
lium is very important in host defense and immune surveillance, since it is the primary cell layer that
506 Laboratory Models for Foodborne Infections
first encounters potentially pathogenic microorganisms.91 Reconstituted human epithelium that mimics
human epithelium is commercially available. It has been used as model for oral and vaginal candidiasis,
but not for foodborne Candida infections. After infecting the tissues, their level of colonization and
invasion and morphological characteristics of Candida can be analyzed by microscopy.92 Epithelial dam-
age can be visualized by histological analysis and quantified by the extracellular activity of LDH.91,92
Besides, the expression patterns of several Candida virulence genes, such as agglutinin-like sequence
(ALS), hyphal wall protein (HWP), epithelial adhesion (EPA), phospholipase B (PLB), PLD, and SAP
genes, can be analyzed by RT-PCR when using such in vitro models.91–93 The lack of certain host immune
responses is a limitation of such epithelium models, while proinflammatory cytokine responses by the
tissue infected with Candida are associated with the yeast strain virulence.91,93
32.5 Conclusions
Several Candida species are commonly present in food and beverages. However, candidal infections are
not normally associated with consumption of food containing Candida. In the last years, some Candida
species found in food are emerging as causative agents of infections. Further researches are thus neces-
sary for elucidating the role of foodborne Candida in human infections.
A variety of methods to detect and diagnose Candida infections have been described, most of which
have been designed for clinical purpose but could also be applied for investigation of foodborne cases.
Phenotypic methods have been traditionally used for detecting Candida; however, they are time con-
suming and can lead ambiguous or inconclusive results. Serological methods have limited sensitivity
and specificity. More recently, molecular-based protocols have become available, although they are still
in need of proper standardization. Consequently, a combination of methods for diagnosis of pathogenic
Candida from both clinical and food origins should be performed for obtaining definitive results.
In order to analyze different aspects of clinical Candida infections, several laboratory models based
on using vertebrate or nonvertebrate animals, cell lines, and commercial reconstituted human epithelium
have been developed. Mice are the most commonly used animal models because of ethical reason and
their relative anatomic and immunological similarity to human beings despite the fact that Candida is
not a natural inhabitant of mice. The use of cell lines such as Caco-2 cells has less ethical concerns and
other advantages compared to animal models. In addition, epithelial organotypic models used for analyz-
ing oral and vaginal candidiasis could be adapted for Candida foodborne investigation.
As most currently available methods have focused on Candida of clinical origin and not on foodborne
Candida, further investigations are required to develop adequate laboratory models for investigating
foodborne Candida infections. These include refining the experimental gastrointestinal models designed
for clinical Candida.
Acknowledgements
This work has been funded by the Spanish Comisión Interministerial de Ciencia y Tecnología with the
projects AGL2013-45729P and Carnisenusa CSD2007-00016, Consolider Ingenio 2010 and GRU09162
of the Junta de Extremadura and FEDER.
REFERENCES
1. Eggimann, P., Garbino, J. and Pittet, D., Epidemiology of Candida species infections in critically ill
non-immunosuppressed patients, Lancet Infect. Dis., 3, 685, 2003.
2. Lewis, P. et al., Candida, in Molecular Detection of Foodborne Pathogens, p. 549, Liu, D. (Ed.), Taylor
and Francis Group LLC, Boca Raton, 2010.
3. Johnson, E.M., Rare and emerging Candida species, Curr. Fungal Infect. Rep., 3, 152, 2009.
4. Hobson, R., The global epidemiology of invasive Candida infections—is the tide turning? J. Hosp.
Infect., 55, 159, 2003.
Candida 507
5. Criseo, G., Scordino, F. and Romeo, O., Current methods for identifying clinically important cryptic
Candida species, J. Microbiol. Methods, 111, 50, 2015.
6. Fleet, G.H. and Balia, R., The public health and probiotic significance of yeasts in foods and bever-
ages, in Yeasts in Food and Beverages, p. 381, Querol, A., Fleet, G.H. (Eds.), Springer-Verlag, Berlin,
Heidelberg, 2006.
7. Boekhout, T. and Robert, V. (Eds.), Yeasts in Food. Beneficial and Detrimental Aspects, Behr’s Verlag,
Hamburg, Germany, 2003.
8. Yapar, N., Epidemiology and risk factors for invasive candidiasis, Ther. Clin. Risk Manag., 10, 95, 2014.
9. Brandt, M.E. and Lockhart, S.R., Recent taxonomic developments with Candida and other opportunis-
tic yeasts, Curr. Fungal Infect. Rep., 6, 170, 2012.
10. Quindós, G., Epidemiology of candidaemia and invasive candidiasis. A changing face, Rev. Iberoam.
Micol., 31, 42, 2014.
11. Fleet, G.H., Yeasts in foods and beverages: impact on product quality and safety, Curr. Opin. Biotechnol.,
18, 170, 2007.
12. Simoncini, N. et al., Dynamics and characterization of yeasts during ripening of typical Italian dry-
cured ham, Food Microbiol., 24, 577, 2007.
13. Núñez, F. et al., Yeast population during ripening of dry-cured Iberian ham, Int. J. Food Microbiol., 29,
271, 1996.
14. Andrade, M.J. et al., Differentiation of yeasts growing on dry-cured Iberian ham by mitochondrial DNA
restriction analysis, RAPD-PCR and their volatile compounds production, Food Microbiol., 26, 578, 2009.
15. Ismail, S.A.S. et al., Presence and changes in populations of yeasts on raw and processed poultry prod-
ucts stored at refrigeration temperature, Int. J. Food Microbiol., 62, 113, 2000.
16. Purriños, L. et al., Study of the counts, species and characteristics of the yeast population during the
manufacture of dry-cured “lacón”. Effect of salt level, Food Microbiol., 34, 12, 2013.
17. Gardini, F. et al., A survey of yeasts in traditional sausages of southern Italy, FEMS Yeast Res., 1, 161,
2001.
18. Encinas, J.P. et al., Yeast populations on Spanish fermented sausages, Meat Sci., 54, 203, 2000.
19. Fadda, M.E. et al., Occurrence and characterization of yeasts isolated from artisanal Fiore Sardo cheese,
Int. J. Food Microbiol., 95, 51, 2004.
20. Prillinger, H. et al., Phenotypic and genotypic identification of yeasts from cheese, Antonie Van
Leeuwenhoek, 75, 267, 1999.
21. Arias, C.R. et al., Yeast species associated with orange juice: evaluation of different identification meth-
ods, Appl. Environ. Microbiol., 68, 1955, 2002.
22. Shokri, H., Genotypic variation and antifungal susceptibly of Candida zeylanoides clinical isolates,
J. Mycol. Med., 24, 179, 2014.
23. EFSA, Scientific Opinion on the maintenance of the list of QPS biological agents intentionally added to
food and feed (2013 update), EFSA J., 11, 3449, 2013.
24. EFSA, Introduction of a qualified presumption of safety (QPS) approach for assessment of selected
microorganisms referred to EFSA, EFSA J., 587, 1, 2007.
25. Beyda, N.D. et al., Treatment of Candida famata bloodstream infections: case series and review of the
literature, J. Antimicrob. Chemother., 68, 438, 2013.
26. Greppi, A. et al., Determination of yeast diversity in ogi, mawè, gowé and tchoukoutou by using culture-
dependent and -independent methods, Int. J. Food Microbiol., 165, 84, 2013.
27. Pedersen, L.L. et al., Biodiversity and probiotic potential of yeasts isolated from Fura, a West African
spontaneously fermented cereal, Int. J. Food Microbiol., 159, 144, 2012.
28. Yan, Y.Z. et al., Microbial composition during Chinese soy sauce koji-making based on culture depen-
dent and independent methods, Food Microbiol., 34, 189, 2013.
29. Alloue-Boraud, W.A.M. et al., Fermentation profile of Saccharomyces cerevisiae and Candida
tropicalis as starter cultures on barley malt medium, J. Food Sci. Technol., 1–7, 2014, doi: 10.1007/
s13197-014-1526-0.
30. Las Heras-Vazquez, F.J. et al., Identification of yeast species from orange fruit and juice by RFLP and
sequence analysis of the 5.8S rRNA gene and the two internal transcribed spacers, FEMS Yeast Res., 3,
3, 2003.
31. Jespersen, L. et al., Occurrence and diversity of yeasts involved in fermentation of West African cocoa
beans, FEMS Yeast Res., 5, 441, 2005.
508 Laboratory Models for Foodborne Infections
32. Nielsen, D.S. et al., Yeast populations associated with Ghanaian cocoa fermentations analysed using
denaturing gradient gel electrophoresis (DGGE), Yeast, 22, 271, 2005.
33. Annan, N.T. et al., Volatile compounds produced by Lactobacillus fermentum, Saccharomyces cere-
visiae and Candida krusei in single starter culture fermentations of Ghanaian maize dough, J. Appl.
Microbiol., 94, 462, 2003.
34. Raju, S.B. and Rajjappa, S., Isolation and identification of Candida from the oral cavity, ISRN Dent,
article ID 487921, 2011.
35. Lewis, P., Perry, M.D. and Barnes, R.A., Candida, in Molecular Detection of Human Fungal Pathogens,
p. 551, Liu, D. (Ed.), CRC Press, Boca Raton, FL 2011.
36. Willinger, B. et al., Performance of Candida ID, a new chromogenic medium for presumptive identifica-
tion of Candida species, in comparison to CHROMagar Candida, J. Clin. Microbiol., 39, 3793, 2001.
37. Odds, F.C. and Bernaerts, R., CHROMagar Candida, a new differential isolation medium for presump-
tive identification of clinically important Candida species, J. Clin. Microbiol., 32, 1923, 1994.
38. Eraso, E. et al., Evaluation of the new chromogenic medium Candida ID 2 for isolation and identifica-
tion of Candida albicans and other medically important Candida species, J. Clin. Microbiol., 44, 3340,
2006.
39. Tornai-Lehoczki, J., Péter, G. and Dlauchy, D., CHROMagar Candida medium as a practical tool for
the differentiation and presumptive identification of yeast species isolated from salads, Int. J. Food
Microbiol., 86, 189, 2003.
40. Malhotra, S. et al., Recent diagnostic techniques in mycology, J. Med. Microbiol. Diagn., 3, 146, 2014.
41. Willinger, B. and Haase, G., State-of-the-art procedures and quality management in diagnostic medical
mycology, Curr. Fungal Infect. Rep., 7, 260, 2013.
42. Ellis, M. et al., Prospective evaluation of mannan and anti-mannan antibodies for diagnosis of invasive
Candida infections in patients with neutropenic fever, J. Med. Microbiol., 58, 606, 2009.
43. León, C., Ostrosky-Zeichner, L. and Schuster, M., What’s new in the clinical and diagnostic manage-
ment of invasive candidiasis in critically ill patients, Intensive Care Med., 40, 808, 2014.
44. Miranda, L.N., et al., Candida colonisation as a source for candidaemia, J. Hosp. Infect., 72, 9, 2009.
45. Andrade, M.J. et al., Efficiency of mitochondrial DNA restriction analysis and RAPD-PCR to charac-
terize yeasts growing on dry-cured Iberian ham at the different geographic areas of ripening, Meat Sci.,
84, 377, 2010.
46. Jamal, W.Y. et al., Comparative evaluation of two matrix-assisted laser desorption/ionization time-
of-flight mass spectrometry (MALDI-TOF MS) systems for the identification of clinically significant
yeasts, Int. J. Infect. Dis., 26, 167 2014.
47. Pulcrano, G. et al., Rapid and reliable MALDI-TOF mass spectrometry identification of Candida non-
albicans isolates from bloodstream infections, J. Microbiol. Methods, 94, 262, 2013.
48. Fricke, S. et al., A real-time PCR assay for the differentiation of Candida species, J. Appl. Microbiol.,
109, 1150, 2010.
49. Arancia, S. et al., Rapid, simple, and low-cost identification of Candida species using high-resolution
melting analysis, Diagn. Microbiol. Infect. Dis., 69, 283, 2011.
50. Decat, E. et al., Rapid and accurate identification of isolates of Candida species by melting peak
and melting curve analysis of the internally transcribed spacer region 2 fragment (ITS2-MCA), Res.
Microbiol., 164, 110, 2013.
51. Ahmad, S. et al., Seminested PCR for diagnosis of candidemia: comparison with culture, antigen detec-
tion, and biochemical methods for species identification, J. Clin. Microbiol., 40, 2483, 2002.
52. Taira, C. L. et al., A multiplex nested PCR for the detection and identification of Candida species in
blood samples of critically ill paediatric patients, BMC Infect. Dis., 14, 406, 2014.
53. Romeo, O. et al., A multiplex PCR protocol for rapid identification of Candida glabrata and its phylogeneti-
cally related species Candida nivariensis and Candida bracarensis, J. Microbiol. Methods, 79, 117, 2009.
54. Carvalho, A. et al., Multiplex PCR identification of eight clinically relevant Candida species, Med.
Mycol., 45, 619, 2007.
55. Loeffler, J. et al., Development and evaluation of the nuclisens basic kit NASBA for the detection of
RNA from Candida species frequently resistant to antifungal drugs, Diagn. Microbiol. Infect. Dis., 45,
217, 2003.
56. Pavlovic, M. et al., MALDI-TOF MS based identification of food-borne yeast isolates, J. Microbiol.
Methods, 106, 123, 2014.
Candida 509
83. Apidianakis, Y. and Rahme, L.G., Drosophila melanogaster as a model for human intestinal infection
and pathology, Dis. Model. Mech., 4, 21, 2011.
84. Glittenberg, M.T. et al., Pathogen and host factors are needed to provoke a systemic host response to
gastrointestinal infection of Drosophila larvae by Candida albicans, Dis. Model. Mech., 4, 515, 2011.
85. Pukkila-Worley, R. et al., Candida albicans hyphal formation and virulence assessed using a
Caenorhabditis elegans infection model, Eukaryot. Cell, 8, 1750, 2009.
86. Nett, J.E., Marchillo, K. and Andes, D.R., Modeling of fungal biofilms using a rat central-vein catheter,
Methods Mol. Biol., 845, 547, 2012.
87. Cencič, A. and Langerholc, T., Functional cell models of the gut and their applications in food
microbiology—a review, Int. J. Food Microbiol., 141, S4, 2010.
88. Sohn, K. et al., An in vitro assay to study the transcriptional response during adherence of Candida
albicans to different human epithelia, FEMS Yeast Res., 6, 1085, 2006.
89. Negri, M. et al., An in vitro evaluation of Candida tropicalis infectivity using human cell monolayers,
J. Med. Microbiol., 60, 1270, 2011.
90. Gácser, A. et al., Induction of human defensins by intestinal Caco-2 cells after interactions with
opportunistic Candida species, Microbes Infect., 16, 80, 2014.
91. Schaller, M. et al., Models of oral and vaginal candidiasis based on in vitro reconstituted human epithe-
lia, Nat. Protoc., 1, 2767, 2006.
92. Alves, C.T. et al., Candida albicans promotes invasion and colonisation of Candida glabrata in a recon-
stituted human vaginal epithelium, J. Infect., 69, 396, 2014.
93. Malic, S. et al., Characterisation of Candida albicans infection of an in vitro oral epithelial model using
confocal laser scanning microscopy, Oral Microbiol. Immunol., 22, 188, 2007.
33
Enterocytozoon bieneusi
CONTENTS
33.1 Structure and Life Cycle................................................................................................................511
33.2 Biochemistry and Physiology.........................................................................................................512
33.3 Immunology...................................................................................................................................512
33.4 Host Range.....................................................................................................................................513
33.5 Transmission..................................................................................................................................513
33.6 Clinical Symptoms and Pathology.................................................................................................514
33.7 Techniques for Detection and Characterization............................................................................514
33.7.1 Light Microscopy..............................................................................................................514
33.7.2 Electron Microscopy.........................................................................................................514
33.7.3 Immunofluorescence.........................................................................................................515
33.7.4 In Vitro Culture and Animal Models................................................................................515
33.7.5 Nucleic-Acid-Based Detection..........................................................................................515
33.7.6 Detection from Environmental Samples...........................................................................516
33.8 Treatment.......................................................................................................................................516
33.9 Conclusion and Perspectives..........................................................................................................517
References................................................................................................................................................517
Microsporidia are a large group of obligate intracellular parasites that infect almost all types of animals,
including humans, domesticated animals, wildlife, fish, and even insects. To date, over 1400 microspo-
ridian species have been described, with new species being discovered every year.1,2 Of these, 14 species
in 8 genera have been recognized as human pathogens.3 Recently, members of the phylum Microsporidia
have been shown to be related to fungi based on phylogenetic analysis of genes coding for tubulin and
mitochondrial-like HSP70, amongst others.4,5
As the most common cause of intestinal microsporidiosis,6 Enterocytozoon bieneusi was the only
known member of the genus Enterocytozoon until the recent identification of E. hepatopenaei from
shrimp.7,8 Since the mid-1980s, E. bieneusi has become an increasingly important opportunistic patho-
gen, especially in HIV/AIDS patients.9,10 While infective to both immunocompetent and immuno-
compromised hosts, E. bieneusi is associated with more persistent and severe diarrhea and wasting in
immunocompromised individuals.11 Given that E. bieneusi is unresponsive to therapies that are effective
against many microsporidia,12 there is an urgent need to utilize tissue culture and small animal models
to better understand the infection, transmission, and pathogenesis of this parasite and to develop more
efficient treatment strategies against E. bieneusi disease.
511
512 Laboratory Models for Foodborne Infections
HO
In host cell HN
N
N
N
HN N
N Early
proliferative HC N
Pt cells N
Sporogonial
ES plasmodium
ENDOGENOUS
stages
HC
Sporoblasts N
GI tract HN N
Spore coat N
EXOGENOUS
Excretion
Ingestion Pt
Spore
FIGURE 33.1 Diagram of Enterocytozoon bieneusi life cycle. HN, host cell nucleus; HC, host cell cytoplasm; N, nucleus
of developing E. bieneusi; Pt, polar tube. (Reprinted from Santin, M. and Fayer, R., Res. Vet. Sci., 90, 363–371, 2011. With
permission from Elsevier.)
begins to divide, resulting in multiple elongated nuclei, and electron-dense inclusions appear in the cell.
In the sporogonial plasmodium stage, polar tube elements are formed in the multinucleate plasmodium.
Later, polar tube complexes and nuclei segregate into sporoblasts, which develop further to become spores
that are shed with the host’s feces. The spores of E. bieneusi are oval, 1–2 μm in size, and resistant to most
environmental stresses. They have a cell wall consisting of three layers (plasma membrane, endospore, and
exospore) and a long coiled tubular extrusion apparatus called the polar tube. The polar tube plays a critical
role in invading and injecting infective material (sporoplasm) into the host cell.6,13 In mature spores, the polar
tubule has five to seven coils in two rows, as determined by transmission electron microscopy.
33.3 Immunology
Prior to the AIDS era, microsporidia were rarely reported in humans. Cell-mediated host immune
responses seem to be key in defense against E. bieneusi. With the widespread outbreaks of AIDS, a
Enterocytozoon bieneusi 513
significant number of microsporidiosis cases have been reported in HIV patients, especially in per-
sons with ≤100 CD4+ T cells per μL of blood.6,13,17,18 Highly activated antiretroviral therapy (HAART)
has dramatically reduced opportunistic infections in HIV/AIDS patients, including those caused by
microsporidia.19 However, intestinal microsporidiosis is still a major problem in some parts of the world
where access to HAART is limited. Additionally, E. bieneusi is being increasingly reported in non-HIV
immunosuppressed patients, including the elderly, patients with diabetes, those with malignant disease
undergoing chemotherapy, and those undergoing organ transplant.20–22 The lack of an animal model
and in vitro culture methods limit the work that can be carried out on E. bieneusi. As such, the detailed
immunological characteristics and host–organism relationship remain unclear.
33.4 Host Range
E. bieneusi was originally only reported in humans, both immunocompromised and immunocompetent
patients. However, many vertebrate animal hosts have now been shown to harbor E. bieneusi.23 There
remain a number of questions regarding the epidemiology of E. bieneusi, as the major host reservoirs and
routes of transmission are not completely understood. Fortunately, recent development of molecular tools
has allowed us to identify a wide range of animal hosts.23,24 Host specificity and zoonotic potential of an
organism can be evaluated by genotyping and phylogenetic analysis targeting the internal transcribed
spacer of the ribosomal RNA gene.23 Zoonotic and animal-specific E. bieneusi genotypes have been
identified from domesticated animals, companion animals, and wildlife, including primates, marmo-
sets, dogs, cats, pigs, cattle, horses, llamas, kudus, foxes, raccoons, otters, guinea pigs, beavers, rabbits,
muskrats, falcons, and some other birds.23 Cats and pigs are common hosts of zoonotic genotypes in
Asian countries.25–28 Phylogenetic analysis of E. bieneusi divides the ITS genotypes into eight groups.1,29
Group 1 is the largest, containing all human-specific, zoonotic, and animal-specific genotypes. Groups
2–8 are mostly only found in specific hosts and wastewater.1
33.5 Transmission
Transmission routes of E. bieneusi are not fully elucidated. In humans, vertical transmission has not been
reported, and the fecal–oral route is considered the main route of transmission. A report of E. bieneusi
in the respiratory tract also suggests the possibility of airborne transmission.30,31 Additionally, the spore
enables the organism to survive in the environment for long periods of time, contributing to both water-
and foodborne transmission. Risk factors associated with human microsporidiosis are homosexual prac-
tices, intravenous drug use, and water contact.32,33 As summarized in Table 33.1, a foodborne outbreak in
Sweden was attributed to contaminated cucumbers.34 E. bieneusi spores also have been identified on other
fresh food products and in milk.35,36 Waterborne transmission is frequently reported, with E. bieneusi
being isolated from surface water, waste water, irrigation water, swimming pools, and river water.25,37–49
In 1999, there was a confirmed waterborne microsporidium outbreak associated with a water distribu-
tion system.50 Numerous genotypes of E. bieneusi have also been identified from animals.23 Cama and
TABLE 33.1
Enterocytozoon bieneusi Detected in Food
Produce Type Country Detection Method Genotype References
Raspberries, bean Poland Microscopy (chromotrope 2R, Unknown 35
sprouts, lettuce calcofluor white), FISH
Cucumbers in salad and Sweden PCR C 34
sandwich
Milk Korea PCR D, I, J, CEbD, type IV 36
PCR, polymerase chain reaction; FISH, fluorescence in situ hybridization.
514 Laboratory Models for Foodborne Infections
colleagues reported the transmission of E. bieneusi between a child and pet guinea pigs,51 whereby an
unusual genotype was identified in both the guinea pigs and a 2-year-old child in the same household.
33.7.1 Light Microscopy
Microsporidia spores are difficult to identify in stool specimens by light microscopy because they are
very small (1–2 μm) and can look similar to bacteria. Chromotrope-based techniques, initially described
by Weber and colleagues,59 and then modified and further improved by other groups,60–63 are commonly
applied. Using a 5-min rapid-hot Gram-chromotrope staining method, the microsporidial spores stain
dark violet against a pale-green background, and a characteristic purple belt-like strip or bar in the
middle or at the end of the body is enhanced. Microsporidian spores can also be visualized by ultraviolet
microscopy using chemofluorescent stains such as Calcofluor White 2MR, Fungi-Fluor, or Uvitex 2B,
which bind chitin in the endospore.64 However, these types of stains are nonspecific and readily interact
with some bacteria, small fungi, and artifactual materials, resulting in false-positive results.1 Giemsa
stains microsporidia a light blue color, but the organisms are difficult to differentiate from other stool
elements, meaning this stain may only be of use in intestinal biopsies.17,65 Histological examination, such
as duodenal biopsy, can be used in some clinical situations. Various staining techniques for histological
examination have been studied including fluorescent Uvitex 2B stain, Gram-derived stain, silver stain-
ing, Giemsa stain, and chromotrope 2R stain.66–69
33.7.2 Electron Microscopy
Transmission electron microscopy (TEM) remains the gold standard for genus and species identification
of microsporidia. Structural features of the spores, intracellular proliferative forms, method of division,
and host–parasite interface are required for diagnosis and differentiation of the species. All developmen-
tal stages are observed in infected tissue samples, but not in feces. Therefore, fecal samples alone are
not sufficient for definitive classification of microsporidia.70 The major disadvantages of TEM are low
sensitivity, high cost, and laborious sample preparation. TEM is also not suitable for routine diagnosis.71
The classification of microsporidia by TEM and detailed characteristics of E. bieneusi are described
elsewhere.6,72
Enterocytozoon bieneusi 515
33.7.3 Immunofluorescence
Fluorescein-tagged antibodies are useful for detecting pathogens in clinical biopsy samples from
humans and animals.73,74 Immunofluorescence techniques, especially the use of polyclonal antibodies,
allow visualization of all E. bieneusi development stages, including spores, intracellular developmental
stages, and extruded polar tubes, whereas histochemical methods only stain the walls of sporoblasts
or spores.75,76 However, polyclonal immunofluorescence staining frequently cross-reacts with yeast and
bacteria in fecal specimens, and the sensitivity of the technique is poor compared with chromotrope or
chemofluorescent stains.77 Monoclonal antibodies against E. bieneusi spores do not appear to cross react
with Encephalitozoon species, but are not available for commercial distribution.78,79
33.7.5 Nucleic-Acid-Based Detection
Nucleic-acid-based detection methods are more sensitive and specific than microscopy, and therefore
have been widely applied.24,87 The most commonly used method is polymerase chain reaction (PCR).
In PCR, the target pathogen DNA is bound by a specific set of primers, and the original few copies
of DNA are amplified across several orders of magnitude, generating millions of copies of a particu-
lar DNA sequence. Molecular diagnostic tests for microsporidia are not routinely available in clinical
diagnostic laboratories despite being widely used in research settings. PCR diagnosis of E. bieneusi
was first reported by Zhu and colleagues.88 PCR methods can also be used for more in-depth analyses,
such as genotypic identification at the subspecies level. The primers generally used for the diagnosis
of E. bieneusi target the small and large subunits and internal transcribed spacer (ITS) region of the
rRNA gene. The ITS region in particular has been used in many studies for detecting and genotyping
E. bieneusi because of the high degree of sequence diversity in this region. Although the ITS sequence
remains the gold standard for the analysis of E. bieneusi, additional gene markers are being sought and,
more recently, a multilocus sequence typing assay targeting three microsatellite and one minisatellite
markers has been developed.23,89
Real-time PCR detects accumulating amplicons in real time via either nonspecific fluorochrome or
specific fluorescently-labeled probes. Real-time PCR has the advantage of being quantitative over a
broad dynamic range, but is relatively expensive. A few real-time PCR analyses have been reported for
detection of E. bieneusi.90,91
Several studies have used fluorescent in situ hybridization (FISH)-based techniques to detect
E. bieneusi. FISH is a cytogenetic technique using a fluorescently-labeled probe that binds to comple-
mentary nucleic acid (DNA or RNA) in the specimen.92 It was used successfully to detect E. bieneusi
from clinical samples, with probes targeting the small subunit or ITS regions of the rRNA.93,94 FISH has
the advantage of providing general morphological information, as the procedure is performed in situ.
The major disadvantages of FISH are the laborious method and lower sensitivity than PCR.
A recently developed oligonucleotide microarray method can simultaneously detect E. bieneusi and
Encephalitozoon species from clinical samples.95 An array of target-complementary DNA fragments
are spotted on a glass slide, and the nucleic acid from the sample is hybridized to the chip after being
fluorescently labeled. This technique is somewhat quantitative; the abundance of the DNA in the sample
is correlated with the intensity of fluorescence. Additionally, this method is high throughput.
516 Laboratory Models for Foodborne Infections
TABLE 33.2
Enterocytozoon bieneusi Detected in Water
Water Source Country Detection Method Genotype References
River France PCR Unknown 41
Surface water, ground USA PCR Unknown 37
water
River Korea Uvitex 2B, PCR Unknown 42
River Ireland FISH, PCR Unknown 43
Lake France PCR Unknown 44
Recreational water USA FISH, PCR Unknown 45
Surface inland and Ireland FISH Unknown 104
coastal waters
Wetlands Ireland FISH, PCR K 46
Waste water Ireland FISH Unknown 47
Waste water China PCR Type IV, EbpA, EbpC, EbpD, 40
Peru6, Peru8, Peru10, C, D,
BEB6, PtEb IV, PigEBITS7,
PigEBITS8, WL4, WL12, WL14,
WW1–WW9
Waste water Tunisia PCR D, IV, etc. 39
Storm water USA PCR WL4, WL6, SW1 to SW3 48
Drinking water, waste Spain Chromotrope stain, PCR C, D, and D-like 49
water
River water China PCR EbpA, EbpB, EbpC, D, CS-8, 38
PtEb IX, Peru8, Peru11,
PigEBITS4, G, O, RWSH1 to
RWSH6
PCR, polymerase chain reaction; FISH, fluorescence in situ hybridization.
33.8 Treatment
Albendazole, a benzimidazole drug used to treat of a variety of parasitic infections, is highly active
against Encephalitozoon species, but has shown only limited efficacy against E. bieneusi. When
albendazole is used in patients infected with E. bieneusi, diarrhea may improve in some patients,
but the excretion of the organism continues and diarrhea exacerbates rapidly after discontinuation of
the drug.97–99 Albendazole works by binding to β-tubulin, and variations in the amino acid sequence
of E. bieneusi β-tubulin may be related to clinical resistance.100 Fumagillin, originally isolated from
Aspergillus fumigatus, is an antimicrobial agent that is effective against E. bieneusi and Entamoeba his-
tolytica. Fumagillin has successfully treated E. bieneusi infection in AIDS and transplant patients.101,102
The main toxicity is thrombocytopenia, which is reversible after cessation of treatment. However, more
severe side effects, such as aseptic meningoencephalitis, have also been reported.103
Enterocytozoon bieneusi 517
REFERENCES
1. Thellier, M. & Breton, J. Enterocytozoon bieneusi in human and animals, focus on laboratory identifica-
tion and molecular epidemiology. Parasite 15, 349–58 (2008).
2. Szumowski, S.C. & Troemel, E.R. Microsporidia-host interactions. Curr Opin Microbiol 26, 10–16
(2015).
3. Didier, E.S. Microsporidiosis: an emerging and opportunistic infection in humans and animals. Acta
Trop 94, 61–76 (2005).
4. Vivares, C.P., Gouy, M., Thomarat, F. & Metenier, G. Functional and evolutionary analysis of a eukary-
otic parasitic genome. Curr Opin Microbiol 5, 499–505 (2002).
5. Keeling, P.J. & Fast, N.M. Microsporidia: biology and evolution of highly reduced intracellular para-
sites. Annu Rev Microbiol 56, 93–116 (2002).
6. Desportes, I. et al. Occurrence of a new microsporidan: Enterocytozoon bieneusi n. g., n. sp., in the
enterocytes of a human patient with AIDS. J Protozool 32, 250–4 (1985).
7. Tourtip, S. et al. Enterocytozoon hepatopenaei sp. nov. (Microsporida: Enterocytozoonidae), a parasite
of the black tiger shrimp Penaeus monodon (Decapoda: Penaeidae): fine structure and phylogenetic
relationships. J Invertebr Pathol 102, 21–9 (2009).
8. Tangprasittipap, A. et al. The microsporidian Enterocytozoon hepatopenaei is not the cause of white
feces syndrome in whiteleg shrimp Penaeus (Litopenaeus) vannamei. BMC Vet Res 9, 139 (2013).
9. Cali, A. General microsporidian features and recent findings on AIDS isolates. J Protozool 38, 625–30
(1991).
10. Weiss, L.M. Microsporidia: emerging pathogenic protists. Acta Trop 78, 89–102 (2001).
11. Tumwine, J.K. et al. Enterocytozoon bieneusi among children with diarrhea attending Mulago Hospital
in Uganda. Am J Trop Med Hyg 67, 299–303 (2002).
12. Didier, E.S. & Weiss, L.M. Microsporidiosis: current status. Curr Opin Infect Dis 19, 485–92 (2006).
13. Orenstein, J.M. Microsporidiosis in the acquired immunodeficiency syndrome. J Parasitol 77, 843–64
(1991).
14. Vavra, J. & Lukes, J. Microsporidia and ‘the art of living together’. Adv Parasitol 82, 253–319 (2013).
15. Weidner, E. The microsporidian spore invasion tube. III. Tube extrusion and assembly. J Cell Biol 93,
976–9 (1982).
16. Keeling, P.J. et al. The reduced genome of the parasitic microsporidian Enterocytozoon bieneusi lacks
genes for core carbon metabolism. Genome Biol Evol 2, 304–9 (2010).
17. van Gool, T. et al. Diagnosis of Enterocytozoon bieneusi microsporidiosis in AIDS patients by recovery
of spores from faeces. Lancet 336, 697–8 (1990).
18. van Gool, T. et al. High prevalence of Enterocytozoon bieneusi infections among HIV-positive individu-
als with persistent diarrhoea in Harare, Zimbabwe. Trans R Soc Trop Med Hyg 89, 478–80 (1995).
518 Laboratory Models for Foodborne Infections
19. Stark, D. et al. Limited genetic diversity among genotypes of Enterocytozoon bieneusi strains isolated
from HIV-infected patients from Sydney, Australia. J Med Microbiol 58, 355–7 (2009).
20. Lono, A.R., Kumar, S. & Chye, T.T. Incidence of microsporidia in cancer patients. J Gastrointest
Cancer 39, 124–9 (2008).
21. Antonios, S.N., Tolba, O.A., Othman, A.A. & Saad, M.A. A preliminary study on the prevalence of
parasitic infections in immunocompromised children. J Egypt Soc Parasitol 40, 617–30 (2010).
22. Sak, B. et al. Unapparent microsporidial infection among immunocompetent humans in the Czech
Republic. J Clin Microbiol 49, 1064–70 (2011).
23. Santin, M. & Fayer, R. Microsporidiosis: Enterocytozoon bieneusi in domesticated and wild animals.
Res Vet Sci 90, 363–71 (2011).
24. Fedorko, D.P., Nelson, N.A. & Cartwright, C.P. Identification of microsporidia in stool specimens by
using PCR and restriction endonucleases. J Clin Microbiol 33, 1739–41 (1995).
25. Li, W. et al. High diversity of human-pathogenic Enterocytozoon bieneusi genotypes in swine in north-
east China. Parasitol Res 113, 1147–53 (2014).
26. Li, W. et al. Genotypes of Enterocytozoon bieneusi in livestock in China: high prevalence and zoonotic
potential. PLoS One 9, e97623 (2014).
27. Mori, H. et al. Presence of zoonotic Enterocytozoon bieneusi in cats in a temple in central Thailand. Vet
Parasitol 197, 696–701 (2013).
28. Karim, M.R. et al. Genetic diversity in Enterocytozoon bieneusi isolates from dogs and cats in China:
host specificity and public health implications. J Clin Microbiol 52, 3297–302 (2014).
29. Karim, M.R. et al. Genetic polymorphism and zoonotic potential of Enterocytozoon bieneusi from
nonhuman primates in China. Appl Environ Microbiol 80, 1893–8 (2014).
30. Didier, E.S. et al. Epidemiology of microsporidiosis: sources and modes of transmission. Vet Parasitol
126, 145–66 (2004).
31. Weber, R. et al. Pulmonary and intestinal microsporidiosis in a patient with the acquired immunodefi-
ciency syndrome. Am Rev Respir Dis 146, 1603–5 (1992).
32. Hutin, Y.J. et al. Risk factors for intestinal microsporidiosis in patients with human immunodeficiency
virus infection: a case-control study. J Infect Dis 178, 904–7 (1998).
33. Dascomb, K., Frazer, T., Clark, R.A., Kissinger, P. & Didier, E. Microsporidiosis and HIV. J Acquir
Immune Defic Syndr 24, 290–2 (2000).
34. Decraene, V., Lebbad, M., Botero-Kleiven, S., Gustavsson, A.M. & Lofdahl, M. First reported food-
borne outbreak associated with microsporidia, Sweden, October 2009. Epidemiol Infect 140, 519–27
(2012).
35. Jedrzejewski, S., Graczyk, T.K., Slodkowicz-Kowalska, A., Tamang, L. & Majewska, A.C. Quantitative
assessment of contamination of fresh food produce of various retail types by human-virulent microspo-
ridian spores. Appl Environ Microbiol 73, 4071–3 (2007).
36. Lee, J.H. Molecular detection of Enterocytozoon bieneusi and identification of a potentially human-
pathogenic genotype in milk. Appl Environ Microbiol 74, 1664–6 (2008).
37. Dowd, S.E., Gerba, C.P. & Pepper, I.L. Confirmation of the human-pathogenic microsporidia
Enterocytozoon bieneusi, Encephalitozoon intestinalis, and Vittaforma corneae in water. Appl Environ
Microbiol 64, 3332–5 (1998).
38. Hu, Y., Feng, Y., Huang, C. & Xiao, L. Occurrence, source, and human infection potential of
Cryptosporidium and Enterocytozoon bieneusi in drinking source water in Shanghai, China, during a
pig carcass disposal incident. Environ Sci Technol 48, 14219–27 (2014).
39. Ben Ayed, L. et al. Survey and genetic characterization of wastewater in Tunisia for Cryptosporidium
spp., Giardia duodenalis, Enterocytozoon bieneusi, Cyclospora cayetanensis and Eimeria spp. J Water
Health 10, 431–44 (2012).
40. Li, N. et al. Molecular surveillance of Cryptosporidium spp., Giardia duodenalis, and Enterocytozoon
bieneusi by genotyping and subtyping parasites in wastewater. PLoS Negl Trop Dis 6, e1809 (2012).
41. Sparfel, J.M. et al. Detection of microsporidia and identification of Enterocytozoon bieneusi in surface
water by filtration followed by specific PCR. J Eukaryot Microbiol 44, 78S (1997).
42. Fournier, S. et al. Detection of microsporidia in surface water: a one-year follow-up study. FEMS
Immunol Med Microbiol 29, 95–100 (2000).
43. Graczyk, T.K. et al. Human waterborne parasites in zebra mussels (Dreissena polymorpha) from the
Shannon River drainage area, Ireland. Parasitol Res 93, 385–91 (2004).
Enterocytozoon bieneusi 519
44. Coupe, S. et al. Detection of Cryptosporidium, Giardia and Enterocytozoon bieneusi in surface water,
including recreational areas: a one-year prospective study. FEMS Immunol Med Microbiol 47, 351–9
(2006).
45. Graczyk, T.K., Sunderland, D., Tamang, L., Lucy, F.E. & Breysse, P.N. Bather density and levels of
Cryptosporidium, Giardia, and pathogenic microsporidian spores in recreational bathing water.
Parasitol Res 101, 1729–31 (2007).
46. Graczyk, T.K. et al. Human zoonotic enteropathogens in a constructed free-surface flow wetland.
Parasitol Res 105, 423–8 (2009).
47. Cheng, H.W., Lucy, F.E., Graczyk, T.K., Broaders, M.A. & Mastitsky, S.E. Municipal wastewater treat-
ment plants as removal systems and environmental sources of human-virulent microsporidian spores.
Parasitol Res 109, 595–603 (2011).
48. Guo, Y. et al. Host specificity and source of Enterocytozoon bieneusi genotypes in a drinking source
watershed. Appl Environ Microbiol 80, 218–25 (2014).
49. Galvan, A.L. et al. Molecular characterization of human-pathogenic microsporidia and Cyclospora
cayetanensis isolated from various water sources in Spain: a year-long longitudinal study. Appl Environ
Microbiol 79, 449–59 (2013).
50. Cotte, L. et al. Waterborne outbreak of intestinal microsporidiosis in persons with and without human
immunodeficiency virus infection. J Infect Dis 180, 2003–8 (1999).
51. Cama, V.A. et al. Transmission of Enterocytozoon bieneusi between a child and guinea pigs. J Clin
Microbiol 45, 2708–10 (2007).
52. Sandfort, J. et al. Enterocytozoon bieneusi infection in an immunocompetent patient who had acute diar-
rhea and who was not infected with the human immunodeficiency virus. Clin Infect Dis 19, 514–6 (1994).
53. Kotler, D.P. & Orenstein, J.M. Clinical syndromes associated with microsporidiosis. Adv Parasitol 40,
321–49 (1998).
54. Remadi, S., Dumais, J., Wafa, K. & MacGee, W. Pulmonary microsporidiosis in a patient with the
acquired immunodeficiency syndrome. A case report. Acta Cytol 39, 1112–6 (1995).
55. Mor, S.M., Tumwine, J.K., Naumova, E.N., Ndeezi, G. & Tzipori, S. Microsporidiosis and malnutrition
in children with persistent diarrhea, Uganda. Emerg Infect Dis 15, 49–52 (2009).
56. Orenstein, J.M., Tenner, M. & Kotler, D.P. Localization of infection by the microsporidian Enterocytozoon
bieneusi in the gastrointestinal tract of AIDS patients with diarrhea. AIDS 6, 195–7 (1992).
57. Schwartz, D.A. et al. Enteric Opportunistic Infections Working Group. The presence of Enterocytozoon
bieneusi spores in the lamina propria of small bowel biopsies with no evidence of disseminated micro-
sporidiosis. Arch Pathol Lab Med 119, 424–8 (1995).
58. Schwartz, D.A., Sobottka, I., Leitch, G.J., Cali, A. & Visvesvara, G.S. Pathology of microsporidiosis:
emerging parasitic infections in patients with acquired immunodeficiency syndrome. Arch Pathol Lab
Med 120, 173–88 (1996).
59. Weber, R. et al. The Enteric Opportunistic Infections Working Group. Improved light-microscopical
detection of microsporidia spores in stool and duodenal aspirates. N Engl J Med 326, 161–6 (1992).
60. Kokoskin, E. et al. Modified technique for efficient detection of microsporidia. J Clin Microbiol 32,
1074–5 (1994).
61. Ryan, N.J. et al. A new trichrome-blue stain for detection of microsporidial species in urine, stool, and
nasopharyngeal specimens. J Clin Microbiol 31, 3264–9 (1993).
62. Ignatius, R. et al. A new acid-fast trichrome stain for simultaneous detection of Cryptosporidium
parvum and microsporidial species in stool specimens. J Clin Microbiol 35, 446–9 (1997).
63. Moura, H. et al. A new and improved “quick-hot Gram-chromotrope” technique that differentially
stains microsporidian spores in clinical samples, including paraffin-embedded tissue sections. Arch
Pathol Lab Med 121, 888–93 (1997).
64. van Gool, T. et al. Diagnosis of intestinal and disseminated microsporidial infections in patients with
HIV by a new rapid fluorescence technique. J Clin Pathol 46, 694–9 (1993).
65. Rijpstra, A.C., Canning, E.U., Van Ketel, R.J., Eeftinck Schattenkerk, J.K. & Laarman, J.J. Use of
light microscopy to diagnose small-intestinal microsporidiosis in patients with AIDS. J Infect Dis 157,
827–31 (1988).
66. Conteas, C.N. et al. Fluorescence techniques for diagnosing intestinal microsporidiosis in stool, enteric
fluid, and biopsy specimens from acquired immunodeficiency syndrome patients with chronic diarrhea.
Arch Pathol Lab Med 120, 847–53 (1996).
520 Laboratory Models for Foodborne Infections
67. Field, A.S., Marriott, D.J. & Hing, M.C. The Warthin-Starry stain in the diagnosis of small intestinal
microsporidiosis in HIV-infected patients. Folia Parasitol (Praha) 40, 261–6 (1993).
68. Franzen, C. et al. Tissue diagnosis of intestinal microsporidiosis using a fluorescent stain with Uvitex
2B. J Clin Pathol 48, 1009–10 (1995).
69. Kotler, D.P., Giang, T.T., Garro, M.L. & Orenstein, J.M. Light microscopic diagnosis of microsporidi-
osis in patients with AIDS. Am J Gastroenterol 89, 540–4 (1994).
70. Didier, E.S. et al. Identification and characterization of three Encephalitozoon cuniculi strains.
Parasitology 111 (Pt 4), 411–21 (1995).
71. Field, A.S. Light microscopic and electron microscopic diagnosis of gastrointestinal opportunistic
infections in HIV-positive patients. Pathology 34, 21–35 (2002).
72. Cali, A. & Owen, R.L. Intracellular development of Enterocytozoon, a unique microsporidian found in
the intestine of AIDS patients. J Protozool 37, 145–55 (1990).
73. Weber, R., Bryan, R.T., Schwartz, D.A. & Owen, R.L. Human microsporidial infections. Clin Microbiol
Rev 7, 426–61 (1994).
74. Weiss, L.M. & Vossbrinck, C.R. Microsporidiosis: molecular and diagnostic aspects. Adv Parasitol 40,
351–95 (1998).
75. Sheoran, A.S. et al. Purification of Enterocytozoon bieneusi from stools and production of specific anti-
bodies. J Clin Microbiol 43, 387–92 (2005).
76. Sheoran, A.S. et al. Monoclonal antibodies against Enterocytozoon bieneusi of human origin. Clin
Diagn Lab Immunol 12, 1109–13 (2005).
77. Garcia, L.S., Shimizu, R.Y. & Bruckner, D.A. Detection of microsporidial spores in fecal specimens
from patients diagnosed with cryptosporidiosis. J Clin Microbiol 32, 1739–41 (1994).
78. Accoceberry, I. et al. Production of monoclonal antibodies directed against the microsporidium
Enterocytozoon bieneusi. J Clin Microbiol 37, 4107–12 (1999).
79. Zhang, Q. et al. Production and characterization of monoclonal antibodies against Enterocytozoon
bieneusi purified from rhesus macaques. Infect Immun 73, 5166–72 (2005).
80. Feng, X. et al. Serial propagation of the microsporidian Enterocytozoon bieneusi of human origin in
immunocompromised rodents. Infect Immun 74, 4424–9 (2006).
81. Tzipori, S. et al. Transmission and establishment of a persistent infection of Enterocytozoon bieneusi,
derived from a human with AIDS, in simian immunodeficiency virus-infected rhesus monkeys. J Infect
Dis 175, 1016–20 (1997).
82. Sestak, K. et al. Quantitative evaluation of Enterocytozoon bieneusi infection in simian immunodefi-
ciency virus-infected rhesus monkeys. J Med Primatol 32, 74–81 (2003).
83. Mansfield, K.G. et al. Localization of persistent Enterocytozoon bieneusi infection in normal rhesus
macaques (Macaca mulatta) to the hepatobiliary tree. J Clin Microbiol 36, 2336–8 (1998).
84. Visvesvara, G. et al. Short-term in vitro culture and molecular analysis of the microsporidian,
Enterocytozoon bieneusi. J Eukaryot Microbiol 42, 506–10 (1995).
85. Desportes-Livage, I. et al. Comparative development of two microsporidian species: Enterocytozoon
bieneusi and Enterocytozoon salmonis, reported in AIDS patients and salmonid fish, respectively.
J Eukaryot Microbiol 43, 49–60 (1996).
86. Visvesvara, G.S. et al. Adenovirus masquerading as microsporidia. J Parasitol 82, 316–9 (1996).
87. Franzen, C. & Muller, A. Molecular techniques for detection, species differentiation, and phylogenetic
analysis of microsporidia. Clin Microbiol Rev 12, 243–85 (1999).
88. Zhu, X. et al. Small subunit rRNA sequence of Enterocytozoon bieneusi and its potential diagnostic role
with use of the polymerase chain reaction. J Infect Dis 168, 1570–5 (1993).
89. Karim, M.R. et al. Multilocus sequence typing of Enterocytozoon bieneusi in nonhuman primates in
China. Vet Parasitol 200, 13–23 (2014).
90. Wumba, R. et al. Enterocytozoon bieneusi identification using real-time polymerase chain reaction
and restriction fragment length polymorphism in HIV-infected humans from Kinshasa Province of the
Democratic Republic of Congo. J Parasitol Res 2012, 278028 (2012).
91. Verweij, J.J., Ten Hove, R., Brienen, E.A. & van Lieshout, L. Multiplex detection of Enterocytozoon
bieneusi and Encephalitozoon spp. in fecal samples using real-time PCR. Diagn Microbiol Infect Dis
57, 163–7 (2007).
92. Langer-Safer, P.R., Levine, M. & Ward, D.C. Immunological method for mapping genes on Drosophila
polytene chromosomes. Proc Natl Acad Sci USA 79, 4381–5 (1982).
Enterocytozoon bieneusi 521
93. Carville, A. et al. Development and application of genetic probes for detection of Enterocytozoon
bieneusi in formalin-fixed stools and in intestinal biopsy specimens from infected patients. Clin Diagn
Lab Immunol 4, 405–8 (1997).
94. Velasquez, J.N. et al. In situ hybridization: a molecular approach for the diagnosis of the microsporidian
parasite Enterocytozoon bieneusi. Hum Pathol 30, 54–8 (1999).
95. Wang, Z., Orlandi, P.A. & Stenger, D.A. Simultaneous detection of four human pathogenic microsporid-
ian species from clinical samples by oligonucleotide microarray. J Clin Microbiol 43, 4121–8 (2005).
96. Borchardt, M.A. & Spencer, S.K. Concentration of Cryptosporidium, microsporidia and other water-
borne pathogens by continuous separation channel centrifugation. J Appl Microbiol 92, 649–56 (2002).
97. Blanshard, C., Ellis, D.S., Tovey, D.G., Dowell, S. & Gazzard, B.G. Treatment of intestinal microspo-
ridiosis with albendazole in patients with AIDS. AIDS 6, 311–3 (1992).
98. Dieterich, D.T., Lew, E.A., Kotler, D.P., Poles, M.A. & Orenstein, J.M. Treatment with albendazole
for intestinal disease due to Enterocytozoon bieneusi in patients with AIDS. J Infect Dis 169, 178–83
(1994).
99. Leder, K., Ryan, N., Spelman, D. & Crowe, S.M. Microsporidial disease in HIV-infected patients: a
report of 42 patients and review of the literature. Scand J Infect Dis 30, 331–8 (1998).
100. Akiyoshi, D.E. et al. Analysis of the β-tubulin genes from Enterocytozoon bieneusi isolates from a
human and rhesus macaque. J Eukaryot Microbiol 54, 38–41 (2007).
101. Champion, L. et al. Fumagillin for treatment of intestinal microsporidiosis in renal transplant recipients.
Am J Transplant 10, 1925–30 (2010).
102. Molina, J.M. et al. Fumagillin treatment of intestinal microsporidiosis. N Engl J Med 346, 1963–9
(2002).
103. Audemard, A. et al. Fumagillin-induced aseptic meningoencephalitis in a kidney transplant recipient
with microsporidiosis. Transpl Infect Dis 14, E147–9 (2012).
104. Lucy, F.E., Graczyk, T.K., Tamang, L., Miraflor, A. & Minchin, D. Biomonitoring of surface and coastal
water for Cryptosporidium, Giardia, and human-virulent microsporidia using molluscan shellfish.
Parasitol Res 103, 1369–75 (2008).
34
Fusarium
CONTENTS
34.1 Introduction................................................................................................................................... 523
34.1.1 The Genus Fusarium........................................................................................................ 523
34.1.2 Mycotoxins of Fusarium Species and Mycotoxicoses..................................................... 524
34.1.2.1 Zearalenone....................................................................................................... 524
34.1.2.2 Fumonisins........................................................................................................ 524
34.1.2.3 Trichothecenes.................................................................................................. 525
34.2 L aboratory Models for Foodborne Fusarium Mycotoxicoses...................................................... 525
34.2.1 Rodents............................................................................................................................. 525
34.2.2 Human and Animal Cell Lines.........................................................................................539
34.3 C onclusions................................................................................................................................... 546
Acknowledgments................................................................................................................................... 546
References............................................................................................................................................... 546
34.1 Introduction
The members of the genus Fusarium are hyaline filamentous fungi and are found largely as saprophytic
organisms in soil. Fusaria cause a range of infections collectively known as fusariosis and have been doc-
umented as etiological agents in localized tissue infections, keratitis, endophthalmitis, septic arthritis,
cystitis, peritonitis, brain abscesses, and breast abscess. The mycotoxins of these fungi are involved in
the infectious processes and may serve as potential virulence factors. Fusarium is also one of the major
fungal genera associated with maize and other cereals throughout the world. Several species are the most
prolific producers of mycotoxins and are frequently associated with mycotoxicoses in both humans and
animals. This chapter intends to provide an overview about the rodents (rats and mice) and mammalian
cell lines that were most recently used as laboratory models to study Fusarium mycotoxicoses.
523
524 Laboratory Models for Foodborne Infections
of devastating diseases, e.g., head blight of wheat and barley, wilt of bananas [4], sudden death syndrome
of soybean [5], crown and root rot of tomato [6], and vascular wilt of tomato [7].
Fusarium spp. are also associated with a broad spectrum of human infections, including superficial
(i.e., keratitis and onychomycosis), invasive localized, and disseminated infections, allergic diseases
(i.e., sinusitis), and mycotoxicosis [8]. Out of the 20 species complexes of the genus, seven comprise
clinically relevant Fusaria: the F. chlamydosporum species complex (FCSC), the Fusarium dimerum
species complex (FDSC), the F. fujikuroi species complex (FFSC), the F. incarnatum-equiseti species
complex (FIESC), the F. oxysporum species complex (FOSC), the F. sambucinum species complex
(FSAMSC, including Fusarium sporotrichioides), and the FSSC. Among them, the FSSC—which
comprises at least 60 haplotypes—is the most common group, accounting for 50%–60% of all fusari-
oses worldwide [9,10].
Certain Fusarium species are also able to produce mycotoxins, which may contribute to plant and
human pathogenesis [11]. Besides the direct negative impact of fungal infections on crop yield, the myco-
toxin contamination of animal feeds also causes significant financial loss and damage to agriculture
worldwide. The major Fusarium mycotoxins frequently occurring in cereal grains and animal feeds are
fumonisins, trichothecenes, and zearalenone.
34.1.2.2 Fumonisins
Fumonisins have been discovered in 1988 in South Africa from cultures of the F. moniliforme strain
MRC 826 (=F. verticillioides, teleomorph: Gibberella moniliformis) [22]. The chemical structure of this
cancer-promoting compound was elucidated in the same by Bezuidenhout et al. [23]. The most impor-
tant fumonisin-producing Fusaria are the members of section Liseola, including the widespread maize
pathogens F. verticillioides and F. proliferatum. Other sections containing fumonisin-producing strains
are Dlaminia, Elegans, and Arthrosporiella. In section Elegans, F. oxysporum produces only C-series
fumonisins [24].
Fusarium 525
Fumonisins are polyketide-derived mycotoxins and can be divided into four groups (A, B, C, and P)
based on their chemical structure with several isomers and stereomers identified during the past decade
[25–28]. The most prevalent and naturally occurring fumonisins belong to type B analogues (FB). In
Fusaria, usually FB1 is predominant, accounting for 70%–80% of the total fumonisin content [29]. The
A and B series of fumonisins consist of a 20 carbon atom long backbone, while the C-type fumonisins
are 19 carbon atoms long. Fumonisins are sphingosine-analog compounds, which disrupt the biosyn-
thesis of sphingolipids by the inhibition of the ceramide synthase enzyme [30]. The altered sphingo-
lipid metabolism could lead to neural tube defects through the disrupted folate uptake [31]. Ingestion of
fumonisin-contaminated feeds is associated with several fatal diseases in domestic animals, for example,
equine leukoencephalomalacia (ELEM) and porcine pulmonary edema (PPE). Fumonisins can cause
nephrotoxicity, hepatotoxicity, and hepatocarcinogenicity in laboratory animals [32]. Fumonisins are
the possible causative agents of esophageal cancer in several countries like China and Transkei in South
Africa [33,34]; therefore, fumonisin B1 is considered as possibly carcinogenic to humans (Group 2B) by
the International Agency for Research on Cancer (IARC) [35].
34.1.2.3 Trichothecenes
Fusaria and other mold fungi such as Mycothecium, Trichoderma, Trichothecium, Stachybotrys,
Verticimonosporium, and Cephalosporium produce trichothecenes, which comprise a family of closely
related low-molecular-weight, nonvolatile, and relatively water-insoluble compounds known as sesquiter-
penoids. They are classified into four types—A, B, C, and D, among which Fusaria produce type A (like
T-2 and HT-2 toxin) and type B [like deoxynivalenol (DON) and nivalenol (NIV)] trichothecenes [36].
Fusarium species are probably the most cited and among the most prolific trichothecene-producing
fungi [37]. Trichothecenes contaminate many field crops across the world, and it is necessary to screen
large number of food samples for the presence of these toxic fungal metabolites [38].
Trichothecenes are powerful inhibitors of eukaryotic protein synthesis and are phytotoxic, insecti-
cidal, and toxic to animals, and some are among the most toxic non-nitrogenous compounds known
to man. Several are commonly found in cereal grains, and the potential health risk from contaminated
animal feed and human food is a major factor in stimulating research into this group of compounds
[39–42]. Common manifestations of trichothecene toxicity are depression of immune responses and
nausea, sometimes vomiting [43].
Wang et al. [44] reported human toxicosis in China caused by moldy rice contaminated with T-2 toxin
of F. heterosporum and F. graminearum. The chief symptoms were nausea, dizziness, vomiting, chills,
abdominal distension, abdominal pain, thoracic stuffiness, and diarrhea. T-2 mycotoxicosis—or “moldy
corn disease”—in pigs is characterized by multiple hemorrhages on the serosa of the liver, stomach, and
esophagus (at necropsy). T-2 also has an important impact on reproductive performance in pigs [45]. The
in vivo toxicity of T-2 and H-2 is explicitly described by EFSA Panel on CONTAM [46]. Wu et al. [47]
stated that in human cell lines, HT-2 and neosolaniol (NEO) are the major metabolites of T-2 toxin. The
T-2 degradation products are less cytotoxic compared to T-2 toxin [48].
DON—or vomitoxin—is one of the most common mycotoxins produced by F. graminearum and
F. culmorum found in grains. When ingested in high doses by agricultural animals, it causes nau-
sea, vomiting, and diarrhea; at lower doses, pigs and other farm animals exhibit weight loss and food
refusal [49]. Human or animal intoxications with either DON or NIV are much less likely to be fatal than
those with T-2 [50].
and—in cases when full-text access was not available—abstracts from the PubMed database. The
search was performed in article titles and abstracts using the following keyword combinations in order
to find specific literature: zearalenone OR fumonisin OR T-2 toxin OR deoxynivalenol AND rat OR
mouse OR mice. The search was limited to publications from the year 2010 onwards in order to focus
on recent literature. Articles and abstracts were screened to include studies with relevant information
(exact definition of the mycotoxin studied and the animal model, details about the way of administra-
tion, and the dosage).
Table 34.1 gives an overview of the analyses of Fusarium mycotoxins utilizing rat models [51–74]. Out
of 24 publications analyzed (1 DON, 10 T-2, 7 ZEA, and 6 FB1), it was found that Sprague Dawley rats
were mostly used, followed by Wistar rats. There is only one study included for DON [58] in 8-week-old
axenic male Sprague Dawley rats, where the concentration of E. coli bacteria decreased in the gut at day
27 after subchronic exposure to DON.
Among the 10 studies analyzed for T-2 toxin, 7 studies utilized Wistar rats to investigate MRI in the
tibial bone abnormalities [66,69], chondrocyte necrosis of Kashin Bede disease (KBD) [67], cardiac
and autonomic nervous effect [68], reproductive toxicity [70], KBD [69,71,72], and apoptotic induction
mechanism [70]. The remaining three studies with Sprague Dawley rats analyzed the effect of T-2 on
lipid peroxidation in brain [53,63] and KBD [57].
Out of seven studies analyzing ZEA in rat models, the most popular model animals were Sprague
Dawley rats in the cases the analyses were related to the effect of ZEA on the reproductive system
[51,52,56,64] and modification of mRNA levels involved in ZEA detoxification [54]. Belli et al. [65]
investigated the involvement of ZEA in breast endocrine disorders using Wistar female rats.
Of six studies analyzing the effects of FB1, four used Sprague Dawley rats for the exploration of serum
enzyme activities and DNA lesions [61], gene expression [62], general toxicity after nixtamalization of
whole kernel corn [59], and gastric ulcer [60]. Riedel et al. [73] reported the effect of FB1 in cancer pro-
motion involving lipid changes utilizing Fischer rats. Pellanda et al. [74] investigated the global histone
modification due to FB1 administration in Wistar female and male rats.
Table 34.2 summarizes a total of 51 studies involving mice as animal models to study the possible
effects of Fusarium mycotoxins viz., DON (22 studies), T-2 (9 studies), ZEA (16 studies), and FB1 (10)
[75–126]. Of 22 mice studies involving DON, B6C3F1 followed by BAL13/3 and C57136/6 mice were
the most popular.
B6C3F1 mice were used for the analysis of the effect of DON on immunomodulation [98,99],
insulin-like growth factor [93], and diet-related issues [94,95,96]. C57BL/6 mice were used to ana-
lyze the effect of DON on inflammation [104], diet issues [102], fetal skeletal development [103],
and mouse thymus [100]. With BALB/c mice, the effects of DON were studied on hepatotoxicity
[87], immunomodulation [76,81], and hemostability [77]. With the ICR mouse model, DON was
analyzed for oocyte development [90,91] and oxidative stress [89]. With Swiss albino mice, Mishra
et al. [115] found that topical application of DON increased cell proliferation, DNA synthesis, and
inflammation, while Kouadio et al. [112] revealed that the oral administration of DON created dis-
orders in systemic targets. Choi et al. [122] reported that DON in drinking water can disrupt the
immune response of porcine parvovirus-vaccinated C3H mice. Behavioral changes due to inflam-
matory cytokines after DON intoxication in PGES wild-type and knockout mice were observed by
Girardet et al. [125].
Chaudhary et al. [108,116] and Agarwal et al. [109] studied the oxidative stress due to T-2, while
Agarwal et al. [110] reported the CC-2 formulation’s effectiveness against topical exposure to T-2 toxin
in Swiss albino mice. Using the BALB/c mouse model, Ahmadi et al. [88] showed that selenium protects
the alteration of B lymphocytes after T-2 toxin exposure, while Maragos et al. [82] developed Mab2–13
antibody detection for T-2 and T-2glc. The harmful effect of T-2 toxin in early embryo development was
explored by Somoskői et al. [123] in BDF1 mice. Yang et al. [118] demonstrated the decrease in the tes-
tosterone biosynthesis due to T-2 toxin in Kunming mice.
BALB/c mice were employed to investigate the hepatotoxicity [79,87] and immunotoxicity [75,78,80]
of ZEA. Zhu et al. [90,92] and Hou et al. [91] explained the reduction in oocyte development, while Hou
et al. [89] studied the oxidative stress induced by ZEA in ICR mice. Boeira et al. [111,113,114] clearly
indicated the impaired testicular functions due to ZEA toxicity among male Swiss albino mice. ZEA was
TABLE 34.1
Fusarium
Studies Using Rat Models for the Examination of Fusarium Mycotoxins Published Since 2010 Based on PubMed
Rat Model MT Administration/Dose Aim of the Study Major Findings References
10-w-old Sprague ZEA Intraperitoneal, 5 mg/kg To investigate the effects of ZEA on ZEA induces apoptosis in germ cells of male rats; this toxicity [51]
Dawley spermatogenesis and possible mechanisms of ZEA is partially mediated through modulation of Fas and
involved in germ cell injury Fas-L systems, though ERα may not play a significant role
8-w-old male ZEA Intraperitoneal, 300 mg/kg To investigate the effects of KRG extract on Impaired spermatogenesis resulting from ZEA treatment was [52]
Sprague Dawley testicular toxicity induced by ZEA prevented by KRG through Fas-Fas L modulation
1-month-old male T-2 Intragastric, 100 and To compare antioxidant capacity and lipid Increasing TBARS and decreasing antioxidants in serum and [53]
Sprague Dawley 200 ng/g b.w./d peroxidation using a novel model cartilage by T-2 toxin treatment with a selenium-deficient
nutritional status may alter oxidative stress in joint tissues and
contribute to the pathological process of cartilage damage
in KBD
Male Sprague ZEA Intraperitoneal, 25 mg/kg To determine the levels of expression of rat The initial modifications in mRNA levels indicate a close [54]
Dawley b.w. proteins that are involved in the ZEA association with microsomal enzyme activity of the CYP2B,
detoxification pathway upon acute ZEA CYP2C, and CYP3A protein families
treatment
6-w-old female ZEA Intragastric, 3 mg/kg To investigate the effect of ZEA ZEA exposure can cause oxidative stress and change common [55]
Sprague Dawley supplementation on rat metabolism systemic metabolic processes, including cell membrane
metabolism, protein biosynthesis, glycolysis, and gut
microbiota metabolism
Pregnant Sprague ZEA Subcutaneous, 0, 1, 50, or To assess the impact of ZEA in adult rats ZEA neonatal exposure could affect the exposure of testis to [56]
Dawley 100 and 0, 0.75, 1.25, or exposed neonatally ABC transporter substrates and negatively influence
2.5 μg/d spermatogenesis and male fertility
1-month-old male T-2 Intragastric, 100 and To observe pathogenic lesions of joint cartilages Rat can be used as a suitable animal model for studying [57]
Sprague Dawley 200 ng/g b.w./d in rats fed with T-2 toxin under a selenium- etiological factors contributing to the pathogenesis
deficiency nutrition status in order to determine (chondronecrosis) observed in human KBD
possible etiological factors causing KBD
8-w-old axenic male DON Gavage feeding, 100 µg/kg To evaluate the impact of a subchronic NOAEL A significant increase of 0.5 log10 was observed for the [58]
Sprague Dawley b.w. dose exposure of DON on the composition of Bacteroides/Prevotella group during the first 3 w of
human gut microbiota administration, concentration levels for Escherichia coli
decreased at d 27
(Continued)
527
TABLE 34.1 (Continued)
528
Studies Using Rat Models for the Examination of Fusarium Mycotoxins Published Since 2010 Based on PubMed
Rat Model MT Administration/Dose Aim of the Study Major Findings References
3-w-old male FB1 FB1 contaminated corn, To assess how nixtamalization of whole kernel Nixtamalization is an effective cooking method for reducing [59]
Sprague Dawley 30 mg/kg corn affects fumonisin toxicity, male rats were the potential toxicity of FB1-contaminated corn
fed diets containing low, mid or high levels of
uncooked (LU, MU, HU) or alkaline-cooked
(LC, MC, HC) FB1-contaminated corn for
3 week
Male Sprague FB1 Gastric subserosa, To investigate whether accumulated ceramide The ceramide pathway, in particular the metabolites of [60]
Dawley 0.036–0.09 g/kg b.w. could serve as the effector molecule of ulcer ceramide, significantly contribute to acetic-acid-induced
formation in a rat model of acetic-acid-induced gastric damage, possibly via enhancing apoptosis
gastric ulcer
4- to 5-w-old adult FB1 FB1-contaminated corn, To assess changes in serum biochemical profile In rats consuming diets containing FB1, there is a time- and [61]
male Sprague 50, 100, and 200 mg/kg and DNA fragmentation of growing male rats dose-dependent increase in serum enzyme activities and DNA
Dawley diet fed a diet with FB1-contaminated corn and the lesions
role of a Lactobacillus delbrueckii spp. lactis
and Pediococcus acidilactici supplementation
in counteracting the FB1 effects in
intoxicated rats
3-month-old FB1 Oral feed—corn oil To evaluate the protective role of PGE against the AFB1 and FB1 have synergistic genotoxic effects; PGE induced [62]
Fusarium
Studies Using Rat Models for the Examination of Fusarium Mycotoxins Published Since 2010 Based on PubMed
Rat Model MT Administration/Dose Aim of the Study Major Findings References
Pregnant female ZEA Subcutaneous injection, (1) Morphometric analysis to evaluate the ZEA could contribute to the induction of breast endocrine [65]
Wistar 0.2 μg/kg to 5 mg/kg development of the mammary glands; disorders
(2) immunohistochemical study of mammary
tissue slides to evaluate differentiation and the
level of cell replication and apoptosis in the
tissue; and (3) to test the presence of possible
lesions in the mammary tissue
4-w-old male and T-2 Intragastric, 0.04 mg/kg/d To investigate magnetic MRI in the tibial The MRI image of the rat epiphyseal plate is altered in the [66]
female weanling epiphyseal growth plate development KBD model rats, and epiphyseal plate MRI appearance has
Wistar been reproduced by using T-2 toxin and KBD-affected feed of
an epidemic-affected district
Male and female T-2 Intragastric, 0.1 mg/kg/d To determine whether giving rats selenium- and This animal model of KBD can be approached by feeding rats [67]
Wistar iodine-deficient food low in protein and made a low-nutrition diet (low levels of selenium, iodine, and
with barley from an area where KBD is protein) and exposing them to T-2 toxin. The pathological and
endemic, in combination with exposing them to radiographic changes observed were very similar to those in
T-2 toxin, would produce the characteristic patients with KBD
chondrocyte necrosis of KBD, to establish an
experimental KBD animal model
10-w-old male T-2 Subcutaneous, 0.1 and To clarify and reevaluate the cardiac and T-2 toxin produced significant cardiac dysfunctions involving [68]
Wistar 0.5 mg/kg autonomic nervous effects of T-2 toxin disturbance of the conduction pathway influenced by the
autonomic nervous activity and also possible direct effects on
cardiac myocytes
4-w-old-male and T-2 0.04 mg/kg/d To characterize the features of radiographic A low-nutrition diet may be involved in the etiology of KBD, [69]
female weanling abnormalities of the tibial bone in rats that have and determination of this should be studied in the future
Wistar been fed T-2 toxin and KBD epidemic district
food
Wistar T-2 0, 1, 10, and 100 nM To investigate the reproductive toxicity and A possible underlying molecular mechanism for T-2 toxin is [70]
cytotoxicity of T-2 toxin and to explore its that it induces the apoptosis signaling pathway in rat
potential apoptotic induction mechanism granulosa cells by regulation of ROS-mediated mitochondrial
pathway
(Continued)
529
TABLE 34.1 (Continued)
530
Studies Using Rat Models for the Examination of Fusarium Mycotoxins Published Since 2010 Based on PubMed
Rat Model MT Administration/Dose Aim of the Study Major Findings References
Male and female T-2 Intragastric, 0.04 mg/kg/d To investigate the effect of KBD-affected feed KBD-affected feed rats had less weight gain than T-2 toxin [71]
weanling Wistar and T-2 toxin on bone development intervention rats, which means there were other etiological
factors in KBD-affected feed
Male and female T-2 Intragastric, 1 mg/kg/d To observe early lesions of rat epiphyseal platesT-2 toxin combined with a low-nutrition diet could lead to [72]
Wistar rats and metaphysis caused by T-2 toxin and T-2 more serious chondrocyte necrosis in the epiphyseal plate and
toxin combined with a low-nutrition diet to disturb metaphyseal trabecular bone formation
determine possible pathogenic factors of KBD
Male Fischer FB1 250 mg/kg diet To characterize the involvement of lipid changes A typical lipid phenotype was observed, including increased [73]
during cancer promotion resulting in the membrane PE and cholesterol content, increased levels of
development of preneoplastic lesions, altered C16:0 and monounsaturated fatty acids in PE and PC, as well
lipid phenotype, and to compare FA profiles of as a decrease in C18:0 and long-chain polyunsaturated fatty
two different cancer-promotion regimens acids in the PC fraction
3-month-old FB1 Gavage feeding, 4 μg/kg To investigate the synergistic impact of prenatal Low doses of FB1 interact with MDD, thus contributing to the [74]
sexually mature b.w. methyl donor deficiency and low dosage of FB1 disruption of the epigenetic landscape
virgin female and administration on the pattern of global histone
male Wistar modifications
ABC, ATP-binding cassette; ADC, activated diatomaceous clay; b.w., body weight; CYP, cytochrome P450; d, day; DON, deoxynivalenol; ER, estrogen receptor; FA, fatty acid; FB1, fumonisin
Fusarium
Studies Using Mouse Models for the Examination of Fusarium Mycotoxins Published Since 2010 Based on PubMed
Mouse Model MT Administration/Dose Aim of the Study Major Findings References
6-w-old female BALB/c ZEA Oral, 40 mg/kg b.w. To investigate the effects of radish extract on the b.w. Radish extract was effective for the protection of high [75]
gain, the lymphoid relative weight organs, dose ZEA-immunotoxication in mice
hematological parameters, and the expression of
antibody level and cytokines production LPS
stimulation on intoxication
6-w-old female BALB/c DON Intraperitoneal To determine the optimal concentrations of antigen/ The detection limit was 0.01–100 μg/mL, and average [76]
inoculation, antibody for DON detection recovery of DON from contaminated grain was
100–250 μg 82%–93%
BALB/c DON 10 μg/kg To elucidate the reason for blood parameters and Significant decrease of hematocrit value and rise of [77]
hemostatic effect after oral administration of DON blood clotting time and bleeding time; DON is a
potential hematotoxin
5-w-old female BALB/c ZEA Oral, 40 mg/kg b.w. To determine the abilities of the living Lactobacillus Both L. plantarum and TM are safe by themselves and [78]
plantarum MON03 cells, TM clay, and their their composite succeeded to exert a potential
composite to accumulate ZEA from a liquid medium prevention by counteracting ZEA-immunotoxicity
and elucidate the preventive effect of their composite
in ZEA-contaminated mice
BALB/c ZEA Oral, 50,100, 200 μg/kg To establish whether ZEA produced hepatotoxicity via ZEA is a potential hepatotoxin when administered by [79]
b.w. oral administration the oral route
8-w-old female BALB/c ZEA Oral, 40 mg/kg b.w. To isolate a new ZEA-binding microorganism for use ZEA induced toxicity in immunologic and [80]
in biological detoxification and to examine its ability hematologic parameters as indicated by the changes
to remove ZEA in liquid medium and its potential for in lymphocyte cell numbers; Lactobacillus paracasei
prevention of ZEA-induced immunomodulation in BEJ01 treatment prevents weight loss and reduces the
mice immunotoxic effects caused by ZEA
Female BALB/c DON Oral, 0, 0.5, or 2 mg/kg To investigate the differential immunomodulatory DON exposure differentially modulated IL-1, IL-10, [81]
effects of DON and TNF-α production; DON can cause various
immunomodulatory effects
Female BALB/c T-2 Injection, 100 μg To develop an antibody capable of interacting with Mab 2–13 will be useful for the simultaneous [82]
T2G-KLH detection of T-2-Glc detection of T-2 toxin and T-2-Glc
7- to 8-w-old male and FB1 0.1 mg/kg To evaluate the genotoxic potentials of FB1 using a FB1 is nongenotoxic in nature, while the reduced ratio [83]
female BALB/c 10 mg/kg simple micronuclei test in a rodent model of PCE/NCE suggests the cytotoxic nature of FB1
(Continued)
531
TABLE 34.2 (Continued)
532
Studies Using Mouse Models for the Examination of Fusarium Mycotoxins Published Since 2010 Based on PubMed
Mouse Model MT Administration/Dose Aim of the Study Major Findings References
Female BALB/c FB1Subcutaneous injection To obtain a monoclonal antibody against FB1 with high Anti-FB1 mcAb excreted by 4G5 can be used to detect [84]
FB1-BSA (0.2 mL, specificity and affinity FB1 in corn and related samples
100 mg)
10-w-old female FB1 Intraperitoneal and To investigate the effect of silymarin on experimental Silymarin ameliorated toxic liver damage [85]
BALB/c gavage, 1.5 mg/kg FB1 liver toxication induced by FB1
4.5 mg/kg FB1
6-w-old male BALB/c T-2 Intraperitoneal, 1, 2, 3, 4, To check the effect of a sublethal dose of T-2 toxin on Selenium could exert an important effect against the [86]
or 5 mg/kg T lymphocyte subpopulation levels and the potential immunotoxic effects of T-2 toxin against T
protective effects from treatment with selenium or lymphocytes
vitamin E
4-w-old female BALB/c DON, 5.0 mg/kg b.w. To assess the individual and combined toxic effects of The combination of AFB1 + DON displayed a [87]
ZEA AFB1, ZEA, and DON within the liver synergistic hepatotoxic effect, while AFB1 + ZEA
displayed an antagonistic hepatotoxic effect
BALB/c T-2 NA To investigate the toxic effect of T-2 toxin on the Selenium plays a pivotal role on altered B lymphocyte [88]
percentage of peripheral blood B lymphocytes and the subset induced by T-2 toxin compared with vitamin E
potential protective role of selenium and vitamin E
4-w-old female ICR DON, Maize feed DON To investigate the regulation of multiple mycotoxins on MT-contaminated diet could result in liver damage, [89]
Fusarium
Studies Using Mouse Models for the Examination of Fusarium Mycotoxins Published Since 2010 Based on PubMed
Mouse Model MT Administration/Dose Aim of the Study Major Findings References
4-w-old female ICR DON, Contaminated maize feed To check the effect of DON on oocyte quality DON and ZEA affect oocyte polarity during meiosis; [91]
ZEA 3.875 mg/kg of DON or abnormal mitochondrial distributions in the oocytes
1,897 μg/kg of ZEA
Female ICR ZEA Germinal vesicle-intact To study the effect of ZEA on mouse egg ZEA can affect chromatin compaction and the cell [92]
oocytes harvested from developmental competence cycle progression of oocytes by reduced H4K20me2
ovaries of mice: low and increased H4K20me3 levels
dose group—10 μM
ZEA
High dose group—50 μM
ZEA
3- to 4-w-old female DON Oral, 0.1–12.5 mg/kg b.w. To test the hypothesis that impairment of the GH axis Oral DON exposure perturbs GH axis by suppressing [93]
B6C3F1 precedes DON-induced growth retardation in the two clinically relevant growth-related proteins,
mouse IGFALS and IGF1
11- to 12-w-old female DON Contaminated pellet feed, To determine whether this mouse strain is similarly DON consumption lowered weight gain and produced [94]
B6C3F1 0, 2, 5, or 10 ppm affected by DON during the process of obesity weight loss in diet-induced obese mice at higher
induction or when in the obese state thresholds than that observed previously in normal
B6C3F1 mice
10- to 11-w-old female, DON Oral, 0, 0.1, 0.5, 1.0, or To check whether the acute administration of DON to Mice had partial resistance to feed refusal when [95]
B6C3F1 mice 2.5 mg/kg the mouse causes a rapid, measurable, and exposed to DON subsequently after 2 d but not after
reproducible reduction in food intake 7 d, suggesting that this modest tolerance was
reversible
11-w-old female adult DON Oral, 10 mg/kg To relate DON-induced b.w. loss in HF-induced obese DON induced rapid decreases in b.w. and fat mass, [96]
B6C3F1 mice to food intake, fat mass, lean mass, and which stabilized to those of the LF control within
obesity-related hormones 11 d; DON-mediated effects on b.w., fat mass, food
intake, and hormonal levels were consistent with a
state of chronic energy restriction
3-w-old female DON 0, 1, 2.5, 5, and 10 ppm To explore the feasibility of using plasma IGFALS as a Plasma IGFALS was significantly depressed; it might [97]
B6C3F1 biomarker of effect for DON be a useful biomarker for DON’s adverse effects on
To demonstrate that in mice fed with 15 ppm DON diet growth
there are significantly less plasma IGFALS than in
mice fed identical amounts of control diet
(Continued)
533
TABLE 34.2 (Continued)
534
Studies Using Mouse Models for the Examination of Fusarium Mycotoxins Published Since 2010 Based on PubMed
Mouse Model MT Administration/Dose Aim of the Study Major Findings References
8- to 10-w-old male DON Oral 5 mg/kg b.w. To identify early protein phosphorylation changes in DON impacted phosphorylation of proteins within [98]
B6C3F1 the immune system of DON-exposed mice and relate diverse immune cell populations, including
these to the toxin’s downstream immunomodulatory monocytes, macrophages, T cells, B cells, dendritic
effects cells, and mast cells
10- to 12-w-old female DON Oral, 2.5 mg/kg b.w. To compare the effects of DON and its congeners on Naturally occurring and synthetic DON congeners to [99]
B6C3F1 (C57BL/6 ×, DON, D3G, 3-ADON, splenic TNF-α, interleukin and chemokine mRNA elicit aberrant mRNA upregulation of cytokines
C3HeN) 15-ADON, FX, NIV, expression in the mouse associated with acute and chronic trichothecene
EN139528 or toxicity
EN139544 in 100 μL
PBS
7-w-old male C57BL/6 DON 5, 10, or 25 mg/kg b.w. To understand the mechanism of action of DON in the DON downregulated genes involved in proliferation, [100]
thymus mitochondria, protein synthesis, and ribosomal
proteins
Early precursor thymocytes, particularly at the
double-positive CD4+ CD8+ stage, are more
vulnerable to DON than very early or late precursor
thymocytes
10-w-old C57BL/6 DON Intraperitoneal injection To investigate the toxic effects of DON on fetal skeletal Various skeletal defects in fetuses, including [101]
Fusarium
Studies Using Mouse Models for the Examination of Fusarium Mycotoxins Published Since 2010 Based on PubMed
Mouse Model MT Administration/Dose Aim of the Study Major Findings References
Male and female CD1 ZEA Oral, 1.3, 3.9, and To analyze the effects on the transcriptome in testes Mono-(2-ethylhexyl)-phthalate and ZEA both [105]
6.6 mg/kg exposed to mono-(2-ethylhexyl) phthalate, ZEA, produced specific alterations of gene signatures
lindane, bisphenol-A, or 17β-estradiol irrespective of the concentration of the toxicant or
the developmental period
8-w-old CD1 FB1 0, 25, 50, or 100 mg/mL To examine toxicity on the development of embryos NSP had an unexpected high adsorption capacity [106]
of NSP and its capacity to prevent teratogenesis-induced in vitro; NSP is a feasible and effective agent for
by FB1 supplementary use in reducing the toxicity of FB1
to animals
CD1 outbred male ZEA Oral, lower dose To assess the effect of treatment with a low dose of Animals exposed to low ZEA concentration were [107]
(0.15 μg/L, higher dose ZEA on the male gonadal pathology, sperm quality, affected considerably more than animals exposed to
(150 μg/L) and expression of selected genes thigh ZEA concentration; a low concentration of
ZEA is able to negatively influence the sperm
parameters and testicular gene expression
Female Swiss albino T-2 Dermal and subcuta- To evaluate the acute toxicity of dermal and Percutaneously and subcutaneously applied T-2 toxin [108]
neous, 5.94 mg/kg subcutaneous exposure of T-2 toxin on brain oxidative can cause brain oxidative damage possibly after
1.54 mg/kg stress crossing blood–brain barrier by altering its
permeability
Male Swiss albino T-2 Percutaneous, 5.94 mg/kg To investigate the biochemical and histological Skin inflammation and cutaneous injury are [109]
b.w. 2.97, 5.94 and alterations behind inflammation and cutaneous injury mediated through oxidative stress, activation of
11.88 mg/kg b.w. caused by T-2 toxin myeloperoxidase, MMP activity, increase in
inflammatory cytokines, activation of p38 MAPK,
and apoptosis of epidermal cells, leading to
degenerative skin histological changes
Male Swiss albino T-2 Topical, 11.8 and To evaluate the protective efficacy of CC-2 formulation CC-2 formulation may be an effective decontaminant [110]
23.76 mg/kg against lethal topical doses; to check the effect of against topical exposure to T-2 toxin if treated
dose of T-2 toxin and time of CC-2 application on within 5–15 min of T-2 toxin exposure
lethality; to evaluate the recovery profile of surviving
animals
90-d-old male Swiss ZEA Gavage, 40 mg/kg To investigate the effect of an acute dose of ZEA on ZEA has acute toxic effects mainly in reproductive [111]
albino reproductive and hematological parameters, as well as system of adult male mice, and its effect is
on markers of oxidative stress in liver, kidney, and probably related to a reduced activity of GST and
testes increase in SOD activity in testes
(Continued)
535
TABLE 34.2 (Continued)
536
Studies Using Mouse Models for the Examination of Fusarium Mycotoxins Published Since 2010 Based on PubMed
Mouse Model MT Administration/Dose Aim of the Study Major Findings References
7- to 8-w-old male and DON, Oral 45 μg/kg To determine whether association of DON and FB1 NOAEL of both DON and FB1 should be lower than [112]
female Swiss FB1 cause an additive or synergistic toxic effect on their 45 μg/kg b.w./d and 110 μg/kg b.w./d; the oral
systemic targets repetitive administration of low dose of DON
revealed disorders in lipid metabolism, renal
filtration disturbance, and renal cell DNA
methylation, rhabdomyolysis, and blood lymphocyte
cell deaths
9-d-old male Swiss ZEA ZEA (40 mg/kg—8% To check the effects of lycopene on reproductive, Lycopene prevented the testicular histopathological [113]
albino of LD50) hematological, histopathological, and oxidative stress damage caused by ZEA, as well as its harmful
parameters following acute exposure to ZEA effects on sperm count and motility
Male Swiss albino ZEA Oral, single dose To study ZEA toxicity effects and the mechanism of ZEA impaired testicular functions (spermatozoa [114]
(40 mg/kg—8% of lycopene’s protective effects following ZEA exposure count and motility) and testosterone levels;
LD50) lycopene can be considered a potential therapeutic
nutrient in protection against male reproductive
toxicity induced by ZEA
6- to 7-w-old female DON Topical application in To understand the cellular events leading to DON- Topical application increased cell proliferation, DNA [115]
Swiss albino concentrations of 84, mediated in vivo dermal toxicity and to investigate synthesis, and inflammation, which are mediated
168, 336, and whether DON causes induction of cell proliferation through PI3K/AKT and MAPK signaling pathways
Fusarium
Studies Using Mouse Models for the Examination of Fusarium Mycotoxins Published Since 2010 Based on PubMed
Mouse Model MT Administration/Dose Aim of the Study Major Findings References
60- to 90-d-old T-2 10 ng/mL To investigate the effects of T-2 toxin on testosterone T-2 toxin can directly decrease the testosterone [118]
Kunming biosynthesis in Leydig cells biosynthesis in the primary Leydig cells derived
from the testis
SWV and LM/Bc FB1 Intraperitoneal, 20 mg/kg To determine the relationship between failure of neural Elevated sphingoid base-1-P after FB1 or FTY720 [119]
tube closure and accumulation of sphingoid bases and suggest a potential role for these bioactive lipid
their phosphorylated metabolites in maternal and ligands and activation of S1P receptor signaling
embryonic tissue after administration of FB1 pathways in the failure of neural tube closure after
FB1 or FTY720; Sa1P may represent a biomarker
for FB1-NTD risk assessment
15- to 18-w-old female FB1 25 mg/kg To investigate whether the consumption of a diet Folate deficiency does not exacerbate NTD induction [120]
acclimated LM/Bc 1 mg/kg deficient in folate and with reduced vitamin B12 would by FB1 in LM/Bc mice; interactions between folate,
exacerbate NTD induction by FB1 other nutritional factors, and FB1 in this mouse
model for NTDs are complex and require further
investigations
Male LM/Bc FB1 Oral gavage, 5, 10, 15, To develop and validate in an animal model, and In both mouse and human, the A270, total protein, [121]
25, and 50 mg/kg b.w. ultimately in humans, a method to estimate the and blood volume were closely correlated and the
volume of blood collected as blood spots on absorbent volume of blood spotted was accurately estimated
paper so as to allow quantification of the molar using only the A270 of the extracts; in mouse blood
concentration of sphingoid base 1-phosphates in spots, as in tissues and embryos, the FB1-induced
blood changes in sphingolipids were correlated with
urinary FB1
6-w-old male C3H DON Contaminated drinking To evaluate immune function of mice exposed to DON; Exposure to DON at 2.0 mg/L via drinking water can [122]
water, 2 mg/mL to study the effects of exposure on the immune disrupt the immune response in vaccinated mice by
response after vaccination with inactivated porcine modulating cytokines and chemokines
parvovirus vaccine
6-w-old BDF1 T-2 0.5, 0.75, and 1 ng/mL To investigate embryotoxicity of T-2 via the in vitro T-2 mycotoxin has a harmful effect on early [123]
development of preimplantation mouse embryos and embryonal development, which results in decreased
its effect on the nuclear chromatin status blastocyst proportion, delayed blastulation, and
increased rate of chromatin damage
(Continued)
537
538
TABLE 34.2 (Continued)
Studies Using Mouse Models for the Examination of Fusarium Mycotoxins Published Since 2010 Based on PubMed
Mouse Model MT Administration/Dose Aim of the Study Major Findings References
Female FB1 150 mg/kg To examine the apoptotic and proliferative activity of Oral administration of FB1 caused atrophy in gastric [124]
gastric mucosa following administration of FB1 mucosa both via increasing of apoptosis and
suppressing the mitotic activity of these cells
mPGES-1 wild-type DON Oral, 6.25, 12.5, and To demonstrate that DON-induced sickness-like Behavioral changes observed after DON intoxication [125]
and knock out adult 25 mg/kg behavior is independent of PGE2 action differ from classical sickness behavior evoked by
male inflammatory cytokines
5- to 7-w-old FB1 5, 50, and 150 mg/kg To compare the responses to chronic dietary FB1 Based on similar responses in p53+/– and p53+/+ [126]
p53+/– and p53+/+ exposure in p53+/– and p53+/+ mice to check mice, p53 and related pathways play a secondary
whether FB1 is a nongenotoxic carcinogen; to assess role in responses to FB1 toxicity and carcinogenesis
confirmed to diminish female fertility in C57BL/6J mice [103]. Using CD1 males, Zatecka et al. [107]
illustrated that ZEA has considerable effect on male fertility, and López-Casas et al. [105] proved the
alterations of gene signatures due to ZEA. Yuan et al. [117] assessed the ZEA toxicity in male germ cells
of Kunming mice.
FB1-induced changes in sphingoids were analyzed by Riley et al. [121] and Gelineau-van Waes et al.
[119] among LM/Bc mice. Voss et al. [120] established that folate deficiency does not exacerbate NTD
induction by FB1 in LM/Bc mice. Hepatotoxicity [85] and cytotoxicity [83] of FB1 were confirmed using
BALB/c mice. Ling et al. [84] utilized anti-FB1 monoclonal antibody from BALB/c mice to detect FB1
in corn and related samples. Liao et al. [106] evidenced that nanosilicate platelets reduced the toxicity
of FB1 in CD1 mice. Kouadio et al. [112] proved that no-observed-adverse effect level (NOAEL) of FB1
should be lower than 110 μg/kg/b.w./day in Swiss albino mice. FB1-induced carcinogenesis was reported
to be similar in both p53 heterozygous cancer-prone model and p53 homozygous mice [126]. Alizadeh
et al. [124] reported that atrophy in gastric mucosa was due to FB1 in female mice.
540
Studies Using Human and Animal Cell Lines for the Examination of Fusarium Mycotoxins Published Since 2010 Based on PubMed
Applied Cell Line MT Administration/Dose Aim of the Study Major Findings References
IPEC-1 cells derived from DON 30 μmol/L To investigate the involvement of MAPK in the DON-induced activation of the p44/42 ERK signaling [127]
small intestine of a DON-induced loss of barrier function pathway inhibits the expression of claudin-4 protein,
newborn unsuckled piglet which leads to impaired intestinal barrier function
IPEC-J2 non-transformed DON 200 and 2000 ng/mL To elucidate the impact of the direction of DON Severity of impact of the DON on the intestinal epithelial [128]
porcine intestinal columnar exposure on epithelial cell behavior and barrier is dependent on route of application
epithelial cells intestinal barrier integrity
IPEC-J2 DON 200 ng/mL To detect potential expression differences and to Apical and basolateral challenge of epithelial cell layers [129]
2000 ng/mL identify candidate genes for further trigger different gene-response profiles paralleled with a
investigations on cellular reaction mechanisms higher susceptibility towards basolateral challenge
IPEC-J2 DON To evaluate the effects of IP6 as a possible Phytic acid decreased the negative effects of DON on the [130]
inhibitor of cellular damage induced by toxic membrane integrity
substances such as MTs
IPEC-J2 DON 10 μM To assess short-term effects of DON on functional A significant decrease in ATP levels was seen at 48 h in a [131]
characteristics of the intestinal epithelial cells dose-dependent manner and demonstrated that DON
has a distinct cytotoxic effect on IPEC-J2 cells
IPEC-J2 DON 1 mg/mL To determine the penetration of FOS in the Nontoxic concentration of DON on IPEC-J2 cells after [132]
541
TABLE 34.3 (Continued)
542
Studies Using Human and Animal Cell Lines for the Examination of Fusarium Mycotoxins Published Since 2010 Based on PubMed
Applied Cell Line MT Administration/Dose Aim of the Study Major Findings References
MTEC1 murine thymic DON 100, 500, 1000, and To investigate the mRNA expression differences Results provided molecular insights into the gene [142]
epithelial cells 2000 ng/mL after treatment with DON in MTEC1 cells expression differences of DON-induced toxic effects
and suggest that p53 signaling pathway may play an
important role in the inhibition of MTEC1 cell
proliferation
MTEC1 DON 500, 1000, and To investigate if DON could induce apoptosis and DON causes activation of p53, increases levels of ROS, [143]
2000 ng/mL to elucidate the possible mechanism of the action and causes the induction of mitochondrial
dysfunction, which may contribute to DON-induced
apoptosis
HepG2 human DON 1 μM To evaluate the impact of MKPs, particularly DUSP1 is a novel target gene of DON, which is [144]
hepatocellular carcinoma 10 μM DUSP1 essential for the prevention of DON-induced
cells apoptosis
HepG2, THP-1 human DON — To investigate the effect of DON on the cytosolic DON induced accumulation of Trx-1 in HepG2 cells, [145]
monocyte-like cells redox state and antioxidative system which plays one of the key roles in protection against
cytotoxicity caused by DON; the mechanism may be
mediated by the antioxidant properties of Trx-1
HepG2 FB1 0–1000 μM To investigate the effect of FB1 on miR-27b CYP1B1 is posttranscriptionally regulated by miR-27b [146]
suppression and its effect on CYP1B1 after HepG2 exposure to FB1; FB1-induced
Fusarium
Studies Using Human and Animal Cell Lines for the Examination of Fusarium Mycotoxins Published Since 2010 Based on PubMed
Applied Cell Line MT Administration/Dose Aim of the Study Major Findings References
HL60 human acute DON — To elucidate the mechanisms underlying the Deoxynivalenol induces the secretion of chemokines, [149]
promyelocytic leukemia NIV toxicities of the trichothecene mycotoxins whereas nivalenol has the opposite effect, clearly
cells deoxynivalenol and nivalenol, their effects on the indicating that the toxicity mechanisms of
secretion of antihematopoietic chemokines, deoxynivalenol and nivalenol differ
macrophage inflammatory protein-1 α (MIP-1α),
and MIP-1β
HL60 T-2 0, 4, 8, 16, and 32 μg/mL To study the effects of T-2 toxin produced by T-2 toxin could inhibit proliferation and induce [150]
Fusarium fungi on proliferation and apoptosis apoptosis in vitro in a dose-dependent manner
Jurkat human DON 0.5 mM Immunocytological and biochemical analyses of Immune cells are more sensitive to DON than other [151]
lymphoblastoid T-cells the effects of DON in Jurkat cells cell types due to the induction of a T-cell activation
response by increased intracellular calcium levels
Jurkat DON 0.5 mM To understand the mechanism of action of DON in Immune cells are more sensitive to DON than other [152]
immune cells; to understand why immune cells cell types due to the induction of a T-cell activation
are more sensitive to DON than most other cell response by increased intracellular calcium levels
types
CTLL-2 murine cytotoxic T DON 1 or 2 mM To assess the usefulness of the mouse CTLL-2 Based on the results for TBTO and DON, the CTLL-2 [153]
lymphocyte cells cell line for immunotoxicity cell line does not yield an added value for
immunotoxicity compared to the human Jurkat T cell
line
U937 human monocyte DON 500–1000 ng/mL To elucidate linkages that exist between the PKR and Hck were critical for DON-induced [154]
cultures ribosome and PKR, Hck, and p38 following ribosomal recruitment of p38, its subsequent
stimulation with DON in human and mouse phosphorylation, and p38-driven proinflammatory
mononuclear phagocytes cytokine expression
HT-29 human colorectal FB1 1.1–69 μM To investigate by dose- and time-dependent Lipid peroxidation followed by modifications to [155]
adenocarcinoma cells experiments the modifications induced by FB1 at membrane microviscosity and inflammatory response
concentrations ranging from 0.25 to 69 μM was the main and most sensitive effect of FB1
AGS gastric epithelial cells FB1 4.5–72 mg/L To evaluate FB1 effects on the production of FB1 increases inflammatory cytokines production [156]
and SW742 human colon inflammatory cytokines
adenocarcinoma cells
HCT116 and HCT116- DON 0.2 mg/mL To analyze the molecular mechanisms of Mitochondria-related caspase-dependent apoptotic [157]
Bax-KO human colon DON-induced toxicity and explore the pathway is involved in this in vitro model of DON
adenocarcinoma cells contribution of mitochondria to cell death induced-cytotoxicity
(Continued)
543
TABLE 34.3 (Continued)
544
Studies Using Human and Animal Cell Lines for the Examination of Fusarium Mycotoxins Published Since 2010 Based on PubMed
Applied Cell Line MT Administration/Dose Aim of the Study Major Findings References
STC-1 murine invasive DON 0–5 mM To test the hypothesis that DON induces hormone DON evokes CCK and GLP-1 secretion in the STC-1 [158]
small intestinal exocytosis in EEC by GPCR-mediated Ca2+ EEC model by activating CaSR- and TRPA1-
neuroendocrine carcinoma signaling mediated Ca2+ signaling pathways
cells
IEC-6 (CRL-1592) rat small DON 0.5–80 mM To test the effects of variable concentrations of Both nivalenol and deoxynivalenol significantly [159]
intestine epithelial cells NIV and/or DON on the nontumorigenic affected IEC-6 viability through a proapoptotic
intestinal epithelial cells, IEC-6 process
EL4 thymoma cells DON 0.5 μM To investigate whether proteomic analysis of DON stimulated the expression levels of several [160]
thymoma cells treated with DON as compared to proteins in thymoma cells
nontreated control cells would reveal differential
protein expression
GT1-7 murine hypothalamic T-2 0.01–1000 ng/mL To find out at how low-dose T-2 toxin influences Low-dose T-2 toxin stimulates GnRH secretion and [161]
neuronal cells GnRH secretion, especially with the presence of alter the expression of associated proteins in vitro, and
kisspeptins the activation of cell response to T-2 toxin is increased
after pretreatment with kisspeptins
MLTC-1 mouse Leydig ZEA 0–200 μg/mL To demonstrate the involvement of ER stress in The activation of an ER stress pathway plays a key role [162]
cells ZEA-induced cell death in ZEA-induced apoptosis
Fusarium
Studies Using Human and Animal Cell Lines for the Examination of Fusarium Mycotoxins Published Since 2010 Based on PubMed
Applied Cell Line MT Administration/Dose Aim of the Study Major Findings References
HEK293 human embryonic ZEA 0–20 μM To investigate the mechanisms of ZEA Oxidative stress does not play a key role in DNA strand [166]
kidney cells nephrotoxicity and DNA damage breaks induced by ZEA; lysosomal injury precedes
DNA strand breaks; the lysosome may be a primary
target for ZEA
Vero African green monkey FB1 1, 5, and 10 μM To study the ROS production using a procedure Significant increase of ROS products in Vero cells at 10 [167]
kidney cells based on an antioxidant sensitive fluorescein IM dose; ROS production by FB1 on renal cells is a
probe H2-DCFDA and to detect intracellular mechanism of fumonisin-mediated toxicity
ROS as early-stage marker for toxin-induced
oxidative stress
H4IIE rat hepatoma cells FB1 10 and 20 μM To evaluate the effects of MTs, alone or AHR pathway activation as a toxicity mechanism of [168]
AFB1 combined, on activation and expression of AFB1 and FB1; FB1 may increase AFB1 bioactivation
CYP1A and its transcription factor AHR
HL-7702 normal human FB1 0.0, 0.1, 1.0, 10.0, and To investigate the effect of FB1 on the cell cycle The underlying mechanism of action is associated with [169]
liver cells 100 μmol/L and the expression of cell-cycle-related genes alterations in the expression levels of cyclin E and
P21 and cyclin E P21 induced by FB1
RTL-W1 rainbow trout liver ZEA 300 ng/mL To investigate the possible metabolization of ZEA Results confirm a lysosomal pathway as a main target [170]
cells, RTgill-W1 gill cells in fish cell lines suggesting that mainly of ZEA in fish cells
and SHK-1 salmon head glucuronidation takes place
kidney cells
ADON, acetyldeoxynivalenol; AFB1, aflatoxin B1; AHR, aryl hydrocarbon receptor; CaSR, GPCR Ca(2+)-sensing receptor; CCK, cholecystokinin; CYP, cytochrome P; DNA, deoxyribonucleic
acid; DON, deoxynivalenol; DUSP1, dual specific phosphatase 1; EEC, enteroendocrine cell; ER, endoplasmic reticulum; ERK, extracellular signal-regulated kinase; GnRH, gonadotropin-
releasing hormone; GLP-1, glucagon-like peptide-17–36 amide; GPCR, G protein-coupled receptor; ENN, enniatin; FOS, fosfomycin; FB1, fumonisin B1; H2-DCFDA,
dichloro-dihydro-fluorescein diacetate; Hck, hematopoietic cell kinase; hER, the human oestrogenic receptor; IMA, imazalil; IP6, phytic acid; MAPK, mitogen activated protein kinases;
MKP, mitogen activated protein kinase phosphatase; MT, mycotoxin; NIV, nivalenol; OTA, ochratoxin A; PCV2, porcine circovirus type 2; PCVAD, porcine circovirus-associated disease;
PKR, RNA-activated protein kinase; RNA, ribonucleic acid; ROS, reactive oxygen species; TBTO, tributyltin oxide; TRPA1, transient receptor potential ankyrin-1 channel; Trx-1, thiore-
doxin 1; ZEA, zearalenone.
545
546 Laboratory Models for Foodborne Infections
epithelial cells [164], MLTC-1 murine testis Leydig cells [162], as well as CYP19-overexpressing MCF-
7aro human breast cancer cells and T98G human glioblastoma cells [163].
34.3 Conclusions
Based on the recent literature search (since 2010), mainly rats, mice, and cell lines—particularly IPEC-J2
nontransformed porcine intestinal epithelial cells—were extensively involved in Fusarium mycotoxin
research. These laboratory models revealed substantial details of scientific and technical information
related to the mycotoxins DON, T-2, ZEA, and FB1. These available data and the particulars discussed in
this review chapter obviously warrant that laboratory animal models and cell lines have the potential for
the detection of fusarial mycotoxins in food and feed, as well as in the research leading to the reduction
and control of these mycotoxins in the products for consumption by humans and animals, which may
otherwise lead to deleterious effects.
Acknowledgments
This work was supported by the Indian National Science Academy and the Hungarian Academy of
Sciences, by project GINOP-2.3.3-15-2016-00006 (Széchenyi 2020 Programme), and by King Saud
University, Deanship of Scientific Research, College of Sciences Research Center.
REFERENCES
1. Link, H.F., Observationes in ordines plantarum naturales, Dissertatio I, Mag. Ges. Naturf. Freunde
Berlin, 3, 3, 1809.
2. O’Donnell, K. et al., Phylogenetic analyses of RPB1 and RPB2 support a middle Cretaceous origin for a
clade comprising all agriculturally and medically important fusaria, Fungal Genet. Biol., 52, 20, 2013.
3. Coleman, J.J. et al., The genome of Nectria haematococca: contribution of supernumerary chromo-
somes to gene expansion, PLoS Genet., 5, e1000618, 2009.
4. Summerell, B.A., Laurence, M.H., Liew, E.C.Y., Leslie, J.F., Biogeography and phylogeography of
Fusarium: a review, Fungal Divers., 44, 3, 2010.
5. Westphal, A., Abney, T.S., Xing, L.J., Shaner, G.E., Sudden death syndrome of soybean, Plant Health
Instr., doi: 10.1094/PHI-I-2008-0102-01, 2008.
6. Huang, C.H., Roberts, P.D., Datnoff, L.E., Silicon suppresses Fusarium crown and root rot of tomato,
J. Phytopathol., 159, 546, 2011.
7. Shanmugam, V., Kanoujia, N., Biological management of vascular wilt of tomato caused by Fusarium
oxysporum f.sp. lycospersici by plant growth-promoting rhizobacterial mixture, Biol. Control, 57, 85,
2011.
8. Garnica, M., Nucci, M., Epidemiology of fusariosis, Curr. Fungal Infect. Rep., 7, 301, 2013.
9. O’Donnell, K. et al., Internet-accessible DNA sequence database for identifying fusaria from human
and animal infections, J. Clin. Microbiol., 48, 3708, 2010.
10. van Diepeningen, A.D., Al-Hatmi, A.M.S., Brankovics, B., de Hoog, G.S., Taxonomy and clinical spec-
tra of Fusarium species: where do we stand in 2014? Curr. Clin. Microbiol. Rep., 1, 10, 2014.
11. Ma, L.J. et al., Fusarium pathogenomics, Annu. Rev. Microbiol., 67, 399, 2013.
12. Zinedine, A., Soriano, J.M., Molto, J.C., Manes, J., Review on the toxicity, occurrence, metabolism,
detoxification, regulations and intake of zearalenone: an oestrogenic mycotoxin, Food Chem. Toxicol.,
45, 1, 2007.
13. Maragos, C.M., Zearalenone occurrence and human exposure, World Mycotoxin J., 3, 369, 2010.
14. Döll, S., Dänicke, S., The Fusarium toxins deoxynivalenol (DON) and zearalenone (ZON) in animal
feeding, Prev. Vet. Med., 102, 132, 2011.
15. Massart, F., Saggese, G., Oestrogenic mycotoxin exposures and precocious pubertal development, Int.
J. Androl., 33, 369, 2010.
Fusarium 547
16. EFSA Panel on Contaminants in the Food Chain (CONTAM), Scientific opinion on the risks for public
health related to the presence of zearalenone in food, EFSA J., 9, 2197, 2011.
17. Wolf, J.C., Mirocha, C.J., Control of sexual reproduction in Gibberella zeae (Fusarium roseum
“Graminearum”), Appl. Environ. Microbiol., 33, 546, 1977.
18. Kościelniak, J., Biesaga-Kościelniak, J., Janeczko, A., Filek, W., Kalaji, H.M., Can the Gibberella zeae
toxin zearalenone affect the photosynthetic productivity and increase yield formation in spring wheat
and soybean plants? Photosynthetica, 47, 586, 2009.
19. Wu, F., Measuring the economic impacts of Fusarium toxins in animal feeds, Anim. Feed Sci. Technol.,
137, 363, 2007.
20. Asci, A., Durmaz, E., Erkekoglu, P., Pasli, D., Bircan, I., Kocer-Gumusel, B., Urinary zearalenone levels
in girls with premature thelarche and idiopathic central precocious puberty, Minerva Pediatr., 66, 571,
2014.
21. Shephard, G., Fusarium mycotoxins and human health, Plant Breed. Seed Sci., 64, 113, 2011.
22. Gelderblom, W.C. et al., Fumonisins—novel mycotoxins with cancer-promoting activity produced by
Fusarium moniliforme, Appl. Environ. Microbiol., 54, 1806, 1988.
23. Bezuidenhout, S.C. et al., Structure elucidation of the fumonisins, mycotoxins from Fusarium monili-
forme, J. Chem. Soc. Chem. Commun., 11, 743, 1988.
24. Seo, J.A., Kim, J.C., Lee, Y.W., Isolation and characterization of two new type C fumonisins produced
by Fusarium oxysporum, J. Nat. Prod., 59, 1003, 1996.
25. Rheeder, J.P., Marasas, W.F.O., Vismer, H.F., Production of fumonisin analogs by Fusarium species,
Appl. Environ. Microbiol., 68, 2101, 2002.
26. Bartók, T., Szécsi, Á., Szekeres, A., Mesterházy, Á., Bartók, M., Detection of new fumonisin myco-
toxins and fumonisin-like compounds by reversed-phase high-performance liquid chromatography/
electrospray ionization ion trap mass spectrometry, Rapid Commun. Mass Spectrom., 20, 2447,
2006.
27. Bartók, T. et al., Detection and characterization of twenty-eight isomers of fumonisin B1 (FB1) myco-
toxin in a solid rice culture infected with Fusarium verticillioides by reversed-phase high-performance
liquid chromatography/electrospray ionization time-of-flight and ion trap mass spectrometry, Rapid
Commun. Mass Spectrom., 24, 35, 2010.
28. Bartók, T. et al., Detection of previously unknown fumonisin P analogue mycotoxins in a Fusarium ver-
ticillioides culture by high-performance liquid chromatography-electrospray ionization time-of-flight
and ion trap mass spectrometry, J. Chromatogr. Sci., 52, 508, 2014.
29. Marasas, W.F., Fumonisins: history, world-wide occurrence and impact, Adv. Exp. Med. Biol., 392, 1,
1996.
30. Wang, E. et al., Inhibition of sphingolipid biosynthesis by fumonisins. Implications for diseases associ-
ated with Fusarium moniliforme, J. Biol. Chem., 266, 14486, 1991.
31. Marasas, W.F. et al., Fumonisins disrupt sphingolipid metabolism, folate transport, and neural tube
development in embryo culture and in vivo: a potential risk factor for human neural tube defects among
populations consuming fumonisin-contaminated maize, J. Nutr., 134, 711, 2004.
32. Stockmann-Juvala, H., Savolainen, K., A review of the toxic effects and mechanisms of action of
fumonisin B1, Hum. Exp. Toxicol., 27, 799, 2008.
33. Rheeder, J.P. et al., Fusarium moniliforme and fumonisins in corn in relation to human esophageal
cancer in Transkei, Phytopathology, 82, 353, 1992.
34. Chu, F.S., Li, G.Y., Simultaneous occurrence of fumonisin B1 and other mycotoxins in moldy corn col-
lected from the People’s Republic of China in regions with high incidences of esophageal cancer, Appl.
Environ. Microbiol., 60, 847, 1994.
35. International Agency for Research on Cancer (IARC), IARC Monographs on the Evaluation of
Carcinogenic Risks to Humans. Vol. 82. Some Traditional Herbal Medicine, Some Mycotoxins,
Naphthalene and Styrene, IARC Press, Lyon, France, 2002.
36. Ueno, Y., Sawano, M., Ishii, K., Production of trichothecene mycotoxins by Fusarium species in shake
culture, Appl. Microbiol., 30, 4, 1975.
37. Rocha, O., Ansari, K., Doohan, F.M., Effects of trichothecene mycotoxins on eukaryotic cells: a review,
Food Addit. Contam., 22, 369, 2005.
38. Lancova, K., Bowens, P., Stroka, J., Gmuender, H., Ellinger, T., Naegeli, H., Transcriptomic-based
bioassays for the detection of type A trichothecenes, World Mycotoxin J., 2, 247, 2009.
548 Laboratory Models for Foodborne Infections
39. Bennet, J.W., Klich, M., Mycotoxins, Clin. Microbiol. Rev., 16, 497, 2003.
40. Grovey, J.F., The trichothecenes and their biosynthesis, Prog. Chem. Org. Nat. Prod., 88, 63, 2007.
41. Bin-Umer, M.A., McLaughlin, J.E., Basu, D., McCormick, S., Tumer, N.E., Trichothecene mycotoxins
inhibit mitochondrial translation—implication for the mechanism of toxicity, Toxins, 3, 1484, 2011.
42. Escrivá, L., Font, G., Manyes, L., In vivo toxicity studies of Fusarium mycotoxins in the last decade: a
review, Food Chem. Toxicol., 78, 185, 2015.
43. Peraica, M., Radić, B., Lucić, A., Pavlović, M., Toxic effects of mycotoxins in humans, Bull. World
Health Organ., 77, 754, 1999.
44. Wang, Z.G., Feng, J.N., Tong, Z., Human toxicosis caused by moldy rice contaminated with Fusarium
and T-2 toxin, Biomed. Environ. Sci., 6, 65, 1993.
45. Kanora, A., Maes, D., The role of mycotoxins in pig reproduction: a review, Ved. Med-Czech., 54, 565, 2009.
46. EFSA Panel on Contaminants in the Food Chain (CONTAM), Scientific opinion on the risks for animal
and public health related to the presence of T-2 and HT-2 toxin in food and feed, EFSA J., 9, 2481, 2011.
47. Wu, Q., Dohnal, V., Kuca, K., Yuan, Z., Trichothecenes: structure-toxic activity relationships, Curr.
Drug Metab., 14, 641, 2013.
48. Beyer, M., Ferse, I., Mulac, D., Würthwein, E.U., Humpf, H.U., Structural elucidation of T-2 toxin
thermal degradation products and investigations toward their occurrence in retail food, J. Agric. Food
Chem., 57, 1867, 2009.
49. Rotter, B.A., Prelusky, D.B., Pestka, J.J., Toxicology of deoxynivalenol (vomitoxin), J. Toxicol. Environ.
Health, 48, 1, 1996.
50. Summerell, B.A., Leslie, J.F., Fifty years of Fusarium: how could nine species have ever been enough?
Fungal Divers., 50, 135, 2011.
51. Jee, Y., Noh, E.M., Cho, E.S., Son, H.Y., Involvement of the Fas and Fas ligand in testicular germ cell
apoptosis by zearalenone in rat, J. Vet. Sci., 11, 115, 2010.
52. Cho, E.S., Ryu, S.Y., Jung, J.Y., Park, B.K., Son, H.Y., Effects of red ginseng extract on zearalenone
induced spermatogenesis impairment in rat, J. Ginseng Res., 35, 294, 2011.
53. Chen, J.H. et al., Oxidant damage in Kashin-Beck disease and a rat Kashin-Beck disease model by
employing T-2 toxin treatment under selenium deficient conditions, J. Orthop. Res., 30, 1229, 2012.
54. Duca, R.C., Mabondzo, A., Bravin, F., Delaforge, M., In vivo effects of zearalenone on the expression of
proteins involved in the detoxification of rat xenobiotics, Environ. Toxicol., 27, 98, 2012.
55. Liu, G. et al., Biological system responses to zearalenone mycotoxin exposure by integrated metabolo-
mic studies, J. Agric. Food Chem., 61, 11212, 2013.
56. Koraïchi, F. et al., Neonatal exposure to zearalenone induces long term modulation of ABC transporter
expression in testis, Toxicology, 310, 29, 2013.
57. Guan, F. et al., Histopathology of chondronecrosis development in knee articular cartilage in a rat model
of Kashin-Beck disease using T-2 toxin and selenium deficiency conditions, Rheumatol. Int., 33, 157,
2013.
58. Saint-Cyr, M.J., Perrin-Guyomard, A., Houée, P., Rolland, J.G., Laurentie, M., Evaluation of an oral
subchronic exposure of deoxynivalenol on the composition of human gut microbiota in a model of
human microbiota-associated rats, PLoS One, 8, e80578, 2013.
59. Voss, K.A., Riley, R.T., Moore, N.D., Burns, T.D., Alkaline cooking (nixtamalisation) and the reduction
in the in vivo toxicity of fumonisin-contaminated corn in a rat feeding bioassay, Food Addit. Contam.
Part A Chem. Anal. Control Expo. Risk Assess., 30, 1415, 2013.
60. Nakashita, M. et al., Attenuation of acetic acid-induced gastric ulcer formation in rats by glucosylce-
ramide synthase inhibitors, Dig. Dis. Sci., 58, 354, 2013.
61. Khalil, A.A., Abou-Gabal, A.E., Abdellatef, A.A., Khalid, A.E., Protective role of probiotic lactic acid
bacteria against dietary fumonisin B1-induced toxicity and DNA-fragmentation in Sprague Dawley rats,
Prep. Biochem. Biotechnol., 45, 530, 2015.
62. Hassan, A.M., Abdel-Aziem, S.H., El-Nekeety, A.A., Abdel-Wahhab, M.A., Panax ginseng extract
modulates oxidative stress, DNA fragmentation and up-regulate gene expression in rats sub chronically
treated with aflatoxin B1 and fumonisin B1, Cytotechnology, 67, 861, 2015.
63. Zhang, R. et al., Combined effects of T-2 toxin and selenium deficiency on lipid peroxidation in rat’s
brain, Wei Sheng Yan Jiu, 43, 6, 2014.
64. Denli, M., Blandon, J.C., Guynot, M.E., Salado, S., Pérez, J.F., Efficacy of activated diatomaceous clay
in reducing the toxicity of zearalenone in rats and piglets, J. Anim. Sci., 93, 637, 2015.
Fusarium 549
65. Belli, P. et al., Fetal and neonatal exposure to the mycotoxin zearalenone induces phenotypic alterations
in adult rat mammary gland, Food Chem. Toxicol., 48, 2818, 2010.
66. Yan, D., Song, Y., Shen, B., Kang, P., Pei, F., Magnetic resonance imaging in the tibial epiphyseal
growth plate development of Wistar rat, J. Orthop. Surg. Res., 9, 39, 2014.
67. Kang, P., Yao, Y., Yang, J., Shen, B., Zhou, Z., Pei, F., An animal model of Kashin–Beck disease induced
by a low-nutrition diet and exposure to T-2 toxin, Osteoarthritis Cartilage, 21, 1108, 2013.
68. Ngampongsa, S., Ito, K., Kuwahara, M., Ando, K., Tsubone, H., Reevaluation of arrhythmias and
alterations of the autonomic nervous activity induced by T-2 toxin through telemetric measurements in
unrestrained rats, Toxicol. Mech. Methods, 22, 662, 2012.
69. Yan, D. et al., Radiographic findings of Wistar rats fed with T-2 toxin and Kashin–Beck disease-affected
diet, Int. J. Rheum. Dis., 14, 92, 2011.
70. Wu, J., Jing, L., Yuan, H., Peng, S.Q., T-2 toxin induces apoptosis in ovarian granulosa cells of rats
through reactive oxygen species-mediated mitochondrial pathway, Toxicol. Lett., 202, 168, 2011.
71. Yan, D. et al., The effect of Kashin–Beck disease-affected feed and T-2 toxin on the bone development
of Wistar rats, Int. J. Rheum. Dis., 13, 266, 2010.
72. Yao, Y.F. et al., Study on the effect of T-2 toxin combined with low nutrition diet on rat epiphyseal plate
growth and development, Int. Orthop., 34, 1351, 2010.
73. Riedel, S., Abel, S., Swanevelder, S., Gelderblom, W.C., Induction of an altered lipid phenotype by two
cancer promoting treatments in rat liver, Food Chem. Toxicol., 78, 96, 2015.
74. Pellanda, H. et al., Fumonisin FB1 treatment acts synergistically with methyl donor deficiency during
rat pregnancy to produce alterations of H3- and H4-histone methylation patterns in fetuses, Mol. Nutr.
Food Res., 56, 976, 2012.
75. Salah-Abbès, J.B., Abbès, S., Abdel-Wahhab, M., Oueslati, R., Immunotoxicity of zearalenone in Balb/c
mice in a high subchronic dosing study counteracted by Raphanus sativus extract, Immunopharmacol.
Immunotoxicol., 32, 628, 2010.
76. Ji, F., Li, H., Xu, J., Shi, J., Enzyme-linked immunosorbent-assay for deoxynivalenol (DON), Toxins
(Basel), 3, 968, 2011.
77. Chatopadhyay, P., Gupta, V., Gogoi, H.K., Singh, L., Hematotoxicity of deoxynivalenol in BALB/c
mice, J. Pharmacol. Pharmacother., 2, 115, 2011.
78. Abbès, S., Ben Salah-Abbès, J., Sharafi, H., Noghabi, K.A., Oueslati, R., Interaction of Lactobacillus plan-
tarum MON03 with Tunisian montmorillonite clay and ability of the composite to immobilize zearalenone
in vitro and counteract immunotoxicity in vivo, Immunopharmacol. Immunotoxicol., 34, 944, 2012.
79. Chatopadhyay, P., Pandey, A., Chaurasia, A.K., Upadhyay, A., Karmakar, S., Singh, L., Hepatic hyper-
plasia and damages induces by zearalenone Fusarium mycotoxins in BALB/c mice, Arq. Gastroenterol.,
49, 77, 2012.
80. Abbès, S., Ben Salah-Abbès, J., Sharafi, H., Oueslati, R., Noghabi, K.A., Lactobacillus paracasei BEJ01
prevents immunotoxic effects during chronic zearalenone exposure in Balb/c mice, Immunopharmacol.
Immunotoxicol., 35, 341, 2013.
81. Islam, M.R., Roh, Y.S., Kim, J., Lim, C.W., Kim, B., Differential immune modulation by deoxynivale-
nol (vomitoxin) in mice, Toxicol. Lett., 221, 152, 2013.
82. Maragos, C.M., Kurtzman, C., Busman, M., Price, N., McCormick, S., Development and evaluation of
monoclonal antibodies for the glucoside of T-2 toxin (t2-glc), Toxins (Basel), 5, 1299, 2013.
83. Karuna, R., Rao, B.S., Lack of micronuclei induction by fumonisin B1 mycotoxin in BALB/c mice,
Mycotoxin Res., 29, 9, 2013.
84. Ling, S. et al., Preparation and identification of monoclonal antibody against fumonisin B1 and develop-
ment of detection by Ic-ELISA, Toxicon, 80, 64, 2014.
85. Sozmen, M., Devrim, A.K., Tunca, R., Bayezit, M., Dag, S., Essiz, D., Protective effects of silymarin on
fumonisin B1-induced hepatotoxicity in mice, J. Vet. Sci., 15, 51, 2014.
86. Salimian, J., Arefpour, M.A., Riazipour, M., Poursasan, N., Immunomodulatory effects of selenium
and vitamin E on alterations in T lymphocyte subsets induced by T-2 toxin, Immunopharmacol.
Immunotoxicol., 36, 275, 2014.
87. Sun, L.H., Lei, M.Y., Zhang, N.Y., Zhao, L., Krumm, C.S., Qi, D.S., Hepatotoxic effects of mycotoxin
combinations in mice, Food Chem. Toxicol., 74, 289, 2014.
88. Ahmadi, A., Poursasan, N., Amani, J., Salimian, J., Adverse effect of T-2 toxin and the protective role
of selenium and vitamin E on peripheral blood B lymphocytes, Iran. J. Immunol., 12, 64, 2015.
550 Laboratory Models for Foodborne Infections
89. Hou, Y.J. et al., Mycotoxin-containing diet causes oxidative stress in the mouse, PLoS One, 8, e60374, 2013.
90. Zhu, C.C. et al., Effect of mycotoxin-containing diets on epigenetic modifications of mouse oocytes by
fluorescence microscopy analysis, Microsc. Microanal., 20, 1158, 2014.
91. Hou, Y.J. et al., Oocyte quality in mice is affected by a mycotoxin-contaminated diet, Environ. Mol.
Mutagen., 55, 354, 2014.
92. Zhu, C.C., Hou, Y.J., Han, J., Cui, X.S., Kim, N.H., Sun, S.C., Zearalenone exposure affects epigenetic
modifications of mouse eggs, Mutagenesis, 29, 489, 2014.
93. Amuzie, C.J., Pestka, J.J., Suppression of insulin-like growth factor acid-labile subunit expression—a
novel mechanism for deoxynivalenol-induced growth retardation, Toxicol. Sci., 113, 412, 2010.
94. Amuzie, C.J., Flannery, B.M., Ulrich, A.M., Pestka, J.J., Effects of deoxynivalenol consumption on
body weight and adiposity in the diet-induced obese mouse, J. Toxicol. Environ. Health A, 74, 658, 2011.
95. Flannery, B.M., Wu, W., Pestka, J.J., Characterization of deoxynivalenol-induced anorexia using mouse
bioassay, Food Chem. Toxicol., 49, 1863, 2011.
96. Kobayashi-Hattori, K., Amuzie, C.J., Flannery, B.M., Pestka, J.J., Body composition and hormonal
effects following exposure to mycotoxin deoxynivalenol in the high-fat diet-induced obese mouse, Mol.
Nutr. Food Res., 55, 1070, 2011.
97. Flannery, B.M., He, K., Pestka, J.J., Deoxynivalenol-induced weight loss in the diet-induced obese
mouse is reversible and PKR-independent, Toxicol. Lett., 221, 9, 2013.
98. Pan, X., Whitten, D.A., Wu, M., Chan, C., Wilkerson, C.G., Pestka, J.J., Early phosphoproteomic changes
in the mouse spleen during deoxynivalenol-induced ribotoxic stress, Toxicol. Sci., 135, 129, 2013.
99. Wu, W. et al., Effects of oral exposure to naturally-occurring and synthetic deoxynivalenol congeners on
proinflammatory cytokine and chemokine mRNA expression in the mouse, Toxicol. Appl. Pharmacol.,
278, 107, 2014.
100. van Kol, S.W., Hendriksen, P.J., van Loveren, H., Peijnenburg, A., The effects of deoxynivalenol on gene
expression in the murine thymus, Toxicol. Appl. Pharmacol., 250, 299, 2011.
101. Zhao, Y. et al., Evaluation of fetal skeletal malformations in deoxynivalenol-treated mice using microar-
ray analysis, Arch. Environ. Contam. Toxicol., 63, 445, 2012.
102. Flannery, B.M., Amuzie, C.J., Pestka, J.J., Evaluation of insulin-like growth factor acid-labile subunit
as a potential biomarker of effect for deoxynivalenol-induced proinflammatory cytokine expression,
Toxicology, 304, 192, 2013.
103. Zhao, F. et al., Postweaning exposure to dietary zearalenone, a mycotoxin, promotes premature onset of
puberty and disrupts early pregnancy events in female mice, Toxicol. Sci., 132, 431, 2013.
104. Tardivel, C. et al., The food born mycotoxin deoxynivalenol induces low-grade inflammation in mice in
the absence of observed-adverse effects, Toxicol. Lett., 232, 601, 2015.
105. López-Casas, P.P., Mizrak, S.C., López-Fernández, L.A., Paz, M., de Rooij, D.G., del Mazo, J., The
effects of different endocrine disruptors defining compound-specific alterations of gene expression pro-
files in the developing testis, Reprod. Toxicol., 33, 106, 2012.
106. Liao, Y.J. et al., Inhibition of fumonisin B1 cytotoxicity by nanosilicate platelets during mouse embryo
development, PLoS One, 9, e112290, 2014.
107. Zatecka, E. et al., Effect of zearalenone on reproductive parameters and expression of selected testicular
genes in mice, Reprod. Toxicol., 45, 20, 2014.
108. Chaudhary, M., Rao, P.V., Brain oxidative stress after dermal and subcutaneous exposure of T-2 toxin in
mice, Food Chem. Toxicol., 48, 3436, 2010.
109. Agrawal, M., Yadav, P., Lomash, V., Bhaskar, A.S., Lakshmana Rao, P.V., T-2 toxin induced skin inflam-
mation and cutaneous injury in mice, Toxicology, 302, 255, 2012.
110. Agrawal, M., Pardasani, D., Lakshmana Rao, P.V., Evaluation of protective efficacy of CC-2 formulation
against topical lethal dose of T-2 toxin in mice, Food Chem. Toxicol., 50, 1098, 2012.
111. Boeira, S.P. et al., Possible role for glutathione-S-transferase in the oligozoospermia elicited by acute
zearalenone administration in Swiss albino mice, Toxicon, 60, 358, 2012.
112. Kouadio, J.H., Moukha, S., Brou, K., Gnakri, D., Lipid metabolism disorders, lymphocytes cells death,
and renal toxicity induced by very low levels of deoxynivalenol and fumonisin B1 alone or in combina-
tion following 7 days oral administration to mice, Toxicol. Int., 20, 218, 2013.
113. Boeira, S.P. et al., Lycopene treatment prevents hematological, reproductive and histopathological
damage induced by acute zearalenone administration in male Swiss mice, Exp. Toxicol. Pathol., 66,
179, 2014.
Fusarium 551
114. Boeira, S.P. et al., Lycopene protects against acute zearalenone-induced oxidative, endocrine,
inflammatory and reproductive damages in male mice, Chem. Biol. Interact., 230, 50, 2015.
115. Mishra, S., Tripathi, A., Chaudhari, B.P., Dwivedi, P.D., Pandey, H.P., Das, M., Deoxynivalenol
induced mouse skin cell proliferation and inflammation via MAPK pathway, Toxicol. Appl.
Pharmacol., 279, 186, 2014.
116. Chaudhary, M., Bhaskar, A.S., Rao, P.V., Differential effects of route of T-2 toxin exposure on hepatic
oxidative damage in mice, Environ. Toxicol., 30, 64, 2015.
117. Yuan, H. et al., Gynostemma pentaphyllum protects mouse male germ cells against apoptosis caused by
zearalenone via Bax and Bcl-2 regulation, Toxicol. Mech. Methods, 20, 153, 2010.
118. Yang, J., Zhang, Y., Jing, A., Ma, K., Gong, Q., Qin, C., Effects of T-2 toxin on testosterone biosynthesis
in mouse Leydig cells, Toxicol. Ind. Health, 30, 873, 2014.
119. Gelineau-van Waes, J. et al., Increased sphingoid base-1-phosphates and failure of neural tube closure
after exposure to fumonisin or FTY720, Birth Defects Res. A Clin. Mol. Teratol., 94, 790, 2012.
120. Voss, K.A., Riley, R.T., Gelineau-van Waes, J., Fumonisin B1 induced neural tube defects were not
increased in LM/Bc mice fed folate-deficient diet, Mol. Nutr. Food Res., 58, 1190, 2014.
121. Riley, R.T. et al., A blood spot method for detecting fumonisin-induced changes in putative sphingolipid
biomarkers in LM/Bc mice and humans, Food Addit. Contam. Part A Chem. Anal. Control Expo. Risk
Assess., 10, 1, 2015.
122. Choi, B.K. et al., Effects of oral deoxynivalenol exposure on immune-related parameters in lymphoid
organs and serum of mice vaccinated with porcine parvovirus vaccine, Mycotoxin Res., 29, 185, 2013.
123. Somoskői, B., Kovács, M., Cseh, S., Effects of T-2 mycotoxin on in vitro development and chromatin
status of mouse embryos being in preimplantation stages, Toxicol. Ind. Health, 32, 1260, 2016.
124. Alizadeh, A.M. et al., Apoptotic and proliferative activity of mouse gastric mucosa following oral
administration of fumonisin B1, Iran. J. Basic Med. Sci., 18, 8, 2015.
125. Girardet, C. et al., Central inflammation and sickness-like behavior induced by the food contaminant
deoxynivalenol: a PGE2-independent mechanism, Toxicol. Sci., 124, 179, 2011.
126. Bondy, G. et al., Effects of long term exposure to the mycotoxin fumonisin B1 in p53 heterozygous and
p53 homozygous transgenic mice, Food Chem. Toxicol., 50, 3604, 2012.
127. Pinton, P., Braicu, C., Nougayrede, J.P., Laffitte, J., Taranu, I., Oswald, I.P., Deoxynivalenol impairs
porcine intestinal barrier function and decreases the protein expression of claudin-4 through a mitogen-
activated protein kinase-dependent mechanism, J. Nutr., 140, 1956, 2010.
128. Diesing, A.K. et al., Vulnerability of polarised intestinal porcine epithelial cells to mycotoxin deoxyni-
valenol depends on the route of application, PLoS One, 6, e17472, 2011.
129. Diesing, A.K. et al., Gene regulation of intestinal porcine epithelial cells IPEC-J2 is dependent on the
site of deoxynivalenol toxicological action, PLoS One, 7, e34136, 2012.
130. Pacheco, G.D., Silva, C.A., Pinton, P., Oswald, I.P., Bracarense, A.P., Phytic acid protects porcine intes-
tinal epithelial cells from deoxynivalenol (DON) cytotoxicity, Exp. Toxicol. Pathol., 64, 345, 2012.
131. Awad, W.A., Aschenbach, J.R., Zentek, J., Cytotoxicity and metabolic stress induced by deoxynivalenol
in the porcine intestinal IPEC-J2 cell line, J. Anim. Physiol. Anim. Nutr. (Berl), 96, 709, 2012.
132. Martínez, G., Pérez, D.S., Soraci, A.L., Tapia, M.O., Penetration of fosfomycin into IPEC-J2 cells in the
presence or absence of deoxynivalenol, PLoS One, 8, e75068, 2013.
133. Wan, L.Y., Turner, P.C., El-Nezami, H., Individual and combined cytotoxic effects of Fusarium tox-
ins (deoxynivalenol, nivalenol, zearalenone and fumonisins B1) on swine jejunal epithelial cells, Food
Chem. Toxicol., 57, 276, 2013.
134. Wan, L.Y., Woo, C.S., Turner, P.C., Wan, J.M., El-Nezami, H., Individual and combined effects of
Fusarium toxins on the mRNA expression of pro-inflammatory cytokines in swine jejunal epithelial
cells, Toxicol. Lett., 220, 238, 2013.
135. Wan, L.Y., Woo, C.S., Allen, K.J., Turner, P.C., El-Nezami, H., Modulation of porcine β-defensins 1 and
2 upon individual and combined Fusarium toxin exposure in a swine jejunal epithelial cell line, Appl.
Environ. Microbiol., 79, 2225, 2013.
136. Gu, M.J., Song, S.K., Park, S.M., Lee, I.K., Yun, C.H., Bacillus subtilis protects porcine intestinal
barrier from deoxynivalenol via improved zonula occludens-1 expression, Asian-Australas. J. Anim.
Sci., 27, 580, 2014.
137. Kolf-Clauw, M. et al., The emerging mycotoxin, enniatin B1, down-modulates the gastrointestinal toxicity of
T-2 toxin in vitro on intestinal epithelial cells and ex vivo on intestinal explants, Arch. Toxicol., 87, 2233, 2013.
552 Laboratory Models for Foodborne Infections
138. Kruber, P., Trump, S., Behrens, J., Lehmann, I., T-2 toxin is a cytochrome P450 1A1 inducer and leads
to MAPK/p38- but not aryl hydrocarbon receptor-dependent interleukin-8 secretion in the human intes-
tinal epithelial cell line Caco-2, Toxicology, 284, 34, 2011.
139. Ribonnet, L. et al., Potential of an in vitro toolbox combined with exposure data as a first step for the risk
assessment of dietary chemical contaminants, Food Addit. Contam. Part A Chem. Anal. Control Expo.
Risk Assess., 28, 1136, 2011.
140. Kadota, T., Furusawa, H., Hirano, S., Tajima, O., Kamata, Y., Sugita-Konishi, Y., Comparative study of
deoxynivalenol, 3-acetyldeoxynivalenol, and 15-acetyldeoxynivalenol on intestinal transport and IL-8
secretion in the human cell line Caco-2, Toxicol. In Vitro, 27, 1888, 2013.
141. Clarke, R., Connolly, L., Frizzell, C., Elliott, C.T., Cytotoxic assessment of the regulated, co-existing
mycotoxins aflatoxin B1, fumonisin B1 and ochratoxin, in single, binary and tertiary mixtures, Toxicon,
90, 70, 2014.
142. Li, D. et al., Gene expression profiling analysis of deoxynivalenol-induced inhibition of mouse thymic
epithelial cell proliferation, Environ. Toxicol. Pharmacol., 36, 557, 2013.
143. Li, D. et al., Deoxynivalenol induces apoptosis in mouse thymic epithelial cells through mitochondria-
mediated pathway, Environ. Toxicol. Pharmacol., 38, 163, 2014.
144. Casteel, M., Nielsen, C., Kothlow, S., Dietrich, R., Märtlbauer, E., Impact of DUSP1 on the apoptotic
potential of deoxynivalenol in the epithelial cell line HepG2, Toxicol. Lett. 199, 43, 2010.
145. Sugiyama, K., Kinoshita, M., Kamata, Y., Minai, Y., Tani, F., Sugita-Konishi, Y., Thioredoxin-1
contributes to protection against DON-induced oxidative damage in HepG2 cells, Mycotoxin Res., 28,
163, 2012.
146. Chuturgoon, A.A., Phulukdaree, A., Moodley, D., Fumonisin B1 modulates expression of human cyto-
chrome P450 1b1 in human hepatoma (Hepg2) cells by repressing Mir-27b, Toxicol. Lett., 227, 50, 2014.
147. Weidner, M., Welsch, T., Hübner, F., Schwerdt, G., Gekle, M., Humpf, H.U., Identification and apoptotic
potential of T-2 toxin metabolites in human cells, J. Agric. Food Chem., 60, 5676, 2012.
148. Weidner, M., Lenczyk, M., Schwerdt, G., Gekle, M., Humpf, H.U., Neurotoxic potential and cellular
uptake of T-2 toxin in human astrocytes in primary culture, Chem. Res. Toxicol., 26, 347, 2013.
149. Nagashima, H., Nakagawa, H., Kushiro, M., Opposite effects of two trichothecene mycotoxins, deoxyni-
valenol and nivalenol, on the levels of macrophage inflammatory protein (MIP)-1α and MIP-1β in HL60
cells, Environ. Toxicol. Pharmacol., 34, 1014, 2012.
150. Hou, Y., Yuan, Z., Guo, W., Wang, H., Effects of T-2 toxin of Fusarium on proliferation and apoptosis
of human acute promyelocytic leukemia cell line HL60, Wei Sheng Yan Jiu, 42, 381, 2013.
151. Katika, M.R., Hendriksen, P.J., Shao, J., van Loveren, H., Peijnenburg, A., Transcriptome analysis of
the human T lymphocyte cell line Jurkat and human peripheral blood mononuclear cells exposed to
deoxynivalenol (DON): new mechanistic insights, Toxicol. Appl. Pharmacol., 264, 51, 2012.
152. Katika, M.R., Hendriksen, P.J., van Loveren, H., Peijnenburg, A.C.M., Characterization of the modes of
action of deoxynivalenol (DON) in the human Jurkat T-cell line, J. Immunotoxicol., 12, 206, 2015.
153. Schmeits, P.C., Volger, O.L., Zandvliet, E.T., van Loveren, H., Peijnenburg, A.A., Hendriksen, P.J.,
Assessment of the usefulness of the murine cytotoxic T cell line CTLL-2 for immunotoxicity screening
by transcriptomics, Toxicol. Lett., 217, 1, 2013.
154. Bae, H., Gray, J.S., Li, M., Vines, L., Kim, J., Pestka, J.J., Hematopoietic cell kinase associates with the
40S ribosomal subunit and mediates the ribotoxic stress response to deoxynivalenol in mononuclear
phagocytes, Toxicol. Sci., 115, 444, 2010.
155. Minervini, F. et al., Toxic mechanisms induced by fumonisin B1 mycotoxin on human intestinal cell
line, Arch. Environ. Contam. Toxicol., 67, 115, 2014.
156. Mahmoodi, M. et al., Impact of fumonisin B1 on the production of inflammatory cytokines by gastric
and colon cell lines, Iran. J. Allergy Asthma Immunol., 11, 165, 2012.
157. Bensassi, F., Gallerne, C., Sharaf El Dein, O., Lemaire, C., Hajlaoui, M.R., Bacha, H., Involvement of
mitochondria-mediated apoptosis in deoxynivalenol cytotoxicity, Food Chem. Toxicol., 50, 1680, 2012.
158. Zhou, H.R., Pestka, J.J., Deoxynivalenol (vomitoxin)-induced cholecystokinin and glucagon-like
peptide-1 release in the STC-1 enteroendocrine cell model is mediated by calcium-sensing receptor and
transient receptor potential ankyrin-1 channel, Toxicol. Sci., 145, 407, 2015.
159. Bianco, G., Fontanella, B., Severino, L., Quaroni, A., Autore, G., Marzocco, S., Nivalenol and deoxyni-
valenol affect rat intestinal epithelial cells: a concentration related study, PLoS One, 7, e52051, 2012.
Fusarium 553
160. Osman, A.M., Pennings, J.L., Blokland, M., Peijnenburg, A., van Loveren, H., Protein expression profil-
ing of mouse thymoma cells upon exposure to the trichothecene deoxynivalenol (DON): implications
for its mechanism of action, J. Immunotoxicol., 7, 147, 2010.
161. Liu, Y. et al., Effects of low dose T-2 toxin on secretion of gonadotropin-releasing hormone in the
immortalized hypothalamic GT1-7 cell line, Toxicon, 100, 67, 2015.
162. Lin, P. et al., Mycotoxin zearalenone induces apoptosis in mouse Leydig cells via an endoplasmic reticu-
lum stress-dependent signalling pathway, Reprod. Toxicol., 52, 71, 2015.
163. Wang, Y., Wong, T.Y., Chan, F.L., Chen, S., Leung, L.K., Assessing the effect of food mycotoxins on
aromatase by using a cell-based system, Toxicol. In Vitro, 28, 640, 2014.
164. Savard, C. et al., Effect of deoxynivalenol (DON) mycotoxin on in vivo and in vitro porcine circovirus
type 2 infections, Vet. Microbiol., 176, 257, 2015.
165. Lei, M., Zhang, N., Qi, D., In vitro investigation of individual and combined cytotoxic effects of afla-
toxin B1 and other selected mycotoxins on the cell line porcine kidney 15, Exp. Toxicol. Pathol., 65,
1149, 2013.
166. Gao, F. et al., Genotoxic effects induced by zearalenone in a human embryonic kidney cell line, Mutat.
Res., 755, 6, 2013.
167. Meca, G., Ruiz, M.J., Fernandez-Franzón, M., Ritieni, A., Manes, J., Isolation, purification, LC-MS/
MS characterization and reactive oxygen species induced by fumonisin B1 in Vero cells, Food Chem.
Toxicol., 48, 2891, 2010.
168. Mary, V.S., Valdehita, A., Navas, J.M., Rubinstein, H.R., Fernández-Cruz, M.L., Effects of aflatoxin B1,
fumonisin B1 and their mixture on the aryl hydrocarbon receptor and cytochrome P450 1A induction,
Food Chem. Toxicol., 75, 104, 2015.
169. Wang, S.K., Liu, S., Yang, L.G., Shi, R.F., Sun, G.J., Effect of fumonisin B1 on the cell cycle of normal
human liver cells, Mol. Med. Rep., 7, 1970, 2013.
170. Pietsch, C., Noser, J., Wettstein, F.E., Burkhardt-Holm, P., Unraveling the mechanisms involved in
zearalenone-mediated toxicity in permanent fish cell cultures, Toxicon, 88, 44, 2014.
35
Penicillium and Talaromyces
CONTENTS
35.1 Introduction....................................................................................................................................555
35.2 Pathogens...................................................................................................................................... 556
35.2.1 General Description......................................................................................................... 556
35.2.2 Diagnosis...........................................................................................................................557
35.2.3 Molecular Epidemiology...................................................................................................557
35.2.4 Treatment...........................................................................................................................558
35.3 Mycotoxins.....................................................................................................................................558
35.3.1 General Description..........................................................................................................558
35.3.2 Invertebrate Organisms in Toxicity Bioassays................................................................. 562
35.3.3 Microorganisms in Toxicity Bioassays............................................................................. 563
35.3.4 Vertebrate Animals in Toxicity Bioassays....................................................................... 564
35.3.5 Cell Cultures in Toxicity Bioassays.................................................................................. 565
35.3.6 Culture Media for Mycotoxin Production........................................................................ 565
35.4 Allergens....................................................................................................................................... 566
35.4.1 Penicillium Allergies........................................................................................................ 566
35.4.2 Animal Models to Study Allergies Caused by Penicillium spp. ..................................... 567
35.4.3 Cell Cultures to Study Allergies Caused by Penicillium................................................. 570
35.5 Conclusions................................................................................................................................... 570
References................................................................................................................................................571
35.1 Introduction
The genus Penicillium is a group of anamorphic fungi in the division Ascomycota, with over 250 rec-
ognized species, many of them ubiquitous in warm and moderate climates. The genus Talaromyces
was described as a sexual state of Penicillium that produces soft-walled ascomata. Recently, the
genus Penicillium has been redefined into Penicillium sensu stricto and Talaromyces, with all species
of the former Penicillium subgenus Biverticillium being transferred to Talaromyces.1,2 Species like
Talaromyces marneffei lack a defined sexual cycle but possesses all genes believed to be required for
mating.3
Penicillium species play important roles in the environment, agriculture, and industry. Some species
of genus Penicillium are of economic importance to the food industry because they contribute to food
ripening, while others are postharvest pathogens or cause spoilage. For example, Penicillium camemberti
and Penicillium roqueforti are used for cheese manufacture; Penicillium nalgiovense and Penicillium
chrysogenum contribute to ripening of dry-cured meat products. On the other hand, Penicillium expan-
sum is the causal agent of blue mold postharvest rots of apples and is also able to produce patulin and
other mycotoxins, as discussed later. Penicillium digitatum and Penicillium italicum are responsible for
postharvest citrus decay. Heat-resistant ascospores produced by various Talaromyces spp. cause spoilage
of pasteurized juices and other fruit-based products.4
555
556 Laboratory Models for Foodborne Infections
Penicillia are generally soil saprophytes or plant pathogens with limited invasive ability for animal
tissue, except for Talaromyces marneffei (formerly Penicillium marneffei2). However, production of toxic
secondary metabolites is common in Penicillium spp. In addition, Penicillium is an environmental aller-
gen of variable significance among asthmatic and atopic individuals.
35.2 Pathogens
35.2.1 General Description
The genera Penicillium and Talaromyces comprise animal pathogens. Talaromyces radicus and
Talaromyces helicus have been described as rare causative agents of infectious diseases in dogs.5,6
Penicillium corylophilum infects red snapper (Lutjanus campechanus), but infection seems to be con-
fined to the swim bladder.7
Penicillium infections in humans are still called penicilliosis, even those due to Talaromyces marneffei.
Members of the P. chrysogenum–Penicillium rubens complex can be responsible for rare opportunistic
keratitis8 and Talaromyces piceum has been rarely reported in chronic granulomatous disease.9 However,
the most outstanding infection by Penicillium or Talaromyces spp. is due to T. marneffei.
T. marneffei is responsible for one of the commonest opportunistic infections in humans with HIV-
infected patients, causing fatal systemic mycosis in Southeast Asian countries. However, infection by
T. marneffei has also been described as a result of other immunodeficiencies, immunosuppressive ther-
apy, and genetic diseases leading to lymphocytes exhibiting defective IFN-γ production.10
T. marneffei was first identified as the causative agent of mycosis of the reticuloendothelial system.11
T. marneffei can cause a focal infection that may lead to a progressive disseminated penicilliosis, mainly
in immunocompromised patients. The focal infection can be cutaneous or bronchopulmonary, and it may
spread to cause localized lymphadenopathy. However, T. marneffei can also be responsible for asymp-
tomatic fungemia in HIV-infected patients.12
The mode of transmission is not well understood but is probably via ingestion or inhalation of the
fungus.13 By analogy with other endemic fungal pathogens, infection is presumably initiated by the
inhalation of airborne conidia that are subsequently phagocytized by pulmonary alveolar macrophages.
After an inflammatory response in macrophages, T. marneffei can lead to granulomatous or suppurative
reactions in persons with normal immunity, whereas immunocompromised patients develop necrotiz-
ing reaction.13 It is thought that the fungus is disseminated throughout the body to result in systemic
infection. In some cases, the disease did not appear until immunosuppression allowed for the growth
and dissemination throughout the host. Thus, unique skin lesions like abscesses and ulcers are more
commonly noted in non-HIV-infected patients than in HIV-infected patients.14 T. marneffei dissemi-
nates hematogenously causing generalized lymphadenopathy, even leading to osteolysis, which indicates
severe systemic disturbance.15
Adaptation to the new environmental conditions during infection of human hosts is key to fungal
survival.16 T. marneffei is the only dimorphic species of Penicillium and Talaromyces that is capable of
growing in a filamentous multinucleate hyphal form at 25°C or as a uninucleate pathogenic yeast form
at 37°C.17 When grown in air at 25°C, the saprophytic hyphae produce asexual spores (conidia), the
infectious agents. Upon invasion of a human host, most environmental factors undergo immediate and
drastic changes, including temperature, pH, nutrient sources, carbon dioxide, and oxygen concentrations.
Temperature adaptation of T. marneffei within the host manifests itself as a morphological change from
hyphal mold to budding yeast.
It seems that T. marneffei possesses the ability to evade a normally functioning immune system
until in vivo conditions, due to immunosuppression, are favorable to dissemination throughout the
host.18 Upon switching to 37°C, T. marneffei undergoes a process termed arthroconidiation. Conidia
germinate with the onset of extensive hyphal branching. Fragmentation occurs to generate single, uni-
nucleated yeast cells that divide by fission. Switching from yeast growth at 37°C to hyphal growth at
25°C requires the polarized growth of yeast cells, septation, and branching, to form the characteristic
Penicillium and Talaromyces 557
multinucleate hyphal mycelium. Genes responsible for growth, morphogenesis, and development of
T. marneffei have been characterized.19 Conidial germination and temperature adaptation require
both HHK and heterotrimeric G-protein-Ras signaling, as well as the establishment of actin-mediated
polarized growth through a Rho GTPase. In addition, the p21-activated kinase pathway is involved in
sensing the environment inside a host cell by T. marneffei, being central to eliciting the appropriate
morphogenetic response to the host environment.20 Systems for detecting and responding to changes in
carbon sources also play a major role in adaptation to the host niche and are essential factors for per-
sistence in a mammalian host. Many of the genes whose expression is upregulated during the mold-to-
yeast transition are related to those genes involved in energy metabolism. The temperature-dependent
regulation of isocitrate lyase, a key component of the glyoxylate cycle, activates the glyoxylate cycle
for utilization of poor carbon sources.16
Other potential virulence factors include yeast phase-specific or upregulated genes that encode
enzymes known to combat oxidative host defense responses. Potential virulence factors related to host-
cell attachment have also been identified.18
Proteins secreted at different developmental stages and noncoding RNAs are suggested to play diverse
roles in mycelium-to-yeast transition. RNA structural transition in response to temperature changes may
be related to the control of thermal dimorphism.21
35.2.2 Diagnosis
The diagnosis of penicilliosis has been reviewed recently.22 Patients suffering from penicilliosis may
have skin, palatal, and pharyngeal lesions such as subcutaneous abscesses, molluscum contagiosum-like
lesions and shallow papule-like ulcers. Respiratory signs include productive cough, dyspnea, and hemop-
tysis. The diagnosis is based on the identification of the organism on microscopy with the confirmation
by the culture method through examination of cytology or biopsy specimens from skin, bone marrow,
or lymph nodes.23 In fungemia, yeast cells may be seen inside monocytes in peripheral blood smear.
Detection of nonbudding yeast cells with characteristic central transverse septum should be confirmed
by isolation of T. marneffei on Sabouraud’s dextrose agar at 25°C. A diffusible dark red pigment that is
produced when grown in the mycelial phase at 37°C is one of the first indications that an isolate may be
T. marneffei.24
Diagnosis may also be made by the detection of antibodies produced against various fungal proteins,
or by the detection of fungal proteins in sera or in urine.25–27 A specific exoantigen test using the immu-
nodiffusion technique was devised to identify T. marneffei cultures.28
Also, specific tests have been developed for detecting circulating T. marneffei antigens in serum and
urine specimens by latex agglutination, enzyme immunoassay, and dot blot enzyme-linked immunosor-
bent assay (ELISA) with polyclonal antibodies; and a monoclonal antibody-based sandwich ELISA.27
Several serologic methods have also been developed for detecting specific antibodies against
T. marneffei antigens in clinical specimens. These include an indirect fluorescent-antibody test for
detecting IgG antibodies produced against germinating conidia and yeast forms, immunoblot assays for
detecting at least two protein antigens produced during the growth phase of the yeast, Western blotting
for three cytoplasmic antigens from yeast-form T. marneffei, and an ELISA-based antibody test devel-
oped with a recombinant T. marneffei mannoprotein.27
Specific oligonucleotide primers from the nuclear ribosomal DNA internal transcribed spacer
region have been designed for the specific and selective amplification of T. marneffei DNA in a
PCR-identification system from clinical material.29 Similarly, oligonucleotide probes based on the
18S rRNA gene of T. marneffei have been used to identify T. marneffei DNA from clinical samples
in PCR-hybridization reactions.27
35.2.3 M
olecular Epidemiology
T. marneffei isolates have been classified using various methods that randomly sample for genetic varia-
tion, including restriction fragment length polymorphisms (RFLP), randomly amplified polymorphic
558 Laboratory Models for Foodborne Infections
DNA (RAPD), and pulsed-field gel electrophoresis (PFGE) using restriction enzymes.27 A number of
types have been identified, but no correlation between restriction patterns and geographic region was
observed. On the other hand, multilocus microsatellite typing allowed discrimination between isolates
occurring within the “eastern” clade, from mainland China, Hong Kong, Indonesia, and Vietnam, or the
“western” clade, from Thailand and India.30
T. marneffei life cycle has been related to the high prevalence of infection in the organs of the bamboo
rats, including hoary bamboo rat (Rhizomys pruinosus), Chinese bamboo rat (Rhizomys sinensis), large
bamboo rat (Rhizomys sumatrensis), and bay bamboo rat (Cannomys badidus), as well as the soil around
their burrows.14 Bamboo rats are the only known nonhuman host of T. marneffei, even though these
animals showed no signs of illness.25
Bamboo rat and human coinfection can occur from a common environmental source, rather than
the patients being infected from the rats.27 However, T. marneffei has never been isolated from soil,
water, vegetation, or air, other than from bamboo rats and their burrows. Multilocus genotypes show
that T. marneffei isolates from humans are similar to those infecting rats and are in some cases identical,
suggesting that bamboo rats can be a vector for human infections by acting as amplifiers of infectious
dispersal stages.31
The asexual nature of T. marneffei would have led to the evolution of niche-adapted genotypes, which
could explain the geographically restricted endemicity of this fungus.18
35.2.4 Treatment
Unlike some other emerging fungal pathogens, this organism remains sensitive to many antifungals.32
T. marneffei is sensitive to amphotericin B, itraconazole, and other types of triazoles. However, long-
term secondary prophylaxis with itraconazole has been recommended for HIV-infected patients to pre-
vent reoccurrence.33
However, itraconazole interacts with antiretrovirals, which is an important issue in HIV-positive
patients.34
35.3 Mycotoxins
35.3.1 General Description
The production of secondary metabolites is a characteristic of Penicillium species. Some of the extrolites
produced by Penicillium spp. are toxic (Table 35.1).
Penicillium growth on foods and feeds poses a hazard to consumers due to these toxic metabolites.
Mycotoxins produce toxic effects in different organisms, including mammals, birds, amphibians, arthro-
pods, crustaceans, unicellular organisms, microorganisms, and plants. Taking advantage of this fact,
different bioassays have been developed to study mycotoxin toxicity in foods and feeds.41
The sensitivity of bioassays is generally lower than that of chromatographic methods. However, bioas-
says do not require standards for the detection of mycotoxins. This is a major point, given that only a
small part of the over 300 compounds documented as mycotoxins are commercially available. Therefore,
analyses based on the biological action of mycotoxins allow testing various foods for the presence of
both known and unknown Penicillium mycotoxins. On the other hand, biological methods are generally
less reproducible, less sensitive, and require more time than physicochemical methods.42
The organisms used in biological methods for mycotoxins detection include microorganisms and
invertebrate and vertebrate animals. The use of mammals for toxicity research is rather limited due to
ethical o bjections as well as high costs for food, habitat, and animal welfare.
Organs and cell cultures have also been used. Cell lines and tissue cultures are the best alternatives
to experimental animals in testing for toxic or pathogenic microorganisms.43 Cell cultures are essential
when seeking information on the effect on DNA or DNA-binding molecules, cell organelles, or cell
membranes. But such studies are usually not necessary to detect toxicity in molds from foods. For this,
simpler and easier tests are used, such as those with microorganisms or invertebrates.
TABLE 35.1
559
TABLE 35.1 (Continued)
560
Mycotoxins and Potentially Toxic Extrolites Produced by Penicillium spp. and Talaromyces spp.
Penicillium spp. and Talaromyces spp. Metabolites
P. expansum Chaetoglobosins, citrinin, cyclopiazonic acid, patulin, penitrem A, roquefortine C
P. flavigenum Penicillin, penitrem A, roquefortine C, secalonic acids
P. formosanum Patulin
P. freii Cyclopenol, penicillic acid, xanthomegnin
T. funiculosusa Patulin
P. glandicola Patulin, penitrem A, roquefortine C
P. gladioli Patulin
P. granulatum Patulin
P. griseofulvum Cyclopiazonic acid, griseofulvin, patulin, roquefortine C
P. hirsutum Cyclopenol, cyclopenin, roquefortine C, terrestric acid
P. hordei Roquefortine C, terrestric acid
T. islandicusa Luteoskirin
P. janczewskii Griseofulvin, penitrem A
P. marinum Chaetoglobosins, patulin, roquefortine C
P. melanoconidium Penicillic acid, penitrem A, verrucosidin, xanthomegnin
P. mononematosum Fumitremorgins, isochromantoxin, verrucologen, viriditoxin
561
562 Laboratory Models for Foodborne Infections
TABLE 35.2
Invertebrate Organisms Used in Bioassay Tests to Detect Penicillium Mycotoxins
Organism Organism Type Mycotoxins/Mold Detected Test Type References
Artemia salina Aquatic crustacean Patulin, citrinin, griseofulvin, Disc screening 44
penicillic acid, rubratoxin B method
Moina macrocopa Aquatic crustacean Ochratoxin A, rubratoxin B Mortality 45
Daphnia magna Aquatic crustacean Patulin Bioluminescent test 46
Cyclops fuscus Aquatic crustacean Rubratoxin B, patulin Mortality 47
Tetrahymena pyriformis Protozoan Patulin Mortality 48–52
Penicillic acid, gliotoxin
Rubratoxin B
Patulin
Colpidium campylum Protozoan Patulin, ochratoxin B, roquefortin Mortality 53
Anagasta kuehniella Insects Penicillium spp. and Penicillium Mortality 41
Attagenus megatoma mycotoxins Growth inhibition
Corcyra cephalonica Reduced larval
Drosophila melanogaster growth
Lucilia sericata
Spodoptera frugiperda
Spodoptera littoralis
Tenebrio molitor
Tribolium confusum
Penicillium and Talaromyces 563
Other invertebrate animals used in toxicological bioassays are insects. Panigrahi41 reviewed the tests
using insects to detect the toxicity of mycotoxins produced by Penicillium spp. These tests are based on
the effect of feeds intentionally contaminated with mycotoxins on growth or mortality of adults or larvae
(Table 35.2).
TABLE 35.3
Microorganisms Used in Bioassay Tests to Detect Penicillium Mycotoxins
Organism
Organism Type Mycotoxins Detected Test Type References
Azospirillum Motile Patulin, rubratoxin, penicillic acid, Swarming inhibition 57
brasilense bacteria citrinin assay
Bacillus thuringiensis Bacteria Kojic acid Cup plate assay 57,58
Citrinin, kojic acid, luteoskyrin, Cup plate assay
ochratoxin A, patulin, penicillic acid
Bacillus subtilis Bacteria Ochratoxin A Growth inhibition 59
Patulin, rubratoxin, penicillic acid,
citrinin
Bacillus cereus Bacteria Ochratoxin A 42
Bacillus megaterium Bacteria Patulin, ochratoxin A
Escherichia coli Bacteria Patulin, penicillic acid, ochratoxin A, Genotoxicity in the 60,61
rubratoxin B, kojic acid, citrinin, PR SOS spot test
toxin
Ochratoxin A
Photobacterium Bacteria Rubratoxin B, penicillic acid, citrinin, Bioluminescence 62
phosphoreum ochratoxin A, PR-toxin, patulin assay
Proteus mirabilis Motile Patulin, rubratoxin, penicillic acid, Swarming inhibition 57
bacteria citrinin assay
Pseudomonas Bacteria Patulin Growth inhibition 52
syringae
Saccharomyces Yeast Rubratoxin B, penicillic acid, citrinin, Mutagenicity test 62
cerevisiae ochratoxin A, PR-toxin, patulin
Salmonella Bacteria Ochratoxin A Ames test 61,63,64,65,66
typhimurium Patulin (mutagenicity)
Ochratoxin A, B, citrinin
Citrinin, patulin, cyclopiazonic acid,
luteoskyrin, griseofulvin,
mycophenolic acid, ochratoxin A,
penicillic acid, secalonic acid D
Several Kojic acid Growth inhibition 67
microorganisms
564 Laboratory Models for Foodborne Infections
Another bioassay with bacteria is based on the motility inhibition of Proteus mirabilis and Azospirillum
brasilense caused by some Penicillium toxins.57 Escherichia coli is widely used in bioassays for toxic-
ity and for genotoxic or mutagenic activity detection. Auffray and Boutibones60 evaluated the genotoxic
activity of some mycotoxins produced by strains of Penicillium using Escherichia coli in the SOS spot
test. When comparing the efficiency to detect toxigenic activity between SOS spot test and other tests,
including mutagenicity to Salmonella typhimurium (Ames test), Bacillus subtilis (Rec assay), and in vivo
carcinogenicity, similar results were obtained for ochratoxin A and rubratoxin B, but different results
were observed for patulin, penicillic acid, kojic acid, citrinin, and PR toxin. Tests with microorganisms
have also been used to evidence the toxicity of kojic acid,67 but mutagenicity could only be evidenced at
high concentrations of the mycotoxin.
The Ames test is a classic assay used to assess the mutagenic potential of molds or its metabolites that
produce genetic damage leading to gene mutations.68 The test uses a number of histidine-auxotrophic
Salmonella typhimurium strains carrying different point mutations in genes of the histidine operon.
These mutations can revert to wild type when a mutagen is present. Many strains of Penicillium or their
mycotoxins led to negative results in Ames test, even when rat liver S9 mix was used as an external
metabolizing enzyme system or when a preincubation of toxins in cultured rat hepatocytes was per-
formed. This enzyme treatment is necessary for some compounds to be mutagenic because they need a
metabolic liver transformation that bacterial systems do not possess. Ochratoxin A showed no evidence
of mutagenic activity by SOS spot and Ames tests,61 in contrast to the results obtained with more sensi-
tive bioassays, such as cell cultures or experimental animals, as discussed later.
Penicillium toxins were tested using the bioassay based on the bacterial bioluminescence of
Photobacterium phosphoreum,62 which is a reliable short-term method for assessing the toxicity of
mycotoxins. The order of toxicity determined by bacterial bioluminescence parallels that reported for
mammalian cell cultures.
Growth of Pseudomonas syringae in presence of patulin was evaluated measuring optical density of
culture broth. The sensitivity of this microorganism to the mycotoxin was even higher than that obtained
with rats.52
TABLE 35.4
Vertebrate Animals Used in Several Bioassay Tests to Detect Penicillium Mycotoxins
Organism
Organism Type Mycotoxins Test Type References
Dog Mammals Kojic acid Rate of death 67
Rats and mice Mammals Ochratoxin A Kidney and liver lesions 61,69,70
Kojic acid Rate of death 67
Chicks Birds Ochratoxin A Kidney and liver lesions 71
Chicken embryos Birds Mycotoxins from Rate of death 67,72
Penicillium spp.
Kojic acid Teratogenic abnormalities
Zebrafish Fish Ochratoxin A, Toxicity in larvae 73–75
(Danio rerio) patulin,
penicillic acid
Citrinin, patulin Nephrotoxicity in embryos
Patulin Cardiotoxicity in embryos
TABLE 35.5
Cell Lines Used in Bioassay Tests to Study Penicillium Mycotoxins
Mycotoxins
Cell lines Cell Type Detected Test Type References
CHO-K1-BH4 Chinese hamster ovary cells Ochratoxin A Cytotoxicity (in vitro 77
micronucleus MN and Comet
assays)
TK6 Human lymphoblastoid cells DNA damage
CHO-K1 Chinese hamster ovary cells Patulin Cytotoxicity (NR and MTT 78
assays)
Vero Green monkey kidney cells Ochratoxin A, citrinin Cytotoxicity (MTT assay) 79
and DNA fragmentation
A-549 Human lung cancer cells Sterigmatocystin, Cytotoxicity (NR assay) 80
Hep-G2 Human hepatocellular verruculogen,
carcinoma cells roquefortine C,
L-929 Murine fibroblasts penitrem A,
mycophenolic acid
Neuro-2a Murine neuroblastoma cells
Hep G2 Human hepatocellular Ochratoxin A Cytotoxicity (MTT assay) 81,82
carcinoma cells Patulin Comet assay
LLC-PK1 Porcine renal cells Ochratoxin A and B, Cytotoxicity (MTT assay) 83
patulin, citrinin
CHOK1 Chinese hamster ovary cells Patulin, citrinin Sister chromatid exchange test 84
HEK293 Human embryonic kidney Cytotoxicity (MTT assay)
cells
V79 cells Chinese hamster lung Ochratoxin A, citrinin Cytotoxicity (NR assay and 85
fibroblasts cells In vitro micronucleus MN
assays)
Rao et al.91 evaluated the effect of 19 synthetic and flour media of several Penicillium species in the
ochratoxin A production. A rice-flour-based medium supported maximum amount of OTA production.
OTA production was stimulated by addition of Zn2+ and Mg2+ to the medium. However, when Fe3+ was
added, OTA production was suppressed.
35.4 Allergens
35.4.1 Penicillium Allergies
Penicillium is among the four major genera of allergenic fungi, and its species are commonly isolated
from indoor environments.92 Some Penicillium species pose a respiratory health hazard in susceptible
populations, and the increasing exacerbation of current asthma symptoms has been associated with
increased levels of Penicillium and other fungal genera.93 According to allergen database of the World
Health Organization and International Union of Immunological Societies94 five Penicillium species pro-
duce a total of 17 allergens (Table 35.6), including proteases, ribosomal proteins, membrane proteins,
enolases, and heat shock proteins. Moreover, some fungal volatile metabolites could act as irritants in
susceptible individuals.95
Penicillia are common in a wide variety of food, mainly those of low and intermediate water activ-
ity, such as dry-cured ham, dry sausages, and ripened cheese. Also, airborne spores can be found in all
environments where these foods are manufactured, handled, or stored. Some operations, such as brush-
ing and coating, spread a high amount of Penicillium spores into the air, which may cause occupational
respiratory diseases or urticaria to food industry workers, including those at pork butcheries,96–99 and
cheese factories.100 The most frequent problems associated with fungal allergy are respiratory disorders,
Penicillium and Talaromyces 567
TABLE 35.6
Allergens from Penicillium Species According to WHO/IUIS Allergen Standardization Committee
Species Allergen Biochemical Name
Penicillium brevicompactum Pen b 13 Alkaline serine protease
Pen b 26 Acidic ribosomal protein P1
Penicillium chrysogenum Pen ch 13 Alkaline serine protease
Pen ch 18 Vacuolar serine protease
Pen ch 20 N-acetyl-glucosaminidase
Pen ch 31 Calreticulin
Pen ch 33 16 kDa allergen
Pen ch 35 Transaldolase
Penicillium citrinum Pen c 3 Peroxisomal membrane protein
Pen c 13 Alkaline serine protease
Pen c 19 Heat shock protein Hsp70
Pen c 22 Enolase
Pen c 24 Elongation factor 1 β
Pen c 30 Catalase
Pen c 32 Pectate lyase
Penicillium crustosum Pen cr 26 60S acidic ribosomal phosphoprotein P1
Penicillium oxalicum Pen o 18 Vacuolar serine protease
Source: WHO/IUIS, Allergen nomenclature. http://www.allergen.org/index.php
TABLE 35.7
Penicillium Species Causing Food Allergies
Species Food References
Penicillium camemberti Cheese 102
Penicillium chrysogenum Dry-sausage 103
Penicillium italicum Dry-sausage and cheese 104
Penicillium spp. Dry-sausage 101,105
including rhinitis and asthma, atopic dermatitis, allergic bronchopulmonary mycosis, allergic fungal
sinusitis, and hypersensitivity pneumonitis.92
Several Penicillium species have also been considered as causes of food allergy, and this occurs on
intake of dry-ripened foods coated with molds, such as dry-fermented sausages and cheese (Table 35.7).
These food allergies can cause facial angioedema, allergic rhinitis, urticaria, oropharyngeal pruritus, or
anaphylactic shock immediately upon ingestion.101–105
In addition, mold-ripened foods can be colonized by Penicillium species such as P. chrysogenum,
P. nalgiovense, P. dipodomis, P. griseofulvum, and P. flavigenum that are well-known penicillin produc-
ers. Small amounts of penicillin in foods have been proposed as a potential causative factor in chronic
urticarial reactions,106 and it can also provoke allergic reaction in penicillin-sensitive individuals.107,108
Sensitivity to mold allergens in individuals can be studied using skin prick and intradermal testing or
in vitro testing for specific IgE antibodies with fungal extracts.92 However, to assess the potential aller-
genic effect of molds, some laboratorial models have been used.
provoking the responses in animals. The main responses evaluated include eosinophil, specific IgE and
IgG antibodies, interferon, and interleukins (ILs) levels; bronchoalveolar and lung lavage fluid produc-
tion; and histopathological lesions. Animal assays have also aided in the characterization of the cell
receptors involved in the immune recognition of fungi.109
In addition to mold species, animal strains, dose, and exposure method are important factors affecting
allergic responses. Animal strains have different ability to respond to fungal allergens depending to their
genetic susceptibility. It has been reported that BALB/c mice exhibited stronger inflammatory response
to Stachybotrys chartarum than other congenic mouse strains, such as C3H/HeJ and C56BL/6.120 In con-
trast, intranasal instillation of viable P. chrysogenum conidia increased serum IgE levels in C57 Black/6
mice but not in BALB/c mice.117
Different exposure and sensitization techniques can be used, including inhalation by aerosol of fungal
spores or extracts in suspension, involuntary aspiration, intranasal instillation, and intratracheal instilla-
tion. The advantages and disadvantages of each type of method with regard to modelling fungal exposure
and allergic sensitization in human have been reviewed by Templeton et al.109 Involuntary aspiration and
intranasal or intratracheal instillation are reproducible and allow for a homogenous suspension of fungi to
be administered, but they are not representative of natural exposure. With aerosol, animals are exposed by
normal breathing, but lung exposure concentration is unknown, and its reproducibility is lower.109
Animal studies have demonstrated the ability of Penicillium spp. to provoke allergic and asthma
response. Most of these studies involve acute or chronic exposure of animals to conidia, spore extracts,
hyphae, subcellular fragments, or fungal proteins from P. chrysogenum.
Female C57 Black/6 mouse model was established to examine the in vivo effect in allergic pro-
cesses of different levels of P. chrysogenum viable and nonviable conidia by nasal instillation.114,116,117
The acute instillation of viable P. chrysogenum conidia induced inflammatory reactions in a dose-
dependent manner, while the instillation of nonviable conidia did not.116 Low levels of P. chrysoge-
num conidia (102 spores) neither provoked lung inflammation nor increased serum immunoglobulins.114
About 18% of the viable conidia instilled intranasally were deposited and retained in the lungs and
could produce substances that induce allergic reactions.116 Viable conidia could remain in the lungs long
enough to produce an unknown protein responsible for the chronic Th2-mediated airway inflammation
by increasing total and specific IgE and IgG1, bronchoalveolar lavage fluid levels of IL-4 and IL-5, and
eosinophilia.117 In contrast, repeated exposures to nonviable conidia of P. chrysogenum induced type
Th1 helper responses in mice, with increases in total serum IgG2a and levels of interferon (IFN)-γ in
bronchoalveolar lavage fluid.117
The allergic and inflammatory response of BALB/c mice to mycelium and spores showed differences
in the threshold dose for allergy induction among species.113 These authors classified molds accord-
ing to their allergenic potential: G1 molds induced low-to-moderate responses requiring higher doses
than house dust mite; G2 molds, including P. chrysogenum, required lower doses to induce a similar
response.113 In this sense, the allergic and inflammatory response of BALB/c mice to intratracheal aspi-
ration of mycelium and spores of P. chrysogenum was more robust than that produced by house dust
mite.111 Therefore, molds must be evaluated individually for allergic or asthmatic potential.
Some experiments have been performed to study the response of animals to different extract or com-
pounds from P. chrysogenum. A dose-dependent allergic asthma-like response was observed after
involuntary aspiration of soluble components of P. chrysogenum by BALB/c mice.110 There are some
differences in the effect between single and multiple exposures to high doses. While single exposure
resulted in edema and cellular damage but not immune responses, multiple exposures showed increased
allergen-triggered immediate respiratory responses.110
C57 Black/6 mice have been used to study the effect of allergen protease extract Pen ch released by
viable P. chrysogenum conidia.114,115 Mice previously sensitized with Pen ch or viable P. chrysogenum
conidia by intraperitoneal injections and exposed to intranasal challenge showed increased allergic air-
way inflammation.114 In these mice, significant increases in serum IgE and IgG1, eosinophilia, and mucus
hyperproduction in bronchoalveolar lavage and lung tissue was recorded.114 Furthermore, intraperitoneal
sensitization and posterior instillation of Pen ch produce a strong allergic inflammatory response charac-
terized by increasing serum IgE and IgG1, eosinophils and neutrophils counts in bronchoalveolar lavage,
as well as mucous production and perivascular inflammation by eosinophils and neutrophils in lungs.115
570 Laboratory Models for Foodborne Infections
Guinea pigs have been used for experimental induction of hypersensitivity pneumonitis by using
aerosol inhalation for 12 weeks of a glycoprotein from P. chrysogenum (formerly P. notatum).119 In
exposed animals, specific serum IgM, IgG, and IgE antibodies and sensitized leukotriene CD4 cells were
detected. Moreover, interstitial infiltrates of macrophages and leukotriene cells, cellular bronchiolitis,
and single nonnecrotizing granulomas were observed in lungs. This response is a typical delayed-type
reaction due to chronic contact with the heterologous glycoprotein of Penicillium.119
Swiss Webster Carworth Farms white (CFW) male mice were used to elucidate the role of bioactive
constituents of spores from P. brevicompactum and P. chrysogenum that mediate allergenic responses.118
Inflammatory and cytotoxic responses to intratracheal instillation of brevianamide A and mycophenolic
acid from P. brevicompactum, and roquefortine C from P. chrysogenum were investigated. High doses
of these three metabolites induced a dose-dependent-like inflammatory response expressed as increased
macrophage, neutrophil, MIP-2, TNF, and IL-6 concentrations in the bronchoalveolar lavage fluid in
exposed mice. The analysis of this fluid revealed some compound-specific toxic responses: brevianamide
A and mycophenolic acid provoked vascular leakage according to albumin concentration, and brevian-
amide A induced cytotoxicity, increasing lactate dehydrogenase (LDH) concentration.118
BALB/c mice have also been used to study the effect of intratracheal inoculation of Pen c 13 protease
from P. citrinum. An increase in airway hyperresponsiveness, inflammatory cell infiltration, mucus over-
production, and collagen deposition in the lung, as well as serum levels of total IgE and Pen c 13-specific
IgE and IgG1 were observed.112 Moreover, the exposure to Pen c 13 provoked changes in lung proteome,
with increase of proteins involved in leukocyte extravasation signalling, oxidative stress response, and
actin cytoskeleton organization, and also decrease of vinculin, a junctional protein between cells.112
Therefore, Pen c 13 exposure causes structure alterations and actin cytoskeletal rearrangements, result-
ing in increased permeability and airway structural changes.112
35.5 Conclusions
Given that penicillia only show a limited invasive ability for animal tissue, they are mainly soil sapro-
phytes or plant pathogens, being responsible for mycotoxin production and postharvest rotting of fruits
and vegetables. However, Penicillium and Talaromyces spp. have been described as rare causative agents
of infectious diseases in humans, dogs, and fish, as well as environmental allergens among asthmatic
and atopic individuals. T. marneffei, the only dimorphic species of this group, is capable of growing
as a uninucleate pathogenic yeast at 37°C upon invasion of a human host, leading to localized reac-
tions in persons with normal immunity, while causing fatal systemic mycosis in immunocompromised
Penicillium and Talaromyces 571
patients. Diagnosis of penicilliosis is based on the confirmation by culture methods through specimen
examination, detection of antibodies produced against various fungal proteins, the detection of fungal
proteins in sera or in urine, or by selective amplification of T. marneffei DNA. Bamboo rats are the only
known nonhuman host of T. marneffei, but these animals show no signs of illness. Therefore, laboratory
models to study the pathogenicity of T. marneffei are extremely limited.
In contrast to pathogenic penicillia, mycotoxins produce toxic effects in various organisms, allow-
ing the use of different bioassays to study the toxicity of such secondary metabolites. Vertebrate ani-
mals have been used to test the toxicity of Penicillium mycotoxins. Tests with microorganisms and
invertebrates are preferred to the use of vertebrates due to ethical objections and high maintenance
costs. Organs and cell cultures have also been used, particularly when obtaining information on the
effect on DNA, cell organelles, or cell membranes. In addition, different culture media have been
tested to study the ability and efficiency of Penicillium spp. to produce mycotoxins, including food
analogues media.
Some Penicillium species produce allergens that pose a respiratory health hazard to susceptible indi-
viduals. Airborne spores from dry-ripened foods may cause food allergies or occupational respiratory
diseases to food industry workers. Sensitivity to penicillia allergens can be studied using common tests
with fungal extracts, but rodents and cell cultures are required to assess organic effects, such as eosino-
phil, specific IgE and IgG antibodies, interferon, and ILs levels; lung lavage fluids production; and his-
topathological lesions. The genetic susceptibility of the different animal strains to allergens makes it
necessary to select the strain exhibiting the adequate response to the mold, according to the exposure
and sensitization techniques used. Different types of respiratory epithelial cell cultures have been used
to study the effect of allergens form Penicillium spp. on secretion of mediators linked to local immune
responses. However, the use of cell cultures is otherwise rather limited.
REFERENCES
1. Houbraken, J. and Samson, R.A., Phylogeny of Penicillium and the segregation of Trichocomaceae into
three families, Stud. Mycol., 70, 1, 2011.
2. Samson, R.A. et al., Phylogeny and nomenclature of the genus Talaromyces and taxa accommodated in
Penicillium subgenus Biverticillium, Stud. Mycol., 70, 159, 2011.
3. Dyer, P.S. and O’Gorman, C.M., A fungal sexual revolution: Aspergillus and Penicillium show the way,
Curr. Opin. Microbiol., 14, 649, 2011.
4. Yilmaz, N. et al., Polyphasic taxonomy of the genus Talaromyces, Stud. Mycol., 78, 175, 2014.
5. Vos, J.P. et al., Disseminated Penicillium radicum infection in a dog, clinically resembling multicentric
malignant lymphoma, Vlaams Diergeneeskundig Tijdschrift, 78, 183, 2009.
6. Tomlinson, J.K. et al., Case report: Granulomatous lymphadenitis caused by Talaromyces helicus in a
labrador retriever, Vet. Clin. Pathol., 40, 553, 2011.
7. Blaylock, R.B., Overstreet, R.M. and Klich, M.A., Mycoses in red snapper (Lutjanus campechanus)
caused by two deuteromycete fungi (Penicillium corylophilum and Cladosporium sphaerospermum),
Hydrobiologia, 460, 221, 2001.
8. Ahearn, D.G. and Stulting, R.D., Fungi associated with drug recalls and rare disease outbreaks, J. Ind.
Microbiol. Biotechnol., 41, 1591, 2014.
9. Santos, P.E. et al., Penicillium piceum infection: Diagnosis and successful treatment in chronic
granulomatous disease, Med. Mycol., 44, 749, 2006.
10. Ho, M.H.K. et al., Penicillium marneffei infection and impaired IFN-g immunity in humans with
autosomal-dominant gain-of-phosphorylation STAT1 mutations, J. Allergy Clin. Immunol., 133, 894, 2014.
11. Segretain, G., Penicillium marneffei agent of mycosis of reticuloendothelial system, Mycopathologia,
11, 327, 1959.
12. Wang, T.K.F., Yuen, K.Y. and Wong, S.S.Y., Asymptomatic Penicillium marneffei fungemia in an
HIV-infected patient, Int. J. Inf. Dis., 11, 280, 2007.
13. Walsh, T.J. et al., Infections due to emerging and uncommon medically important fungal pathogens,
Eur. Soc. Clin. Microbiol. Infect. Dis., CMI, 10 (Suppl. 1), 48, 2004.
14. Plonga, R. et al., Penicilliosis marneffei infection in non-HIV-infected patients, J. Infect. Dis.
Antimicrob. Agents, 28, 71, 2011.
572 Laboratory Models for Foodborne Infections
15. Qiu, Y. et al., Retrospective analysis of 14 cases of disseminated Penicillium marneffei infection with
osteolytic lesions, BMC Infect. Dis., 15, 47, 2015.
16. Cooney, N.M. and Klein, B.S., Fungal adaptation to the mammalian host: It is a new world, after all,
Curr. Opin. Microbiol., 11, 511, 2008.
17. Andrianopoulos, A., Control of morphogenesis in the human fungal pathogen Penicillium marneffei,
Int. J. Med. Microbiol., 292, 331, 2002.
18. Cooper, C.R. and Vanittanakom, N., Insights into the pathogenicity of Penicillium marneffei, Future
Microbiol., 3, 43, 2008.
19. Boyce, K.J. and Andrianopoulos, A., Morphogenetic circuitry regulating growth and development in the
dimorphic pathogen Penicillium marneffei, Eukaryot. Cell, 12, 154, 2013.
20. Boyce, K.J. and Andrianopoulos, A., A p21-activated kinase is required for conidial germination in
Penicillium marneffei, PLoS Pathog., 3, e162, 2007.
21. Yang, E. et al., Signature gene expression reveals novel clues to the molecular mechanisms of dimorphic
transition in Penicillium marneffei, PLoS Genet., 10, 1, 2014.
22. Deepa, A.G. et al., Uncommon opportunistic fungal infections of oral cavity: A review, J. Oral
Maxillofac. Pathol., 18, 235, 2014.
23. Supparatpinyo, K. et al., Disseminated Penicillium marneffei infection in Southeast Asia, Lancet, 344,
110, 1994.
24. Lupi, O., Tyring, S.K. and McGinnis, M.R., Tropical dermatology: Fungal tropical diseases, J. Am.
Acad. Dermatol., 53, 931, 2005.
25. Sirisanthana, T. and Supparatpinyo, K., Epidemiology and management of penicilliosis in human
immunodeficiency virus-infected patients, Int. J. Infect. Dis., 3, 48, 1998.
26. Hamilton, A., Penicillium marneffei: Penicilliosis and the red peril in the east, Mycologist, 17, 84, 2003.
27. Vanittanakom, N., Penicillium marneffei infection and recent advances in the epidemiology and
molecular biology aspects, Clin. Microbiol. Rev., 19, 95, 2006.
28. Sekhon, A.S., Li, J.S.K. and Garg, A.K., Penicilliosis marneffei: Serological and exoantigen studies,
Mycopathologia, 77, 51, 1982.
29. LoBuglio, K.F. and Taylor, J.W., Phylogeny and PCR identification of the human pathogenic fungus
Penicillium marneffei, J. Clin. Microbiol., 33, 85, 1995.
30. Fisher, M.C. et al., Multilocus microsatellite typing system for Penicillium marneffei reveals spatially
structured populations, J. Clin. Microbiol., 42, 5065, 2004.
31. Cao, C. et al., Common reservoirs for Penicillium marneffei infection in humans and rodents, China,
Emerg. Infect. Dis., 17, 209, 2011.
32. Steinbach, W.J. and Perfect, J.R., Newer antifungal therapy for emerging fungal pathogens, Int. J. Infect.
Dis., 7, 5, 2003.
33. Sirisanthana, T. et al., Amphotericin B and itraconazole for treatment of disseminated Penicillium marnef-
fei infection in human immunodeficiency virus-infected patients, Clin. Infect. Dis., 26, 1107, 1998.
34. Wong, S.Y.N. and Wong, K.F., Penicillium marneffei infection in AIDS, Pathol. Res. Int., 2011, 1, 2011.
35. Moss, M.O., The environmental factors controlling mycotoxin formation, in Mycotoxins and Animal
Foods, p. 37, Smith J.F. and Henderson R.S. (Eds.), CRC Press, Boca Raton, 1991.
36. Mantle, P.G., Miscellaneous toxigenic fungi, in Mycotoxins and Animal Foods, p. 141, Smith J.E. and
Henderson R.S. (Eds.), CRC Press, Boca Raton, 1991.
37. Bullerman, L.B., Fusaria and toxigenic molds and other Aspergilli and Penicillia, in Food Microbiology:
Fundamentals and Frontiers, p. 419, Doyle M.P., Beuchat L.R. and Montville T.J. (Eds.), ASM Press,
Washington, 1997.
38. Pitt, J.I., Toxigenic Penicillium species, in Food Microbiology: Fundamentals and Frontiers, pp. 406,
Doyle M.P., Beuchat L.R. and Montville T.J. (Eds.), ASM Press, Washington, 1997.
39. Frisvad, J.C. and Samson, R.A., Polyphasic taxonomy of Penicillium subgenus Penicillium: A guide to iden-
tification of food and air-borne terverticillate Penicillia and their mycotoxins, Stud. Mycol., 49, 1, 2004.
40. Barkai-Golan, R., Penicillium mycotoxins, in Mycotoxins in Fruits and Vegetables, p. 153, B arkai-Golan
R. and Paster N. (Eds.), Academic Press, San Diego, 2008.
41. Panigrahi, S., Bioassay of mycotoxins using terrestrial and aquatic, animal and plant species, Food
Chem. Toxicol., 31, 767, 1993.
42. Watson, D.H. and Lindsay, D.G., A critical review of biological methods for the detection of fungal
toxins in foods and foodstuffs, J. Sci. Food Agric., 33, 59, 1982.
Penicillium and Talaromyces 573
43. Lagadic, L. and Caquet, T., Invertebrates in testing of environmental chemicals: Are they alternatives?
Environ. Health Perspect., 106 (Suppl 2), 593, 1998.
44. Harwig, J. and Scott, P.M., Brine shrimp (Artemia salina L.) larvae as a screening system for fungal
toxins, Appl. Microbiol., 21, 1011, 1971.
45. Tanaka, K., Goto, T. and Manabe, M., Biological test using Moina macrocopa Strauss (Part 1). Rearing
conditions of Moina macrocopa Strauss and influence of mycotoxins on this animal plankton, Rep. Nat.
Food Res. Inst., 48, 36, 1986.
46. Starodub, N.F. et al., Express control of toxicity and content of patulin by optical biosensors, in
Nanotechnology 2010: Bio Sensors, Instruments, Medical, Environment and Energy, p. 137, Nano
Science and Technology Institute, Danville, 2010.
47. Reiss, J., Toxic effects of mycotoxins, aflatoxin B1, rubratoxin B, patulin, and diacetoxyscirpenol on the
crustacean Cyclops fuscus, J. Assoc. Off. Anal. Chem., 55, 895, 1972.
48. Escoula, L., More, J. and Baradat, C., The toxins of Byssochlamys nivea Westling. 1. Acute toxicity of
patulin in adult rats and mice, Ann. Rech. Vet., 8, 41, 1977.
49. McKinley, E.R. and Carlton, W.W., Patulin mycotoxicosis in Swiss ICR mice, Food Cosmet Toxicol., 18,
181, 1980.
50. Gräbsch, C. et al., Cytotoxicity assessment of gliotoxin and penicillic acid in Tetrahymena pyriformis,
Environ. Toxicol., 21, 111, 2006.
51. Hayes, A.W., Antiprotozoal activity of rubratoxin B, Antimicrob. Agents Chemother., 4, 80, 1973.
52. Skrobek, A. et al., Evaluation of different biological test systems to assess the toxicity of metabolites
from fungal biocontrol agents, Toxicol. Lett., 161, 43, 2006.
53. Dive, D., Moreau, S. and Cacan, M., Use of a ciliate protozoan for fungal toxins studies, Bull. Environ.
Contam. Toxicol., 19, 489, 1978.
54. Bijl, J.P. et al., Potentials of a synchronized culture of Tetrahymena pyriformis for toxicity studies of
mycotoxins, J. Assoc. Off. Anal. Chem., 71, 282, 1988.
55. Rao, V.K. and Girisham, S., Antimicrobial and DNA damaging activity of ochratoxin A extracted from
Penicillium species, Int. J. Pharm. Bio Sci., 5, 335, 2014.
56. Gharaei-Fathabad, E., Tajick-Ghanbary, M.A. and Shahrokhi, N., Antimicrobial properties of
Penicillium species isolated from agricultural soils of northern Iran, Res. J. Toxins, 6, 1, 2014.
57. Lenz, P. and Süssmuth, R., A highly sensitive bacterial assay for toxins based on swarming inhibition,
and comparison with the cup plate assay based on growth inhibition, Toxicology, 45, 185, 1987.
58. Lenz, P., Süssmuth, R. and Seibel, E., Development of sensitive bacterial tests, exemplified by two
mycotoxins, Toxicology, 40, 199, 1986.
59. Skaug, M.A., Eduard, W. and Størmer, F.C., Ochratoxin A in airborne dust and fungal conidia,
Mycopathologia, 151, 93, 2000.
60. Auffray, Y. and Boutibones, P., Evaluation of the genotoxic activity of some mycotoxins using
Escherichia coli in the SOS spot test, Mutat. Res., 171, 79, 1986.
61. Haighton, L.A. et al., A reassessment of risk associated with dietary intake of ochratoxin A based on a
lifetime exposure model, Crit. Rev. Toxicol., 42, 147, 2012.
62. Yates, I.E. and Porter, J.K., Bacterial bioluminescence as a bioassay for mycotoxins, Appl. Environ.
Microbiol., 44, 1072, 1982.
63. Föllmann, W. and Lucas, S., Effects of the mycotoxin ochratoxin A in a bacterial and a mammalian in
vitro mutagenicity test system, Arch. Toxicol., 77, 298, 2003.
64. Puel, O., Galtier, P. and Oswald, I.P., Biosynthesis and toxicological effects of patulin, Toxins, 2, 613, 2010.
65. Wurgler, F.E., Friederich, U. and Schlatter, J., Lack of mutagenicity of ochratoxin A and B, citrinin,
patulin and cnestine in Salmonella typhimurium TAI02, Mutat. Res., 261, 209, 1991.
66. Wehner, F.C. et al., Mutagenicity to Salmonella typhimurium of some Aspergillus and Penicillium
mycotoxins, Mutat. Res., 58, 193, 1978.
67. Burdock, G.A., Soni, M.G. and Carabin, I.G., Evaluation of health aspects of kojic acid in food. Regul.
Toxicol. Pharmacol., 33, 80, 2001.
68. Mortelmans, K. and Zeiger, E., The Ames Salmonella/microsome mutagenicity assay, Mutat. Res., 455,
29, 2000.
69. Huff, J., Carcinogenicity of ochratoxin A in experimental animals, in Mycotoxins, Endemic Nephropathy
and Urinary Tract Tumours, p. 229, Castegnaro, M., Plestina, R., Dirheimer, G., Chernozemsky, I.N.
and Bartsch, H. (Eds.), International Agency for Research on Cancer, Lion, 1991.
574 Laboratory Models for Foodborne Infections
70. Vettorazzi, A., González-Peñas, E. and López de Cerain, A., Ochratoxin A kinetics: A review of
analytical methods and studies in rat model, Food Chem. Toxicol., 72, 273, 2014.
71. Stoev, S.D., Studies on carcinogenic and toxic effects of ochratoxin A in chicks, Toxins, 2, 649, 2010.
72. Vesely, D., Vesela, D. and Jelinek, R., Use of chick embryo in screening for toxin-producing fungi,
Mycopathologia, 88, 135, 1984.
73. Abedi, Z.H. and Scott P.M., Detection of toxicity of aflatoxins, sterigmatocystin and other fungal toxins
by lethal action on zebra fish larvae, J. Assoc. Off. Anal. Chem., 52, 963, 1969.
74. Wu, T.-S. et al., Evaluation of nephrotoxic effects of mycotoxins, citrinin and patulin, on zebrafish
(Danio rerio) embryos, Food Chem. Toxicol., 50, 4398, 2012.
75. Wu, T.-S. et al., Cardiotoxicity of mycotoxin citrinin and involvement of microRNA-138 in zebrafish
embryos, Toxicol. Sci., 136, 402, 2013.
76. Borenfreund, E., Babich, H. and Martin-Alguacil, N., Comparisons of two in vitro cytotoxicity assays-
the neutral red (NR) and tetrazolium (MTT) tests, Toxicol. In Vitro, 2, 1, 1988.
77. Ali, R. et al., Comparative analysis of micronuclei and DNA damage induced by Ochratoxin A in two
mammalian cell lines, Mutat. Res., 723, 58, 2011.
78. Ferrer, E. et al., Reactive oxygen species induced by beauvericin, patulin and zearalenone in CHO-K1
cells, Toxicol. In Vitro, 23, 1504, 2009.
79. Bouslimi, A. et al., Individual and combined effects of ochratoxin A and citrinin on viability and
DNA fragmentation in cultured Vero cells and on chromosome aberrations in mice bone marrow cells,
Toxicology, 251, 1, 2008.
80. Bünger, J. et al., Cytotoxicity of occupationally and environmentally relevant mycotoxins, Toxicology,
202, 199, 2004.
81. Corcuera, L.A. et al., Ochratoxin A reduces aflatoxin B1 induced DNA damage detected by the comet
assay in Hep G2 cells, Food Chem. Toxicol., 49, 2883, 2011.
82. Zhou, S. et al., Patulin-induced oxidative DNA damage and p53 modulation in HepG2 cells, Toxicon,
55, 390, 2010.
83. Heussner, A.H., Dietrich, D.R. and Brien, E.O., In vitro investigation of individual and combined cytotoxic
effects of ochratoxin A and other selected mycotoxins on renal cells, Toxicol. In Vitro, 20, 332, 2006.
84. Liu, B. et al., Evaluation of genotoxic risk and oxidative DNA damage in mammalian cells exposed to
mycotoxins, patulin and citrinin, Toxicol. Appl. Pharmacol., 191, 255, 2003.
85. Föllmann, W., Behm, C., and Degen, G.H., Toxicity of the mycotoxin citrinin and its metabolite dihy-
drocitrinone and of mixtures of citrinin and ochratoxin A in vitro, Arch. Toxicol., 88, 1097, 2014.
86. Simpson, L.J. and Sale, J.E., Sister chromatid exchange assay, Subcell. Biochem., 40, 399, 2006.
87. Kirsch-Volders, M. et al., In vitro genotoxicity testing using the micronucleus assay in cell lines, human
lymphocytes and 3D human skin models, Mutagenesis, 26, 177, 2011.
88. Collins, A.R., The comet assay for DNA damage and repair. Principles, applications, and limitations,
Mol. Biotechnol., 26, 249, 2004.
89. Dombrink-Kurtzman, M.A. and Blackburn, J.A., Evaluation of several culture media for production of
patulin by Penicillium species, Int. J. Food Microbiol., 98, 241, 2005.
90. Kokkonen, M., Jestoi, M. and Rizzo, A., The effect of substrate on mycotoxin production of selected
Penicillium strains, Int. J. Food Microbiol., 99, 207, 2005.
91. Rao, V.K. et al., Culture media and factors influencing ochratoxin A production by two species of
Penicillium isolated from poultry feeds, Natl. Acad. Sci. Lett., 36, 101, 2013.
92. Portnoy, J.M. and Jara, D., Mold allergy revisited, Ann. Allergy Asthma Immunol., 114, 83, 2015.
93. Sharpe, R.A. et al., Indoor fungal diversity and asthma: A meta-analysis and systematic review of risk
factors, J. Allergy Clin. Immunol., 135, 110, 2015.
94. WHO/IUIS, Allergen nomenclature. http://www.allergen.org/index.php (Accessed May 27, 2015), 2015.
95. Gunschera, J. et al., Formation and emission of chloroanisoles as indoor pollutants, Environ. Sci. Pollut.
Res. Int., 11, 147, 2004.
96. Feo, F. et al., Penicillium nalgiovense as an occupational and contact allergen, J. Allergy Clin. Immunol.,
112, 213, 2003.
97. Merget, R. et al., Occupational immunoglobulin E-mediated asthma due to Penicillium camemberti in
a dry-sausage packer, Respiration, 76, 109, 2008.
98. Rouzaud, P. et al., Symptoms and serum precipitins in workers exposed to dry sausage mould:
Consequences of exposure to sausage mould, Int. Arch. Occup. Environ. Health, 74, 371, 2001.
Penicillium and Talaromyces 575
99. Wantke, F. et al., Contact dermatitis caused by salami skin, Contact Dermat., 64, 111, 2011.
100. Campbell, J.A. et al., Cheese worker’s hypersensitivity pneumonitis, Am. Rev. Respir. Dis., 127, 495, 1983.
101. Bobolea, I. et al., Allergy to dry fermented sausage, J. Invest. Allergol. Clin. Immunol., 19, 324, 2009.
102. Boisnault, A. et al., Allergie alimentaire à une moisissure présente sur une croûte de fromage: À propos
d’une observation pédiatrique, Rev. Fr. d’Allergol. d’Immunol. Clin., 45, 597, 2005.
103. González-de-Olano, D. et al., Different patterns of sensitization in allergy to dry fermented sausage,
J. Invest. Allergol. Clin. Immunol., 22, 152, 2012.
104. Guillet, M.H. et al., Urticaire et choc anaphylactique par allergie alimentaire à Penicillium italicum,
Rev. Fr. d’Allergol. d’Immunol. Clin., 43, 520, 2003.
105. Morisset, M. et al., Food allergy to moulds: Two cases observed after dry fermented sausage ingestion,
Allergy Eur. J. Allergy Clin. Immunol., 58, 1203, 2003.
106. Ormerod, A.D., Reid, T.M.S. and Main, R.A., Penicillin in milk—its importance in urticaria, Clin. Exp.
Allergy, 17, 229, 1987.
107. Padinjakara, R.N.K., Ashawesh, K. and Patel, V., Allergic reaction to blue cheese: Serendipity or actual
causation? N. Z. Med. J., 121, 102, 2008.
108. Raison-Peyron, N. et al., Anaphylaxis to beef in penicillin-allergic patient, Allergy, 56, 796, 2001.
109. Templeton, S.P. et al., Murine models of airway fungal exposure and allergic sensitization, Med. Mycol.,
48, 217, 2010.
110. Chung, Y.J. et al., Dose-dependent allergic responses to an extract of Penicillium chrysogenum in
BALB/c mice, Toxicology, 216, 73, 2005.
111. Ward, M.D.W. et al., A comparison of the allergic responses induced by Penicillium chrysogenum and
house dust mite extracts in a mouse model, Indoor Air, 20, 380, 2010.
112. Chen, J.C. et al., The protease allergen Pen c 13 induces allergic airway inflammation and changes in
epithelial barrier integrity and function in a murine model, J. Biol. Chem., 286, 26667, 2011.
113. Ward, M.D.W. et al., Assessing the allergenic potential of molds found in water-damaged homes in a
mouse model, Inhal. Toxicol., 26, 474, 2014.
114. Schwab, C.J. et al., Characterization of exposure to low levels of viable Penicillium chrysogenum
conidia and allergic sensitization induced by a protease allergen extract from viable P. chrysogenum
conidia in mice, Int. Arch. Allergy Immunol., 130, 200, 2003.
115. Schwab, C.J. et al., Allergic inflammation induced by a Penicillium chrysogenum conidia-associated
allergen extract in a murine model, Allergy Eur. J. Allergy Clin. Immunol., 59, 758, 2004.
116. Cooley, J. et al., Cellular and humoral responses in an animal model inhaling Penicillium chrysogenum
conidia, in Bioaerosols, Fungi and Mycotoxins : Health Effects, Assessment, Prevention and Control,
p. 403, Johanning, E. (Ed.), Boyd Printing Company Inc., Albany, NY, 1999.
117. Cooley, J.D. et al., An animal model for allergic penicilliosis induced by the intranasal instillation of
viable Penicillium chrysogenum conidia, Thorax, 55, 489, 2000.
118. Rand, T.G. et al., Inflammatory and cytotoxic responses in mouse lungs exposed to purified toxins from
building isolated Penicillium brevicompactum Dierckx and P. chrysogenum Thom, Toxicol. Sci., 87,
213, 2005.
119. Alonso, A. et al., Hypersensitivity pneumonitis induced by Penicillium notatum antigens in guinea pigs,
J. Invest. Allergol. Clin. Immunol., 8, 52, 1998.
120. Lichtenstein, J.H.R. et al., Strain differences influence murine pulmonary responses to Stachybotrys
chartarum, Am. J. Respir. Cell Mol. Biol., 35, 415, 2006.
121. Tai, H.Y. et al., Pen ch 13 allergen induces secretion of mediators and degradation of occludin protein of
human lung epithelial cells, Allergy Eur. J. Allergy Clin. Immunol., 61, 382, 2006.
Section V
Dongyou Liu
CONTENTS
36.1 Introduction................................................................................................................................... 579
36.1.1 Classification, Morphology, and Genomics..................................................................... 580
36.1.1.1 Classification..................................................................................................... 580
36.1.1.2 Morphology....................................................................................................... 580
36.1.1.3 Genomics...........................................................................................................581
36.1.2 Life Cycle and Epidemiology........................................................................................... 582
36.1.2.1 Life Cycle.......................................................................................................... 582
36.1.2.2 Epidemiology.................................................................................................... 582
36.1.3 Clinical Features and Pathogenesis.................................................................................. 583
36.1.3.1 Amoebic (Acanthamoebic) Keratitis................................................................. 583
36.1.3.2 Granulomatous Amoebic Encephalitis............................................................. 583
36.1.3.3 Cutaneous Acanthamoebiasis and Sinusitis..................................................... 583
36.1.4 Diagnosis.......................................................................................................................... 584
36.1.5 Treatment and Prevention................................................................................................. 585
36.2 Laboratory Models........................................................................................................................ 585
36.2.1 Animal Models................................................................................................................. 585
36.2.1.1 Mice.................................................................................................................. 585
36.2.1.2 Rats................................................................................................................... 585
36.2.1.3 Chinese Hamsters............................................................................................. 586
36.2.1.4 Rabbits.............................................................................................................. 586
36.2.1.5 Pigs.................................................................................................................... 586
36.2.2 In Vitro Models................................................................................................................. 586
36.3 Conclusion..................................................................................................................................... 586
References............................................................................................................................................... 586
36.1 Introduction
Acanthamoeba is a unicellular organism that was first detected by Castellani from Cryptococcus
pararoseus cultures. As a free-living bacterivore, Acanthamoeba is present in a diversity of
environments, including soil, dust, water, air, plants, animals, and humans. However, Acanthamoeba
has the ability to take advantage of temporary weakness (trauma or suppressed immune function) in
human host, causing keratitis, encephalitis, and skin infections, often with severe consequences. In
view of the limited treatment options for Acanthamoeba infections at present, there is a need to improve
our understanding of the pathogenicity of this devastating disease, and to devise novel, highly effective
intervention strategies. Utilization of laboratory models to Acanthamoeba infections will be undoubt-
edly beneficial in this regard.
579
580 Laboratory Models for Foodborne Infections
36.1.1.2 Morphology
Acanthamoeba trophozoites (usually <30 μm, rarely >65 μm in size) are amoeboid in shape (or wide
and tongue-shaped), with irregular margins and short and fine pseudopodia (or acanthopodia; Greek
“acanth” means “spikes”). Continuously forming and reabsorbing, acanthopodia (formed by bundles of
actin microfilaments extending as rigid cores) protrude from every area of the body’s surface, contribut-
ing to Acanthamoeba locomotion. However, in A. astronyxis and A. comandoni, the acanthopodia may
be quite long.
Under adverse environmental conditions (e.g., food deprivation, desiccation, and changes in tempera-
ture and pH), Acanthamoeba forms a double-walled wrinkled cyst (of 13–20 μm in size). The outer
wall is made up of proteins and polysaccharides, and the inner wall contains cellulose, with the two
walls being separated by a space forming the so-called ectocyst and endocyst. Cysts are resistant to
Acanthamoeba 581
TABLE 36.1
Correlation of Acanthamoeba T-Types with Clinical Diseases
T-Type Species Clinical Disease
T1 Acanthamoeba sp. Encephalitis
T2 A. palestinensis, A. pustulosa Keratitis, encephalitis, sinusitis
T3 A. griffini, A. pearcei Keratitis
T4 A. castellanii, A. divionensis, A. lugdunensis, A. Keratitis, encephalitis
mauritaniensis, A. polyphaga, A.quina, A. rhysodes
T5 A. lenticulata Keratitis, encephalitis
T6 Acanthamoeba sp. Keratitis
T7 A. astronyxis Unknown
T8 A. tubiashi Unknown
T9 A. comandoni Unknown
T10 A. culbertsoni, Acanthamoeba sp. Keratitis, encephalitis
T11 A. hatchetti, A. stevensoni Keratitis
T12 A. healyi Encephalitis
T13 Acanthamoeba sp. Unknown
T14 Acanthamoeba sp. Unknown
T15 A. jacobsi Keratitis
T16 Acanthamoeba sp. Unknown
T17 Acanthamoeba sp. Unknown
T18 A. byersi Encephalitis, skin infection
Sources: Crary, M.J. Genetic variability and its relationship to Acanthamoeba pathogenesis. Thesis,
The Ohio State University, 2012; Sente, C., et al., Parasite Vectors, 9(1), 127, 2016.
biocides, chlorination, and antibiotics and survive low temperatures (0°C–2°C). There is evidence
that Acanthamoeba cysts remain viable for more than 24 years after storage in water at 4°C and sus-
tain desiccation for more than 20 years. However, Acanthamoeba cysts are sensitive to treatment with
Freon or methylene oxide or autoclaving. When condition improves [e.g., inoculation on nonnutrient
agar (NNA) containing bacteria], excystment takes place with trophozoites emerging from the cyst. As
Acanthamoeba cyst formation (encystment) and excystation may be induced using nonnutrient media,
Acanthamoeba offers a valuable tool to study cellular differentiation [12].
Electron microscopy examination reveals the presence of a Golgi complex, smooth and rough endo-
plasmic reticula, free ribosomes, digestive vacuoles, mitochondria, and microtubules in Acanthamoeba
trophozoites. Apart from the existence of a trilaminar plasma membrane surrounding the cytoplasmic
contents, characteristic spiny surface projections (i.e., acanthopodia) are observable. Within the cyto-
plasm are one or more prominent contractile vacuoles with osmotic function, and a nucleus (with a large
central nucleolus) that is approximately one-sixth the size of trophozoite. Other notable types of vacuoles
in the cytoplasm include lysosomes, digestive vacuoles, and glycogen-containing vacuoles [2].
36.1.1.3 Genomics
From the sequence data of 14 Acanthamoeba species available in GenBank, it is clear that
Acanthamoeba genomes vary from 42.02 to 120.42 Mb in size, with GC contents ranging from 42.7%
to 59.3% (Table 36.2). Interestingly, A. castellanii, a solitary free-living amoebozoan of T4 genotype,
possesses a genome of 42.02 Mb including 15,455 compact intron-rich genes. Besides encoding vari-
ous tyrosine kinase signaling proteins, A. castellanii genome also generates a diverse repertoire of
predicted pattern recognition receptors, many of which show predicted orthologous functions in the
innate immune systems of higher organisms. The size of A. castellanii mitochondrial DNA genome
is 41,591 bp [13].
582 Laboratory Models for Foodborne Infections
TABLE 36.2
Genomic Comparison of Acanthamoeba Species
Species T-Type Genome Size (Mb) GC Content (%)
Acanthamoeba astronyxis T7 83.43 42.9
Acanthamoeba castellanii T4 42.02–45 57.8–58.4
Acanthamoeba culbertsoni T10 55.54 50.4
Acanthamoeba divionensis T4 84.77 42.7
Acanthamoeba healyi T12 75.32 58.7
Acanthamoeba lenticulata T3 66.03 56.9
Acanthamoeba lugdunensis T4 99.42 59.1
Acanthamoeba mauritaniensis T4 106.84 58.7
Acanthamoeba palestinensis T2 103.48 59.1
Acanthamoeba pearcei T3 115.61 59
Acanthamoeba polyphaga T4 120.42 59.3
Acanthamoeba quina T11 83.59 59.2
Acanthamoeba rhysodes T4 75.82 57.9
Acanthamoeba royreba T4 79.54 51.3
36.1.2.2 Epidemiology
As a free-living organism, Acanthamoeba is ubiquitously distributed in nature and has been found in
the environment (e.g., soil, dust, air, natural and treated water, seawater, swimming pools, sewage, sedi-
ments, air-conditioning units, domestic tap water, drinking water treatment plants, and bottled water),
health care facilities (e.g., dental treatment units, hospitals and dialysis units, eyewash stations, contact
Acanthamoeba 583
lenses, and lens cases), vegetation, animals (e.g., fish, amphibia, reptiles, and mammals), and humans
(skin, nasal cavities, throat, intestines, brain, lungs, and cornea) [14–16].
In addition, Acanthamoeba may harbor various microbial endosymbionts (e.g., Candidatus Caedibacter
acanthamoebae, Candidatus Odyssella thessalonicensis, Candidatus Paracaedibacter acantham-
oebae, Candidatus Paracaedibacter symbiosus, Comamonas acidovorans, Legionella pneumophila,
Pseudomonas aeruginosa, mimivirus, megavirus, and pandoravirus). Further, a large number of other
bacterial species (e.g., Aeromonas, Bacillus cereus, Bartonella, Burkholderia, Campylobacter jejuni,
Chlamydia pnuemoniae, Coxiella burnetii, Cytophaga, E. coli O157:H7, Flavobacterium, Francisella
tularensis, Helicobacter pylori, Listeria, Mycobacterium, Pasteurella multocida, Prevotella interme-
dia, Porphyromonas gingivalis, Rickettsia, Salmonella Typhimurium, Shigella, Simkania negevensis,
Staphylococcus aureus, Vibrio, and Waddlia chondrophila) have been shown to survive and multiply
within Acanthamoeba. This highlights the potential role of Acanthamoeba in serving as bacterial reser-
voirs for human infections [2,12].
36.1.4 Diagnosis
Traditionally, identification of Acanthamoeba is dependent on morphological characterization through
microscopic examination of clinical specimens [direct wet mounts of cerebrospinal fluid (CSF) or bron-
choalveolar lavage (BAL) fluid cytospin preparations, and stained smears of CSF sediment, brain, cuta-
neous lesion scrape/biopsy, or corneal scrape/biopsy]. The sizes and shape of trophozoites and cysts
as well as distinct nuclear structure [characterized by a prominent nucleolus, contractile vacuole, and
cytoplasmic vacuoles, which may be visualized more readily using trichrome or hematoxylin and eosin
(H&E) stains on fixed preparations after cytocentrifugation] help distinguish Acanthamoeba from host
macrophages and other immune cells. In addition, Acanthamoeba cyst wall turns red after periodic
acid-Schiff staining, and its cyst becomes black after Gomori methenamine silver staining. Calcofluor
white (a chemofluorescent dye with an affinity for the polysaccharide polymers) stains amebic cyst walls
bright apple green, which, with Evans blue counterstaining the background, is useful for identifica-
tion of Acanthamoeba cysts in brain or corneal tissue. Acridine orange staining of corneal scrapings
or CSF represents another simple and reliable method for rapid histological diagnosis of AK or GAE.
Furthermore, transmission electron microscopy of infected tissues, immunofluorescent or immunoper-
oxidase cytochemical staining of cryostat sections, or infected tissues embedded in paraffin offer other
approaches for identification of Acanthamoeba [26].
To enhance its detection and identification, Acanthamoeba is grown on NNA, 1.5% containing a
lawn of E. coli or E. aerogenes, axenically in PYG medium (2% proteose peptone, 0.2% yeast extract,
and 0.1 M glucose), or in Oxoid medium (Cline medium, containing serum and hemin) at 28°C–35°C
for 10 days or more (to allow sufficient time for excystment). Alternatively, Acanthamoeba may be cul-
tured on mammalian cell monolayers [e.g., African green monkey kidney (Vero), human embryonic
lung (HEL), human embryonic kidney (HEK), HeLa, B103 rat neuroblastoma, and L929 fibroblasts].
Acanthamoeba ingestion of bacteria or cells produces clear plaques after a week. Microscopic examina-
tion of Acanthamoeba culture isolates provides valuable confirmation [2].
With molecular techniques moving from laboratory bench to clinical setting, a variety of nucleic acid
amplification procedures have been developed for Acanthamoeba identification. These methods exploit
variations of complete or partial nuclear 18S rRNA gene, complete mitochondrial 16S rRNA, complete
mitochondrial genome, and randomly amplified polymorphic DNA patterns. In particular, analysis of
nuclear 18S ribosomal RNA (18S rRNA) gene sequence has made fast, reliable, and repeatable identifica-
tion of Acanthamoeba feasible. By setting a cutoff of <5% sequence divergences, the nuclear 18S ribo-
somal RNA-based method has enabled separation of Acanthamoeba isolates into 18 genotypes (T-types
1–18 or T1–18). Typically, primers CRN5 (5′-TGGTTGATCCTGCCAGTAG-3′) and SSU2-TRUN
Acanthamoeba 585
36.2 Laboratory Models
36.2.1 Animal Models
36.2.1.1 Mice
Mice represent a model of choice for CNS infection, as mice infected via intranasal inoculation of
Acanthamoeba develop symptoms of head tilt, circling, twirling, limb paresis, and convulsive seizures,
along with amebic rhinitis and pneumonitis. In the capillaries of the lungs and brains, trophozoites and
cysts are present. In addition, mice may also be used for modeling AK, with neutrophil being the pre-
dominant cell type during early stage of AK pathogenesis [33].
36.2.1.2 Rats
Wistar rats are a preferred model for AK because of a low death rate and larger corneas for inoculation.
Intrastromal injection of Acanthamoeba or coinjection with bacteria (e.g., Corynebacterium) provides
an efficient way to initiate AK [34–36].
586 Laboratory Models for Foodborne Infections
36.2.1.3 Chinese Hamsters
Application of “contact lenses” containing Acanthamoeba to the abraded corneal surfaces of Chinese
hamsters for at least 5–7 days leads to acute and self-limiting corneal infection, with clinical features
of neutrophil infiltration, epithelial ulceration, edema, corneal opacity, and neovascularization [37–40].
36.2.1.4 Rabbits
Microinjection of 1 × 104/100 μL Acanthamoeba healyi trophozoites between the corneal epithelium
and Bowman’s layer, anterior to the corneal stroma of New Zealand white rabbits, enabled successful
establishment of AK, which displayed an efficient immune response with less severe pathology and was
strikingly similar to AK in humans [41,42].
36.2.1.5 Pigs
Due to the anatomical similarities of the pig eye to the human eye, pigs offer a useful model for ocular
infections such as AK. However, spontaneous resolution of AK in pigs occurs in 8–10 weeks, in contrast
to the prolonged infection in humans [43].
36.3 Conclusion
The genus Acanthamoeba consists of more than 25 species of free-living protozoa, which are charac-
terized by the generation of pseudopodia (acanthopodia) for locomotion during trophozoite stage and
the formation of a double-walled wrinkled cyst under adverse environmental conditions. While being
seemingly harmless creatures in the environment, certain Acanthamoeba species are surprisingly dan-
gerous, with the ability to take advantage of the temporary weakness in human host for its own gain,
and cause painful, sight-threatening AK in contact lens wearers and deadly GAE and skin infections in
immunocompromised individuals. In addition, Acanthamoeba has the potential to serve as reservoirs
for a number of human bacterial pathogen due to the fact that many bacteria have been shown to survive
and multiply within the parasite. Following the development of molecular test targeting nuclear 18S
rRNA gene, rapid and reliable identification of Acanthamoeba is possible [45]. However, considering the
current lack of options for treating and controlling Acanthamoeba infections, especially GAE, further
research using laboratory models to uncover the molecular details of Acanthamoeba pathogenesis is
critical.
REFERENCES
1.
Qvarnstrom Y, Nerad TA, Visvesvara GS. Characterization of a new pathogenic Acanthamoeba spe-
cies, A. byersi n. sp., isolated from a human with fatal amoebic encephalitis. J Eukaryot Microbiol.
2013;60(6):626–33.
2.
Marciano-Cabral F, Cabral G. Acanthamoeba spp. as agents of disease in humans. Clin Microbiol Rev.
2003;16:273–307.
Acanthamoeba 587
3. Crary MJ. Genetic variability and its relationship to Acanthamoeba pathogenesis. PhD Thesis, The Ohio
State University, 2012. Available at https://etd.ohiolink.edu/rws_etd/document/get/osu1343831120/inline.
4. Corsaro D, Venditti D. Phylogenetic evidence for a new genotype of Acanthamoeba (Amoebozoa
Acanthamoebida). Parasitol Res. 2010;107(1):233.
5. Fuerst PA, Booton GC, Crary M. Phylogenetic analysis and the evolution of the 18S rRNA gene typing
system of Acanthamoeba. J Eukaryot Microbiol. 2015;62(1):69–84.
6. Booton GC, Visvesvara GS, Byers TJ, Kelly DJ, Fuerst PA. Identification and distribution of Acanthamoeba
species genotypes associated with nonkeratitis infections. J Clin Microbiol. 2005;43(4):1689–93.
7. Booton GC, Joslin CE, Shoff M, Tu EY, Kelly DJ, Fuerst PA. Genotypic identification of Acanthamoeba
sp. isolates associated with an outbreak of Acanthamoeba keratitis. Cornea. 2009;28(6):673–6.
8. Kong HH. Molecular phylogeny of Acanthamoeba. Korean J Parasitol. 2009;47:S21–8.
9. Nuprasert W, Putaporntip C, Pariyakanok L, Jongwutiwes S. Identification of a novel T17 genotype of
Acanthamoeba from environ-mental isolates and T10 genotype causing keratitis in Thailand. J Clin
Microbiol. 2010;48(12):4636–40.
10. Maciver SK, Asif M, Simmen MW, Lorenzo-Morales J. A systematic analysis of Acanthamoeba geno-
type frequency correlated with source and pathogenicity: T4 is confirmed as a pathogen-rich genotype.
Eur J Protistol. 2013;49(2):217.
11. Mirjalali H, Niyyati M, Abedkhojasteh H, Babaei Z, Sharifdini M, Rezaeian M. Pathogenic assays of
Acanthamoeba belonging to the T4 genotype. Iran J Parasitol. 2013;8(4):530–5.
12. Siddiqui R, Khan NA. Biology and pathogenesis of Acanthamoeba. Parasite Vectors. 2012;5:6.
13. Clarke M, et al. Genome of Acanthamoeba castellanii highlights extensive lateral gene transfer and
early evolution of tyrosine kinase signaling. Genome Biol. 2013;14(2):R11.
14. Lasjerdi Z, et al. Potentially pathogenic free-living amoebae isolated from hospital wards with immu-
nodeficient patients in Tehran Iran. Parasitol Res. 2011;109(3):575.
15. Rahdar M, et al. Isolation and genotyping of Acanthamoeba strains from environmental sources in
Ahvaz city, Khuzestan province, southern Iran. Iran J Parasitol. 2012;7(4):22–6.
16. Sente C, et al. Occurrence and genetic characterisation of Acanthamoeba spp. from environmental and
domestic water sources in Queen Elizabeth Protected Area, Uganda. Parasite Vectors. 2016;9(1):127.
17. Polat ZA, Ozcelik S, Vural A, Yildiz E, Cetin A. Clinical and histologic evaluations of experimental
Acanthamoeba keratitis. Parasitol Res. 2007;101(6):1621–5.
18. Knickelbein JE, Kovarik J, Dhaliwal DK, Chu CT. Acanthamoeba keratitis: a clinicopathologic case
report and review of the literature. Hum Pathol. 2013;44(5):918–22.
19. McKellar MS, et al. Fatal granulomatous Acanthamoeba encephalitis mimicking a stroke, diagnosed by
correlation of results of sequential magnetic resonance imaging, biopsy, in vitro culture, immunofluo-
rescence analysis, and molecular analysis. J Clin Microbiol. 2006;44(11):4265–9
20. Khan NA. Acanthamoeba invasion of the central nervous system. Int J Parasitol. 2007;37(2):131–8.
21. Mortazavi PN, Goldsworthy G, Kirk R, Khan NA. Acanthamoeba produces disseminated infection
in locusts and traverses the locust blood-brain barrier to invade the central nervous system. BMC
Microbiol. 2010;10:186.
22. Clarke DW, Niederkorn JY. The pathophysiology of Acanthamoeba keratitis. Trends Parasitol.
2006;22(4):175–80.
23. Panjwani N. Pathogenesis of Acanthamoeba keratitis. Ocul Surf. 2010;8(2):70–9.
24. Khan NA. Pathogenicity, morphology, and differentiation of Acanthamoeba. Curr Microbiol.
2001;43(6):391–5.
25. Khan NA. Pathogenesis of Acanthamoeba infections. Microb Pathog. 2003;34(6):277–85.
26. Walochnik J, Obwaller A, Aspock H. Correlations between morphological, molecular biological, and
physiological characteristics in clinical and nonclinical isolates of Acanthamoeba spp. Appl Environ
Microbiol. 2000;66(10):4408.
27. Gooi P, Lee-Wing M, Brownstein S, El-Defrawy S, Jackson WB, Mintsioulis G. Acanthamoeba
keratitis: persistent organisms without inflammation after 1 year of topical chlorhexidine. Cornea.
2008;27:246–8.
28. Roberts CW, Henriquez FL. Drug target identification, validation, characterization and exploitation for
treatment of Acanthamoeba (species) infections. Ext Parasite. 2010; 126(1):91–6.
29. Kashiwabuchi RT, et al. Assessing efficacy of combined riboflavin and UV-A light (365 nm) treatment
of Acanthamoeba trophozoites. Invest Ophthalmol Vis Sci. 2011;52(13):9333–8.
588 Laboratory Models for Foodborne Infections
30. Berra M, et al. Treatment of Acanthamoeba keratitis by corneal cross-linking. Cornea. 2013;32(2):174–8.
31. Polat ZA, Walochnik J, Obwaller A, Vural A, Dursun A, Arici MK. Miltefosine and polyhexamethylene
biguanide: a new drug combination for the treatment of Acanthamoeba keratitis. Clin Exp Ophthalmol.
2014;42(2):151–8.
32. Walia R, Montoya JG, Visvesvera GS, Booton GC, Doyle RL. A case of successful treatment of cutane-
ous Acanthamoeba infection in a lung transplant recipient. Transpl Infect Dis. 2007;9(1):51–4.
33. Massilamany C, et al. SJL mice infected with Acanthamoeba castellanii develop central nervous sys-
tem autoimmunity through the generation of cross-reactive T cells for myelin antigens. PLoS One.
2014;9(5):e98506.
34. Ren M, Wu X. Evaluation of three different methods to establish animal models of Acanthamoeba
keratitis. Yonsei Med J. 2010;51(1):121–7.
35. Ren MY, Wu XY. Toll-like receptor 4 signalling pathway activation in a rat model of Acanthamoeba
keratitis. Parasite Immunol. 2011;33(1):25–33.
36. Veríssimo Cde M, Maschio VJ, Correa AP, Brandelli A, Rott MB. Infection in a rat model reactivates
attenuated virulence after long-term axenic culture of Acanthamoeba spp. Mem Inst Oswaldo Cruz.
2013;108(7):832–5.
37. Van Klink F, Leher H, Jager MJ, Alizadeh H, Taylor W, Niederkorn JY. Systemic immune response to
Acanthamoeba keratitis in the Chinese hamster. Ocul Immunol Inflamm. 1997;5:235–44.
38. Hurt M, Apte S, Leher H, Howard K, Niederkorn J, Alizadeh H. Exacerbation of Acanthamoeba
keratitis in animals treated with anti-macrophage inflammatory protein 2 or antineutrophil antibodies.
Infect Immun. 2001;69:2988–95.
39. Polat ZA, Obwaller A, Vural A, Walochnik J. Efficacy of miltefosine for topical treatment of
Acanthamoeba keratitis in Syrian hamsters. Parasitol Res. 2012;110(2):515–20.
40. Suryawanshi A, Cao Z, Sampson JF, Panjwani N. IL-17A-mediated protection against Acanthamoeba
keratitis. J Immunol. 2015;194(2):650–63.
41. Said NA, Shoeir AT, Panjwani N, Garate M, Cao Z. Local and systemic humoral immune response
during acute and chronic Acanthamoeba keratitis in rabbits. Curr Eye Res. 2004;29:429–39.
42. Feng X, Zheng W, Wang Y, Zhao D, Jiang X, Lv S. A Rabbit model of Acanthamoeba keratitis that
better reflects the natural human infection. Anat Rec (Hoboken). 2015;298(8):1509–17.
43. Alizadeh H, et al. Successful immunization against Acanthamoeba keratitis in a pig model. Cornea.
1995;14:180–6.
44. De Jonckheere JF. Growth characteristics, cytopathic effect in cell culture, and virulence in mice of 36
type strains belonging to 19 different Acanthamoeba spp. Appl Environ Microbiol. 1980;39(4):681–5.
45. Niyyati M, et al. Genotyping of Acanthamoeba isolates from clinical and environmental specimens in
Iran. Exp Parasitol. 2009;121(3):242–5.
37
Cryptosporidium
Dongyou Liu
CONTENTS
37.1 Introduction.................................................................................................................................... 589
37.1.1 Classification, Morphology, and Genome Structure........................................................ 589
37.1.1.1 Classification..................................................................................................... 589
37.1.1.2 Morphology....................................................................................................... 590
37.1.1.3 Genome Structure............................................................................................. 590
37.1.2 Life Cycle and Epidemiology............................................................................................591
37.1.3 Clinical Features and Pathogenesis.................................................................................. 592
37.1.4 Diagnosis.......................................................................................................................... 593
37.1.5 Treatment and Prevention................................................................................................. 593
37.2 Laboratory Models........................................................................................................................ 594
37.2.1 Animal Models................................................................................................................. 594
37.2.2 In Vitro Models................................................................................................................. 595
37.3 Conclusion..................................................................................................................................... 595
References............................................................................................................................................... 595
37.1 Introduction
The genus Cryptosporidium comprises nearly 30 known species of apicomplexan protozoans that infect
birds, reptiles, fish, and mammals, including humans. Of these, C. parvum and C. hominis along with
a few other species are responsible for producing watery diarrhea (intestinal cryptosporidiosis) with or
without a persistent cough (respiratory cryptosporidiosis) in both immunocompetent and immunodefi-
cient humans.
Although Cryptosporidium was first identified from the stomach of mice in 1907, its role in human
infection was only established in 1976. Since then, cryptosporidiosis has been recognized as a major
cause of chronic diarrhea in patients with AIDS, as a cause of zoonotic and waterborne outbreaks of
diarrhea, and as a cause of diarrhea and unexplained cough in immunocompetent children.
589
590 Laboratory Models for Foodborne Infections
37.1.1.2 Morphology
Cryptosporidium oocysts are round to oval in shape and measure about 4–6 μm in diameter (Table 37.1).
Under a phase-contrast microscope, Cryptosporidium oocysts are highly refractile in wet smear and con-
tain prominent dark granules (black dot) or small vacuoles, in contrast to yeast, which are not refractile
and contain no granules. Further, Cryptosporidium oocysts do not stain with Lugol’s iodine solution, but
yeast do. Excystment of Cryptosporidium oocysts release four sporozoites each.
37.1.1.3 Genome Structure
The genome sequences of C. parvum IOWA, C. hominis TU502, and C. muris were released on CryptoDB
in 2003 and 2011, respectively [11–13]. Composed of eight chromosomes each, these genomes are of
9.1–92 Mb in size, with 26%–30% GC content and 94%–97% nucleotide identity. Altogether, about 4000
Cryptosporidium 591
TABLE 37.1
Morphological and Biological Characteristics of Human-Infecting Cryptosporidium Species
Main
Former Oocyst Dimension Oocyst Length/ Susceptible Predilection
Designation (μm) Width Ratio Hosts Site
Cryptosporidium Bovine genotype, 5.0 × 4.5 (4.5–5.4 × 1.16 or 1.15 Cattle (especially Small intestine
parvum senso genotype 2 4.2–5.0) or 5.2 × 4.6 (1.04–1.22) preweaned
stricto (4.8–5.6 × 4.2–4.8) calves), sheep,
goats, deer, and
humans
Cryptosporidium Human genotype, 5.2 × 4.9 (4–6 × 4–5) 1.1 (1.0–1.1) Humans, dugong, Small intestine
hominis genotype 1, and lamb
genotype H
Cryptosporidium Dog genotype 4.95 × 4.71 1.05 (1.04–1.06) Dogs, coyotes, Small intestine
canis (3.68–5.88 × foxes, and
3.68–5.88) humans
Cryptosporidium 5 × 4.5 (6.0–5.0 × 1.19 Cats Small intestine
felis 5.0–4.5)
Cryptosporidium 4.6 × 4.2 (4.9–4.4 × 1.1 Pigs and humans Small and large
suis 4.0–4.3) intestine
Cryptosporidium 5.2 × 4.6 (4.5–6.0 × 1.13 (1.00–1.33) Chicken and Small intestine
meleagridis 4.2–5.3) turkey
Cryptosporidium 7.4 × 5.6 (7–8 × 1.3 (1.1–1.5) or 1.4 Rodents, other Stomach
muris 5–6.5) or 8.4 × 6.3 mammals, and
(7.5–10 × 5.5–7) humans
Cryptosporidium C. muris-like 7.4 × 5.5 (6.0–8.1 × 1.35 Cattle, bactrian Abomasum
andersoni 5.0–6.5) camels, sheep,
rodents, and
humans
protein-encoding genes are present. Cryptosporidium genomes appear to be unusual among eukaryotes
in having a degenerate “mitosome” (located in the posterior end of sporozoites) instead of a mitochon-
drion, with accompanied loss of many mitochondrial proteins, including those required for the TCA
cycle, oxidative phosphorylation, and fatty acid oxidation as well as de novo biosynthesis of amino acids,
nucleotides, and sugars. These gene losses render Cryptosporidium species heavily reliant on scavenging
nutrients from the host rather than de novo biosynthesis [12–15]. In addition, sequencing analyses of C.
hominis IbA10G2 (the most virulent subtype responsible for all outbreaks in Europe and Australia) and
IaA28R4 (a dominant outbreak subtype in the United States) uncovered major differences in the 5′ and
3′ ends of chromosome 6 and the putative virulence determinant gp60 region, suggesting that genetic
recombination plays a potential role in the emergence of hypertransmissible C. hominis subtypes [16,17].
thin, incomplete wall, rupture in the intestine, and release four infectious sporozoites, resulting in a new
cycle of infection (endogenous autoinfection). The autoinfection allows Cryptosporidium to sporulate
and persist within the same host indefinitely, accounting for the severe chronic forms of cryptosporidi-
osis in immunocompromised persons in the absence of exogenous reinfection. In respiratory cryptospo-
ridiosis, as seen in immune-deficient patients, the infective oocysts spread via nasal secretions into the
environment [1,2].
Cryptosporidium oocysts are resistant to environmental adversaries and are highly infectious; as few
as 10 C. hominis oocysts are able to produce disease in healthy adults. Although fecal–oral transmission
represents the main route of infection, transmission via coughing and fomites is also possible [1,2].
Most (90%) of the human infections are caused by C. hominis and C. parvum, with C. hominis being
found almost exclusively in humans, while C. parvum is readily transmitted between humans and ani-
mals as well as between humans. Other Cryptosporidium species (C. meleagridis, C. muris, C. felis,
C. canis, C. suis, and C. andersoni) may also be involved in human infections [1,2].
As a most common cause of severe diarrhea in humans, cryptosporidiosis poses a serious public health
risk worldwide. In industrialized nations, cryptosporidiosis is often associated with recreational water
use, animals on petting farms, and day care centers. In developing countries (e.g., sub-Saharan Africa,
Asia, and Latin America), cryptosporidiosis may result from contaminated food or drinking water, or
contaminated recreational water (as in swimming pools) and occurs mostly in children younger than
5 years (with peak occurrence in children younger than 2 years) [1,2].
The population groups with an elevated risk of being exposed to Cryptosporidium include: (1) people
who swim regularly in pools with insufficient sanitation (as some Cryptosporidium strains are chlorine-
resistant); (2) child-care workers; (3) parents of infected children; (4) people caring for others with cryp-
tosporidiosis; (5) backpackers, hikers, and campers who drink unfiltered, untreated water; (6) people,
including swimmers, who swallow water from contaminated sources; (7) people handling infected cat-
tle; and (8) people exposed to human feces [1,2].
37.1.4 Diagnosis
Traditional method for diagnosing human intestinal cryptosporidiosis is based on microscopic detection
of Cryptosporidium oocysts in stool samples with acid-fast, auramine, or indirect immunofluorescence
stains. Use of fluorescence microscopy (e.g., light-emitting diode light sources) together with fluorescent
stains (e.g., auramine-rhodamine, which makes oocysts appear yellow-orange) provides a more sensitive
detection than the modified acid-fast stain [e.g., modified Ziehl–Neelsen (hot) or Kinyoun (cold), which
demonstrates the acid resistance of oocysts]. Further, application of formalin-ether or formalin-ethyl
acetate sedimentation concentration procedures, or Sheather’s flotation-concentration method (using
a sucrose gradient) improves the sensitivity of microscopic detection. Nonetheless, Cryptosporidium
oocysts (of 4–6 µm in diameter) must be differentiated from other partially acid-fast organisms (e.g.,
Cyclospora cayetanensis) and other similar looking yeast or fungal spores [23,24].
Similarly, human respiratory cryptosporidiosis may be confirmed by microscopic detection of
Cryptosporidium organisms in respiratory secretions [e.g., sputum, tracheal aspiration, bronchoalveolar
lavage (BAL)], bronchial and lung biopsy, or autopsy specimens. Examination of respiratory secretions
using acid-fast, auramine or indirect immunofluorescence stains may show thick-walled Cryptosporidium
oocysts and invasive forms of the parasite (sporozoites and merozoites). Sporozoites and merozoites may
be also detected from BAL fluid specimens using Giemsa stain. Hematoxylin–eosin staining of tis-
sue specimens may uncover sporozoites, merozoites, and oocysts lining the mucosal epithelium at the
luminal surface of the trachea, bronchi, and bronchioles, as well as within bronchial mucous glands.
Bronchial and lung biopsy specimens may yield intracellular and extracellular cryptosporidia, including
sporozoites, merozoites, or oocysts on the bronchoepithelial surface [25].
Serological assays are important for detection of specific antibodies in both symptomatic and
asymptomatic infections. Although antibodies to Cp23 correlate with distant infection, those to Cp17
(also called gp15) suggest recent infection, and those to P2 are associated with repeated infection. In
addition, several serological assays [e.g., direct immunofluorescence assay (DFA), enzyme immune
assays (e.g., EIA, ELISA), and immunochromatographic assays] allow detection of oocyst-specific
antigens [26].
In recent years, nucleic amplification techniques [e.g., polymerase chain reaction (PCR), PCR-
restriction fragment length polymorphism (RFLP) analysis] are increasingly used for sensitive detection
and subtyping of Cryptosporidium species [27–30]. This involves disruption of oocysts by bead-beating,
freeze-thaw, boiling, or chemical lysis for nucleic acid extraction and use of oligonucleotide primers
from selective gene targets [e.g., 18S rRNA, actin, oocyst wall protein (COWP), thrombospondin-related
adhesive protein 1 (TRAP-C1), tubulin, HSP70, gp60, and gp900 (also known as polythreonine protein or
poly-T) in addition to noncoding internal transcribed spacer 1 and microsatellites] for a mplification, and
detection of amplified products on various platforms. Application of molecular procedures has enabled
the differentiation of C. andersoni from C. muris, the discrimination of C. canis from C. parvum, and
the establishment of C. hominis as a species distinct from C. parvum [1,2].
the relatively high cost of nitazoxanide limits its clinical adoption. Further, nitazoxanide is ineffective
against respiratory cryptosporidiosis and has proven unsuccessful in eradicating the parasite from the
guts of chronically infected children with HIV [31].
In individuals coinfected with HIV, antiretroviral therapy (ART) is useful for controlling chronic
diarrhea and wasting due to cryptosporidiosis. Essentially, ART helps restore immune function of
human patients, leading to parasitological clearance after treatment for several months. Protease
inhibitors, including indinavir, have also been shown to directly interfere with Cryptosporidium
development. Alternatively, paromomycin may be considered for AIDS patients with respiratory cryp-
tosporidiosis [31].
Prevention of human cryptosporidiosis should aim to interrupt fecal–oral transmission of
Cryptosporidium oocysts through provision of clean water and sanitation, maintenance of scrupulous
hygiene in communal settings such as day care centers, implementation of food safety practice, and
development of effective vaccines [32].
Use of filtration technologies (e.g., slow sand filters, diatomaceous earth filter, membranes, bag- and
cartridge-filter products) is useful for the removal of Cryptosporidium in water supply. Ultrafiltration and
UV irradiation are also efficient. However, high-rate filtration and chlorine disinfection are largely inef-
fective in the reduction of Cryptosporidium oocysts from water supply. For wastewater treatment, stabili-
zation ponds and constructed wetlands are valuable for the elimination of Cryptosporidium oocysts [33].
37.2 Laboratory Models
Given the ability of Cryptosporidium to perpetuate in human hosts, use of animal models and in vitro
cultivation system is vital for characterization of Cryptosporidium life cycle, elucidation of host immune
mechanisms, evaluation of potential drugs, and development of vaccines [34–36].
37.2.1 Animal Models
A number of laboratory animals are susceptible to Cryptosporidium infection, although numerous
parameters (e.g., isolate employed, dosage, oocyst age, oocyst storage conditions, chemical pretreat-
ments, and host genetics) may have a bearing on the outcome.
Out of the 19 different strains of adult mice examined, C. parvum has been shown to produce the
highest levels of infection in the beige mouse (C57BL/6J-bgJ). In addition, similar acute patterns of
C. parvum infection in C57BL/6 wild-type and T- and B cell-deficient Rag2–/– newborn mice were also
observed. Using dexamethasone-treated or untreated adult severe combined immunodeficiency (SCID)
mice, it was shown that C. parvum strains (of both animal and human origins) are capable of inducing
intraepithelial neoplasia and invasive adenocarcinoma in the stomach, ileocecal region, and intrahepatic
biliary tree [37]. Further, use of the interleukin-12 (IL-12) knockout mouse model, which mimics acute
human cryptosporidiosis, facilitated the identification of a compound (P131) with significant antipara-
sitic activity [36]. Use of various animal models has helped to confirm that C. hominis at high doses may
also infect calves, lambs, and piglets.
C. muris oocysts obtained from the gastric glands of wild rat stomach have been successfully transmit-
ted to uninfected rats, mice, guinea pigs, rabbits, dogs, and cats. Indeed, C. muris seems to be infective to
hamsters, squirrels, Siberian chipmunks, wood mice (Apodemus sylvaticus), bank voles (Clethrionomys
glareolus), Dolichotis patagonum, rock hyrax, bactrian camels, mountain goats, cynomolgus monkeys,
and humans. Interestingly, C. muris demonstrates notable differences in prepatent and patent periods
in two laboratory rodents—BALB/c mice and the southern multimammate rat (Mastomys coucha).
Specifically, C. muris infection progressed more rapidly in BALB/c mice (with a prepatent period of
7.5–10 days) than M. coucha (with prepatent period of 18–21 days) [38].
In contrast to C. muris, C. andersoni (previously C. muris-like) is not infective to outbred, inbred, neo-
natal, or immunocompetent mice (including common field mice, BALB/c mice, and SCID mice) as well
as common voles, bank voles, desert gerbils, guinea pigs, rats, rabbits, or goats. However, C. andersoni
has the capacity to infect Mongolian gerbils.
Cryptosporidium 595
37.3 Conclusion
Of nearly 30 recognized species within the genus Cryptosporidium, C. parvum and C. hominis are
important waterborne and foodborne pathogens of humans. These parasites cause villus atrophy, crypt
hyperplasia, infiltration of the lamina propria, chloride hypersecretion, glucose malabsorption, and
reduced barrier function, leading to diarrhea, wasting, stunt growth, and explained cough. Considering
its high infectivity (as few as 10 oocysts are sufficient to establish infection in immunocompetent per-
sons) and strong resistance to chlorine disinfection, large-scale outbreaks of human cryptosporidiosis
attributable to contaminated drinking or recreational water have been reported. In order to develop
improved control measures, it is essential to uncover the molecular mechanisms of Cryptosporidium
infection. Application of laboratory models will be fundamental to achieving this goal.
REFERENCES
1. Xiao L, Fayer R, Ryan U, Upton SJ. Cryptosporidium taxonomy: recent advances and implications for
public health. Clin Microbiol Rev. 2004;17:72–97.
2. Xiao L, Feng Y. Zoonotic cryptosporidiosis. FEMS Immunol Med Microbiol. 2008;52(3):309–23.
3. Li X, et al. Cryptosporidium rubeyi n. sp. (Apicomplexa: Cryptosporidiidae) in multiple Spermophilus
ground squirrel species. Int J Parasitol Parasites Wildl. 2015;4(3):343–50.
4. Ryan U, et al. Cryptosporidium huwi n. sp. (Apicomplexa: Eimeriidae) from the guppy (Poecilia reticu-
lata). Exp Parasitol. 2015;150:31–5.
5. Ryan U, Fayer R, Xiao L. Cryptosporidium species in humans and animals: current understanding and
research needs. Parasitology. 2014;141(13):1667–85.
6. Ryan U, Hijjawi N. New developments in Cryptosporidium research. Int J Parasitol. 2015;45(6):367–73.
7. Nakamura AA, Meireles MV. Cryptosporidium infections in birds—a review. Rev Bras Parasitol Vet.
2015;24(3):253–67.
8. Jiang Y, et al. Cryptosporidium andersoni as a novel predominant Cryptosporidium species in outpa-
tients with diarrhea in Jiangsu Province, China. BMC Infect Dis. 2014;14:555.
9. Vinayak S, et al. Genetic modification of the diarrhoeal pathogen Cryptosporidium parvum. Nature.
2015;523(7561):477–80.
10. Kváč M, et al. Cryptosporidium proliferans n. sp. (Apicomplexa: Cryptosporidiidae): molecular
and biological evidence of cryptic species within gastric Cryptosporidium of mammals. PLoS One.
2016;11(1):e0147090.
11. Puiu D, Enomoto S, Buck GA, Abrahamsen MS, Kissinger JC. CryptoDB: the Cryptosporidium genome
resource. Nucleic Acids Res. 2004;32:D329–31.
596 Laboratory Models for Foodborne Infections
12. Widmer G, Sullivan S. Genomics and population biology of Cryptosporidium species. Parasite
Immunol. 2012;34(2–3):61–71.
13. Widmer G, Lee Y, Hunt P, Martinelli A, Tolkoff M, Bodi K. Comparative genome analysis of two
Cryptosporidium parvum isolates with different host range. Infect Genet Evol. 2012;12(6):1213–21.
14. Abrahamsen MS, et al. Complete genome sequence of the apicomplexan Cryptosporidium parvum.
Science. 2004;304:441–5.
15. Xu P, et al. The genome of Cryptosporidium hominis. Nature. 2004;431:1107–12.
16. Guo Y, et al. Comparative genomic analysis reveals occurrence of genetic recombination in virulent
Cryptosporidium hominis subtypes and telomeric gene duplications in Cryptosporidium parvum. BMC
Genomics. 2015;16:320.
17. Hadfield SJ, et al. Generation of whole genome sequences of new Cryptosporidium hominis and
Cryptosporidium parvum isolates directly from stool samples. BMC Genomics. 2015;16:650.
18. Sponseller JK, Griffiths JK, Tzipori S. The evolution of respiratory cryptosporidiosis: evidence for
transmission by inhalation. Clin Microbiol Rev. 2014;27(3):575–86.
19. Kothavade RJ. Challenges in understanding the immunopathogenesis of Cryptosporidium infections in
humans. Eur J Clin Microbiol Infect Dis. 2011;30(12):1461–72.
20. Zhou R, Feng Y, Chen XM. Non-coding RNAs in epithelial immunity to Cryptosporidium infection.
Parasitology. 2014;141(10):1233–43.
21. Petry F, Jakobi V, Tessema TS. Host immune response to Cryptosporidium parvum infection. Exp
Parasitol. 2010;126(3):304–9.
22. Halliez MC, Buret AG. Gastrointestinal parasites and the neural control of gut functions. Front Cell
Neurosci. 2015;9:452.
23. Chalmers RM, Katzer F. Looking for Cryptosporidium: the application of advances in detection and
diagnosis. Trends Parasitol. 2013;29(5):237–51.
24. Laude A, et al. Is real-time PCR-based diagnosis similar in performance to routine parasitological
examination for the identification of Giardia intestinalis, Cryptosporidium parvum/Cryptosporidium
hominis and Entamoeba histolytica from stool samples? Evaluation of a new commercial multiplex PCR
assay and literature review. Clin Microbiol Infect. 2016;22(2):190.e1–8.
25. Checkley W, et al. A review of the global burden, novel diagnostics, therapeutics, and vaccine targets for
Cryptosporidium. Lancet Infect Dis. 2015;15(1):85–94.
26. Kothavade RJ. Potential molecular tools for assessing the public health risk associated with waterborne
Cryptosporidium oocysts. J Med Microbiol. 2012;61(8):1039–51.
27. Bankier AT, et al. Integrated mapping, chromosomal sequencing and sequence analysis of
Cryptosporidium parvum. Genome Res. 2003;13:1787–99.
28. Skotarczak B. Progress in the molecular methods for the detection and genetic characterization of
Cryptosporidium in water samples. Ann Agric Environ Med. 2010;17(1):1–8.
29. Robinson G, Chalmers RM. Assessment of polymorphic genetic markers for multi-locus typing of
Cryptosporidium parvum and Cryptosporidium hominis. Exp Parasitol. 2012 132(2):200–15.
30. Lendner M, Daugschies A. Cryptosporidium infections: molecular advances. Parasitology.
2014;141(11):1511–32.
31. Gargala G. Drug treatment and novel drug target against Cryptosporidium. Parasite. 2008;15(3):275–81.
32. Mead JR. Prospects for immunotherapy and vaccines against Cryptosporidium. Hum Vaccines
Immunother. 2014;10(6):1505–13.
33. Nasser AM. Removal of Cryptosporidium by wastewater treatment processes: a review. J Water Health.
2016;14(1):1–13.
34. Qin HY, Wu JC, Tong XD, Sung JJ, Xu HX, Bian ZX. Systematic review of animal models of post-
infectious/post-inflammatory irritable bowel syndrome. J Gastroenterol. 2011;46(2):164–74.
35. Costa LB, et al. Novel in vitro and in vivo models and potential new therapeutics to break the vicious
cycle of Cryptosporidium infection and malnutrition. J Infect Dis. 2012;205(9):1464–71.
36. Gorla SK, et al. Validation of IMP dehydrogenase inhibitors in a mouse model of cryptosporidiosis.
Antimicrob Agents Chemother. 2014;58(3):1603–14.
37. Benamrouz S, et al. Cryptosporidium parvum-induced ileo-caecal adenocarcinoma and Wnt signaling
in a mouse model. Dis Model Mech. 2014;7(6):693–700.
38. Melicherová J, Ilgová J, Kváč M, Sak B, Koudela B, Valigurová A. Life cycle of Cryptosporidium muris
in two rodents with different responses to parasitization. Parasitology. 2014;141(2):287–303.
Cryptosporidium 597
39. Borowski H, Thompson RC, Armstrong T, Clode PL. Morphological characterization of Cryptosporidium
parvum life-cycle stages in an in vitro model system. Parasitology. 2010;137(1):13–26.
40. Hijjawi N, Estcourt A, Yang R, Monis P, Ryan U. Complete development and multiplication of
Cryptosporidium hominis in cell-free culture. Vet Parasitol. 2010;169(1–2):29–36.
41. Castellanos-Gonzalez A, Cabada MM, Nichols J, Gomez G, White AC Jr. Human primary intestinal
epithelial cells as an improved in vitro model for Cryptosporidium parvum infection. Infect Immun.
2013;81(6):1996–2001.
42. Varughese EA, Bennett-Stamper CL, Wymer LJ, Yadav JS. A new in vitro model using small intestinal
epithelial cells to enhance infection of Cryptosporidium parvum. J Microbiol Methods. 2014;106:47–54.
43. Morada M, et al. Continuous culture of Cryptosporidium parvum using hollow fiber technology. Int
J Parasitol. 2016;46(1):21–9.
44. Huang L, et al. An in vitro model of infection of chicken embryos by Cryptosporidium baileyi. Exp
Parasitol. 2014;147:41–7.
45. Zhang S, et al. Chick embryo tracheal organ: a new and effective in vitro culture model for
Cryptosporidium baileyi. Vet Parasitol. 2012;188(3–4):376–81.
38
Cystoisospora belli
CONTENTS
38.1 Introduction................................................................................................................................... 599
38.1.1 Classification and Phylogeny............................................................................................ 600
38.1.2 Biology, Morphology, and Life Cycle.............................................................................. 600
38.1.3 Epidemiology.................................................................................................................... 603
38.1.4 Pathology and Clinical Aspects....................................................................................... 604
38.1.5 Laboratory Diagnosis....................................................................................................... 605
38.1.5.1 Stool Examination............................................................................................ 605
38.1.5.2 Examination of Duodenal Content................................................................... 606
38.1.5.3 Histopathological Diagnosis............................................................................. 606
38.1.5.4 Molecular Diagnosis......................................................................................... 607
38.1.6 Treatment and Prevention................................................................................................. 607
38.2 Laboratory Models........................................................................................................................ 608
38.2.1 Animal Models................................................................................................................. 608
38.2.2 In Vitro Growth in Mammalian Cells.............................................................................. 608
38.2.2.1 Preparation of Infective Oocysts...................................................................... 608
38.2.2.2 Inoculation of Sporozoites into Cell Lines........................................................610
38.2.2.3 Inoculation of Sporozoites into Human and Murine Macrophages..................610
38.3 Conclusion and Future Perspectives..............................................................................................611
References................................................................................................................................................612
38.1 Introduction
Cystoisospora belli, formerly known as Isospora belli, is an important enteric coccidian protozoa that
is responsible for diarrheal illness in humans. Belonging to phylum Apicomplexa, coccidians parasit-
ize diverse species of hosts ranging from higher invertebrates to vertebrates. First observed as globu-
lar and ovoidal structures in the bile of a cow by Leeuwenhoek in 1674 [1], coccidian protozoa of the
apicomplexan parasites are known to cause diseases in humans. These include the genera Plasmodium
and Babesia infecting mainly erythrocytes, Toxoplasma and Sarcocystis inhabiting various tissues, and
enteric coccidia in the genera Cystoisospora, Cyclospora, and Cryptosporidium, mainly involving intes-
tinal epithelial cells. All enteric coccidian protozoa produce gastrointestinal symptoms indistinguishable
from one another; therefore, laboratory investigation is crucial to make definite diagnosis that is man-
datory for a proper management. Although a number of species in the genus Cystoisospora are known
to infect mammalian hosts, C. belli is the only known pathogenic species to humans, causing cystoiso-
sporiasis. The description of characteristic oocysts of C. belli in stools was initially observed among
military personnel stationed in Egypt, the Middle East, and eastern Mediterranean countries during
1914 and 1921 [2]. Nowadays, it is well recognized that cystoisosporiasis has a cosmopolitan distribu-
tion. Although oocysts of Isospora natalensis were reportedly found in human fecal samples, several
studies have failed to reaffirm the presence of these oocysts as a causative agent of diarrheal illness in
599
600 Laboratory Models for Foodborne Infections
humans since its first report in 1953, raising the possibility that it may not be a valid pathogenic species in
humans [3]. Most of the enteric coccidian parasites of humans are considered to be opportunistic patho-
gens except the genus Sarcocystis. Thus, the prevalence of these coccidian infections seems to increase
as the number of immunocompromised patients increases, especially after AIDS became pandemic. In
general, severity of illness caused by C. belli seems to be more aggressive in patients with compromised
immunity than those with normal immune status. However, chronic emaciating infections have been
reported in immunocompetent patients, suggesting that host immunity per se may not determine the
severity of cystoisosporiasis [4,5]. Meanwhile, difference in intrinsic virulence of this organism has not
yet been explored.
Eimeriidae Sarcocystidae
Cystoisospora felis L76471
Neospora caninum U03069
Hammondia hammondi AF096498
Toxoplasma gondii M97703
Sarcocystis neurona U07812
Caryospora bigenetica AF060975
Isospora robini AF080612
Isospora gryphoni AF080613
Cyclospora cayetanensis U40261
Eimeria tenella U67121
0.02
FIGURE 38.1 Phylogeny of Cystoisospora and other apicomplexan protozoa inferred by using the Maximum Likelihood
method based on the small subunit ribosomal RNA sequences. Tree was constructed by using the MEGA6 program [15].
Scale indicates the number of base substitutions per site. GenBank accession numbers are listed after species.
? ?
Monozoic tissue cyst
Sporogony in
environment Endodyogeny/
schizogony
Microgametes
Immature oocyst
with sporoblasts
Fertilization
FIGURE 38.2 Life cycle of Cystoisospora belli depicting asexual multiplication, sexual development, and sporogony.
(Illustration by Urassaya Pattanawong.)
stage of C. belli is characterized by an oval or rugby-shaped oocyst with a wide range of size variation
in terms of the maximum length and width, both within and between isolates. The average length mea-
sured from more than 700 oocysts is 28.3 μm (range = 17–37). A remarkable narrowing with a neck-like
appearance may be observed at one end of some oocysts, whereas most oocysts are nearly symmetric
at both ends. The maximum width of oocysts averages 13.5 μm (range = 8–21). The mean shape index
or the ratio of length to width of oocysts is 2.1 (range = 1.3–3.3) [9]. The oocyst wall is bilayer, thin,
602 Laboratory Models for Foodborne Infections
smooth, and transparent for all stages of development. The inner wall is membranous, and the outer
wall is rigid and relatively impermeable to external fluids [16]. When freshly passed in feces, the oocyst
remains immature, containing a spherical or slightly elongated, oval-shaped sporoblast that develops
within another rigid cyst wall called sporocyst. Occasionally, some freshly passed oocysts in stool may
contain two sporocysts, each of which possesses one sporoblast. The sporocyst of C. belli lacks a pro-
teinaceous plug, known as a Stieda body, a structure existing in the genus Isospora. The dimension of
sporocyst is 12–14 μm × 7–9 μm.
Sporogony, the process of producing infective sporozoites of Cystoisospora, usually occurs outside the
host (Figure 38.2). The maturation period of oocysts usually completes within 24 h to 10 days [9]. The
mature oocyst possesses two sporocysts, each with four crescentic or banana-shaped sporozoites cluster-
ing together with a clump of granular mass or residual bodies surrounded by sporocyst wall (Figure 38.3).
Detailed investigation of ex vivo development has shown that the percentage of oocyst maturation varies
between isolates. In vitro studies reveal that only 27%–30% of oocysts excreted from stools of patients
become fully sporulated at ambient temperature. However, sporulation process requires optimum condi-
tions that could be variable among studies. Importantly, despite the frequently identified characteristic
mature oocysts containing two sporocysts, about 5% of the oocysts that undergo the maturation pro-
cess possess eight sporozoites in a single sporocyst akin to oocysts of Caryospora, a member of fam-
ily Eimeriidae [9,17]. The formation of Caryospora-like oocysts requires 5–14 days after excretion in
feces [9]. Both types of mature oocysts contain a number of granular residual bodies clumping together.
Environmental factors such as moisture, temperature, and oxygen composition in the atmosphere have
been suggested to influence the maturation of Cystoisospora oocysts. It is noteworthy that incubation
of C. rivolta oocysts at 50°C for 5 min enhances development of Caryospora-like oocysts, and they are
infective to mice and cats [18]. It is therefore likely that Caryospora-like oocysts of C. belli could also
be infective to humans.
Sporozoites are released from the oocysts and become free in the lumen of small intestine, espe-
cially around distal duodenum and jejunum. Thereafter, sporozoites invade small intestinal epithelial
cells where asexual multiplication ensues. All endogenous stages of Cystoisospora reside within para-
sitophorous vacuoles in the cytoplasm of host cells. Sporozoites of mammalian Cystoisospora species,
for example, C. suis, C. rivolta, and C. felis, initially undergo asexual multiplication by endodyogeny,
a cellular division process in which two daughter cells are generated from a mother cell so that these
daughter cells are surrounded by the mother cell membrane [19–21]. Repeated endodyogeny probably
occurs and generates at least three structurally distinct types of merozoites in C. suis, whereas schizo-
gonic development with repeated nuclear division within a single cytoplasmic mass possibly occurs in
epithelium of upper small intestine [22,23]. However, the number of generations of merozoites remains
unknown. Eventually, some merozoites develop into sexual stages consisting of female and male game-
tocytes. Although detailed knowledge on asexual development of C. belli remains to be elucidated,
it could resemble these porcine and feline species of Cystoisospora. In vitro studies have shown that
endodyogenic development of C. belli occurs after inoculation of sporozoites into cell cultures resulting
in the formation of merozoites, while histopathological studies demonstrated multinucleate schizonts
in infected enterocytes [11–13,24–27]. Fully developed merozoites disintegrate host cells, invade other
epithelial cells, and undergo further asexual multiplication cycles (Figure 38.4).
FIGURE 38.3 Oocysts of Cystoisospora belli. (A) Oocyst from freshly passed stool containing one sporocyst. (B) and
(C) Developing oocysts with two sporoblasts. (D) Mature oocyst with two sporocysts, each with four sporozoites.
(E) Caryospora-like oocyst with eight sporozoites in one sporocyst. Scale = 10 μm.
Cystoisospora belli 603
FIGURE 38.4 Upper intestinal biopsy showing (A) shortening of villi, (B) and (C) endogenous stages of Isospora belli in
epithelium cells. Single and broken arrows indicate merozoites/schizonts and oocysts, respectively.
Extraintestinal monozoic tissue cysts of C. belli have been found in sections of lymph nodes, spleen,
and liver at autopsy of AIDS patients, suggesting that these stages may represent hypnozoites or dormant
forms that could be responsible for relapse of symptoms [11,12,28–30]. Ultrastructural studies reveal
that the extraintestinal monozoic tissue cyst is surrounded by a parasitophorous vacuole membrane and
characterized by oval or ellipsoidal body measuring about 11 μm × 8 μm. A conspicuous cyst wall is
observed with a thickness varying from 0.7 to 4 μm, enclosing a crescent-shaped zoite. Occasionally,
the surface of monozoic tissue cyst possesses grooves or small projections. The zoite inside monozoic
tissue cyst resembles the sporozoite stage and contains a single rather than multiple nuclei. The anterior
part of these zoites possesses polysaccharide-like and lipid-like granules, and apical organelles consist-
ing of rhoptries, micronemes, dense bodies, and a conoid. Each zoite is surrounded by a parasitopho-
rous vacuole and granular materials forming the cyst wall. Monozoic tissue cysts enlarge over time
without cellular division. The monozoic tissue cysts of C. canis in mouse, a paratenic host, are infective
to dogs similar to ingestion of oocysts [31]. Although the origin of this developmental process remains
elusive, abundant numbers of tissue cysts found at autopsy favor that they probably originated from
merozoites rather than sporozoites from the oocyst stage [28–30]. It is noteworthy that extraintestinal
stages and monozoic tissue cysts have also been found in C. felis after oral inoculation of the oocysts
into kittens [10,32,33].
Sexual reproduction or gametogony usually occurs after initiation of the asexual cycle for about
1 week. Female gamont develops intracellularly without nuclear division and becomes macrogamete,
whereas male gamont undergoes multiple nuclear and cytoplasmic divisions, resulting in the production
of a number of microgametes. Microgametes disrupt host cells and enter into other epithelial cells where
macrogametes are located. After fertilization, zygote is formed within the epithelial cell and sequentially
develops into an oocyst. Intraepithelial oocysts containing granular bodies are excreted in feces upon
rupture of host cell. The oocysts are resistant to the hostile environment, including common disinfec-
tants, and remain viable in cool and moist condition for months [14].
38.1.3 Epidemiology
Transmission of C. belli is solely anthroponotic because humans are the only known natural hosts.
Before 1935, the prevalence of human cystoisosporiasis seemed to be have been low as only 200 cases
were reported [34]. However, in certain circumstances when living condition is overcrowded or sani-
tation is below the standard level, cases of C. belli infections could be encountered. The majority of
infected cases were identified mainly among military or related personnel during 1914–1921 [2]. Until
1961, after awareness of the infection along with the competency of microscopic detection, more than
800 cases were detected in various localities in South America [34]. Before the pandemic of HIV infec-
tions, sporadic or endemic cystoisosporiasis has been reported from several countries both in tropical
and temperate zones. The infection rates of C. belli among immunocompetent individuals are usually
604 Laboratory Models for Foodborne Infections
<2%, while a remarkable higher rate was observed among those with underlying immunodeficiency.
The prevalence of cystoisosporiasis in AIDS patients in Zaire, Haiti, Venezuela, Brazil, and Thailand
is 19%, 15%, 14%, 9.9%, and 7.6%, respectively [9,35–38]. Nevertheless, endemicity, public sanitation,
preexisting anticoccidial drug treatment, and other factors may also affect such prevalence. An 8-year
surveillance of intestinal infections among AIDS patients in Los Angeles County revealed the presence
of C. belli in 1% of pathogen-positive stool samples [39]. Interestingly, most of the C. belli-positive
samples were from foreign-born patients, especially those from Central America. Travel-related infec-
tions may be a contributing factor.
TABLE 38.1
Comparative Characteristics of Cystoisospora belli, Cyclospora cayetanensis, Cryptosporidium spp., and
Sarcocystis hominis
Feature C. belli C. cayetanensis Cryptosporidium spp. S. hominis
First report in humans 1914 1977–1978 1974 1925
Location in enterocytes of Intracytoplasmic in Intracytoplasmic in Intracellular, Lamina propria
small intestine apical supranuclear apical extracytoplasmic at
region supranuclear luminal surface
region
Zoonotic potential No Unknown Yes Yes
Complete life cycle within Yes Yes Yes No
humans
Average size of oocyst 13.5 × 28.3 μm 8–10 μm 4.5 to >6 μm 13.5 × 19.5 μm
(sporocyst: 9.3 ×
14.7 μm)
Acid-fast staining of Acid-fast Variably acid-fact Acid-fast Not acid-fast (due
oocysts to thick
sporocyst wall)
Number of sporocysts in 2 (1 for Caryospora-like 2 No sporocyst 2
mature oocyst oocyst)
Number of sporozoites per 8 4 4 8
sporulated oocyst
Sporulation outside host Yes Yes No No
Autofluorescence of Yes Yes No Unknown. Oocyst
oocyst wall usually disrupted
when passed in
stool
Number of oocysts in Low Low to moderate Low to very high Low
stools
Infectivity of oocysts in No No Yes Yes
freshly passed stool
Resistance of oocysts to Yes Yes Yes Unknown
chlorine
Source: Herwaldt, B.L., Clin. Infect. Dis., 31, 1040, 2000.
Laboratory test frequently shows eosinophilia in the majority of infected cases, but a remarkable
a lteration of other hematological profiles is not observed. Patients with normal immune status tend
to have a higher level of eosinophils than those with suppressed immune system [9]. Charcot–Leyden
crystals are frequently observed in stools of cystoisosporiasis patients, indicating active proliferation and
degradation of eosinophils in gut tissues. Because endogenous stages of C. belli neither cause apparent
ulceration of intestinal epithelial cells nor induce intense inflammatory response, a remarkable number
of erythrocyte or leucocyte is not found in stool. Identification of characteristic oocysts in stool allows
definite diagnosis in routine laboratory practice. The efficiency of detection depends on various factors
such as the microscopist’s experience, shedding pattern of oocysts that may be fluctuating during the
course of infection, as well as the choice of diagnostic techniques. Some differential features of coccid-
ian protozoa that shed their oocysts in host stool samples are listed in Table 38.1 [47].
38.1.5 Laboratory Diagnosis
38.1.5.1 Stool Examination
Definite diagnosis of cystoisosporiasis relies on identification of characteristic oocysts in stool or intesti-
nal content. Collection of stool samples containing or suspicious of having oocysts of C. belli essentially
follows standard guidelines for parasitological diagnosis. Although immediate examination of stool
606 Laboratory Models for Foodborne Infections
specimen is not a crucial issue as those for amoebic trophozoites, it is recommended to examine the
stool samples within 24 h after passage. If examination of stool sample is not possible within 1 day, stool
should be kept at less than 10°C, but not frozen, in order to minimize bacterial overgrowth and decay
of fecal materials [48]. Although coccidian oocysts can be preserved in 5% or 10% formalin solution
for microscopy-based diagnostic purpose, preservation of coccidian oocysts in ethanol also gives good
results for subsequent acid-fast stain. Structures of C. belli and Cryptosporidium oocysts kept in 80%
ethanol at ambient temperature for more than 2 years are also well preserved for acid-fast stains [9,49].
Microscopic examination of stool samples should be done by both direct smear and concentration
methods. For fresh stool samples containing mucus, direct smear sampling from the mucus portion
may yield better discovery than the watery part of stool as coccidian oocysts tend to stick to mucoid
substance. Meanwhile, C. belli oocysts usually do not excrete in large amount; therefore, they are
often diluted in voluminous diarrhea precluding efficient diagnosis by direct smear method. Oocysts of
C. belli can be concentrated without morphological alteration by formalin-ethylacetate (or ether) sedi-
mentation procedure to increase diagnostic yield [50]. Despite superiority of formalin-ethylacetate sedi-
mentation to simple smear method, simple sedimentation by centrifugation of previously sieved stool
samples may yield higher diagnostic probability than conventional formalin-ethylacetate sedimentation,
because some oocysts are lost to the fatty layer in the latter method [51]. It is noteworthy that oocysts of
C. belli can be overlooked in unstained stool samples because of their transparency when adjustment of
microscope does not make good contrast. Alternatively, various acid-fast staining methods such as modi-
fied Kinyoun acid-fast and modified Ziehl–Neelsen stains that intentionally deployed for detection of
Cryptosporidium spp. and Cyclospora oocysts are also useful for diagnosing C. belli oocysts. Specimens
can be versatile ranging from fresh stool, sediment from formalin-ethylacetate sedimentation, ethanol-
preserved stool sample, duodenal fluid, and other body fluids [52].
Oocysts of C. belli, Cyclospora cayetanensis, and Cryptosporidium spp. become autofluorescent
bright green under violet excitation (405 nm), bluish violet autofluorescent under ultraviolet excitation
(365 nm), and green autofluorescent under blue-violet light excitation (436 nm) [53]. Faint reddish fluo-
rescence is also observed under green light (546 nm). Characteristic size, shape, and structure of C. belli
oocysts under autofluorescence microscopy differ from those of C. cayetanensis and Cryptosporidium
spp., and thus, the method is useful for diagnosis. Autofluorescence is proven to be more sensitive than
bright-field microscopy in detecting Cystoisospora oocysts [54,55].
38.1.5.3 Histopathological Diagnosis
Biopsy material from upper small intestine during endoscopic examination provides a useful diagnostic
sample for C. belli as well as other pathogens that parasitize these regions of the gastrointestinal tract
Cystoisospora belli 607
such as G. intestinalis, Cryptosporidium spp., and C. cayetanensis. Biopsy material should never be
allowed to dry but should be fixed with 10% formalin solution, embedded in paraffin or equivalent mate-
rials, and stained with hematoxylin-eosin. Biopsy materials are also good sample sources for molecular
detection provided that they should be kept frozen at less than –20°C without preservatives. The endog-
enous stages of C. belli consisting of asexual and sexual development can be seen in the cytoplasmic
mass of enterocytes or at the lamina propria of upper small intestine, although involvement of biliary
ducts and large intestine has been reported [11,12,28]. Duodenal mucosal architecture of patients with
chronic cystoisosporiasis usually presents with blunting of villi and hypertrophied crypts. Inflammatory
responses in upper intestinal tissues of both cystoisosporiasis and cyclosporiasis are similar, showing
mixed infiltration of polymorphonuclear cells, plasma cells, lymphocytes, and eosinophils, although
the eosinophilic infiltration seems to be more prominent in cystoisosporiasis [40,57]. The presence of
characteristic immature oocysts of C. belli in the intestinal epithelium cells assists in definite diagnosis.
Meanwhile, all endogenous stages of Cryptosporidium spp. are located at the intracellular rim at the
luminal surface of mucosal epithelium, and the diameters of all stages do not exceed 6 μm, making them
easily differentiable from C. belli [58,59]. Extraintestinal stages of C. belli, existing predominantly in
mesenteric lymph nodes, liver, and spleen of some AIDS patients, are mostly crescent-shaped, centrally
located, and contain only one zoite in the tissue cyst. The presence of longitudinal grooves or projections
of the zoite surface may represent simultaneous invasion of two zoites in the same host cell [60]. Neither
multinucleate nor sexual stages exist in the tissue cyst of cystoisosporiasis [30]. Although stool exami-
nation often gives more sensitive results than histopathology, the sampling areas of biopsy are limited
and may not include the affected regions. However, a large study involving 118 adult AIDS patients who
suffered from chronic diarrhea identified C. belli from duodenal biopsy samples in two patients who had
negative results in stool examinations [28].
38.1.5.4 Molecular Diagnosis
Amplification of a specific region in the small subunit ribosomal RNA gene (18S rDNA) of C. belli
by nested polymerase chain reaction (PCR) has been successfully applied to duodenal biopsy and bile
and stool samples without cross-reactivity to other enteric protozoa [8,61]. PCR targeting the 5.8S
ribosomal RNA gene and the internal transcribed spacer II (ITS2) region is proven to be more sensi-
tive than microscopy in diagnosing C. belli [62]. Likewise, quantitative PCR assay with melting curve
analysis using primers derived from the 18S rDNA can differentiate human enteric coccidian proto-
zoa [63]. Diagnosis of C. belli infection by PCR involves the isolation of C. belli DNA from clinical
specimens that can be stool, duodenal fluid, tissue sample, or other potential sources of infections. The
source of C. belli DNA is usually from oocyst stage in stool, while tissue sample contains any stages
of endogenous development. Freshly collected stool specimen, frozen stool sample, or stool preserved
in 80% ethanol can be used for extraction of C. belli DNA. Ethanol in preserved stool samples should
be removed before disruption of oocysts. Duodenal fluid or tissue samples can be kept at −20°C, and
ethanol should not be used for preservation of these samples. Because the oocyst wall of coccidian
protozoa is solid and resistant to diverse environmental conditions, it is recommended that the oocyst
wall should be ruptured so that internal contents, either sporoblasts or sporozoites, are exposed to DNA
extraction solution. Examples of simple procedures to disrupt oocyst wall of C. belli are mechanical
disruption and freeze-thaw method.
38.2 Laboratory Models
38.2.1 Animal Models
A number of animals have been tested for the susceptibility of infection with orally administered
oocysts of C. belli. These include kittens, puppies, guinea pigs, mice, rats, rabbits, and rhesus macaques
[14,68,69]. However, no consistent evidence of infection has been demonstrated in these studies.
1. Removal of 2.5% potassium dichromate from sporulated oocyst suspension is done by adding
more than one volume of phosphate buffer saline solution (PBS, pH 7.2) containing 2% Tween
20 and is centrifuged at 1500 × g for 10 min at 4°C.
2. After discarding supernatant, PBS-Tween 20 solution and diethylether (2:1 v/v) are added to the
pellet for removal of lipid portion of the sample, and centrifugation is repeated under the same
condition. The supernatant is discarded.
Cystoisospora belli 609
Alternatively, the following similar protocol has been described by Siripanth et al. for purification of
infective oocysts of C. belli [25].
1. Fecal samples containing oocysts of C. belli that have been sporulated in 2.5% potassium
dichromate solution are filtered through a few layers of gauze or cheesecloth. Subsequently,
oocysts are purified from the remaining fecal materials by sugar flotation. Preparation of sugar
solution is done by dissolving 454 g of table sugar in 355 mL water using gentle or indirect
heat. To keep the viability of oocysts, the originally recommended preservatives such as form-
aldehyde and phenol are not added in the sugar solution. The specific gravity of sugar solution
should be about 1.27. After adding filtered stool sample to a 15 mL centrifuge tube, the tube
is filled with sugar solution until the level reaches below the top of the tube. A lid or cap is
attached if the tube has screw cap, and it is then centrifuged at 280 × g for 5 min. A centrifuge
that has swinging cup should be used. Slowly sugar solution is added by carefully inserting tip
of pipette below the surface to create a slightly inverted meniscus. Oocysts at the surface of the
solution can be subsequently harvested [71].
2. After obtaining the oocysts from the flotation procedure, the sample is washed thrice by adding
distilled water and is centrifuged at 1000 × g for 10 min.
3. 2% sodium hypochlorite solution is added to the sediment containing oocysts and is left on ice
for 10 min.
4. The sample with PBS is washed by centrifugation at 1000 × g for 10 min. This step is repeated
twice.
610 Laboratory Models for Foodborne Infections
5. The sediment is resuspended in Eagle’s MEM medium containing 80 μg/mL gentamicin and
kept at 4°C until use.
6. A Teflon-coated tissue grinder is used to break oocyst wall for releasing sporocysts.
7. The sporocysts are resuspended in sterile 3% sodium taurocholate solution containing 1% tryp-
sin in MEM medium. It is then incubated at 37°C in a CO2 incubator for 2 h.
8. Sporozoites are harvested by centrifugation at 1000 × g for 10 min and washed twice with PBS.
9. The number of oocysts is estimated using Neubauer chamber.
10. Sporozoite viability is checked by using trypan blue exclusion test.
1. The frozen cells are thawed by placing the vial immediately in a water bath preset at 37°C.
2. Before opening, the vial is cleaned thoroughly with 70% ethanol.
3. The content is transferred from the vial to a 15 mL tube containing appropriate culture medium.
4. It is centrifuged at 200 × g for 5 min and supernatant is discarded.
5. The remaining pellets containing cells are transferred to a new culture flask preadded with
appropriate cell culture medium. It is mixed by gently pipetting.
6. It is incubated at 37°C. The culture flask is examined using inverted microscope.
4. After centrifugation, four layers appear consisting of plasma at the top, followed by a thin layer
of peripheral blood mononuclear cells (PBMCs), Ficoll-Hypaque layer, and erythrocytes mixed
with granulocytes at the bottom. PBMCs are collected either by gently removing the upper
plasma layer until the PBMCs can be harvested or by inserting the pipette tip directly through
the upper plasma layer to collect cells at the interface. The PBMCs are kept in a new tube.
5. The harvested cells are washed with RPMI-1640 supplemented with 20% fetal calf serum and
centrifuged at 200 × g for 20 min.
6. After discarding the supernatant, the cell pellet is resuspended with RPMI-1640 supplemented
with 20% fetal calf serum and kept in culture flask for 20 days or until complete differentiation
to macrophages with a density of 3 × 106 cells per 12.5 cm2 culture flask.
About 105–106 sporozoites are inoculated to the cell culture flask when host cells reach appropriate den-
sity. The culture flask is incubated at 37°C in a CO2 incubator. Growth and development of sporozoites
should be monitored every day under inverted microscope until no further development can be observed.
Infected cells can be examined by histopathological and ultrastructural studies.
of host cells. Undoubtedly, identification of natural cryptic optional intermediate or paratenic hosts for
C. belli has significant biological and medical implications.
Developmental cycle of C. belli consists of both asexual and sexual reproductions in a single host.
Freshly excreted oocysts from infected human stools are not readily infective but require some time in
environment to reach maturation, producing eight infective, crescentic-shaped sporozoites per oocyst.
The oocysts are environmentally resistant and can retain viability for months at low temperature.
Invasion of upper small intestinal enterocytes by sporozoites initiates asexual developmental cycle where
endodyogeny and possibly schizogony ensue. However, extraintestinal unicellular zoites or monozoic tis-
sue cysts, found mostly in lymphoid tissues of some AIDS patients, are suggestive of dormant stage that
could be responsible for relapse in almost half of symptomatic isosporiasis cases. The high rate of relapse
in C. belli infection suggests the presence of resistant cryptic stage that is not responsive to treatment. It
remains to be explored whether monozoic tissue cysts of C. belli directly contribute to relapses and what
reactivates their development. Although the majority of multiple relapses in cystoisosporiasis occurred
in humans with compromised immunity, some immunocompetent individuals reportedly suffered from
recurrent relapsing symptoms. It seems that the interplay between host immunity and parasite virulent
factors determines clinical course of C. belli infection. However, the lack of natural reservoir host for
C. belli has hindered experimental investigation to elucidate this issue. Therefore, modern molecular
biology techniques will be important alternative strategies to unravel several unknown aspects of patho-
genesis of human cystoisosporiasis. However, establishment of complete developmental cycle of this
coccidian parasite in cell culture system is mandatory to shed light on this neglected but important
human enteric coccidian pathogen.
REFERENCES
1. Manwell, R.D. Coccidia. In Manwell, R.D. (ed.), Introduction to Protozoology, 1st ed. St. Martin’s
Press, New York, pp. 515–538, 1961.
2. Wenyon, C.M. Coccidiosis of cats and dogs and the status of the Isospora of man. Ann. Trop. Med.
Parasitol. 17, 231, 1923.
3. Elsdon-Dew, R. Isospora natalensis (sp. Nov.) in man. J. Trop. Med. Hyg. 56, 149, 1953.
4. Ravenel, J.M., Suggs, J.L., and Legerton, C.W. Human coccidiosis. Recurrent diarrhea of 26 years dura-
tion due to Isospora belli: A case report. J. S. Carolina Med. Assoc. 72, 217, 1976.
5. Jongwutiwes, S., Sampatanukul, P., and Putaporntip, C. Recurrent isosporiasis over a decade in an
immunocompetent host successfully treated with pyrimethamine. Scand. J. Infect. Dis. 34, 859, 2002.
6. Barta, J.R. et al. The genus Atoxoplasma (Garnham 1950) as a junior objective synonym of the genus
Isospora (Schneider 1881) species infecting birds and resurrection of Cystoisospora (Frenkel 1977) as
the correct genus for Isospora species infecting mammals. J. Parasitol. 91, 726, 2005.
7. Franzen, C. et al. Taxonomic position of the intestinal protozoan parasite Isospora belli as based on
ribosomal RNA sequences. Parasitol. Res. 86, 669, 2000.
8. Morrison, D.A. et al. The current status of the small subunit rRNA phylogeny of the coccidia (Sporozoa).
Int. J. Parasitol. 34, 501, 2004.
9. Jongwutiwes, S. et al. Morphologic and molecular characterization of Isospora belli oocysts from
patients in Thailand. Am. J. Trop. Med. Hyg. 77, 107, 2007.
10. Dubey, J.P., and Frenkel, J.K. Extra-intestinal stages of Isospora felis and I. rivolta (Protozoa:
Eimeriidae) in cats. J. Protozool. 19, 89, 1972.
11. Restrepo, C., Macher, A.M., and Radany, E.H. Disseminated extraintestinal isosporiasis in a patient
with acquired immune deficiency syndrome. Am. J. Clin. Pathol. 87, 536, 1987.
12. Michiels, J.F. et al. Intestinal and extraintestinal Isospora belli infection in an AIDS patient. A second
case report. Pathol. Res. Pract. 190, 1089, 1994.
13. Frenkel, J.K. Besnoitia wallacei of cats and rodents: With a reclassification of other cyst-forming
isosporoid coccidia. J. Parasitol. 63, 611, 1977.
14. Lindsay, D.S., Dubey, J.P., and Blagburn, B.L. Biology of Isospora spp. from humans, nonhuman
primates, and domestic animals. Clin. Microbiol. Rev. 10, 19, 1997.
15. Tamura, K. et al. MEGA6: Molecular evolutionary genetics analysis version 6.0. Mol. Biol. Evol. 30,
2725, 2013.
Cystoisospora belli 613
16. Beaver, P.C., Jung, R.C., and Cupp, E.W. Coccidia, Microsporidia and Pneumocystis. In Beaver, P.C.,
Jung, R.C., and Cupp, E.W. (eds.), Clinical Parasitology, 9th ed. Lea and Febiger, Philadelphia, PA,
pp. 149–173, 1984.
17. Zaman, V. Observations on human Isospora. Trans. R. Soc. Trop. Med. Hyg. 62, 556, 1968.
18. Matsui, T. et al. Infectivity and sporogony of Caryospora-type oocyst of Isospora rivolta obtained by
heating. Parasitol. Res. 79, 599, 1993.
19. Dubey, J.P. Life cycle of Isospora rivolta (Grassi, 1879) in cats and mice. J. Protozool. 26, 433, 1979.
20. Ferguson, D.J.P. et al. Ultrastructural observations on multiplication of Cystoisospora (Isospora) felis
by endodyogeny. Z. Parasitenkd. 63, 289, 1980.
21. Matuschka, F.R. Ultrastructural evidence of endodyogeny in Isospora suis from pigs. Z. Parasitenkd.
67, 27, 1982.
22. Shah, H.L. The life cycle of Isospora felis Wenyon, 1923, a coccidium of the cat. J. Protozool. 18, 3,
1971.
23. Fayer, R., and Thompson, D.E. Isospora felis: Development in cultured cells with some cytological
observations. J. Parasitol. 60, 160, 1974.
24. Oliveira-Silva, M.B. et al. Cystoisospora belli: In vitro multiplication in mammalian cells. Exp.
Parasitol. 114, 189, 2006.
25. Siripanth, C. et al. Development of Isospora belli in Hct-8, Hep-2, human fibroblast, BEK and Vero
culture cells. Southeast Asian J. Trop. Med. Public. Health. 35, 796, 2004.
26. Resende, D.V. et al. Experimental infection of murine and human macrophages with Cystoisospora
belli. Acta. Trop. 111, 177, 2009.
27. Resende, D.V. et al. Ultrastructural aspects of Cystoisospora belli (syn. Isospora belli) in continuous cell
lines. Microsc. Res. Tech. 77, 472, 2014.
28. Velásquez, J.N. et al. Isosporosis and unizoite tissue cysts in patients with acquired immunodeficiency
syndrome. Hum. Pathol. 32, 500, 2001.
29. Frenkel, J.K. et al. Isospora belli infection: Observation of unicellular cysts in mesenteric lymphoid tissues
of a Brazilian patient with AIDS and animal inoculation. J. Eukaryot. Microbiol. 50(Suppl), 682, 2003.
30. Lindsay, D.S. et al. Examination of extraintestinal tissue cysts of Isospora belli. J. Parasitol. 83, 620,
1997.
31. Dubey, J.P. Experimental Isospora canis and Isospora felis infection in mice, cats, and dogs. J. Protozool.
22, 416, 1975.
32. Houk, A.E., and Lindsay, D.S. Cystoisospora canis (Apicomplexa: Sarcocystidae): Development of
monozoic tissue cysts in human cells, demonstration of egress of zoites from tissue cysts, and demon-
stration of repeat monozoic tissue cyst formation by zoites. Vet. Parasitol. 197, 455, 2013.
33. Mitchell, S.M., Zajac, A.M., and Lindsay, D.S. Development and ultrastructure of Cystoisospora canis
Nemeséri, 1959 (syn, Isospora canis) monozoic cysts in two noncanine cell lines. J. Parasitol. 95, 793,
2009.
34. Faust, E.C. et al. Human isosporosis in the western hemisphere. Am. J. Trop. Med. Hyg. 10, 343, 1961.
35. Henry, M.C. et al. Parasitological observations of chronic diarrhoea in suspected AIDS adult patients in
Kinshasa (Zaire). Trans. R. Soc. Trop. Med. Hyg. 80, 309, 1986.
36. DeHovitz, J.A. et al. Clinical manifestations and therapy of Isospora belli infection in patients with the
acquired immunodeficiency syndrome. N. Engl. J. Med. 315, 87, 1986.
37. Certad, G. et al. Isosporiasis in Venezuelan adults infected with human immunodeficiency virus:
Clinical characterization. Am. J. Trop. Med. Hyg. 69, 217, 2003.
38. Lainson, R., and da Silva, B.A. Intestinal parasites of some diarrhoeic HIV-seropositive individuals
in North Brazil, with particular reference to Isospora belli Wenyon, 1923 and Dientamoeba fragilis
Jepps & Dobell, 1918. Mem. Inst. Oswaldo. Cruz. 94, 611, 1999.
39. Sorvillo, F.J. et al. Epidemiology of isosporiasis among persons with acquired immunodeficiency syn-
drome in Los Angeles County. Am. J. Trop. Med. Hyg. 53, 656, 1995.
40. Brandborg, L.L., Goldberg, S.B., and Breidenbach, W.C. Human coccidiosis—A possible cause of mal-
absorption. N. Engl. J. Med. 283, 1306, 1970.
41. Trier, J.S. et al. Chronic intestinal coccidiosis in man: Intestinal morphology and response to treatment.
Gastroenterology 66, 923, 1974.
42. Matsubayashi, H., and Nozawa, T. Experimental infection of Isospora hominis in man. Am. J. Trop.
Med. 28, 633, 1948.
614 Laboratory Models for Foodborne Infections
43. McCracken, A.W. Natural and laboratory-acquired infection by Isospora belli. South. Med. J. 65, 800,
1972.
44. Syrkis, I. et al. A case of severe human coccidiosis in Israel. Israel. J. Med. Sci. 1, 373, 1975.
45. Pape, J.W., Verdier, R.I., and Johnson, W.D. Jr. Treatment and prophylaxis of Isospora belli infection in
patients with the acquired immunodeficiency syndrome. N. Engl. J. Med. 320, 1044, 1989.
46. de Otazu, R.D. et al. Endometrial coccidiosis. J. Clin. Pathol. 57, 1104, 2004.
47. Herwaldt, B.L. Cyclospora cayetanensis: A review, focusing on the outbreaks of cyclosporiasis in the
1990s. Clin. Infect. Dis. 31, 1040, 2000.
48. Garcia, L.S. Collection, preservation, and shipment of fecal specimens. In Garcia, L.S. (ed.), Diagnostic
Medical Parasitology, 4th ed. ASM Press, Washington, DC, pp. 723–740, 2001.
49. Jongwutiwes, S. et al. Simple method for long-term copro-preservation of Cryptosporidium oocysts for
morphometric and molecular analysis. Trop. Med. Int. Health. 7, 257, 2002.
50. Garcia, L.S. Macroscopic and microscopic examination of fecal specimens. In Garcia, L.S. (ed.),
Diagnostic Medical Parasitology, 4th ed. ASM Press, Washington, DC, pp. 741–785, 2001.
51. Pacheco, F.T. et al. Differences in the detection of Cryptosporidium and Isospora (Cystoisospora) oocysts
according to the fecal concentration or staining method used in a clinical laboratory. J. Parasitol. 99,
1002, 2013.
52. Ma, P., and Soave, R. Three-step stool examination for cryptosporidiosis in 10 homosexual men with
protracted watery diarrhea. J. Infect. Dis. 147, 824, 1983.
53. Varea, M. et al. Fuchsin fluorescence and autofluorescence in Cryptosporidium, Isospora and Cyclospora
oocysts. Int. J. Parasitol. 28, 1881, 1998.
54. Daugschies, A. et al. Autofluorescence microscopy for the detection of nematode eggs and protozoa, in
particular Isospora suis, in swine feces. Parasitol. Res. 87, 409, 2001.
55. Bialek, R. et al. Comparison of autofluorescence and iodine staining for detection of Isospora belli in
feces. Am. J. Trop. Med. Hyg. 67, 304, 2002.
56. Garcia, L.S. Sputum, aspirates, and biopsy material. In Garcia, L.S. (ed.), Diagnostic Medical
Parasitology, 4th ed. ASM Press, Washington, DC, pp. 809–828, 2001.
57. Sun, T. et al. Light and electron microscopic identification of Cyclospora species in the small intestine.
Evidence of the presence of asexual life cycle in human host. Am. J. Clin. Pathol. 105, 216, 1996.
58. Nime, F.A. et al. Acute enterocolitis in a human being infected with the protozoan Cryptosporidium.
Gastroenterology 70, 592, 1976.
59. Meisel, J.L. et al. Overwhelming watery diarrhea associated with a Cryptosporidium in an immunosup-
pressed patient. Gastroenterology 70, 1156, 1976.
60. Lindsay, D.S. et al. Developmental biology of Cystoisospora (Apicomplexa: Sarcocystidae) monozoic
tissue cysts. J. Parasitol. 100, 392, 2014.
61. Müller, A. et al. Detection of Isospora belli by polymerase chain reaction using primers based on small-
subunit ribosomal RNA sequences. Eur. J. Clin. Microbiol. Infect. Dis. 19, 631, 2000.
62. Taniuchi, M. et al. Multiplex polymerase chain reaction method to detect Cyclospora, Cystoisospora,
and microsporidia in stool samples. Diagn. Microbiol. Infect. Dis. 71, 386, 2011.
63. Lalonde, L.F., Reyes, J., and Gajadhar, A.A. Application of a qPCR assay with melting curve analysis for
detection and differentiation of protozoan oocysts in human fecal samples from Dominican Republic.
Am. J. Trop. Med. Hyg. 89, 892, 2013.
64. Petri, W.A. Sulfonamides, trimethoprim-sulfamethoxazole, quinolones, and agents for urinary tract
infections. In Brunton, L.L., Chabner, B.A., and Knollmann, B.C. (eds.), Goodman & Gilman’s: The
Pharmacological Basis of Therapeutics, 12th ed. McGraw-Hill, New York, NY, pp. 1462–1476, 2011.
65. Gangjee, A., Kurup, S., and Namjoshi, O. Dihydrofolate reductase as a target for chemotherapy in para-
sites. Curr. Pharm. Des. 13, 609, 2007.
66. Capasso, C.L., and Supuran, C.T. Sulfa and trimethoprim-like drugs—Antimetabolites acting as car-
bonic anhydrase, dihydropteroate synthase and dihydrofolate reductase inhibitors. J. Enzyme Inhib.
Med. Chem. 29, 379, 2014.
67. Bushby, S.R., and Hitchings, G.H. Trimethoprim, a sulphonamide potentiator. Br. J. Pharmacol.
Chemother. 33, 72, 1968.
68. Foner, A. An attempt to infect animals with Isospora belli. Trans. R. Soc. Trop. Med. Hyg. 33, 357, 1939.
Cystoisospora belli 615
69. Jeffery, G.M. Human coccidiosis in South Carolina. J. Parasitol. 42, 491, 1956.
70. Duszynski, D.W., and Conder, G.A. External factors and self-regulating mechanisms which may influ-
ence the sporulation of oocysts of the rat coccidium, Eimeria nieschulzi. Int. J. Parasitol. 7, 83, 1977.
71. Ryley, J.F. et al. Methods in coccidiosis research: Separation of oocysts from faeces. Parasitology 73,
311, 1976.
72. Dubey, J.P., and Streitel, R.H. Isospora felis and I. rivolta infections in cats induced by mouse tissue or
oocysts. Br. Vet. J. 132, 649, 1976.
39
Entamoeba histolytica
CONTENTS
39.1 Introduction....................................................................................................................................617
39.1.1 Human Amoebiasis...........................................................................................................617
39.1.2 Life Cycle of E. histolytica................................................................................................618
39.1.3 Isolation and Culture Conditions.......................................................................................618
39.1.4 Virulence Factors..............................................................................................................618
39.2 Animal Models of Amoebiasis......................................................................................................619
39.2.1 Intestinal Amoebiasis........................................................................................................619
39.2.1.1 Rats..................................................................................................................619
39.2.1.2 G uinea Pigs and Hamsters.............................................................................. 622
39.2.1.3 R abbits............................................................................................................ 622
39.2.1.4 G erbils............................................................................................................. 622
39.2.1.5 P igs.................................................................................................................. 622
39.2.1.6 M ice Models................................................................................................... 623
39.2.1.7 C3H/HeJ Mice................................................................................................ 623
39.2.1.8 C 57BL/6 IL-10-Deficient Mouse.................................................................... 623
39.2.1.9 SCID-HU-INT Mouse.................................................................................... 623
39.2.1.10 Human Colonic Explants Model.................................................................... 623
39.2.2 Amoebic Liver Abscess.................................................................................................... 624
39.2.3 Animal Models for Amoebiasis Treatment...................................................................... 626
39.2.4 Animal Models for Amoebic Vaccine Research.............................................................. 627
39.3 Conclusions and Future Directions............................................................................................... 628
Acknowledgments................................................................................................................................... 629
References............................................................................................................................................... 629
39.1 Introduction
39.1.1 Human Amoebiasis
Amoebiasis is a parasitic infection that is caused by an extracellular enteric protozoan named Entamoeba
histolytica. This parasite is able to coexist with the human intestine through a tolerogenic/hyporespon-
sive immune reaction, and the intestinal barrier is not broken. Through an unknown mechanism, the
host switches perception of the parasites from commensal to invaders and mounts an acute inflammatory
response that leads to colonic lesions of diverse magnitude. The symptoms of intestinal amoebiasis (IA)
are abdominal pain, dysentery, and ulcerative colitis with mucous and blood, which can, in some cases,
evolve into appendicitis and amoeboma, which is an acute manifestation of the disease. Rarely, the
parasites can spread through portal circulation to the liver and produce the main extraintestinal infec-
tion, amoebic liver abscess (ALA). E. histolytica can also reach the lungs and the brain, primarily in
617
618 Laboratory Models for Foodborne Infections
immunocompromised patients. Both IA and ALA can be fatal if left untreated. Amoebiasis is the third
leading cause of death due to parasites after malaria and schistosomiasis. This disease presents a high
index of morbidity and mortality, mainly in developing countries with poor hygiene conditions, where
it can be endemic. It can also affect travelers who visit those countries. According to the World Health
Organization (WHO) [1], 500 million people are infected with amoebae worldwide; 10% of them have
virulent E. histolytica, which leads to 40,000–100,000 deaths annually. Epidemiological studies dem-
onstrated the presence of antibodies (Abs) against E. histolytica in the populations of Mexico, Brazil,
Venezuela, Bangladesh, South Africa, and Egypt, showing the high prevalence of this parasite [2]. The
closely related nonpathogenic species E. dispar and E. moshkovskii are morphologically identical to
E. histolytica in standard stool microscopy analysis and are the cause of asymptomatic amoebiasis [3–8].
Studies of colon microbiota have been valuable in understanding the interplay among amoeba, the host,
and disease status [9–11]. Other host factors also influence the pathogenicity of amoeba, such as diet,
sex, and age [12].
39.1.4 Virulence Factors
E. histolytica is an active protist parasite that can adhere to the human epithelial mucosa and extracel-
lular matrix proteins, phagocytose erythrocytes and cell debris, secrete toxins and proteolytic enzymes,
cause apoptosis in target cells, and scavenge iron and other nutrients from the host [17–21]. Amoebiasis
is a multifactorial disease because host pathogen recognition receptors (PRRs), immune cells, amoeba
virulence factors, and microenvironment all contribute to the damage of the intestinal mucosa [4,22].
The contact between amoebae and target cells appears to be the first step in cell lysis and phagocytosis.
Intestinal flask-shaped ulcers, which are a hallmark of amoebic colitis, are characterized by severe dam-
age to enteric cells and migration to the lamina propria and blood vessels; an inflammatory reaction is
also present [20,23,24].
A wide variety of molecules have been described as E. histolytica virulence factors that act in con-
cert and favor amoebae invasion. For example, disruption of the colonic mucosa through amoebic
enzymes leads to direct contact between the parasites and the epithelium mainly through the amoebic
Gal/GalNAc lectin (Gal-lectin), which activates the inflammasome. The IL-1β released during invasion
Entamoeba histolytica 619
produces a severe inflammatory response [25]. Amoebapore polypeptides can lyse bacterial and host
cell membranes and are involved in liver abscess and cell apoptosis [26–29]. Several membrane and
secreted cysteine proteases (CPs) that degrade host proteins have been described in E. histolytica;
the most studied are CP1 and CP5 [30–33]. Currently, more than 50 genes are known to encode CPs,
and their expression depends on conditions such as the amoeba strain and culture conditions [34,35].
Lipophosphopeptidoglycan is a surface molecule that activates NKT cells in a CD1d-dependent manner,
and this interaction limits ALA development [25,36,37].
39.2.1 Intestinal Amoebiasis
The first in vivo model of IA was developed by Lesh in 1875 [42]. He produced intestinal lesions in a
dog inoculated with feces from a patient suffering from dysentery. Since then, many studies have been
performed in different animals (dogs, cats, rabbits, hamsters, and monkeys); however, the use of these
models is limited at present due to several factors, including a lack of uniformity, difficulty in handling,
and an insufficient number of animals [41]. IA in animals has yielded inconsistent and poor results,
and in many cases, this difficulty has been simply attributed to their natural immune resistance [41,43].
Moreover, although experimental animals are inoculated with several million trophozoites of a known
specific virulent strain, this inoculum then mixes with poorly characterized intestinal flora consisting
of additional protozoa, bacteria, and other microorganisms. This mixture of intestinal organisms, and
our lack of knowledge of the physicochemical characteristics of the intestinal milieu, are important
factors that have not been considered in the past and are likely the primary causes of the heterogeneity
of results [41].
Differences in susceptibility have been reported by different groups, even when using similar labora-
tory animals under apparently identical conditions. Despite these limitations, interesting information
regarding the early stages of IA, pathology, and host immune response has been obtained [41,44–46].
At the molecular level, various research groups have studied the genes or gene products of E. histolytica
related to those of humans through the evaluation of severe combined immunodeficient (SCID) and
SCID-modified mice, microarray analysis, and examination of several aspects of the mechanisms of the
immune response. In the forthcoming sections, we analyze some of the models used to establish IA [43].
Table 39.1 presents the foremost models used in IA.
39.2.1.1 Rats
Wistar strain rats (mainly weanling), or colon loops of adult Sprague Dawley rats, have been used to
model IA. However, due to the great variability of the macroscopic appearance related to the damage
induced by E. histolytica trophozoites, this model is no longer used [47,48].
TABLE 39.1
620
The Foremost Models Used to Study Intestinal Amoebiasis
Animal Model Method Advantages/Main Findings Limitations Reference
Dogs Inoculation with feces from a patient suffering Intestinal lesions in some animals Results were not reproducible, the animals [42]
from dysentery resolved the infection
Newborn guinea pigs, hamsters Intracecal inoculation axenic cultures Lesions in cecum Results were not reproduced in a larger [49]
number of animals
C3H/HeCr, BALB/c, NZB/BIN, Intracecal inoculation of axenic cultures Susceptibility of mice to infection Only early states of ameobiasis can be [56]
B10.A, DBA/2, and C57BL/6 depended on the genetic charge of the studied
inbred mice strains
Conventionally raised animals Inoculation of axenic or monoxenic cultures Early stages of amoeba invasion inside the Only early states of amoebiasis can be [50]
(hamsters or Guinea pigs) on a closed loop of the cecum mucosal and submucosal layers with a studied
substantial inflammatory reaction
Adult Sprague Dawley rats Inoculation of axenic cultures into in vivo The mucus blanket provided a significant Only early states of amoebiasis can be [47]
colon loops barrier to trophozoite access to intestinal studied
epithelium target
Washed-closed cecal loop model An artificial cecal loop inoculated with It helped reported the possible role of Only early states of amoebiasis can be [24,50]
axenic or monoxenic cultures leukocytes in the development of the studied
amoebic ulcer
Balb/c mice Intracecal inoculation of axenic cultures in Trophozoites did not survive in the Some mice were resistant to disease [54]
Entamoeba histolytica
The Foremost Models Used to Study Intestinal Amoebiasis
Animal Model Method Advantages/Main Findings Limitations Reference
Mongolian gerbils Intracecal inoculation of gerbils with Microulcerative mucosal lesions appeared Normal cecal mucosa at 96 h postinfection [51]
monoxenic cultures 24–72 h postinoculation, inflammatory
infiltrates and edema of the lamina
propria
C3H/HeJ mice The cecum was directly inoculated with Amoebae release toxic factors that Only intestinal amoeabiasis developed [125]
monoxenic cultures contributed to the inflammatory disease
SCID mouse-human intestinal Human intestinal xenografts were infected Extensive tissue damage, an early Only early intestinal amoebiasis can be [59]
xenograft model with monoxenic cultures inflammatory response composed studied
primarily of neutrophils. IL-1 and IL-8
were produced
Porcine colonic fragments Trophozoites cocultured with porcine Severe acute ulcerative jejunitis, with Lesions in two of the four animals [52]
colonic fragments large hemorrhagic lesions. Typical
large-sized hepatic abscesses
Human colon model ex vivo Parasite penetration into the mucus Human colonic explants are far less [63]
and mucosa, cell lysis, and an available
inflammatory response
621
622 Laboratory Models for Foodborne Infections
39.2.1.3 Rabbits
To develop a model capable of reproducing the events that occur during the initial interaction of E. his-
tolytica trophozoites with the mucosa of the large intestine, Navarro-García used full-thickness rabbit
colon preparations (0.28 cm2) mounted in Ussing-type chambers. The untreated samples had electro-
physiological properties (potential difference, short-circuit current, and electrical resistance) that were
similar in magnitude and duration to those reported for stripped colonic mucosa. However, in samples
exposed to amoeba lysates for up to 80 min, dose-dependent lesions in the colon were observed and
consisted of (1) increased decay rates for potential difference, short-circuit current, and transmural resis-
tance and (2) mucosal lesions involving vacuolization at the bases and shortening of epithelial cells, loss
of intercellular junctions, destruction of microvilli, and necrosis of interglandular epithelial zones. The
authors discussed the specificity and speed of the electrophysiological effects and their correlation with
the microscopic lesions and suggested that this model will help increase our understanding of the initial
pathogenic events of IA [44].
39.2.1.4 Gerbils
By using monoxenic cultures of E. histolytica trophozoites inoculated in Mongolian gerbils, Shibayama
observed that an increase in mucus production occurred during the first hours of the interaction. Furthermore,
microulcerative mucosal lesions appeared 24–72 h postinoculation. Inflammatory infiltrate and edema of the
lamina propria were associated with necrotic foci. Unfortunately, at 96 h, the cecal mucosa was normal,
amoebae were no longer detected, and the gerbils healed spontaneously [51]. However, overall, it was con-
cluded that gerbils are useful as experimental models for studying the early stages of invasive IA.
39.2.1.5 Pigs
More recently, assays with trophozoites of virulent E. histolytica cocultured with porcine colonic frag-
ments have shown that outbred pigs can be used to reproduce some lesions associated with human IA.
A detailed analysis showed that loops inoculated with virulent amoebae showed severe acute ulcerative
jejunitis 14 days postinoculation, with large hemorrhagic areas associated with the presence of amoebae in
the depth of the mucosa in two of the four animals [52]. Furthermore, typical large hepatic abscesses were
observed in the liver of one animal at 7 days postinfection after inoculation by the portal vein or directly
into the liver parenchyma, which showed that the pig model could be useful in simultaneously studying IA
and ALA. Moreover, human colonic explants have been identified as valuable assets in the study of host–
parasite interactions. However, because human colonic explants are far less available, porcine colonic
explants have been investigated as an alternative to human tissues. Porcine colonic explants cultured with
virulent E. histolytica (HM1:IMSS) or an avirulent strain (Rahman) showed that explants cultured with
virulent trophozoites react similar to their human counterparts, including tissue invasion by amoebae and
the triggering of a typical innate immune response against the parasite. In contrast, explants cultured with
avirulent amoebae were healthy. The authors suggest that this study opens the way for the use of porcine
colonic explants in the study of the complex interactions between the parasite and the host [53].
Entamoeba histolytica 623
39.2.1.6 Mice Models
In the past, mice were not commonly used as a model for amoebiasis, primarily since these rodents were
always considered naturally resistant to E. histolytica infection [54]. Depending on the strain, mice pre-
sented different susceptibilities to developing IA; thus, the authors concluded that a genetic factor could
be involved in mouse susceptibility to intestinal infection. It was also postulated that other factors such
as a cholesterol-rich diet, association with bacteria, and the presence of other protozoa could influence
the results found in the different mice strains [43,54]. A neutropenic BALB/c mouse model was devel-
oped, in which the production of a granulomatous inflammatory reaction in the intestinal wall of mice
inoculated with axenically cultured amoebae was reported for the first time [55]. Assays in other strains
corroborated that the establishment of IA depends on the mouse strain; for example, C57BL/6 mice are
highly resistant, whereas C3H/HeJ mice are relatively susceptible. The amoebic colitis in these mice was
limited to the cecum, and the morphology of the inflammatory infiltrate was similar to that observed
in humans. This model of resistant versus susceptible mice could provide useful insight into the human
variability of parasite clearance when compared with invasive disease [56]. In the next section, we ana-
lyze the mice strains that are susceptible to intestinal amoebiasis.
39.2.1.7 C3H/HeJ Mice
In 2002, it was shown that C3H/HeJ mice, which have a mutation at the lipopolysaccharide response
locus, were 60% infected with amoeba after intracecal inoculation, whereas C57BL/6 or BALB/c mice
were resistant, including mice that were deficient for IL-12, IFN-γ, or inducible NO synthase. Infection
was a chronic cecum inflammation that pathologically mirrored the human disease. This model revealed
important immune factors that influence susceptibility to infection and, for the first time, established the
pathological contribution of the host immune response in amoebiasis [57]. The early steps in IA, such as
parasite adhesion to the mucosa, can be investigated in this model, and chronic infection can be obtained
after mechanical injury of the cecal epithelium.
39.2.1.9 SCID-HU-INT Mouse
Infection of the SCID mouse-human intestinal xenograft (SCID-HU-INT) model of disease with amoe-
bae resulted in extensive tissue damage, which was associated with an early inflammatory response
composed primarily of neutrophils [59]. In this model, it was evidenced that human intestinal epithelial
cells can produce inflammatory cytokines in response to infection in vivo, and it was established as a
system for studying the interactions between E. histolytica and the human intestine [59,60]. Furthermore,
research on an E. histolytica substrain in which the expression of several CPs was downregulated by an
antisense transcript showed that these enzymes play a major role in the advancement of IA in the SCID-
HU-INT mouse [61]. One of them, EhCP-A5, which is not expressed in E. dispar, has been shown to
degrade the cysteine-rich domains of the MUC2 mucin, which are the major structural component of the
colonic mucus gel in the human digestive tract [61].
compare them with those observed after infection with the nonpathogenic parasite E. dispar [63]. Based
on recent results in work with this model, the Gal-lectin and amoebapores are not required for the inva-
sion of human colon explants, thus suggesting that CP-A5, which is involved in extracellular matrix deg-
radation, is not required for crossing the mucus but rather contributes directly or indirectly to penetrating
the lamina propria and inducing inflammation [64].
(A) (B) h
h
40x
(C) h (D)
h
40x n 20x
(E) ec (F)
n ly
40x 20x
FIGURE 39.1 (a) Amoebic liver abscess. Macroscopic aspect. Whitish lesions covering the entire right lobule (visceral
face); 7 days postintrahepatic inoculation with 1 × 106 trophozoites of Entamoeba histolytica. (b–f) Histopathological
analysis. (b) Three hours postinfection. Trophozoites (arrows), inflammatory cells (arrow-heads), and hepatocytes (h). 40×.
(c) Six hours postinoculation. The inflammatory foci increase in number; amoeba (arrow), inflammatory reaction (arrow-
heads) and hepatocytes (h) 40×. (d) Two days postintrahepatic inoculation. The necrotic areas are evident (n), the amoe-
bae increase in number (arrows), inflammatory cells (arrow-head). Some hepatocytes appear apparently normal (h). 20×.
(e) Granulomatous reaction. Five days postinfection. Necrotic center (n), epithelioid cells (ec), and trophozoites (arrows).
40×. (f) Seven days postinoculation. The necrotic areas increased in the liver. Some amoebae are seen (arrows) intermixed
with the lytic necrosis (ly), inflammation (arrow-heads). 20×.
TABLE 39.2
Amoebiasis: Susceptibility in Rodents
Intestinal Liver
Hamster − +
Gerbil +/− +
Guinea pig +/− −
Mouse +/− −
Rat +/− −
lymphocytes are important in the resistance to hepatic amoebiasis [69,71]. More recently, studies using
Balb/c mice treated with a monoclonal Ab (RB6–8C5) against neutrophils were challenged by intrahe-
patic administration of amoebae. The animals were sacrificed at different times postinoculation, and liv-
ers were analyzed histologically. The results showed that the neutropenic animals developed more severe
ALA than did the controls. The authors concluded that neutrophils are important to natural resistance in
amoebiasis, in contrast to the results found in hamster and gerbil models that develop ALA [72].
626 Laboratory Models for Foodborne Infections
Information about the participation of cytokines, complement, and Abs is still incomplete. Cytokines
such as IFN-γ, TNF-α, and MCSF-1 increase the amoebicidal effect of macrophages (peritoneal and
Kupffer cells) [73]. Proteinases and amoebapores of E. histolytica are also involved in ALA produc-
tion. To actively invade, amoebae must degrade different components of the extracellular matrix, which
occurs via the action of proteinases secreted by the parasite. These enzymes degrade collagen, elastin,
fibrinogen, fibronectin, and laminin [21,30]. Another factor that is known to contribute to tissue inva-
sion is the inflammatory reaction produced by the presence of amoebae in the liver. The expression of
TNF-α, IFN-γ, IL-1β, IL-8, and IL-10 was studied throughout ALA evolution in hamster. The authors
showed that neutrophils and macrophages that infiltrate the liver parenchyma in the acute and chronic
stages of ALA are also responsible for the tissue damage. IL-10 does not regulate the local production of
proinflammatory cytokines. The results showed that the exacerbated inflammatory milieu contributes to
damage and probably supports the survival of E. histolytica [74].
It is important to mention that the nonpathogenic E. dispar strain SAW760 cultured in axenic condi-
tions showed early inflammatory reaction around the amoebae. However, hepatic lesions did not progress
to ALA formation. In other recent studies using different strains of E. dispar in xenic or monoxenic
culture conditions, the production of amoebic liver lesions has been reported [75,76].
a high-throughput screening for compounds effective against amoeba identified auranofin, an FDA-
approved drug used therapeutically against rheumatoid arthritis. Auranofin is 10 times more potent than
Mtz, which makes it a promising therapy for amoebiasis [100]. The innate immune response by pro-
tein lactoferrin, an iron-chelating molecule that is responsible for avoiding pathogens to acquire iron in
mucosae and infection sites, has been tested in ALA and IA [101,102]. Remarkably, bovine lactoferrin
was able to cure ALA and synergize with Mtz in biological action. This observation is particularly
important given that a reduced dose of Mtz can be used if combined with lactoferrin, with the same effect
on the parasite, thus diminishing the side effects and toxicity of the drug. In addition, other properties of
lactoferrin such as its anti-inflammatory activity can help resolve the amoebic abscesses [101].
Other possible vaccine candidates are antigenic proteins, such as Gal-lectin adhesin, which is
the best characterized E. histolytica protein. This protein is a 260-kDa heterodimer localized on
the amoeba surface [115]. Gal-lectin is an attractive candidate for a vaccine because of its immu-
nogenicity and its importance in the development of the disease. The first experiment to use puri-
fied Gal-lectin showed 86% protection against ALA in the gerbil model [116]. Another potential
protein target is the serine-rich E. histolytica protein (SREHP). Previous studies of this protein have
reported its success as an antigen to protect gerbils against ALA [68]. Intradermal immunization
with SREHP as a maltose-binding protein induced 100% protection against ALA in gerbils [68]. The
E. histolytica 29-kDa molecule (Eh29) is also considered an important antigen for an amoebiasis
vaccine. Eh29 is an alkyl hydroperoxide reductase that is involved in the detoxification of reactive
oxygen species secreted by microflora or immune cells [117]. One study of this protein in the ham-
ster model showed 54% protection against ALA [118]. Another study using Eh-29 conjugated with
cholera toxin B subunit (CTB) conferred protection against intracecal amoebiasis in C3H/heJ mice
(IA), which was associated with anti-Eh29 IgA Abs in the intestine and anti-Eh29 IgG-specific Abs
in the serum [119].
The development of DNA vaccines is recent and involves the induction of DNA of known
sequences of some antigens of interest into a bacterial plasmid. DNA vaccines have been shown
to exert strong humoral and cell-mediated responses and were successful in conferring protection
against parasites. One example of this method is the generation of a codon-optimized DNA vaccine
encoding a portion of the Gal-lectin of E. histolytica. When Balb/c mice were vaccinated intrader-
mally with the DNA plasmid, the vaccine stimulated a Th1-type Gal-lectin-specific cellular immune
response as well as the development of serum Abs that recognized a recombinant portion of the
heavy subunit and inhibited the adherence of trophozoites to target cells in vitro [120]. Another type
of DNA vaccine is the multivalent vaccine. One example of this is the EhCPADH complex, which is
formed by two surface molecules: CP112 (EhCP112) and an adhesin (EhADH112). Compared with
immunization with each plasmid alone (EhCP112) or EhADH112), the coimmunization of hamsters
with the two plasmids induced a significantly greater level of anti-E. histolytica IgG. Interestingly,
protection against liver abscesses was detected only in animals that received the plasmid mixture,
and no protection was observed in hamsters independently inoculated with plasmid pcDNA-Ehcp112
or pcDNA-Ehadh112 [121].
There are other amoebic targets such as the heparin sulfate-binding protein (HSBP) and the 30-kDa
collagen-binding protein (CBP30) recombinant fused to portions of the Trypanosoma cruzi heat shock
protein of 70 kDa, which have been assayed as vaccine antigens in guinea pigs [122] and in hamsters
[123], respectively. Beyond rodent models, only one study has explored the utility of vaccines in a non-
human primate model of amoebiasis [124]. In this model, Gal-lectin was administered with CTB as an
adjuvant in baboons by colonoscopy into the lumen of small bowel and cecum. This vaccine resulted in
a moderate level of protection against E. histolytica reinfection. However, the nonhuman primate study
represents a significant advance toward an anti-E. histolytica vaccine. It is evident that more studies are
needed in this field. The identification of E. histolytica immunogenic proteins and the correct combina-
tion of doses, boosts, and adjuvants is a priority to obtain long-term immunological memory in experi-
mental animal models to advance beyond the preclinical stage in humans.
Acknowledgments
This work was supported by grants No. 1674231 (Serrano-Luna), 179251 (de la Garza), and 237523
(Shibayama-Salas) from Conacyt, México.
REFERENCES
1. W.H.O., Amebiasis. Wkly. Epidemiol. Rec., 72: 97–9, 1997.
2. Chacin-Bonilla, L., Current pharmacotherapy of amebiasis, advances in new drugs, and design of a
vaccine. Invest. Clin., 53: 301–14, 2012.
3. Ximenez, C., Epidemiology of amebiasis in Mexico: a molecular approach. Arch. Med. Res., 37: 263–5,
2006.
4. Espinosa-Cantellano, M. and Martinez-Palomo, A., Pathogenesis of intestinal amebiasis: from mol-
ecules to disease. Clin. Microbiol. Rev., 13: 318–31, 2000.
5. Stanley, S.L., Jr., Amoebiasis. Lancet, 361: 1025–34, 2003.
6. Haque, R., et al., Amebiasis. N. Engl. J. Med., 348: 1565–73, 2003.
7. Ali, I.K., Clark, C.G., and Petri, W.A., Jr., Molecular epidemiology of amebiasis. Infect. Genet. Evol., 8:
698–707, 2008.
8. Mortimer, L. and Chadee, K., The immunopathogenesis of Entamoeba histolytica. Exp. Parasitol., 126:
366–80, 2010.
9. Phillips, B.P., et al., Studies on the ameba-bacteria relationship in amebiasis; comparative results of the
intracecal inoculation of germfree, monocontaminated, and conventional guinea pigs with Entamoeba
histolytica. Am. J. Trop. Med. Hyg., 4: 675–92, 1955.
10. Mirelman, D., Ameba-bacterium relationship in amebiasis. Microbiol. Rev., 51: 272–84, 1987.
11. Galvan-Moroyoqui, J.M., et al., The interplay between Entamoeba and enteropathogenic bacteria mod-
ulates epithelial cell damage. PLoS Negl. Trop. Dis., 2: e266, 2008.
12. Soh, C.T., Factors influencing the pathogenicity of Entamoeba histolytica. Yonsei. Med. J., 29: 1–10, 1988.
13. Martinez-Palomo, A., The pathogenesis of amoebiasis. Parasitol. Today, 3: 111–8, 1987.
14. Diamond, L.S., Axenic cultivation of Entamoeba hitolytica. Science, 134: 336–7, 1961.
15. Diamond, L.S., Harlow, D.R., and Cunnick, C.C., A new medium for the axenic cultivation of Entamoeba
histolytica and other Entamoeba. Trans. R. Soc. Trop. Med. Hyg., 72: 431–2, 1978.
16. De la Torre, M., et al., Cultivos axénicos de Entamoeba histolytica. Arch. Invest. Med., 2: 165–172, 1971.
17. Mann, B.J., Mirelman, D., and Petri, W.A., Jr., The d-galactose-inhibitable lectin of Entamoeba histo-
lytica. Carbohydr. Res., 213: 331–8, 1991.
18. Lopez-Soto, F., et al., Use and endocytosis of iron-containing proteins by Entamoeba histolytica tropho-
zoites. Infect. Genet. Evol., 9: 1038–50, 2009.
19. Christy, N.C. and Petri, W.A., Jr., Mechanisms of adherence, cytotoxicity and phagocytosis modulate
the pathogenesis of Entamoeba histolytica. Future Microbiol., 6: 1501–19, 2011.
20. Ralston, K.S. and Petri, W.A., Jr., Tissue destruction and invasion by Entamoeba histolytica. Trends
Parasitol., 27: 254–63, 2011.
21. Pina-Vazquez, C., et al., Host-parasite interaction: parasite-derived and -induced proteases that degrade
human extracellular matrix. J. Parasitol. Res., 2012: 748206, 2012.
22. Faust, D.M. and Guillen, N., Virulence and virulence factors in Entamoeba histolytica, the agent of
human amoebiasis. Microbes Infect., 14: 1428–41, 2012.
23. Tsutsumi, V., et al., Cellular bases of experimental amebic liver abscess formation. Am. J. Pathol., 117:
81–91, 1984.
24. Tsutsumi, V., Anaya-Velazquez, F., and Martinez-Palomo, A., Experimental intestinal amebiasis: inva-
sion and extension of the amebic lesion. Arch. Invest. Med. (Mex), 21 Suppl 1: 47–52, 1990.
25. Mortimer, L., et al., Gal-lectin-dependent contact activates the inflammasome by invasive Entamoeba
histolytica. Mucosal Immunol., 7: 829–41, 2014.
26. Leippe, M., Amoebapores. Parasitol. Today, 13: 178–83, 1997.
27. Bracha, R., Nuchamowitz, Y., and Mirelman, D., Transcriptional silencing of an amoebapore gene in
Entamoeba histolytica: molecular analysis and effect on pathogenicity. Eukaryot. Cell, 2: 295–305,
2003.
630 Laboratory Models for Foodborne Infections
28. Leippe, M., et al., Ancient weapons: the three-dimensional structure of amoebapore A. Trends Parasitol.,
21: 5–7, 2005.
29. Andra, J., Herbst, R., and Leippe, M., Amoebapores, archaic effector peptides of protozoan origin,
are discharged into phagosomes and kill bacteria by permeabilizing their membranes. Dev. Comp.
Immunol., 27: 291–304, 2003.
30. Que, X. and Reed, S.L., Cysteine proteinases and the pathogenesis of amebiasis. Clin. Microbiol. Rev.,
13: 196–206, 2000.
31. Stanley, S.L., Pathophysiology of amoebiasis. Trends Parasitol., 17: 280–5, 2001.
32. Bruchhaus, I., et al., The intestinal protozoan parasite Entamoeba histolytica contains 20 cysteine protease
genes, of which only a small subset is expressed during in vitro cultivation. Eukaryot. Cell, 2: 501–9, 2003.
33. Serrano-Luna, J., et al., Proteases from Entamoeba spp. and pathogenic free-living amoebae as viru-
lence factors. J. Trop. Med., 2013: 890603, 2013.
34. Loftus, B., et al., The genome of the protist parasite Entamoeba histolytica. Nature, 433: 865–8, 2005.
35. Loftus, B.J. and Hall, N., Entamoeba: still more to be learned from the genome. Trends Parasitol., 21:
453, 2005.
36. Wong-Baeza, I., et al., The role of lipopeptidophosphoglycan in the immune response to Entamoeba
histolytica. J. Biomed. Biotechnol., 2010: 254521, 2010.
37. Padilla-Vaca, F. and Anaya-Velazquez, F., Insights into Entamoeba histolytica virulence modulation.
Infect. Disord. Drug Targets, 10: 242–50, 2010.
38. Perez-Tamayo, R., Martinez-Villegas, J.E., and Perez-Montfort, R., Effect of cellular immunity on the
interaction between the peritoneal cell and the ameba in vitro. Arch. Invest. Med. (Mex), 17 Suppl 1:
259–67, 1986.
39. Tsutsumi, V. and Martinez-Palomo, A., Inflammatory reaction in experimental hepatic amebiasis. An
ultrastructural study. Am. J. Pathol., 130: 112–9, 1988.
40. Chadee, K. and Meerovitch, E., Entamoeba histolytica: diffuse liver inflammation in gerbils (Meriones
unguiculatus) with experimentally induced amebic liver abscess. J. Protozool., 36: 154–8, 1989.
41. Tsutsumi, V. and Shibayama, M., Experimental amebiasis: a selected review of some in vivo models.
Arch. Med. Res., 37: 210–20, 2006.
42. Lesh, F.A., Massive development of amebas in the large intestine. Fedor Aleksandrovich Lesh (Losch).
Am. J. Trop. Med. Hyg., 24: 383–92, 1975.
43. Ivory, C., Kammanadiminti, S., and Chadee, K., Innate resistance to Entamoeba histolytica in murine
models. Trends Parasitol., 23: 46–8, 2007.
44. Navarro-Garcia, F., et al., Model of intestinal amebiasis: structural and functional lesions to the rabbit
colon mucosa by Entamoeba histolytica lysates. Arch. Med. Res., 23: 197–201, 1992.
45. Ghosh, P.K., Castellanos-Barba, C., and Ortiz-Ortiz, L., Intestinal amebiasis: cyclic suppression of the
immune response. Parasitol. Res., 81: 475–80, 1995.
46. Ghosh, P.K., et al., Experimental amebiasis: immunohistochemical study of immune cell populations.
J. Eukaryot. Microbiol., 47: 395–9, 2000.
47. Leitch, G.J., et al., Entamoeba histolytica trophozoites in the lumen and mucus blanket of rat colons
studied in vivo. Infect. Immun., 47: 68–73, 1985.
48. Neal, R.A. and Harris, W.G., Proceedings: attempts to infect inbred strains of rats and mice with
Entamoeba histolytica. Trans. R. Soc. Trop. Med. Hyg., 69: 429–30, 1975.
49. Diamond, L.S., Tanimoto Weki, M., and Martinez-Palomo, A., Production of cecal lesions in newborn
guinea pigs with axenically cultivated Entamoeba histolytica. Arch. Invest. Med. (Mex), 9 Suppl 1:
223–8, 1978.
50. Anaya-Velazquez, F., et al., Intestinal invasive amebiasis: an experimental model in rodents using axenic
or monoxenic strains of Entamoeba histolytica. Am. J. Trop. Med. Hyg., 34: 723–30, 1985.
51. Shibayama-Salas, M., Tsutsumi, V., and Martinez-Palomo, A., Early invasive intestinal amebiasis in
Mongolian gerbils. Arch. Med. Res., 23: 187–90, 1992.
52. Girard-Misguich, F., et al., Towards the establishment of a porcine model to study human amebiasis.
PLoS One, 6: e28795, 2011.
53. Girard-Misguich, F., et al., Porcine colon explants in the study of innate immune response to Entamoeba
histolytica. Vet. Immunol. Immunopathol., 145: 611–7, 2012.
54. Anaya-Velazquez, F. and Underdown, B.J., Early expression in mice of genetic resistance to intestinal
amebiasis. Arch. Invest. Med. (Mex), 21 Suppl 1: 53–6, 1990.
Entamoeba histolytica 631
55. Rivero-Nava, L., et al., Entamoeba histolytica: acute granulomatous intestinal lesions in normal and
neutrophil-depleted mice. Exp. Parasitol., 101: 183–92, 2002.
56. Ghadirian, E. and Kongshavn, P.A., Genetic control of susceptibility of mice to infection with E. histo-
lytica. Parasite Immunol., 6: 349–60, 1984.
57. Houpt, E.R., et al., The mouse model of amebic colitis reveals mouse strain susceptibility to infection
and exacerbation of disease by CD4+ T cells. J. Immunol., 169: 4496–503, 2002.
58. Hamano, S., et al., Resistance of C57BL/6 mice to amoebiasis is mediated by nonhemopoietic cells but
requires hemopoietic IL-10 production. J. Immunol., 177: 1208–13, 2006.
59. Seydel, K.B., et al., Human intestinal epithelial cells produce proinflammatory cytokines in response to
infection in a SCID mouse-human intestinal xenograft model of amebiasis. Infect. Immun., 65: 1631–9,
1997.
60. Seydel, K.B., et al., Epithelial cell-initiated inflammation plays a crucial role in early tissue damage in
amebic infection of human intestine. Gastroenterology, 115: 1446–53, 1998.
61. Zhang, Z., et al., Entamoeba histolytica cysteine proteinases with interleukin-1 β converting enzyme
(ICE) activity cause intestinal inflammation and tissue damage in amoebiasis. Mol. Microbiol., 37:
542–8, 2000.
62. Bansal, D., et al., An ex-vivo human intestinal model to study Entamoeba histolytica pathogenesis.
PLoS Negl. Trop. Dis., 3: e551, 2009.
63. Thibeaux, R., et al., Identification of the virulence landscape essential for Entamoeba histolytica inva-
sion of the human colon. PLoS Pathog., 9: e1003824, 2013.
64. Thibeaux, R., et al., The parasite Entamoeba histolytica exploits the activities of human matrix metal-
loproteinases to invade colonic tissue. Nat. Commun., 5: 5142, 2014.
65. Reinertson, J.W. and Thompson, P.E., Experimental amebic hepatitis in hamsters. Proc. Soc. Exp. Biol.
Med., 76: 518–21, 1951.
66. Tanimoto, M., Sepúlveda, B., and Vázquez, J.A., Lesiones producidas en el hígado de hámsteres por
inoculación de E. histolytica cultivada en medio axénico. Arch. Invest. Med. (Mex), 2: 275–84, 1971.
67. Chadee, K. and Meerovitch, E., The pathogenesis of experimentally induced amebic liver abscess in the
gerbil (Meriones unguiculatus). Am. J. Pathol., 117: 71–80, 1984.
68. Zhang, T. and Stanley, S.L., Jr., Protection of gerbils from amebic liver abscess by immunization with a
recombinant protein derived from the 170-kilodalton surface adhesin of Entamoeba histolytica. Infect.
Immun., 62: 2605–8, 1994.
69. Cieslak, P.R., Virgin, H.W.T., and Stanley, S.L., Jr., A severe combined immunodeficient (SCID) mouse
model for infection with Entamoeba histolytica. J. Exp. Med., 176: 1605–9, 1992.
70. Stern, J.J., Graybill, J.R., and Drutz, D.J., Murine amebiasis: the role of the macrophage in host defense.
Am. J. Trop. Med. Hyg., 33: 372–80, 1984.
71. Wijesundera Mde, S., Hepatic amoebiasis in immunodepressed mice. Trans. R. Soc. Trop. Med. Hyg.,
74: 216–20, 1980.
72. Velazquez, C., et al., Role of neutrophils in innate resistance to Entamoeba histolytica liver infection in
mice. Parasite Immunol., 20: 255–62, 1998.
73. Denis, M. and Chadee, K., Cytokine activation of murine macrophages for in vitro killing of Entamoeba
histolytica trophozoites. Infect. Immun., 57: 1750–6, 1989.
74. Pacheco-Yepez, J., et al., Expression of cytokines and their regulation during amoebic liver abscess
development. Parasite Immunol., 33: 56–64, 2011.
75. Shibayama, M., et al., A Brazilian species of Entamoeba dispar (ADO) produces amoebic liver abscess
in hamsters. Ann. Hepatol., 6: 117–8, 2007.
76. Dolabella, S.S., et al., Amoebic liver abscess production by Entamoeba dispar. Ann. Hepatol., 11: 107–
17, 2012.
77. Jones, W.R., The experimental infection of rats with Entamoeba histolytica; with a method for evaluat-
ing the anti-amoebic properties of new compounds. Ann. Trop. Med. Parasitol., 40: 130–40, 1946.
78. Hernandez-Lopez, H.R. and Escobedo-Salinas, A., Effects of metronidazole on amebic hepatic abscess
in hamsters. Arch. Invest. Med. (Mex), 1: Suppl:125–8, 1970.
79. Kradolfer, F. and Jarumilinta, R., Ciba 32,644-Ba, a new systemically active amoebicide. Ann. Trop.
Med. Parasitol., 59: 210–8, 1965.
80. Cuckler, A.C., Malanga, C.M., and Conroy, J., Therapeutic efficacy of new nitroimidazoles for experi-
mental trichomoniasis, amebiasis, and trypanosomiasis. Am. J. Trop. Med. Hyg., 19: 916–25, 1970.
632 Laboratory Models for Foodborne Infections
81. Ray, D.K., et al., Comparative studies on the amoebicidal activity of known 5-nitroimidazole derivatives
and CG 10213-Go in golden hamsters, Mesocricetus auratus, infected in the liver or caecum or both
with trophozoites of Entamoeba histolytica. Ann. Trop. Med. Parasitol., 77: 287–91, 1983.
82. Pargal, A., et al., Pharmacokinetics and amoebicidal activity of (±)-(E)-3-(4-methylsulphinylstyryl)-
1,2,4-oxadiazole (BTI 2286E) and its sulphone metabolite (BTI 2571E) in the golden hamster,
Mesocricetus auratus. J. Antimicrob. Chemother., 32: 109–15, 1993.
83. Pargal, A., et al., Comparative pharmacokinetics and amoebicidal activity of metronidazole and satra-
nidazole in the golden hamster, Mesocricetus auratus. J. Antimicrob. Chemother., 32: 483–9, 1993.
84. Kradolfer, F., Jarumilinta, R., and Sackmann, W., The amoebicidal, trichomonicidal, and antibacterial
effects of niridazole in laboratory animals. Ann. N. Y. Acad. Sci., 160: 740–8, 1969.
85. Roe, F.J., Toxicologic evaluation of metronidazole with particular reference to carcinogenic, mutagenic,
and teratogenic potential. Surgery, 93: 158–64, 1983.
86. Dobias, L., et al., Genotoxicity and carcinogenicity of metronidazole. Mutat. Res., 317: 177–94, 1994.
87. Kitchen, L.W., Case studies in international travelers. Am. Fam. Physician, 60: 471–4, 1999.
88. Showler, A.J. and Boggild, A.K., Entamoeba histolytica. Can. Med. Assoc. J., 185: 1064, 2013.
89. Upcroft, P. and Upcroft, J.A., Drug targets and mechanisms of resistance in the anaerobic protozoa.
Clin. Microbiol. Rev., 14: 150–64, 2001.
90. Sharma, G.L. and Bhutani, K.K., Plant based antiamoebic drugs; Part II. Amoebicidal activity of
parthenin isolated from Parthenium hysterophorus. Planta Med., 54: 120–2, 1988.
91. Sohni, Y.R., Kaimal, P., and Bhatt, R.M., The antiamoebic effect of a crude drug formulation of herbal
extracts against Entamoeba histolytica in vitro and in vivo. J. Ethnopharmacol., 45: 43–52, 1995.
92. Sawangjaroen, N., Sawangjaroen, K., and Poonpanang, P., Effects of Piper longum fruit, Piper sar-
mentosum root and Quercus infectoria nut gall on caecal amoebiasis in mice. J. Ethnopharmacol., 91:
357–60, 2004.
93. Avila-Blanco, M.E., et al., Amoebicidal activity of essential oil of Dysphania ambrosioides (L.)
Mosyakin & Clemants in an amoebic liver abscess hamster model. Evid. Based Complement. Alternat.
Med., 2014: 930208, 2014.
94. Ghosh, S., et al., Effects of bisphosphonates on the growth of Entamoeba histolytica and Plasmodium
species in vitro and in vivo. J. Med. Chem., 47: 175–87, 2004.
95. Ordaz-Pichardo, C., et al., Antiamoebic and toxicity studies of a carbamic acid derivative and its thera-
peutic effect in a hamster model of hepatic amoebiasis. Antimicrob. Agents Chemother., 49: 1160–8,
2005.
96. Chatterjee, D.K., Iyer, N., and Ganguli, B.N., Antiamoebic activity of chonemorphine, a steroidal alka-
loid, in experimental models. Parasitol. Res., 74: 30–3, 1987.
97. Bhopale, K.K., et al., A comparative study of experimental caecal amoebiasis and the evaluation of
amoebicides. Ann. Trop. Med. Parasitol., 89: 253–9, 1995.
98. Bhopale, K.K., et al., Additive effect of diloxanide furoate and metronidazole (Entamizole) in experi-
mental mouse caecal amoebiasis. Indian. J. Exp. Biol., 33: 73–4, 1995.
99. Sato, D., et al., Cytotoxic effect of amide derivatives of trifluoromethionine against the enteric proto-
zoan parasite Entamoeba histolytica. Int. J. Antimicrob. Agents, 35: 56–61, 2010.
100. Debnath, A., et al., A high-throughput drug screen for Entamoeba histolytica identifies a new lead and
target. Nat. Med., 18: 956–60, 2012.
101. Ordaz-Pichardo, C., et al., Effect of bovine lactoferrin in a therapeutic hamster model of hepatic amoe-
biasis. Biochem. Cell. Biol., 90: 425–34, 2012.
102. Leon-Sicairos, N., et al., Oral lactoferrin treatment resolves amoebic intracecal infection in C3H/HeJ
mice. Biochem. Cell. Biol., 90: 435–41, 2012.
103. Stanley, S.L., Jr., Vaccines for amoebiasis: barriers and opportunities. Parasitology, 133 Suppl 2: S81–6,
2006.
104. Quach, J., St-Pierre, J., and Chadee, K., The future for vaccine development against Entamoeba histo-
lytica. Hum. Vaccines Immunother., 10: 1514–21, 2014.
105. Lamm, M.E., Interaction of antigens and antibodies at mucosal surfaces. Annu. Rev. Microbiol., 51:
311–40, 1997.
106. Abd-Alla, M.D., et al., Mucosal immunity to asymptomatic Entamoeba histolytica and Entamoeba
dispar infection is associated with a peak intestinal anti-lectin immunoglobulin A antibody response.
Infect. Immun., 74: 3897–903, 2006.
Entamoeba histolytica 633
107. Haque, R., et al., Innate and acquired resistance to amebiasis in Bangladeshi children. J. Infect. Dis.,
186: 547–52, 2002.
108. Sanchez-Guillen Mdel, C., et al., Differentiation of Entamoeba histolytica/Entamoeba dispar by PCR
and their correlation with humoral and cellular immunity in individuals with clinical variants of amoe-
biasis. Am. J. Trop. Med. Hyg., 66: 731–7, 2002.
109. Haque, R., et al., Amebiasis and mucosal IgA antibody against the Entamoeba histolytica adherence
lectin in Bangladeshi children. J. Infect. Dis., 183: 1787–93, 2001.
110. Ravdin, J.I., et al., Intestinal antilectin immunoglobulin A antibody response and immunity to
Entamoeba dispar infection following cure of amebic liver abscess. Infect. Immun., 71: 6899–905,
2003.
111. Blessmann, J., et al., Epidemiology of amebiasis in a region of high incidence of amebic liver abscess in
central Vietnam. Am. J. Trop. Med. Hyg., 66: 578–83, 2002.
112. Jackson, T.F., Anderson, C.B., and Simjee, A.E., Serological differentiation between past and present
infection in hepatic amoebiasis. Trans. R. Soc. Trop. Med. Hyg., 78: 342–5, 1984.
113. Jain, P., Sawhney, S., and Vinayak, V.K., Experimental amoebic infection in guinea-pigs immunized
with low grade amoebic infection. Trans. R. Soc. Trop. Med. Hyg., 74: 347–50, 1980.
114. Bujanover, S., et al., A virulence attenuated amoebapore-less mutant of Entamoeba histolytica and its
interaction with host cells. Int. J. Parasitol., 33: 1655–63, 2003.
115. Petri, W.A., Jr., et al., Subunit structure of the galactose and N-acetyl-d-galactosamine-inhibitable
adherence lectin of Entamoeba histolytica. J. Biol. Chem., 264: 3007–12, 1989.
116. Petri, W.A., Jr. and Ravdin, J.I., Protection of gerbils from amebic liver abscess by immunization with
the galactose-specific adherence lectin of Entamoeba histolytica. Infect. Immun., 59: 97–101, 1991.
117. Bruchhaus, I., Richter, S., and Tannich, E., Removal of hydrogen peroxide by the 29 kDa protein of
Entamoeba histolytica. Biochem. J., 326 (Pt 3): 785–9, 1997.
118. Soong, C.J., et al., Protection of gerbils from amebic liver abscess by immunization with recombinant
Entamoeba histolytica 29-kilodalton antigen. Infect. Immun., 63: 472–7, 1995.
119. Carrero, J.C., et al., Protection against murine intestinal amoebiasis induced by oral immunization with
the 29 kDa antigen of Entamoeba histolytica and cholera toxin. Exp. Parasitol., 126: 359–65, 2010.
120. Gaucher, D. and Chadee, K., Construction and immunogenicity of a codon-optimized Entamoeba his-
tolytica Gal-lectin-based DNA vaccine. Vaccine, 20: 3244–53, 2002.
121. Madriz, X., et al., Expression in fibroblasts and in live animals of Entamoeba histolytica polypeptides
EhCP112 and EhADH112. Microbiology, 150: 1251–60, 2004.
122. Kaur, U., et al., Immunogenicity and protective efficacy of heparan sulphate binding proteins of
Entamoeba histolytica in a guinea pig model of intestinal amoebiasis. Exp. Parasitol., 135: 486–96,
2013.
123. Gonzalez-Vazquez, M.C., et al., Obtaining of three recombinant antigens of Entamoeba histolytica and
evaluation of their immunogenic ability without adjuvant in a hamster model of immunoprotection.
Acta. Trop., 122: 169–76, 2012.
124. Abd Alla, M.D., et al., Efficacy of a Gal-lectin subunit vaccine against experimental Entamoeba histo-
lytica infection and colitis in baboons (Papio sp.). Vaccine, 30: 3068–75, 2012.
125. Ghosh, P.K., Mancilla, R., and Ortiz-Ortiz, L., Intestinal amebiasis: histopathologic features in experi-
mentally infected mice. Arch. Med. Res., 25: 297–302, 1994.
40
Giardia lamblia
CONTENTS
40.1 Introduction................................................................................................................................. 635
40.2 History of G. lamblia.................................................................................................................. 635
40.3 Life Cycle of G. lamblia and Diagnosis..................................................................................... 636
40.4 Morphology of G. lamblia.......................................................................................................... 636
40.5 Epidemiology.............................................................................................................................. 639
40.6 Encystation.................................................................................................................................. 640
40.7 Excystation.................................................................................................................................. 640
40.8 Adhesion..................................................................................................................................... 640
40.9 Pathology..................................................................................................................................... 641
40.10 Immunology................................................................................................................................ 643
40.11 Antigenic Variation in G. lamblia.............................................................................................. 645
40.12 Giardia and Arginine................................................................................................................. 645
40.13 Drug Resistance in Giardia........................................................................................................ 646
40.14 Conclusion................................................................................................................................... 647
References............................................................................................................................................... 647
40.1 Introduction
The protozoan parasite Giardia lamblia (Protista, Diplomonadida, Hexamitidae, syn. G. intestinalis,
G. lamblia) is the etiologic agent of giardiasis, a common diarrheal disease worldwide. G. lamblia can
infect humans and many other mammals, with prevalence rates in humans ranging from 2% to 7% in
developed countries to 20%–30% in developing countries and 100% prevalence reported in some popu-
lations.1,2 Human outbreaks of G. lamblia are most often associated with fecal contamination of drinking
water. However, several foodborne outbreaks have been documented.3 Until recently, giardiasis was not
the subject of significant scientific investigation, but after its inclusion in the Neglected Disease Initiative
by WHO in 2004 and its reemergence in developed countries, the landscape has changed significantly.4–6
In a bibliometric review covering all studies on giardiasis published between 1971 and 2010, Escobedo
et al.7 reported that especially in the last decade, there has been an increased number of publications and
clinical research studies involving giardiasis. Comparative studies, clinical trials, and pharmacotherapy
assessment were the main identified research areas. This chapter will discuss several aspects of the biol-
ogy of Giardia and the pathology associated with infection while highlighting the laboratory models
currently used to study this parasite.
40.2 History of G. lamblia
Historically, G. lamblia was so common that it was once considered a commensal organism of the
human intestine. G. lamblia was first discovered in 1681 by Antony van Leeuwenhoek during an exami-
nation of his own stool.5 Almost 200 years later in 1859, the protozoan was given the name Cercomonas
635
636 Laboratory Models for Foodborne Infections
intestinalis by Lambl and then later renamed Giardia lamblia by Stiles in 1915 in honor of Professor
A. Giard of Paris and Dr F. Lambl of Prague, who both described the parasite.8 The first association
between G. lamblia and the disease was found in 1954, when it was demonstrated that the ingestion of
G. lamblia cysts by humans could cause diarrheal disease even after ingestion of as few as 10 cysts.9 It
was not until 1981, 300 years after its first description, that G. lamblia was added to the WHO’s list of
parasitic pathogens.8 Soon after its addition to the WHO’s list of parasitic pathogens, Koch’s postulates
were fulfilled, confirming G. lamblia as a pathogen of humans.10
40.4 Morphology of G. lamblia
The two forms of the parasite, the trophozoite and the cyst, are highly specialized for survival
within the host and within the environment, respectively. Therefore, by studying the morphology
of G. lamblia, we can better understand both infection and transmission of the parasite. Most of
what we know about G. lamblia morphology is drawn from studies utilizing electron microscopy
to visualize the surface, cytoskeletal, and intracellular components of the parasite. More gross fea-
tures of the parasite can easily be visualized using light microscopy. These studies rely on in vitro
culture techniques, which allow for the production of both trophozoites and cysts outside of a host.
The trophozoites of G. lamblia can be grown in an axenic medium originally created for cultivation
of Entamoeba histolytica.17 Axenization of new strains can be somewhat problematic. Protocols for
excystation of cysts in vitro (Sections 40.6 and 40.7) can be used to initiate cultures, although a more
common procedure is to collect trophozoites from the small intestines of infected animals.18 Addition
of antibiotics to culture media then helps propagation of newly axenized strains.19 Cyst production is
more complicated, but several protocols exist (Table 40.1).
Giardia lamblia 637
1
i = Infective stage i
d
d = Diagnostic stage d Cyst
3 4 5
Cyst Trophozoites
FIGURE 40.1 Life cycle of Giardia. Transition from noninfectious, rapidly multiplying trophozoites to infectious, envi-
ronmentally hardy cysts. (Courtesy of US CDC.)
TABLE 40.1
Methods Used for Encystation of Giardia lamblia In Vitro
Pre-encystation Medium Encystation Medium Timing References
TYI-S33 without bile TYI-S33 + 10 mM lactic acid, pH 7.8 72 h pre-encystation 52,54
and 10 mg/mL porcine bile 48 h encystation
None TYI-S33 + 12.5 mg/mL bovine bile, no 72 h encystation 55
lactic acid
None TYI-S33 with delipidated serum 72 h encystation 57
None TYI-S33, pH 7.8 + 10 mg/mL bovine bile 96 h encystation 165
638 Laboratory Models for Foodborne Infections
(B)
500 nm
(A) VD AF
VD
VLF
VF
CF
PF (C)
N
500 nm
Ax
FIGURE 40.2 Electron micrographs of Giardia lamblia trophozoites. (A) Scanning electron micrograph of the ven-
tral surface showing the ventral disc (VD), anterior (AF), ventral (VF), caudal (CF), and posterior (PF) flagellar pairs.
(B and C) Transmission electron micrographs showing the ventrolateral flange (VLF), ventral disc (VD), flagellar
axonemes (Ax), and nuclei (N).
Trophozoites of G. lamblia have a distinctive pear shape and exhibit bilateral symmetry. Each cell is
12–15 μm in length and 5–9 μm in width with a flattened ventral surface, a rounded dorsal surface, and
four pairs of flagella: anterior, posterior, caudal, and ventral (Figure 40.2). A major component of the
G. lamblia cytoskeleton is the ventral disc. It is a concave structure that covers the ventral surface of
the trophozoite and is thought to play a major role in attachment of the parasite to the intestinal mucosa.
Another prominent feature of the trophozoite cytoskeleton is the median body, which serves as the
microtubule-organizing center of the cell. Differences in appearance of the median body contribute to
distinction among the species of Giardia. Other morphological features of the trophozoite include the
lateral crest and ventrolateral flange, which are structures with unknown function, although believed to
be participating in adhesion.20–22
Like other eukaryotic cells, Giardia also contain numerous cytoplasmic organelles. In the trophozo-
ites, mitosomes, peripheral vesicles, and ribosome granules can be visualized.23 Interestingly, while a
Golgi complex is easily visualized during the process of encystation, vegetatively growing trophozoites
contain only vesicular structures suggestive of a Golgi complex.24–26 Unlike most other eukaryotic cells,
trophozoites have two nuclei that are localized anteriorly and are symmetric across the longitudinal axis
of the cell. During encystation, the nuclei replicate to produce mature cysts with four nuclei. Following
excystation, each cyst gives rise to two trophozoites.
The transformation of trophozoites of G. lamblia into cysts occurs in response to physiological
stimuli, leading to a process characterized by decreased cell metabolic rate, internalization of fla-
gella, cytoplasmic condensation, peripheral vesicle formation, and production of a cystic membrane.22,27
Encystation is fundamental for survival, transmission, and pathogenesis of G. lamblia. It is consid-
ered an important virulence factor because it allows G. lamblia to survive outside the host and to be
transmitted.23,28
Cysts are ellipsoid or oval and have a size of 7–12 μm. They have a refractive cyst wall, which
is 0.3–0.5 μm thick and is composed of an outer layer and an inner filamentous membranous layer.
Median bodies and structural elements of flagella, the axonemes, and four nuclei in mature and infec-
tious cysts are found in the cytoplasm.29,30 The cyst wall is composed mainly of N-acetylgalactosamine
(GalNAc) homopolymers.31 The precursor for the GalNAc homopolymer is UDP-GalNAc, an amino
sugar phosphate, which is produced from glucose. Enzymes required for the synthesis of UDP-GalNAc
include glucosamine-6-phosphate isomerase, phosphoacetyl-glucosamine mutase, UDP-GlcNAc
49-epimerase, and glucosamine-6-phosphate N-acetylase.32 During encystation, transcript levels and
Giardia lamblia 639
enzyme activity for some of these enzymes increase significantly.33 The external portion of the cyst
wall is covered by a network of filaments, 7–20 nm in diameter, composed primarily of GalNAc homo-
polymer, along with cyst wall proteins. Considering that this is the stable life cycle stage, the cysts
have a metabolic rate of only 10%–20% relative to trophozoites.34 Some cysts are resistant to chlorina-
tion and radiation. Boiling is the most efficient way to eliminate cysts; freezing requires several days
to reduce cyst viability.35–37
40.5 Epidemiology
Within the genus Giardia, there are currently six recognized species:38 G. agilis (a parasite of amphib-
ians), G. ardeae and G. psittaci (both of which are found in birds), G. microti (found in voles and
muskrats), G. muris (found in rodents), and G. lamblia (which infects a wide range of mammalian spe-
cies including humans). The aforementioned taxonomic divisions are based on morphological differ-
ences that have been historically used to define the six Giardia species.38 More recently, G. lamblia,
the only species known to infect humans, has been divided into assemblages A–H based on molecular
evidence.5 Assemblages A and B are known to infect a wide range of mammals including humans,
but assemblages C–G appear to have some degree of host specificity.39 Assemblages C and D are most
commonly found in dogs, assemblage E in hoofed animals, assemblage F in cats, and assemblage G in
rats.40 The most recent addition, assemblage H, was identified in marine animals.39 The classification
of these assemblages is supported by studies using both enzyme electrophoretic data and phylogenetic
analyses of molecular sequence data.5
There are striking genetic differences between the two assemblages, assemblage A and assem-
blage B, associated with human infections. Whole genome sequencing of two strains of G. lamblia
isolated from humans, WB (assemblage A strain) and GS (assemblage B strain), revealed that these
two assemblages share only 77% nucleotide identity and 78% amino acid identity in protein-coding
regions.41 Because these two assemblages are so genetically different, it is tempting to speculate that
they should contribute to different geographical distributions or clinical outcomes in humans. However,
data to support these geographical and clinical differences are often conflicting. Reports from Asia,
Europe, Africa, and the Americas all demonstrate that each assemblage is found in pockets through-
out these areas.42 Symptomatic differences attributable to assemblage follow a similar trend. While
assemblage A is found to account for more symptomatic infection in some studies, assemblage B is
found to have a greater association with symptoms in others, with some studies reporting no associa-
tion between patient symptoms and assemblage causing the infection.42 In a laboratory infection model
using adult mice, it was demonstrated that while the GS (assemblage B) strain caused measurable
disaccharidase deficiency, infections with the WB (assemblage A) strain did not.43 Similarly, another
assemblage B strain, H3, caused chronic infections that contributed to enhanced weight loss in mice on
a protein-deficient diet, while the assemblage A strain, WB, did not.44 The use of antibiotics to allow
the WB strain to infect mice in the former paper43 and the use of H3 cysts rather than WB trophozoites
to initiate infections in the latter paper44 require caution in interpreting these results. Nevertheless, they
suggest that mouse models may be useful for identifying strain- and assemblage-specific differences
in virulence. Better cohort studies in humans could also help to distinguish the potential differences in
pathogenicity of these two assemblages.
Because G. lamblia infects such a diverse range of mammals, it is conceivable that zoonotic trans-
mission could occur. However, the zoonotic risk of G. lamblia infection is unclear. Some studies have
found an increased risk for human infection in households that harbor domestic pets.45,46 Molecular
evidence has linked human infections to assemblages found only in cattle, cats, and dogs, indicating that
cross-species transmission can occur.47–50 A large-scale European study found that only 1% of human
G. lamblia infection could be attributed to assemblages C, D, E, and F, indicating that the zoonotic
potential of G. lamblia is low for these assemblages.51 Interestingly, this same study demonstrated that
while assemblage B infections are mostly restricted to humans, assemblage A appears quite frequently
in companion animals, wildlife, and livestock. So, while humans may serve as the main source of assem-
blage B, assemblage A could have a greater zoonotic potential.
640 Laboratory Models for Foodborne Infections
40.6 Encystation
Encystation is the process of differentiation of the trophozoite into the resistant cyst, which occurs due to
external stimuli. During encystation, the main transformations that occur are internalization of flagella
creating an oval shape, formation of encystation-specific vesicles (ESVs), the appearance of a refractory
cyst wall, and replication of the nuclei. The cell also has diminished adherence capacity owing to frag-
mentation of the ventral disc.22 Much of the work on G. lamblia encystation has been done by in vitro
manipulation of axenically cultured trophozoites. By altering the culture conditions of trophozoites, it is
possible to mimic some of the signals a cell might encounter in the host small intestine that are neces-
sary to drive encystation (Table 40.1). In vitro studies have shown that encystation begins when important
modifications of the environment occur such as shifts in pH, concentrations of bile salts, availability of
lipids, and production of lactic acid by intestinal bacteria.52 In a suckling mouse model of G. lamblia
infection, cysts were found in the mid to lower portion of the jejunum, while later the majority of cysts
were in the large intestine and cecum, suggesting that bile concentration may be an important component
of the encystation process.53 It is thought that bile stimulates encystation by sequestering cholesterol.
Common methods used to achieve encystation are based on parasite growth in conventional culture
media without bile, and after 2–3 days, the parasites are switched to a culture media containing a high
concentration of porcine bile with lactic acid and a pH of 7.8.52,54,55 Using these methods, trophozoites
can be induced to transform to viable cysts. However, the ability of in vitro-derived cysts to excyst, either
in animals or in culture, is often poor.
G. lamblia has little capacity for synthesizing lipid molecules de novo and instead depends on envi-
ronmental sources. It has been hypothesized that most of the lipid uptake by the cells is then used for
energy production and biosynthesis of organelles.56 Because lipid metabolism is central to the process
of encystation, these pathways are of interest as a potential drug target.52,57 Recently, attention has been
focused on glycosylceramide pathways in encystation.58 Conversely, specific lipid products from the host
have also been shown to kill trophozoites.59,60
The role of ESVs in the encystation process and their relationship with the normal functions of the Golgi
complex have been the subject of significant interest. Stefanic et al.61 used two-dimensional electrophore-
sis to analyze Golgi-like vesicles isolated from encysting G. lamblia. These vesicles are highly dynamic
spaces that act as hosts of the cyst wall materials and increase only during the induction of encystation. The
authors have shown that the vesicles do not have sorting functions characteristic of mature Golgi, but do
retain protein quality control functions. They also suggested that the ESVs can be involved in anterograde
and retrograde trafficking with the endoplasmic reticulum, similar to the function of the Golgi.61
40.7 Excystation
In vivo, excystation starts following ingestion by the host when the cysts are exposed to the acidic pH
of the stomach. Mobile parasites emerge from within the cyst wall in the host small intestine, but at this
stage, the ventral disc is not fully reorganized. Soon after emergence, the cell divides rapidly generating
two trophozoites capable of adhering to intestinal cells by the ventral disc, which is now fully reorga-
nized.62 Parasite viability requires that cysts produce trophozoites that can emerge from the cyst wall,
perform cytokinesis, and adhere to the intestinal epithelium of a newly infected host. Several protocols
for excystation have been published (e.g.,54,63–65), but the success of the excystation process may vary
depending on whether the source of the cysts is feces or in vitro generation.
40.8 Adhesion
The ability of the parasite to adhere to the intestinal epithelium is an essential factor in its ability to
reproduce and cause disease. In vivo, Giardia trophozoites can adhere to the surface of the small intes-
tine and in vitro to surfaces like glass and polymers.66 Understanding the mechanisms that mediate
Giardia lamblia 641
parasite adhesion can explain how Giardia cells can attach to many surfaces as well as the intestinal
wall of many host animals.67 Several approaches have been developed to evaluate Giardia cell adhesion.
These include adhesion of trophozoites to different glass surfaces, scanning electron microscopy to ana-
lyze attachment, and a microarray-based approach.67–69
Many studies have been published in an attempt to elucidate the adhesion mechanisms of Giardia
cells. The structure responsible for the attachment is the ventral disc, a spiral array of microtubules
associated with microribbons and found just below the plasma membrane in the anterior and ventral por-
tion of the trophozoite.70 Several auxiliary proteins are found associated with the ventral disc including
β-giardin, γ-giardin, δ-giardin, and SALP-1. The periphery of the disc has contractile proteins including
actin, myosin, and tropomyosin, which may serve as the biochemical basis for contraction of the disc, a
fundamental process in parasite adhesion. Hagen et al. used a proteomic approach to detect more than
100 protein candidates associated with the ventral disc as well as the first proteins localized to the lateral
flange.71 Because adhesion is central to parasite viability and these structures are unique to the parasite,
strategies targeting these molecules could be developed as novel therapeutics.
40.9 Pathology
Studies of the pathology associated with giardiasis often use animal models that aim to recapitulate
human infection conditions. Several animal models for studying G. lamblia exist including mouse, ger-
bil, hamster, rat, and recently, nematodes (Table 40.2). Infection with G. muris is a common model of
human giardiasis because the host specificity of this species in mice and rats can be useful for simpli-
fying infection protocols. Both human assemblages of G. lamblia are commonly used in experimental
giardiasis, which can be useful for understanding assemblage-level differences in the human host. In
vitro models are also used to study G. lamblia pathology by exposing human intestinal cell lines to
live parasites or their byproducts. There is a bias in the literature for mouse infections performed with
TABLE 40.2
Common Models Used for Studies of Giardiasis
Model Advantages Disadvantages
Axenic culture of • Rapid • Limited applicability in vivo
trophozoites • Inexpensive
Coculture of Giardia with • More rapid than in vivo • Limited applicability in vivo
epithelial cell lines • Can test hypotheses without
using animals
• Easy to analyze interactions
Adult mouse or rat • Robust infections • Limited applicability to human infection
infection with G. muris • Natural infection cycle • G. muris cannot be cultured axenically—cyst
stocks may have fecal contamination
Neonatal mouse infection • Can use human relevant strains • Limited immunity in neonatal mice
with G. lamblia • Can study more complex
host–parasite interactions
Adult mouse infection • Abundant reagents for analysis • Not all findings translate to human infections
with G. lamblia of immunity
• Many molecular tools available • Infections have little overt pathologya
Gerbil infection • Highly susceptible to infection • Fewer reagents and mutant strains available
and development of symptoms
Hamster infection • Highly susceptible to infection • Fewer reagents and mutant strains available
and development of symptoms
C. elegans • Fewer ethical considerations • Limited applicability to human infection
compared to vertebrates
a This is actually similar to most human infections, however.
642 Laboratory Models for Foodborne Infections
assemblage B strains (particularly GS) and for in vitro experiments performed with assemblage A strains
(e.g., Portland-1 and WB).
There are several pathological mechanisms thought to contribute to the diarrheal disease caused by
G. lamblia infection. These include epithelial barrier breakdown, defects in the epithelial brush border,
and increased intestinal motility.1,2 Changes in epithelial barrier function have been linked to both tight
junction proteins and epithelial apoptosis in epithelial cell lines and in human infections.72–74 Parasite
strain could be important in determining the severity of this pathology. When the human epithelial
cell line HCT-8 was exposed to different assemblages of G. lamblia, the assemblage was seen to influ-
ence both epithelial barrier integrity and apoptosis.73 Similarly, Chin et al. found that some strains of
G. lamblia were superior to others in inducing apoptosis in the SCBN cell line (reported at the time to be
a human small intestinal cell, but later genotyped as being of canine origin).75 Hardin et al. first showed
increased macromolecular uptake in Giardia-infected gerbils.76 Zhou et al. showed that transepithelial
electrical resistance of mucosa isolated from infected mice was reduced compared to mucosa from non-
infected animals, and Chen et al. showed epithelial barrier breakdown and translocation of commensal
bacteria in infected mice.77,78 Reductions in transepithelial electrical resistance were first shown in vitro
using cell lines exposed to Giardia, and these changes were further associated with alterations in tight
junction proteins.79 Parasite factors responsible for changes to the intestinal epithelium have not been
identified at the molecular level, although new structures related to adhesion and potential toxins have
been suggested.68,80,81
Giardiasis is also associated with reduced disaccharidase activity in humans, and this has been associ-
ated with shortening of the intestinal microvilli in animal models. This enzyme deficiency can contrib-
ute to diarrhea by creating a gradient that drives water out of the tissue and into the intestinal lumen.
In a mouse infection model using the murine-specific species G. muris, infection and CD8+ T cell
responses were shown to contribute to microvillous shortening and a deficiency in disaccharidases.82,83
This same deficiency in small intestinal disaccharidases was confirmed in mice using human strains of
G. lamblia.43 In the latter study, parasite assemblage was again shown to be an important factor contrib-
uting to intestinal pathology, as disaccharidase deficiency was observed in assemblage-B-infected mice
but not assemblage-A-infected mice.43
G. lamblia infection has been linked with other changes in bowel function in humans and animal
models. Humans infected with Giardia are prone to postinfectious irritable bowel syndrome.1 Gerbils
infected with G. lamblia had increased intestinal transit and hypercontractility of intestinal smooth
muscle.84 Studies using G. muris and G. lamblia infections in adult mice showed that increased
intestinal transit required adaptive immune responses as well as the neuronal isoform of nitric oxide
synthase.85,86 These studies also showed that increased transit helped facilitate elimination of the para-
site. Follow-up studies using the adult mouse model of G. lamblia infection found that altered motility
is due at least in part to increased contractility of intestinal smooth muscle and that the gut hormone
cholecystokinin (CCK) contributes to this phenotype.87 CCK normally acts to induce contraction of
the gall bladder and release of bile into the digestive tract in response to ingestion of fats. Giardia
requires bile for growth and, thus, appears to have evolved a mechanism to induce the host to provide
it with an essential nutrient.
Infection with G. lamblia is also associated with long-term health consequences. These diseases can
manifest well after infection clearance and have a range of severity. G. lamblia has been linked to several
metabolic outcomes including lowered cognitive function, lower weight and height, failure to thrive, and
nutritional deficiencies.1 Furthermore, acute G. lamblia disease severity is associated with nutritional
deficiency in animals as both mice and gerbils on nutrient-limited diets presented with worsened intesti-
nal pathology as compared to non-nutrient-limited controls.44,88 These studies indicate that the relation-
ship between G. lamblia infection and disease could be complex and bidirectional. The long-term health
consequences of G. lamblia infection represent an intriguing area of investigation as these associations
can be strong but mechanisms are lacking. For example, one study of humans with giardiasis found that
one-third of infected individuals presented with extraintestinal manifestations of infections including
symptoms of the eye, skin, joints, and urinary tract.89 G. lamblia has also been linked with postinfec-
tion development of allergies, muscular complications, chronic fatigue syndrome, and irritable bowel
syndrome,1 but good animal models of these long-term sequelae have not been developed.
Giardia lamblia 643
40.10 Immunology
G. lamblia infection leads to pathological alterations of the host’s intestinal epithelia, yet intestinal
inflammation is not often seen during infection.90 This could indicate that the host immune response
is protective against inflammatory effects. Furthermore, several studies demonstrate the host immune
response having a role in driving pathology in the intestine during infection.43,83 Thus, it is important to
understand which elements of the host immune response contribute to pathology versus those which aid
in clearance and protection during G. lamblia infection.
To successfully colonize the small intestine, G. lamblia must contend with the hosts’ natural barriers
to infection. Both intestinal mucus and epithelial cell turnover could serve as obstacles for trophozoite
attachment, and in vitro studies have shown that mucin can inhibit the ability of trophozoites to attach to
a substrate.91 Recent work has also demonstrated that the host microbiome can play an important role in
successful colonization. Infections of mice from different suppliers with G. lamblia showed that intes-
tinal microbiota conferred protection against infection and that this protective effect could be transmit-
ted through cohousing of mice.92 This same study demonstrated that antibiotic treatment could ablate the
protective effect of the host microbiome, demonstrating that intestinal bacteria composition plays a role in
infection susceptibility.92 Whether the influence of the host microbiome on G. lamblia infection is direct or
a result of bacteria-driven immune stimulation remains unclear. Lactobacillus johnsonii culture superna-
tants have been shown in vitro to have a cytostatic effect on G. lamblia.93 This same bacterium was shown
to inhibit G. lamblia infection of gerbils.94 In a different study, mouse infection with Enterococcus faecium
led to immunological stimulations that decreased G. lamblia colonization success in infected mice, demon-
strating that bacteria could alter host immunity to prevent establishment of G. lamblia infection.95
The innate immune system serves as an early line of defense against pathogens as it does not require
the specificity of adaptive immunity to exert its protective effects. The importance of innate immunity
in the control of G. lamblia infection is not well characterized. However, several studies have shown that
innate immune effectors do have a role in G. lamblia infection. Antimicrobial peptides like defensins
and lactoferrin have been shown to kill G. lamblia in vitro.96,97 Another potent antimicrobial, nitric
oxide (NO), has also been shown to have inhibitory effects on G. lamblia growth and survival in an
in vitro model using human intestinal epithelia cells.98 Studies in G. lamblia-infected mice have shown
that while NO produced by inducible NO synthase (NOS2) may not be effective in controlling infection
alone, it may have a redundant role with MMP-7, a protease required for cleavage of defensins.85,99 These
studies demonstrated that while a loss of either NOS2 or MMP-7 did not affect infection clearance, mice
lacking both MMP-7 and NOS2 had a defect in parasite clearance.
Interestingly, it has also been shown that G. lamblia can inhibit innate cellular immune responses.
G. lamblia extracts are able to inhibit the production of proinflammatory cytokines by murine
bone-marrow-derived dendritic cells stimulated with LPS and other innate immune stimuli.100 At the
same time, production of the anti-inflammatory cytokine IL-10 was increased. The importance of
dendritic cells in stimulating anti-Giardia immunity in vivo was shown using cell transfer systems.101
Giardia infection can also modulate macrophage responses in the intestinal lamina propria. We recently
showed that adult mice infected with G. lamblia have an increase in the proportion of macrophages in
the small intestine and that these cells express both NOS2 and arginase (ARG1).102 In addition, these
macrophages express more IL-10 and less TNF following infection, consistent with an anti-inflammatory
phenotype (JM and SMS, unpublished observation). Giardia has also been reported to actively block
neutrophil recruitment by degrading the chemokine IL-8, which was induced in epithelial cells follow-
ing exposure to the parasite in vitro.103 Combined analysis using in vitro and animal models continue to
elucidate interactions between the host and parasite.
Another important role of the innate immune response is to begin to mobilize the adaptive immune
response. Innate immune recognition of a pathogen drives the release of chemical signals that recruit and
activate other immune cells. In vitro studies have shown that several intestinal chemokines are secreted
in response to G. lamblia infection. CCL2, CCL20, and CXCL1-3 are all secreted when Caco-2 cells, a
colon cell line, are stimulated with G. lamblia.104 The in vivo importance of these chemokines remains
to be explored.
644 Laboratory Models for Foodborne Infections
Communication between the parasite and host is bidirectional, and parasites alter their behavior in
response to intestinal epithelial cells. Ringqvist et al. cocultured G. lamblia trophozoites with the human
adenocarcinoma line, CaCo2, and used proteomics to identify three parasite proteins (arginine deimi-
nase, ornithine carbamoyl transferase, and enolase) in culture medium.105 All three proteins were pre-
viously known as antigens recognized by giardiasis patient sera. The authors showed that G. lamblia
contact with epithelial cells triggers release of metabolic enzymes, which would facilitate effective para-
site colonization of the human small intestine.105
Adaptive immunity has been shown to be essential for control of Giardia infection, with both antibod-
ies and T cells having important roles. Studies using a mouse model of G. muris infection have dem-
onstrated that B cells and specifically IgA produced by these cells are essential for parasite control and
elimination.106–109 B cells appear to be less important for resolving G. lamblia infection. In a study using
B-cell- and T-cell-deficient mice, it was demonstrated that while T cells were required for elimination of
G. lamblia infection in mice, B cells were not.110 However, in a study on the role of IL-6 in G. lamblia
infection, IL-6-deficient mice had a defect in parasite clearance until 60 days after infection when para-
site clearance correlated with the production of antibodies that were reactive to a diverse population of
parasites.111 These studies indicate that while host antibody may not be required for G. lamblia clear-
ance, it could play an important role in parasite population control throughout the course of infection.
These results are consistent with studies on the role of antibody in human infection where patients with
antibody deficiency are at only a slightly increased risk for G. lamblia infection.112 Unlike B cells, T cells
seem to be required for the clearance of G. lamblia infection in mice. In fact, CD4 T cells have been
shown to be essential for parasite clearance as mice with specific deficiency in CD4 T cells are unable
to control infections.43,110,113
The role of several T cell cytokines has been analyzed in animal models. IFN-γ has been shown to
be important for parasite clearance during mouse infection with either G. muris or G. lamblia.110,114,115
IFN-γ production and T cell proliferation were also observed in ex vivo stimulations of human lympho-
cytes with G. lamblia, supporting a role for this cytokine in human infections.116 TNF-α has also been
shown to be important in early control of G. lamblia infection as mice with defects in TNF-α production
demonstrated increased parasite burden during infection.77 Recent studies on the inflammatory cytokine
IL-17 have demonstrated that this cytokine is important for clearance of infection in mouse models of
both G. muris and G. lamblia.117,118 The role of IL-6 in Giardia infection has been analyzed in IL-6-
deficient mice. G. lamblia infections of IL-6-deficient mice have demonstrated that this cytokine is
required for infection control and parasite clearance.111,119 As mentioned earlier, these mice did not have
a defect in antibody production, but instead, defects in dendritic cell function may be responsible for the
observed phenotype.101
Intestinal mast cells also have important functions in control of Giardia infection. Mouse infection
models using G. muris and G. lamblia have demonstrated that mast cell deficiencies lead to deficiencies
in infection clearance.120,121 Mast cell hyperplasia has also been observed in Mongolian gerbils infected
with G. muris.76 Furthermore, G. lamblia-infected mast cell-deficient mice produce less IL-6 mRNA
and IgA than their wild-type counterparts.121 Mast-cells-also appear to contribute to intestinal smooth
muscle contractions during G. lamblia infection in mice and could further support a protective role for
these cells in infection.87
One important goal of immunological studies of G. lamblia is the development of a human vaccine.
Currently, no human Giardia vaccine exists; however, a veterinary vaccine, GiardiaVax, has been used
in a variety of domestic mammals. Studies in humans have found that previous Giardia infection could
convey some level of protection.122,123 This protection indicates that immunological memory to Giardia is
possible in humans and supports the development of a human vaccine. Recently, several studies in mice
have been done with the goal of better understanding how to develop a human vaccine. A reinfection
model in mice with G. lamblia was able to recapitulate the protection observed in humans, indicating
that mouse models could serve as a useful tool for human vaccine development.124 In a mouse model of
G. lamblia infection, a vaccine against parasite cyst wall protein was able to reduce cyst shedding.125–127
In another mouse vaccine study, vaccination against a G. lamblia protein, α1-giardin, was able to confer
protection against infection.128 This protein is highly conserved between both human assemblages of
G. lamblia, indicating it could provide coverage against multiple strains of the parasite.128 The ability
Giardia lamblia 645
of Giardia to undergo antigenic variation of the variant-specific surface proteins (VSPs) (discussed in
Section 40.11) may be a major impediment to the design of a successful vaccine. Rivero et al. used para-
sites engineered to express multiple VSPs simultaneously as a vaccine in gerbils and achieved significant
levels of protection.129 Combinations of approaches may be necessary to generate an effective vaccine
for humans and/or animals.
NO production in vivo is unclear. Recent studies indicate that host-produced NO may be redundant with
α-defensins as only a loss of the expression of enzymes required for the production of both influenced
infection outcomes in mice.85,99 Furthermore, it is likely that G. lamblia influences arginine availability
within the host indirectly. Stimulation of a mouse macrophage cell line with parasite lysate leads to
increased expression of arginase 1 (ARG1), a host-produced arginine-consuming enzyme.102 This same
study found that small intestinal tissue from infected mice also showed increased ARG1 expression
compared to uninfected controls.102
The consumption of arginine through parasite ADI is advantageous for the G. lamblia in potentially
two ways. First, it serves as a means of energy production through conversion of arginine to ATP, and
second, it provides a means by which Giardia might evade the host immune response by limiting NO
production. Limiting arginine availability in the gut could also affect the host immune system in other
ways. For example, it is possible that T cell function is altered as arginine is an important modulator of
T cell proliferation. Arginine depletion could result in the limitation of T cell proliferation and may also
cause an impairment of CD3ζ chain turnover of the T cell receptor after antigen stimulation, inhibiting
signaling through the T cell receptor complex.149 Both of these scenarios would result in limiting the
T cell response during Giardia infection, providing an additional benefit for the parasite through argi-
nine depletion. Thus, giardial ADI or host ARG1 may directly interfere with the host’s ability to gener-
ate a strong, protective cellular immune response against the parasite while limiting the innate immune
response by inhibiting NO production.
40.14 Conclusion
The relationship between G. lamblia and its host is complex and multifactorial with host immunity,
intestinal microbial composition, nutritional status, and parasite strain all having potential roles in host
susceptibility and infection severity. As a disease of the developing world, many of these aspects of
infection remain understudied and poorly understood. Studies that aim to illuminate these relationships
will provide information that could lead to new strategies for combating parasitic disease. These types
of investigations could yield not only better treatments but also better control of a disease with a global
distribution that infects millions of humans and other animals.
REFERENCES
1. Halliez, M.C. & Buret, A.G. Extra-intestinal and long term consequences of Giardia duodenalis infec-
tions. World J Gastroenterol 19, 8974–85 (2013).
2. Solaymani-Mohammadi, S. & Singer, S.M. Giardia duodenalis: the double-edged sword of immune
responses in giardiasis. Exp Parasitol 126, 292–7 (2010).
3. Dawson, D. Foodborne protozoan parasites. Int J Food Microbiol 103, 207–27 (2005).
4. Savioli, L., Smith, H. & Thompson, A. Giardia and Cryptosporidium join the “Neglected Diseases
Initiative.” Trends Parasitol 22, 203–8 (2006).
5. Monis, P.T., Caccio, S.M. & Thompson, R.C. Variation in Giardia: towards a taxonomic revision of the
genus. Trends Parasitol 25, 93–100 (2009).
6. Thompson, R.C. & Monis, P.T. Variation in Giardia: implications for taxonomy and epidemiology. Adv
Parasitol 58, 69–137 (2004).
7. Escobedo, A.A. et al. A bibliometric study of international scientific productivity in giardiasis covering
the period 1971–2010. J Infect Dev Ctries 9, 76–86 (2015).
8. Lipoldová, M. Giardia and Vilém Dušan Lambl. PLoS Negl Trop Dis 8, e2686 (2014).
9. Rendtorff, R.C. & Holt, C.J. The experimental transmission of human intestinal protozoan parasites. IV.
Attempts to transmit Endamoeba coli and Giardia lamblia cysts by water. Am J Hyg 60, 327–38 (1954).
10. Nash, T.E., Herrington, D.A., Losonsky, G.A. & Levine, M.M. Experimental human infections with
Giardia lamblia. J Infect Dis 156, 974–84 (1987).
11. Robertson, L.J. & Lim, Y.A.L. Waterborne and environmentally-borne giardiasis. in Giardia: A Model
Organism (eds. Lujan, H.D. & Svard, S.) 29–69 (Springer-Verlag, Vienna, 2011).
12. Guy, R.A., Payment, P., Krull, U.J. & Horgen, P.A. Real-time PCR for quantification of Giardia and
Cryptosporidium in environmental water samples and sewage. Appl Environ Microbiol 69, 5178–85
(2003).
13. Upjohn, M. et al. Prevalence, molecular typing and risk factor analysis for Giardia duodenalis infec-
tions in dogs in a central London rescue shelter. Vet Parasitol 172, 341–6 (2010).
14. Berkman, D.S., Lescano, A.G., Gilman, R.H., Lopez, S.L. & Black, M.M. Effects of stunting, diarrhoeal
disease, and parasitic infection during infancy on cognition in late childhood: a follow-up study. Lancet
359, 564–71 (2002).
15. Nematian, J., Gholamrezanezhad, A. & Nematian, E. Giardiasis and other intestinal parasitic infections
in relation to anthropometric indicators of malnutrition: a large, population-based survey of schoolchil-
dren in Tehran. Ann Trop Med Parasitol 102, 209–14 (2008).
16. Gardner, T.B. & Hill, D.R. Treatment of giardiasis. Clin Microbiol Rev 14, 114–28 (2001).
17. Keister, D.B. Axenic culture of Giardia lamblia in TYI-S-33 medium supplemented with bile. Trans R
Soc Trop Med Hyg 77, 487–8 (1983).
18. Meloni, B.P. & Thompson, R.C. Comparative studies on the axenic in vitro cultivation of Giardia of
human and canine origin: evidence for intraspecific variation. Trans R Soc Trop Med Hyg 81, 637–40
(1987).
19. Kulakova, L. et al. Discovery of novel antigiardiasis drug candidates. Antimicrob Agents Chemother 58,
7303–11 (2014).
20. Benchimol, M. The nuclei of Giardia lamblia—new ultrastructural observations. Arch Microbiol 183,
160–8 (2005).
648 Laboratory Models for Foodborne Infections
21. Piva, B. & Benchimol, M. The median body of Giardia lamblia: an ultrastructural study. Biol Cell 96,
735–46 (2004).
22. Midlej, V. & Benchimol, M. Giardia lamblia behavior during encystment: how morphological changes
in shape occur. Parasitol Int 58, 72–80 (2009).
23. Adam, R.D. Biology of Giardia lamblia. Clin Microbiol Rev 14, 447–75 (2001).
24. Gillin, F.D., Reiner, D.S. & McCaffery, J.M. Cell biology of the primitive eukaryote Giardia lamblia.
Annu Rev Microbiol 50, 679–705 (1996).
25. Lanfredi-Rangel, A., Kattenbach, W.M., Diniz, J.A., Jr. & de Souza, W. Trophozoites of Giardia lamblia
may have a Golgi-like structure. FEMS Microbiol Lett 181, 245–51 (1999).
26. Soltys, B.J., Falah, M. & Gupta, R.S. Identification of endoplasmic reticulum in the primitive eukaryote
Giardia lamblia using cryoelectron microscopy and antibody to Bip. J Cell Sci 109 (Pt 7), 1909–17
(1996).
27. Sulemana, A., Paget, T.A. & Jarroll, E.L. Commitment to cyst formation in Giardia. Microbiology 160,
330–9 (2014).
28. Svard, S.G., Hagblom, P. & Palm, J.E. Giardia lamblia—a model organism for eukaryotic cell differen-
tiation. FEMS Microbiol Lett 218, 3–7 (2003).
29. Adam, R.D. The biology of Giardia spp. Microbiol Rev 55, 706–32 (1991).
30. Carpenter, M.L., Assaf, Z.J., Gourguechon, S. & Cande, W.Z. Nuclear inheritance and genetic exchange
without meiosis in the binucleate parasite Giardia intestinalis. J Cell Sci 125, 2523–32 (2012).
31. Jarroll, E.L., Manning, P., Lindmark, D.G., Coggins, J.R. & Erlandsen, S.L. Giardia cyst wall-specific
carbohydrate: evidence for the presence of galactosamine. Mol Biochem Parasitol 32, 121–31 (1989).
32. Macechko, P.T., Steimle, P.A., Lindmark, D.G., Erlandsen, S.L. & Jarroll, E.L. Galactosamine-
synthesizing enzymes are induced when Giardia encyst. Mol Biochem Parasitol 56, 301–9 (1992).
33. Lopez, A.B., Sener, K., Jarroll, E.L. & van Keulen, H. Transcription regulation is demonstrated for five
key enzymes in Giardia intestinalis cyst wall polysaccharide biosynthesis. Mol Biochem Parasitol 128,
51–7 (2003).
34. Paget, T.A., Jarroll, E.L., Manning, P., Lindmark, D.G. & Lloyd, D. Respiration in the cysts and tropho-
zoites of Giardia muris. J Gen Microbiol 135, 145–54 (1989).
35. Kasprzak, W. & Majewska, A.C. Infectivity of Giardia sp. cysts in relation to eosin exclusion and excys-
tation in vitro. Tropenmed Parasitol 34, 70–2 (1983).
36. Ortega, Y.R. & Adam, R.D. Giardia: overview and update. Clin Infect Dis 25, 545–9; quiz 550
(1997).
37. Wolfe, M.S. Giardiasis. Clin Microbiol Rev 5, 93–100 (1992).
38. Ballweber, L.R., Xiao, L., Bowman, D.D., Kahn, G. & Cama, V.A. Giardiasis in dogs and cats: update
on epidemiology and public health significance. Trends Parasitol 26, 180–9 (2010).
39. Lasek-Nesselquist, E., Welch, D.M. & Sogin, M.L. The identification of a new Giardia duodenalis
assemblage in marine vertebrates and a preliminary analysis of G. duodenalis population biology in
marine systems. Int J Parasitol 40, 1063–74 (2010).
40. Caccio, S.M. & Ryan, U. Molecular epidemiology of giardiasis. Mol Biochem Parasitol 160, 75–80
(2008).
41. Franzen, O. et al. Draft genome sequencing of Giardia intestinalis assemblage B isolate GS: is human
giardiasis caused by two different species? PLoS Pathogens 5, e1000560 (2009).
42. Laishram, S., Kang, G. & Ajjampur, S.S. Giardiasis: a review on assemblage distribution and epidemiol-
ogy in India. Indian J Gastroenterol 31, 3–12 (2012).
43. Solaymani-Mohammadi, S. & Singer, S.M. Host immunity and pathogen strain contribute to intestinal
disaccharidase impairment following gut infection. J Immunol 187, 3769–75 (2011).
44. Bartelt, L.A. et al. Persistent G. lamblia impairs growth in a murine malnutrition model. J Clin Invest
123, 2672–84 (2013).
45. Traub, R.J. et al. Epidemiological and molecular evidence supports the zoonotic transmission of Giardia
among humans and dogs living in the same community. Parasitology 128, 253–62 (2004).
46. Sackey, M.E., Weigel, M.M. & Armijos, R.X. Predictors and nutritional consequences of intestinal
parasitic infections in rural Ecuadorian children. J Trop Pediatr 49, 17–23 (2003).
47. Souza, S.L. et al. Molecular identification of Giardia duodenalis isolates from humans, dogs, cats and
cattle from the state of Sao Paulo, Brazil, by sequence analysis of fragments of glutamate dehydrogenase
(gdh) coding gene. Vet Parasitol 149, 258–64 (2007).
Giardia lamblia 649
48. Thompson, R.C. The zoonotic significance and molecular epidemiology of Giardia and giardiasis. Vet
Parasitol 126, 15–35 (2004).
49. Lalle, M., Jimenez-Cardosa, E., Caccio, S.M. & Pozio, E. Genotyping of Giardia duodenalis from
humans and dogs from Mexico using a β-giardin nested polymerase chain reaction assay. J Parasitol 91,
203–5 (2005).
50. Johnson, D. et al. Serum biochemistry, serology, and parasitology of boreal caribou (Rangifer tarandus
caribou) in the Northwest Territories, Canada. J Wildl Dis 46, 1096–107 (2010).
51. Sprong, H., Caccio, S.M., van der Giessen, J.W., & ZOOPNET network & partners. Identification of
zoonotic genotypes of Giardia duodenalis. PLoS Negl Trop Dis 3, e558 (2009).
52. Gillin, F.D., Boucher, S.E., Rossi, S.S. & Reiner, D.S. Giardia lamblia: the roles of bile, lactic acid, and
pH in the completion of the life cycle in vitro. Exp Parasitol 69, 164–74 (1989).
53. Gillin, F.D. et al. Encystation and expression of cyst antigens by Giardia lamblia in vitro. Science 235,
1040–3 (1987).
54. Boucher, S.E. & Gillin, F.D. Excystation of in vitro-derived Giardia lamblia cysts. Infect Immun 58,
3516–22 (1990).
55. Sun, C.H., McCaffery, J.M., Reiner, D.S. & Gillin, F.D. Mining the Giardia lamblia genome for new
cyst wall proteins. J Biol Chem 278, 21701–8 (2003).
56. Yichoy, M. et al. Lipid metabolism in Giardia: a post-genomic perspective. Parasitology 138, 267–78
(2011).
57. Lujan, H.D., Mowatt, M.R., Byrd, L.G. & Nash, T.E. Cholesterol starvation induces differentiation of
the intestinal parasite Giardia lamblia. Proc Natl Acad Sci USA 93, 7628–33 (1996).
58. Mendez, T.L. et al. Glucosylceramide transferase activity is critical for encystation and viable cyst pro-
duction by an intestinal protozoan, Giardia lamblia. J Biol Chem 288, 16747–60 (2013).
59. Reiner, D.S., Wang, C.S. & Gillin, F.D. Human milk kills Giardia lamblia by generating toxic lipolytic
products. J Infect Dis 154, 825–32 (1986).
60. Das, S. et al. Killing of Giardia lamblia trophozoites by human intestinal fluid in vitro. J Infect Dis 157,
1257–60 (1988).
61. Stefanic, S., Palm, D., Svard, S.G. & Hehl, A.B. Organelle proteomics reveals cargo maturation mecha-
nisms associated with Golgi-like encystation vesicles in the early-diverged protozoan Giardia lamblia.
J Biol Chem 281, 7595–604 (2006).
62. Bernander, R., Palm, J.E. & Svard, S.G. Genome ploidy in different stages of the Giardia lamblia life
cycle. Cell Microbiol 3, 55–62 (2001).
63. Bingham, A.K. & Meyer, E.A. Giardia excystation can be induced in vitro in acidic solutions. Nature
277, 301–2 (1979).
64. Rice, E.W. & Schaefer, F.W., III. Improved in vitro excystation procedure for Giardia lamblia cysts.
J Clin Microbiol 14, 709–10 (1981).
65. Hautus, M.A., Kortbeek, L.M., Vetter, J.C. & Laarman, J.J. In vitro excystation and subsequent axenic
growth of Giardia lamblia. Trans R Soc Trop Med Hyg 82, 858–61 (1988).
66. Holberton, D.V. Mechanism of attachment of Giardia to the wall of the small intestine. Trans R Soc Trop
Med Hyg 67, 29–30 (1973).
67. Hansen, W.R., Tulyathan, O., Dawson, S.C., Cande, W.Z. & Fletcher, D.A. Giardia lamblia attachment
force is insensitive to surface treatments. Eukaryot Cell 5, 781–3 (2006).
68. Maia-Brigagao, C. & de Souza, W. Using environmental scanning electron microscopy (ESEM) as a
quantitative method to analyse the attachment of Giardia duodenalis to epithelial cells. Micron 43,
494–6 (2012).
69. Pickering, H., Wu, M., Bradley, M. & Bridle, H. Analysis of Giardia lamblia interactions with polymer
surfaces using a microarray approach. Environ Sci Technol 46, 2179–86 (2012).
70. Holberton, D.V. Arrangement of subunits in microribbons from Giardia. J Cell Sci 47, 167–85 (1981).
71. Hagen, K.D. et al. Novel structural components of the ventral disc and lateral crest in Giardia intestina-
lis. PLoS Negl Trop Dis 5, e1442 (2011).
72. Buret, A.G., Mitchell, K., Muench, D.G. & Scott, K.G. Giardia lamblia disrupts tight junctional ZO-1
and increases permeability in non-transformed human small intestinal epithelial monolayers: effects of
epidermal growth factor. Parasitology 125, 11–9 (2002).
73. Koh, W.H. et al. Giardia duodenalis assemblage-specific induction of apoptosis and tight junction dis-
ruption in human intestinal epithelial cells: effects of mixed infections. J Parasitol 99, 353–8 (2013).
650 Laboratory Models for Foodborne Infections
74. Troeger, H. et al. Effect of chronic Giardia lamblia infection on epithelial transport and barrier function
in human duodenum. Gut 56, 328–35 (2007).
75. Chin, A.C. et al. Strain-dependent induction of enterocyte apoptosis by Giardia lamblia disrupts epithe-
lial barrier function in a caspase-3-dependent manner. Infect Immun 70, 3673–80 (2002).
76. Hardin, J.A., Buret, A.G., Olson, M.E., Kimm, M.H. & Gall, D.G. Mast cell hyperplasia and increased
macromolecular uptake in an animal model of giardiasis. J Parasitol 83, 908–12 (1997).
77. Zhou, P., Li, E., Shea-Donohue, T. & Singer, S.M. Tumour necrosis factor α contributes to protection
against Giardia lamblia infection in mice. Parasite Immunol 29, 367–74 (2007).
78. Chen, T.L. et al. Persistent gut barrier damage and commensal bacterial influx following eradication of
Giardia infection in mice. Gut Pathog 5, 26 (2013).
79. Teoh, D.A., Kamieniecki, D., Pang, G. & Buret, A.G. Giardia lamblia rearranges F-actin and α-actinin
in human colonic and duodenal monolayers and reduces transepithelial electrical resistance. J Parasitol
86, 800–6. (2000).
80. Rodriguez-Fuentes, G.B. et al. Giardia duodenalis: analysis of secreted proteases upon trophozoite-
epithelial cell interaction in vitro. Mem Inst Oswaldo Cruz 101, 693–6 (2006).
81. Shant, J., Bhattacharyya, S., Ghosh, S., Ganguly, N.K. & Majumdar, S. A potentially important
excretory-secretory product of Giardia lamblia. Exp Parasitol 102, 178–86 (2002).
82. Scott, K.G., Logan, M.R., Klammer, G.M., Teoh, D.A. & Buret, A.G. Jejunal brush border microvillous
alterations in giardia muris-infected mice: role of T lymphocytes and interleukin-6. Infect Immun 68,
3412–8 (2000).
83. Scott, K.G., Yu, L.C. & Buret, A.G. Role of CD8+ and CD4+ T lymphocytes in jejunal mucosal injury
during murine giardiasis. Infect Immun 72, 3536–42 (2004).
84. Deselliers, L.P., Tan, D.T., Scott, R.B. & Olson, M.E. Effects of Giardia lamblia infection on gastroin-
testinal transit and contractility in Mongolian gerbils. Dig Dis Sci 42, 2411–9 (1997).
85. Li, E., Zhou, P. & Singer, S.M. Neuronal nitric oxide synthase is necessary for elimination of Giardia
lamblia infections in mice. J Immunol 176, 516–21 (2006).
86. Andersen, Y.S., Gillin, F.D. & Eckmann, L. Adaptive immunity-dependent intestinal hypermotility
contributes to host defense against Giardia spp. Infect Immun 74, 2473–6 (2006).
87. Li, E., Zhao, A., Shea-Donohue, T. & Singer, S.M. Mast cell mediated changes in smooth muscle con-
tractility during mouse giardiasis. Infect Immun 75, 4514–8 (2007).
88. Ventura, L.L. et al. Impact of protein malnutrition on histological parameters of experimentally infected
animals with Giardia lamblia. Exp Parasitol 133, 391–5 (2013).
89. Cantey, P.T. et al. Study of nonoutbreak giardiasis: novel findings and implications for research. Am J
Med 124, 1175 e1–8 (2011).
90. Oberhuber, G., Kastner, N. & Stolte, M. Giardiasis: a histologic analysis of 567 cases. Scand J
Gastroenterol 32, 48–51 (1997).
91. Roskens, H. & Erlandsen, S.L. Inhibition of in vitro attachment of Giardia trophozoites by mucin. J
Parasitol 88, 869–73 (2002).
92. Singer, S.M. & Nash, T.E. The role of normal flora in Giardia lamblia infections in mice. J Infect Dis
181, 1510–2 (2000).
93. Perez, P.F. et al. Inhibition of Giardia intestinalis by extracellular factors from Lactobacilli: an in vitro
study. Appl Environ Microbiol 67, 5037–42 (2001).
94. Humen, M.A. et al. Lactobacillus johnsonii La1 antagonizes Giardia intestinalis in vivo. Infect Immun
73, 1265–9 (2005).
95. Benyacoub, J. et al. Enterococcus faecium SF68 enhances the immune response to Giardia intestinalis
in mice. J Nutr 135, 1171–6 (2005).
96. Aley, S.B., Zimmerman, M., Hetsko, M., Selsted, M.E. & Gillin, F.D. Killing of Giardia lamblia by
cryptdins and cationic neutrophil peptides. Infect Immun 62, 5397–403 (1994).
97. Eckmann, L. Mucosal defences against Giardia. Parasite Immunol 25, 259–70 (2003).
98. Eckmann, L. et al. Nitric oxide production by human intestinal epithelial cells and competition for argi-
nine as potential determinants of host defense against the lumen-dwelling pathogen Giardia lamblia.
J Immunol 164, 1478–87 (2000).
99. Tako, E.A., Hassimi, M.F., Li, E. & Singer, S.M. Transcriptomic analysis of the host response to Giardia
duodenalis infection reveals redundant mechanisms for parasite control. MBio 4 (6), e00660–13 (2013).
Giardia lamblia 651
100. Kamda, J.D. & Singer, S.M. Phosphoinositide 3-kinase-dependent inhibition of dendritic cell
interleukin-12 production by Giardia lamblia. Infect Immun 77, 685–93 (2009).
101. Kamda, J.D., Nash, T.E. & Singer, S.M. Giardia duodenalis: dendritic cell defects in IL-6 deficient mice
contribute to susceptibility to intestinal infection. Exp Parasitol 130, 288–91 (2012).
102. Maloney, J., Keselman, A., Li, E. & Singer, S.M. Macrophages expressing arginase 1 and nitric oxide
synthase 2 accumulate in the small intestine during Giardia lamblia infection. Microbes Infect 17,
462–7 (2015).
103. Cotton, J.A. et al. Giardia duodenalis cathepsin B proteases degrade intestinal epithelial interleukin-8
and attenuate interleukin-8-induced neutrophil chemotaxis. Infect Immun 82, 2772–87 (2014).
104. Roxstrom-Lindquist, K., Ringqvist, E., Palm, D. & Svard, S. Giardia lamblia-induced changes in gene
expression in differentiated Caco-2 human intestinal epithelial cells. Infect Immun 73, 8204–8 (2005).
105. Ringqvist, E. et al. Release of metabolic enzymes by Giardia in response to interaction with intestinal
epithelial cells. Mol Biochem Parasitol 159, 85–91 (2008).
106. Snider, D.P., Gordon, J., McDermott, M.R. & Underdown, B.J. Chronic Giardia muris infection in
anti-IgM-treated mice. I. Analysis of immunoglobulin and parasite-specific antibody in normal and
immunoglobulin-deficient animals. J Immunol 134, 4153–62 (1985).
107. Skea, D.L. & Underdown, B.J. Acquired resistance to Giardia muris in X-linked immunodeficient mice.
Infect Immun 59, 1733–8 (1991).
108. Langford, T.D. et al. Central importance of immunoglobulin A in host defense against Giardia spp.
Infect Immun 70, 11–8 (2002).
109. Davids, B.J. et al. Polymeric immunoglobulin receptor in intestinal immune defense against the lumen-
dwelling protozoan parasite Giardia. J Immunol 177, 6281–90 (2006).
110. Singer, S.M. & Nash, T.E. T-cell-dependent control of acute Giardia lamblia infections in mice. Infect
Immun 68, 170–5 (2000).
111. Zhou, P. et al. Role of interleukin-6 in the control of acute and chronic Giardia lamblia infections in
mice. Infect Immun 71, 1566–8 (2003).
112. Lai Ping So, A. & Mayer, L. Gastrointestinal manifestations of primary immunodeficiency disorders.
Semin Gastrointest Dis 8, 22–32 (1997).
113. Heyworth, M.F., Carlson, J.R. & Ermak, T.H. Clearance of Giardia muris infection requires helper/
inducer T lymphocytes. J Exp Med 165, 1743–8 (1987).
114. Venkatesan, P., Finch, R.G. & Wakelin, D. A comparison of mucosal inflammatory responses to Giardia
muris in resistant B10 and susceptible BALB/c mice. Parasite Immunol 19, 137–43 (1997).
115. Jimenez, J.C. et al. Antibody and cytokine responses to Giardia excretory/secretory proteins in Giardia
intestinalis-infected BALB/c mice. Parasitol Res 113, 2709–18 (2014).
116. Ebert, E.C. Giardia induces proliferation and interferon γ production by intestinal lymphocytes. Gut 44,
342–6 (1999).
117. Dreesen, L. et al. Giardia muris infection in mice is associated with a protective interleukin 17A response
and induction of peroxisome proliferator-activated receptor alpha. Infect Immun 82, 3333–40 (2014).
118. Dann, S.M. et al. IL-17A promotes protective IgA responses and expression of other potential effectors
against the lumen-dwelling enteric parasite Giardia. Exp Parasitol 156, 68–78 (2015).
119. Bienz, M., Dai, W.J., Welle, M., Gottstein, B. & Muller, N. Interleukin-6-deficient mice are highly sus-
ceptible to Giardia lamblia infection but exhibit normal intestinal immunoglobulin A responses against
the parasite. Infect Immun 71, 1569–73 (2003).
120. Erlich, J.H., Anders, R.F., Roberts-Thomson, I.C., Schrader, J.W. & Mitchell, G.F. An examination of
differences in serum antibody specificities and hypersensitivity reactions as contributing factors to
chronic infection with the intestinal protozoan parasite, Giardia muris, in mice. Aust J Exp Biol Med
Sci 61, 599–615 (1983).
121. Li, E., Zhou, P., Petrin, Z. & Singer, S.M. Mast cell-dependent control of Giardia lamblia infections in
mice. Infect Immun 72, 6642–9 (2004).
122. Istre, G.R., Dunlop, T.S., Gaspard, G.B. & Hopkins, R.S. Waterborne giardiasis at a mountain resort:
evidence for acquired immunity. Am J Public Health 74, 602–4 (1984).
123. Isaac-Renton, J.L., Lewis, L.F., Ong, C.S. & Nulsen, M.F. A second community outbreak of waterborne
giardiasis in Canada and serological investigation of patients. Trans R Soc Trop Med Hyg 88, 395–9
(1994).
652 Laboratory Models for Foodborne Infections
124. Li, E., Liu, M. & Singer, S.M. Resistance to reinfection in mice as a vaccine model for giardiasis. Hum
Vaccines Immunother 10, 1536–43 (2014).
125. Abdul-Wahid, A. & Faubert, G. Mucosal delivery of a transmission-blocking DNA vaccine encoding
Giardia lamblia CWP2 by Salmonella typhimurium bactofection vehicle. Vaccine 25, 8372–83 (2007).
126. Abdul-Wahid, A. & Faubert, G. Characterization of the local immune response to cyst antigens during
the acute and elimination phases of primary murine giardiasis. Int J Parasitol 38, 691–703 (2008).
127. Lee, P., Abdul-Wahid, A. & Faubert, G.M. Comparison of the local immune response against Giardia
lamblia cyst wall protein 2 induced by recombinant Lactococcus lactis and Streptococcus gordonii.
Microbes Infect 11, 20–8 (2009).
128. Jenikova, G. et al. α1-giardin based live heterologous vaccine protects against Giardia lamblia infection
in a murine model. Vaccine 29, 9529–37 (2011).
129. Rivero, F.D. et al. Disruption of antigenic variation is crucial for effective parasite vaccine. Nat Med 16,
551–7 (2010).
130. Prucca, C.G., Rivero, F.D. & Lujan, H.D. Regulation of antigenic variation in Giardia lamblia. Annu
Rev Microbiol 65, 611–30 (2011).
131. Adam, R.D. et al. Antigenic variation of a cysteine-rich protein in Giardia lamblia. J Exp Med 167,
109–18 (1988).
132. Aggarwal, A., Merritt, J.W., Jr. & Nash, T.E. Cysteine-rich variant surface proteins of Giardia lamblia.
Mol Biochem Parasitol 32, 39–47 (1989).
133. Aggarwal, A. & Nash, T.E. Antigenic variation of Giardia lamblia in vivo. Infect Immun 56, 1420–3
(1988).
134. Nash, T.E., Herrington, D.A., Levine, M.M., Conrad, J.T. & Merritt, J.W., Jr. Antigenic variation of
Giardia lamblia in experimental human infections. J Immunol 144, 4362–9 (1990).
135. Nash, T.E. Antigenic variation in Giardia lamblia and the host’s immune response. Philos Trans R Soc
Lond B Biol Sci 352, 1369–75 (1997).
136. Nash, T.E., Banks, S.M., Alling, D.W., Merritt, J.W., Jr. & Conrad, J.T. Frequency of variant antigens in
Giardia lamblia. Exp Parasitol 71, 415–21 (1990).
137. Prucca, C.G. et al. Antigenic variation in Giardia lamblia is regulated by RNA interference. Nature
456, 750–4 (2008).
138. Saraiya, A.A., Li, W., Wu, J., Chang, C.H. & Wang, C.C. The microRNAs in an ancient protist repress
the variant-specific surface protein expression by targeting the entire coding sequence. PLoS Pathog 10,
e1003791 (2014).
139. Prucca, C.G. & Lujan, H.D. Antigenic variation in Giardia lamblia. Cell Microbiol 11, 1706–15 (2009).
140. Emery, S.J., Lacey, E. & Haynes, P.A. Quantitative proteomic analysis of Giardia duodenalis assem-
blage A: a baseline for host, assemblage, and isolate variation. Proteomics 15(13), 2281–5 (2015).
141. Nash, T.E., Merritt, J.W., Jr. & Conrad, J.T. Isolate and epitope variability in susceptibility of Giardia
lamblia to intestinal proteases. Infect Immun 59, 1334–40 (1991).
142. Singer, S.M., Elmendorf, H.G., Conrad, J.T. & Nash, T.E. Biological selection of variant-specific surface
proteins in Giardia lamblia. J Infect Dis 183, 119–24 (2001).
143. Astiazaran-Garcia, H., Inigo-Figueroa, G., Quihui-Cota, L. & Anduro-Corona, I. Crosstalk between
zinc status and Giardia infection: a new approach. Nutrients 7, 4438–52 (2015).
144. Emery, S.J., van Sluyter, S. & Haynes, P.A. Proteomic analysis in Giardia duodenalis yields insights
into strain virulence and antigenic variation. Proteomics 14, 2523–34 (2014).
145. Schofield, P.J., Costello, M., Edwards, M.R. & O’Sullivan, W.J. The arginine dihydrolase pathway is
present in Giardia intestinalis. Int J Parasitol 20, 697–9 (1990).
146. Popovic, P.J., Zeh, H.J., III & Ochoa, J.B. Arginine and immunity. J Nutr 137, 1681S–86S (2007).
147. Fernandes, P.D. & Assreuy, J. Role of nitric oxide and superoxide in Giardia lamblia killing. Braz J Med
Biol Res 30, 93–9 (1997).
148. Stadelmann, B., Merino, M.C., Persson, L. & Svard, S.G. Arginine consumption by the intestinal para-
site Giardia intestinalis reduces proliferation of intestinal epithelial cells. PLoS One 7, e45325 (2012).
149. Choi, B.S. et al. Differential impact of L-arginine deprivation on the activation and effector functions of
T cells and macrophages. J Leukoc Biol 85, 268–77 (2009).
150. Lindquist, H.D. Induction of albendazole resistance in Giardia lamblia. Microb Drug Resist 2, 433–4
(1996).
Giardia lamblia 653
151. Upcroft, J., Mitchell, R., Chen, N. & Upcroft, P. Albendazole resistance in Giardia is correlated with
cytoskeletal changes but not with a mutation at amino acid 200 in β-tubulin. Microb Drug Resist 2,
303–8 (1996).
152. Upcroft, J.A., Campbell, R.W. & Upcroft, P. Quinacrine-resistant Giardia duodenalis. Parasitology 112
(Pt 3), 309–13 (1996).
153. Arguello-Garcia, R., Cruz-Soto, M., Romero-Montoya, L. & Ortega-Pierres, G. Variability and varia-
tion in drug susceptibility among Giardia duodenalis isolates and clones exposed to 5-nitroimidazoles
and benzimidazoles in vitro. J Antimicrob Chemother 54, 711–21 (2004).
154. Arguello-Garcia, R., Cruz-Soto, M., Romero-Montoya, L. & Ortega-Pierres, G. In vitro resistance to
5-nitroimidazoles and benzimidazoles in Giardia duodenalis: variability and variation in gene expres-
sion. Infect Genet Evol 9, 1057–64 (2009).
155. Boreham, P.F., Phillips, R.E. & Shepherd, R.W. Altered uptake of metronidazole in vitro by stocks of
Giardia intestinalis with different drug sensitivities. Trans R Soc Trop Med Hyg 82, 104–6 (1988).
156. Upcroft, J.A. & Upcroft, P. Drug resistance and Giardia. Parasitol Today 9, 187–90 (1993).
157. Upcroft, J.A. & Upcroft, P. Drug susceptibility testing of anaerobic protozoa. Antimicrob Agents
Chemother 45, 1810–4 (2001).
158. Leitsch, D., Schlosser, S., Burgess, A. & Duchene, M. Nitroimidazole drugs vary in their mode of action
in the human parasite Giardia lamblia. Int J Parasitol Drugs Drug Resist 2, 166–70 (2012).
159. Muller, J., Sterk, M., Hemphill, A. & Muller, N. Characterization of Giardia lamblia WB C6 clones
resistant to nitazoxanide and to metronidazole. J Antimicrob Chemother 60, 280–7 (2007).
160. Sterk, M., Muller, J., Hemphill, A. & Muller, N. Characterization of a Giardia lamblia WB C6 clone
resistant to the isoflavone formononetin. Microbiology 153, 4150–8 (2007).
161. Escobedo, A.A., Hanevik, K., Almirall, P., Cimerman, S. & Alfonso, M. Management of chronic
Giardia infection. Expert Rev Anti Infect Ther 12, 1143–57 (2014).
162. Uzlikova, M. & Nohynkova, E. The effect of metronidazole on the cell cycle and DNA in metronidazole-
susceptible and -resistant Giardia cell lines. Mol Biochem Parasitol 198, 75–81 (2014).
163. Ansell, B.R. et al. Drug resistance in Giardia duodenalis. Biotechnol Adv 33(6), 888–901 (2015).
164. Paz-Maldonado, M.T., Arguello-Garcia, R., Cruz-Soto, M., Mendoza-Hernandez, G. & Ortega-
Pierres, G. Proteomic and transcriptional analyses of genes differentially expressed in Giardia duode-
nalis clones resistant to albendazole. Infect Genet Evol 15, 10–7 (2013).
165. Kane, A.V., Ward, H.D., Keusch, G.T. & Pereira, M.E. In vitro encystation of Giardia lamblia: large-
scale production of in vitro cysts and strain and clone differences in encystation efficiency. J Parasitol
77, 974–81 (1991).
41
Toxoplasma: Animal and In Vitro
Models on Toxoplasmosis
CONTENTS
41.1 Introduction................................................................................................................................... 655
41.1.1 The Disease...................................................................................................................... 655
41.1.2 Biology of the Parasite..................................................................................................... 656
41.2 Laboratory Models to Study Toxoplasma gondii......................................................................... 658
41.2.1 In Vitro Models................................................................................................................. 658
41.2.2 Animal Models................................................................................................................. 660
41.2.2.1 Detection of the Parasite in Experimental Infected Animals...........................661
41.2.2.2 Rodent............................................................................................................... 662
41.2.2.3 Pigs.................................................................................................................... 665
41.2.2.4 Cat..................................................................................................................... 666
41.2.2.5 Chicken............................................................................................................. 667
41.2.2.6 Fish.................................................................................................................... 668
41.3 Concluding Remarks..................................................................................................................... 668
Acknowledgments................................................................................................................................... 669
References............................................................................................................................................... 669
41.1 Introduction
41.1.1 The Disease
Toxoplasmosis is a disease that infects nearly all warm-blooded vertebrates.1,2 It is estimated that about
a third of the world’s human population is serologically positive for this infection.2–4 Transmission of
the parasite has been historically implicated by the ingestion of undercooked or raw meat containing
the parasite in tissue cysts.5 However, in the last two decades, accumulated evidence point also to the
parasite infection by oocysts ingestion through contaminated water, soil, or food—mainly vegetables
and fruits.5,6 Food consumption is culturally based; thus, the venue of infection may vary greatly in dif-
ferent human populations, explaining the variance in different geographical locations.3 Toxoplasmosis
outbreaks through contaminated water in human populations have been reported in detail7–10 showing
how relevant environmental contamination can be.
Most often, toxoplasmosis is asymptomatic in immunocompetent individuals.2–4,11 However, depend-
ing on the Toxoplasma gondii strain,2 the genetic constitution of the host, parasite load, and the mode
of transmission,12,13 infection can cause severe clinical manifestations including ocular toxoplasmosis,14
cerebral toxoplasmosis,15 and congenital toxoplasmosis.1 When acquired by healthy individuals, toxo-
plasmosis most often causes lymphadenopathy, usually of the head and neck, but pneumonia, retino-
choroiditis, myocarditis, neurological disorders, and severe ocular infection have also been reported in
655
656 Laboratory Models for Foodborne Infections
severe cases especially in French Guiana16 and in Brazil.17 Congenital toxoplasmosis can cause mental
retardation, encephalitis, neonatal mortality, and abortion of the fetuses.2,17,18 In immunocompromised
patients, the reactivation of the parasite into fast replicating forms in the central nervous system (CNS)
causes cerebral toxoplasmosis.5 In addition, T. gondii infection has been implicated in schizophrenia and
other neurological disorders.15,19 Domestic animals also develop toxoplasmosis upon infection, causing
relevant economic losses, and wild animals are also infected by T. gondii.1,20 Due to its severe mani-
festations, worldwide distribution, and diverse host infection, toxoplasmosis is a relevant public health
problem.21,22
(A) (B)
(C) (D)
FIGURE 41.1 Tachyzoites infecting ostrich macrophages. Crescent shape of the parasite can be noted after infection
(2 h—A), replication (24 h—B; 48 h—C) and egress (48 h—D). During egress, host cells are usually destroyed (D). Bar: 20 μm.
Toxoplasma: Animal and In Vitro Models on Toxoplasmosis 657
FIGURE 41.2 Toxoplasma gondii cyst (arrow) found in the brain of mice inoculated with heart tissue of an infected pig.
Bar: 50 μm.
Sporulated
oocysts contaminate
the environment
Carnivorism
Carnivorism
FIGURE 41.3 Schematic representation of the Toxoplasma gondii life cycle. In the enterocytes of the members of the
Felidae family, the parasite differentiates in gametocytes that generate a zygote that is shed in the feces as unsporulated
oocysts. After 2–5 days in the environment, oocysts sporulate to form two esporocysts, each with four sporozoites that
contaminate the environment. Hosts get infected by the ingestion of oocysts. Oocysts are digested liberating sporozoites
that first infect enterocytes differentiating in tachyzoites that later infect lamina propria cells and multiply rapidly. These
forms disseminate through the organism by the circulatory system. Tachyzoites convert into bradyzoites forming tissue
cysts. Tachyzoites can migrate to the placenta in pregnant hosts causing congenital infection. Carnivorism among the hosts
disseminates the parasite through the ingestion of bradyzoites in tissue cysts. As with oocysts, tissue cysts are digested
liberating bradyzoites that are resistant to this process. These forms infect host enterocytes and differentiate in tachyzoites
similar to sporozoites.
by meiosis and mitosis in the environment and disseminates. Infection of intermediate hosts occurs by
ingestion of oocysts through the consumption of contaminated water or food, or by the ingestion of tis-
sue cysts by carnivorism. In these hosts, the infective forms, sporozoites or bradyzoites, pass through the
enterocytes and convert into tachyzoites38 that infect and multiply in cells of the lamina propria. At this
stage, tachyzoites disseminate through the organism and then convert into bradyzoites, forming tissue
cysts. During their life cycle, tachyzoites may pass from the blood to the placenta and infect the fetus
causing congenital toxoplasmosis (Figure 41.3).
658 Laboratory Models for Foodborne Infections
Sexual reproduction of the parasite in Felidae provides genetic variability that results in different
recombinant strains.24,39,40 Isolated strains from North America and Europe have an unusual population
structure with basically three clonal lineages (types I, II, and III).41 However, recombinant strains are
found in different parts of the world and have diverse virulence.20,39,42 Studies on isolates from Brazil,
Colombia, and French Guiana have shown that most T. gondii strains are recombinant, suggesting greater
sexual recombination of this parasite in South America,16,39,43 thus explaining the wider spectra of clini-
cal manifestations.16,17,43 This indicates that the parasite population structure varies in different locations
around the world, being composed by distinct genotypes with important implications to infection, clini-
cal manifestations, and development of distinct diagnosis and treatment strategies.
than Trypanosoma cruzi.65 This ROS resistance was correlated with high catalase activity of T. gondii,65
which was later confirmed by the activity of superoxide dismutase showing the robust antioxidant arsenal
of this parasite.66,67 As a result, production of ROS is not detected in the T. gondii macrophage contact sites,
explaining parasite survival, persistence, and multiplication in host cells that are professional ROS produc-
ers.68,69 All this work was done with mammalian macrophages, but the similar result was obtained when
chicken macrophages were examined,70 suggesting that the same mechanism operates in birds. Although
this parasite can evade from host cell ROS production in vitro, it was later demonstrated that these radicals
are implicated in the in vivo control of T. gondii71 suggesting that this system still needs further work.
More recently, it has been demonstrated that IRGs (immunity-related GTPases) are involved in the growth
control of T. gondii in mice.72–75 This is significant because most of the data from different aspects of T. gon-
dii biology, including IRGs, point to rodents as the most relevant intermediate host for the parasite, in a clear
example of coevolution of host cell resistance mechanisms and T. gondii virulence factors.73,75 Interferon-
gamma (IFN-γ) induces IRGs in rodent host cells of myeloid and nonmyeloid origin. If an IFN-γ-activated
cell is infected by T. gondii, the GTPases hierarchically and cooperatively accumulate in the PV membrane,
changing their form until disruption, allowing T. gondii to end up in the cytosol where it is killed.73,76
However, some T. gondii strains can escape the IRG system.40,75 Virulence of T. gondii strains to mice is
directly related to its repertory of rhoptry proteins kinases and pseudokinases against the IRGs microbi-
cidal system.40 Rhoptry is a secretory organelle of T. gondii that releases its protein contents into the host
cell during invasion. However, there is high polymorphism of these proteins between strains.77,78 Virulent
strains of T. gondii, such as type I, have rhoptry proteins that efficiently phosphorylate IRGs components,
impairing their association to the PV, resulting in parasite survival and virulence capacity.40,79 Less-virulent
strains have fewer or no effective rhoptry proteins against IRGs, being destroyed by this system. In virulent
strains, these proteins form complexes that inactivate IRGs.80 These complexes also involve proteins from
another secretory organelle of the parasite, the dense granules.81 The composition of these secretory protein
complexes and how they deactivate IRGs is currently an important area of T. gondii research.
In the last decade, it has also been shown that T. gondii hijacks cells from the immune system, allow-
ing the parasite to cross host barriers such as gut epithelial barrier, blood–brain, and blood–retina barri-
ers in a mechanism known as Trojan horse.82–84 Some of the molecular events underlying this mechanism
have been described by our group. In vitro infection of unstimulated murine macrophages leads to down-
regulation of integrin alphaL, alpha4, and alpha5 immediately after infection, allowing cells to de-attach
from extracellular matrix components.82 On the other hand, T. gondii infection of human or murine
macrophages impairs upregulation of CD86, CD80, CD40, and CD1a, while maintaining their migra-
tory ability.85 T. gondii seems to regulate the migratory machinery similar to what metastatic cells do
as demonstrated by increased expression of host metalloproteinases MT1-MMP and ADAM-10 upon
infection of murine macrophages, in parallel with increased secretion of active MMP-9.84 Active MMP-9
is secreted in association with its cell membrane acceptor CD44, and with uPAR and TIMP-1.86 In fact,
secretion of this multiprotein complex is partially dependent on the extracellular plasminogen urokinase
pathway (uPA/uPAR)86 and ERK.84 In vivo expression of MMP-9 is also increased in the ilea and lungs
of orally infected C57Bl/6 mice (Pimentel, P. M., et al., unpublished data). Adoptive transfer experiments
of MMP knockdown cells infected with T. gondii have been developed in order to access its contribution
to in vivo dissemination of host cells carrying the parasite.
Nitric oxide (NO) produced by inducible NO synthase (iNOS or NOS2) expressed in classically acti-
vated macrophages is another microbicidal molecule of vertebrates87,88 involved in T. gondii control.
Infection of iNOS knockout mice or mice treated with iNOS inhibitors (aminoguanidine) have shown
that NO is important to control T. gondii growth, especially during the chronic phase of the disease.89–91
However, NO may also induce host death in the acute phase, being one of the responsible events of
the classical histopathology found in the ileum.91,92 Macrophages in culture classically activated by
IFN-γ and lipopolysaccharide express iNOS and produce high amounts of NO that control T. gondii
growth.90,93,94 After gut infection, T. gondii disseminates throughout the host, and immune response
initiated locally becomes systemic and, depending on the strain, controls parasite growth leading to the
latent infection. During this phase, tachyzoite will come across activated macrophages producing high
amounts of NO. How do these parasites cope with these encounters? We have been working on this
question by studying T. gondii infection of activated macrophages of mice and chickens in vitro. It was
660 Laboratory Models for Foodborne Infections
shown that tachyzoites of T. gondii inhibit NO production after infection of activated macrophages.70,95–99
NO production inhibition is caused by iNOS degradation.70,96,97 Interestingly enough, this degradation
occurs right at the beginning (2 h) of the infection70,97,99 and is caused by the proteasome pathway.99 It was
determined that an autocrine secretion of TGF-β1 caused by the parasite infection was responsible for
iNOS degradation.97 Similar to amastigotes of Leishmania amazonensis,100 T. gondii tachyzoites expose
phosphatidylserine (PS) without cell death, which, when blocked by annexin-V, abolishes the inhibition
of NO production caused by the infection.97 This indicates that tachyzoites of T. gondii may, indeed, use
what is known as “apoptotic mimicry,” a concept established for L. amazonensis that basically states
that the parasites that expose PS mimic the anti-inflammatory response induced by apoptotic cells.101 We
then separated the PS-positive and -negative tachyzoite subpopulations using annexin-V conjugated with
magnetic beads and found that only the PS-positive subpopulation was able to inhibit NO production in
infected macrophages.98 In addition, mice infected with positive or negative PS subpopulations died faster
than the ones infected with both populations. Mice infected with the positive PS subpopulation died of high
parasite burden, while the ones infected with the negative PS subpopulation died of uncontrolled inflam-
matory response.98 These results indicate that both subpopulations are necessary for a balanced response
of the host, allowing survival and parasite transmission. Recently, we have found that four strains of T.
gondii with distinct virulence exposed PS and inhibited iNOS expression immediately after the infection
of activated macrophages (Damasceno-Sá, J. C. et al., unpublished data). Thus, it seems that apoptotic
mimicry is a general escape mechanism to NO microbicidal system that is present in mammals and birds.
Tachyzoite conversion to bradyzoite and formation of tissue cysts may be a naturally occurring phe-
nomenon (not necessarily induced by the immune response) in the course of T. gondii’s life cycle and
occurs mainly in the CNS and in skeletal and cardiac muscles, defining the latent stage of T. gondii infec-
tion.13,31,32,102 Thus, there is great interest in the understanding of this conversion and the need of a well-
established model in order to test drugs, because most treatments are not effective against the bradyzoite
stage. An interesting in vitro model involves the infection of primary cultures of embryonic-derived
skeletal muscle cells by T. gondii.103 The most impressive result concerns the spontaneous capacity of
some T. gondii strains to convert tachyzoites into bradyzoites, forming in vitro tissue cyst-like struc-
tures in these muscular cells.31,32 The infection of these cells with bradyzoites, instead of tachyzoites,
accelerates the formation of cyst-like structures.102 Skeletal muscle cells become microbicidal against T.
gondii after activation with IFN-γ, controlling parasite growth by NO production and IRGs, indicating
that these cells may also help to control T. gondii growth in mice.104 This model has also been used to
show the induction of lipid bodies by T. gondii infection in these cells and its association to the PV.105
The PV association to lipid bodies was also seen in macrophages infected with T. gondii in a lipid body
model developed by our group.106 Embryonic-derived skeletal muscle cells should be further explored as
a model of in vitro infection to study other aspects of the biology of this parasite.
In vitro models lead to important findings on T. gondii. The lytic cycle and different aspects of the parasite
biology are well studied using tachyzoites to infect distinct cell types in culture. Tachyzoite is the most easily
obtained T. gondii form, which explains why this is, by far, the most used model. Infection of culture cells
with bradyzoites has been used to study the formation of tissue cyst-like structures in vitro (see paragraph
above). Bradyzoites are obtained from mice but may be replaced by an efficient in vitro system as earlier
stated. More recently, spontaneous cystogenesis was demonstrated in an established cell line of renal feline
kidney cells infected with bradyzoites, suggesting that cell culture system may eventually replace infected
animals for studies with cysts and bradyzoites.107 The enteroepithelial part of the life cycle of T. gondii in
Felidae is less well known because there is no established cell culture model, with most findings deriving
from cat experiments.35 However, primary cultures of cat intestinal cells have been infected with bradyzoites
resulting in syncytial parasite structures suggesting the in vitro reproduction of schizogony.108 These results
indicate that it is a matter of time for the different parts of T. gondii cell cycle to be obtained in vitro.
41.2.2 Animal Models
The use of animal models to study toxoplasmosis is extremely important. T. gondii infects a plethora
of warm-blooded animals; thus, most animal infections are unique and important to understand the
parasite cell cycle and the coevolution of host mechanisms that control the parasite and of virulence
Toxoplasma: Animal and In Vitro Models on Toxoplasmosis 661
strategies for the evasion of these mechanisms. Owing to ethical reasons, the use of animals is always
controversial and must follow standard and strict protocols. In addition, some host vertebrates are
difficult and expensive to maintain, including wild, and large or medium-sized domestic animals.
Many different hosts have been used to study infections by T. gondii, but the mouse is by far the most
common (Table 41.1). A PubMed search using common host names (chosen by the authors) and the
terms “toxoplasma” and later “experimental” or “infection” or “epidemiology” (Table 41.1) showed
the highest number of articles for the mouse followed by the rat, cat, pig, chicken, Calomys, and fish.
The term “infection” had the highest number of articles, and since infection encompasses “epidemiol-
ogy,” this term was also included in the survey to better understand the numbers of published articles.
This reveals how important the cat and the pig are in the study of T. gondii epidemiology (Table 41.1).
Here, we will refer to some relevant findings of the models mentioned in Table 41.1.
TABLE 41.1
Number of Published Articles in a PubMed Search (September 2015)
Crossing the Animal Models of the First Column with “Toxoplasma”
and One of the Terms on the Second, Third, or Fourth Column
Animal Host Experimental Infection Epidemiology
Mousea 520 2358 442
Rats 81 263 76
Cat 70 746 437
Pigs 53 315 270
Chicken 14 90 90
Calomys 8 14 1
Fish 3 28 34
a The use of plural or singular for the animal host denomination was based on
the higher number of articles obtained on the “experimental” column.
662 Laboratory Models for Foodborne Infections
41.2.2.2 Rodent
Rodents are abundantly present in different parts of the world.113 Species of this order are usually infected
with T. gondii, although depending on the location, the prevalence varies and is probably related to the
lack of seropositivity in congenital infected animals of this order.114,115 These animals are probably the
most common intermediate hosts of T. gondii,73,75 with small- or medium-sized Felidae being the fur-
thermost natural definitive hosts.116,117 In addition, the mouse is the most used laboratory animal, and
numerous articles describe experimental infections with T. gondii. As a result, most of what is known
about T. gondii biology comes from laboratory models involving rodents, especially mice.
41.2.2.2.1 Mice
Infection of mice has been crucial for the understanding of many aspects of T. gondii. One of the greatest
advantages of mice as animal models for parasitic infection is the availability of a vast number of mice
strains with different genetic backgrounds, deficiencies, and the capacity to genetically manipulate these
animals.118 This has allowed the discovery of important aspects of T. gondii biology. Some relevant find-
ings concerning characteristics of the T. gondii infection will be described in the following paragraphs.
IFN-γ is crucial to control T. gondii infection.119 The first evidence that this cytokine was important
came from in vitro studies showing that the presence of this cytokine increases the microbicidal potential
of different cell types after infection.26,90,93,94,104 Studies using mice treated with the IFN-γ120 and anti-
bodies that neutralize this cytokine121 proved that IFN-γ is essential to control T. gondii growth in vivo.
The importance of IFN-γ in controlling T. gondii infection was also shown in knockout mice that died
rapidly after infection.122
The interleukin (IL)-12/IFN-γ axis was first shown in mice with severe combined immunodeficiency
(SCID) with the demonstration that IL-12 was mainly produced by dendritic cells (DCs) upon IL-1β
stimulation, resulting in the induction of IFN-γ production by NK cells in this model.123,124 The impor-
tance of IFN-γ and IL-12 and their signaling pathway was demonstrated by the infection of mice lacking
the transcription factor interferon consensus sequence binding protein (ICSBP) with an avirulent strain
of T. gondii that lead mice to rapid death after 14 days of infection.125 Also, the regulatory role of IL-10
was demonstrated in knockout mice that succumbed to infection after 2 weeks due to elevated circu-
lating IFN-γ and IL-12 and intense liver pathology.126 The importance of TLRs in immune response
to T. gondii was revealed by the use of knockout mice to MyD88, a crucial adaptor molecule for TLR
signaling transduction.127,128
Another interesting use of the mous model for T. gondii infection came from studies showing that oral
infection of these animals causes death by a typical pathology of the ileum that involves tumor necrosis
factor-α (TNF-α), IFN-γ, and NO production.92 This is relevant because oral infection is the natural route
of T. gondii infection. Depending on the strain of the parasite, the genetic background of the mice, and the
parasite burden, the ilea pathology varies. When the C57BL/6 mice receive 100 cysts of the ME-49 strain
of T. gondii by the oral route, a potent Th1 response is induced leading to the destruction of the ileum
by intense necrosis. The typical ileum villus morphology is lost due to the high inflammatory response
(Figure 41.4). Eventually, the ileum ruptures, and the mice die by sepsis. The infection of C57BL/6 mice
with the ME-49 strain of T. gondii has been proposed as a model for intestinal bowel disease.129 In addi-
tion, this pathology is not exclusive to mice, since it has also been described in different hosts.130
Although TNF-α and IFN-γ are involved in the ileum pathology, another crucial player is the gut flora.131
Oral infection causes an increase in Gram-negative bacteria, antibiotic treatment reduces IFN-γ levels and
NO production in the small intestine, and gnotobiotic mice do not develop the ileum pathology.131 Thus,
the presence of LPS from the gut flora and the increase in IFN-γ, and possibly of TNF-α, caused by the
immune response against the parasite, induces a Th1 response that ends up destroying the ileum. In this
model, IL-22 induced MMP-2 expression in an IL-17-independent manner, and infection of MMP-2 KO
mice showed decreased immunopathology scores of the gut.132 However, oral infection of MIF KO mice
led to reduced pathogenesis in the ileum, with decreased amounts of IL-12, IFN-γ, and TNF-α and also
MMP-9, whereas no modification in MMP-2 expression was observed.133 Our group has demonstrated
that oral infection of C57BL/6 with the ME-49 T. gondii strain induced high amounts of MMP-9 but no
MMP-2 in the ilea and lungs after 7 days of infection (Pimentel, P. M., et al., unpublished data).
Toxoplasma: Animal and In Vitro Models on Toxoplasmosis 663
(A) (C)
(B) (D)
FIGURE 41.4 General aspects of the ilea of C57BL/6 mice infected orally with tissue cysts of Toxoplasma gondii of the
ME-49 strain. (A) Noninfected ileum with apparent normal villi. (B) Higher magnification of the box image “A.” (C) Ileum
from infected mice with necrotic areas disrupting the villi architecture (arrows). (D) Higher magnification of the box of
image “C”; parasites can be seen in the inset. Bars = 200 μm and 10 μm in inset.
Mice models have recently been used to further explore the role of the microbiota and IL-17 in the
gut upon T. gondii infection. Some models show that IL-17 is involved in the immunopathogenesis
at the gut mucosal barrier.134,135 However, it has not been clearly identified whether IL-17 is produced
by Th17 or ROR-γ innate lymphoid cells or both. Elimination of Paneth cells by IFN-γ produced by
Th1 cells induces decreased amounts of antimicrobial peptides, leading to expansion of bacteria of the
Enterobacteriaceae family, amplifying immunopathology at the gut after infection.136 On the other hand,
mice models have also identified mechanisms of containment of spreading of microflora during infec-
tion. Translocation into the luminal space of inflammatory macrophages and neutrophils is observed
early in T. gondii infection, where Fpr1 (N-formyl peptide receptor)-expressing neutrophils promote
encapsulation of γ-proteobacteria,137 leading to bacterial death probably by NETosis.138
Finally, it would be interesting to see in the coming years the exploration of diverse mice models
combined with infection with transgenic parasites designed to verify the cross-talk between T. gondii
virulence factors and host immune response.
41.2.2.2.2 Rats
Infection of rats with T. gondii has been performed since the 1950s. Most studies on rats are per-
formed with two species, Rattus norvegicus and R. rattus, with the latter being the most used owing
to the different strains available.139 Rats may have an important epidemiological role for toxoplasmosis,
since they can be a source of infection to different hosts, especially pigs and possibly the domestic
cats.114,140 Epidemiological studies involving rats are complex, since T. gondii has been isolated by bio-
assay (in mice) from tissue of serologically negative rodents140 including from experimental congenital
infections.114 In addition, various cases have been reported where inoculation with different strains of
T. gondii resulted in different serology findings, the antibodies titers varied greatly with the type of
664 Laboratory Models for Foodborne Infections
test used, and negative serology was not uncommon.140 Thus, epidemiological studies of rats for T. gondii
based on serology must be analyzed with great care.
Most T. gondii strains have been isolated by bioassay with mice using pepsin-digested tissue from dis-
tinct hosts. This standard procedure to isolate T. gondii strains directly classifies the isolates as virulent or
avirulent to mice. Naturally, these strains have been used to experimentally infect other hosts, such as rats.
Rats have a natural resistance to T. gondii that has been known for a long time.56,140–142 Strains that are vir-
ulent to mice may not necessarily cause the disease in other host species. Immunocompetent rats infected
with tachyzoites or tissue cysts of different strains of T. gondii do not develop clinical manifestations being
resistant to the parasite,140 but infection with oocysts of the VAG strain induced clinical manifestations
and, depending on the dosage, killed the rats.143 Because immunocompetent humans are normally resis-
tant to T. gondii infection, the rat T. gondii infection model is considered by different authors to be a closer
animal model for human toxoplasmosis. The rat T. gondii infection model has been used as a congenital
transmission model showing that the parasite first colonizes the placenta and later the fetus.140 It was
found that the transmission to the fetus may be low,140 but higher transmission has also been reported,144,145
although the lactation route is not relevant.144 Differences in the transmission to the fetus in the distinct
congenital rat T. gondii models may be related to the strain of rat and to the strain of parasite used.
One of the most interesting outcomes with the rat T. gondii infection model is the heterogeneous resis-
tance of the different strains of rats to this parasite. After experimental infection, no parasite cysts are
found in the brain of the LEW strain in contrast to the Fisher strain.146 The lack of parasite cysts in the brain
of the LEW rats was further confirmed, and no detectable humoral response was found when compared
to the T. gondii susceptible Fisher and Brown Norway strains where a clear IgG response was observed.147
With the aid of bone marrow chimeras between these rats strains, the resistance of the LEW rat was linked
to cells derived from the myeloid lineage.146 Using congenic lines of the LEW and Brown Norway rats, the
resistance of the LEW strain was determined to be on chromosome 10 in a locus named Toxo1.148 In addi-
tion, T. gondii grew in infected fibroblasts of all congenic rat strains of this study but not in macrophages
from rats with the LEW Toxo1 locus, indicating that these cells are the main immune component related to
the resistance of this rat strain,148 and that possible genetic polymorphisms of the genes at this locus exist
in the rat strains. Although the locus responsible for LEW resistance was found, the gene or genes were not
characterized at that time, and the killing mechanism was not determined, but it was not NO-dependent.148
By using classical and molecular genetic tools with the rat strains that are susceptible or resistant for
T. gondii and distinct studies on infected macrophages, two research groups have recently suggested the
identity of a gene in the Toxo1 locus related to the capacity of the resistant rat macrophage to kill the par-
asite.149,150 These groups found that the NOD-like receptor 1 (Nlrp1), which is part of the inflammasome,
is essential for the resistant rat against the parasite. After bone marrow macrophage infection, this intra-
cellular receptor senses T. gondii and activates the inflammasome, host cells die, the parasite is killed,
and the tissue becomes inflamed. Inflammasomes are high-molecular-weight cytosolic multimeric pro-
tein complexes, which, upon assemblage, activate caspase-1 leading to a lytic cell death program (pyrop-
tosis) and also to the secretion of IL-1β and IL-18, caused by the cleavage of their precursors, inducing
a tissue proinflammatory response that controls infections.151 It was found that Nlrp1 sequence varies
among rat strains and that a variant from the resistant rat strains was responsible for macrophage pyrop-
tosis upon infection and parasite killing; on the other hand, two other variants from T. gondii susceptible
rat strains allowed macrophage survival and parasite growth.150 Activation of caspase-1 of infected mac-
rophages depends on the Nlrp1 variants, with the ones from resistant rats releasing higher levels of IL-18
and IL-1β. Knockdown and over expression of the Nlrp1 variant from the resistant rat in macrophages
establish the importance of this receptor in pyroptosis initiated by the infection and subsequent killing of
T. gondii.150 Genetic and infection studies using the different susceptible or resistant T. gondii rat strains
and their congenics also predicted the Nlrp1 gene to confer the rat macrophages resistance to T. gondii.149
In vitro infection caused parasite and peritoneal macrophage death only of resistant rat strains. The death
of the infected macrophages of the resistant rats was neither apoptotic nor autophagic. Higher production
of ROS and cleavage of caspase-1 were shown on infected macrophages of the resistant rat strain, clearly
correlating T. gondii death with host cells death by inflammasome activation.149 The use of pharmaco-
logical inhibitors of caspase-1 cleavage abrogated parasite killing in the resistance rat macrophages, but
the growth was not totally restored, indicating that the control of T. gondii in resistance rat macrophages
Toxoplasma: Animal and In Vitro Models on Toxoplasmosis 665
involves more mechanisms.149 The identification of the Nlrp1 gene in the Toxo1 locus of the rat and its
possible relation to resistance rat to T. gondii infection is an example of how different host models are
important to better understand parasite biology.
Inflammasome is also important in T. gondii control in mice.152,153 In addition, human Nlrp1 has been
implicated in human congenital toxoplasmosis154,155 and monocyte capacity to control T. gondii growth.153
In addition, GRA15 from a specific T. gondii strain was responsible for triggering IL-1β release from
infected human macrophages.156 Thus, inflammasome activation by sensing different strains of the para-
sites in distinct hosts resulting in broad killing potential seems to be a widespread strategy to control
T. gondii growth that varies within host species. The killing of T. gondii by the inflammasome is rapid
and may explain the low serology results seen in rodent epidemiological surveys. If this microbicidal
mechanism is also operating in humans and varies depending on gene polymorphism, there may be low
estimations of T. gondii infection in the human population as well.
41.2.2.2.3 Calomys callosus
The genus Calomys (Waterhouse, 1937) (Rodentia, Sigmodontinae) comprises small rodents that are
endemic in South America, originally being observed from northern Argentina to Peru, including Brazil,
Bolivia, and Paraguay. Currently, 10 species are described by considering morphology and karyotype.157
Calomys callosus mainly inhabits the central region of Brazil and has the capacity to live in different
biomes, giving it a great adaptive capacity.158
Such a rodent has been successfully domesticated and presents advantages as an animal model being
well adapted to the animal-housing environment.159 They are very prolific throughout the year and are
easy to handle. Common infections of rats, mice, and guinea pigs (Cavia aperea) do not affect this
rodent, which is a robust animal.159
High genetic diversity has been observed in isolates of T. gondii from South America.17 Recombinant
strains isolated in Brazil were more pathogenic to mice than clonal isolates from North America and
Europe.160 Because C. callosus has been found in environments with more pathogenic recombinant
strains of T. gondii, this model becomes quite interesting for the study of the parasite–host relationship.161
C. callosus is highly susceptible to infection with the RH strain of T. gondii,162 which proliferates in
large numbers in the newly implanted trophoblasts in pregnant females,163 and especially in the labyrin-
thine zone of the placenta.164 The same applies if the females are in the acute phase of the infection with
the ME-49 strain. However, females with chronic infection with the ME-49 strain are, at least tempo-
rarily, protected against RH infection with no transplacental infection.165 In contrast, the same chronic
infection does not protect against transplacental migration when reinfection is performed with Brazilian
recombinant strains.166 Thus, this is a good model to study neonatal infection.167
Few studies have been done with blood leukocytes in this model, thus demonstrating great poten-
tial for further research. Intraperitoneal infection with the RH strain of T. gondii induces migration of
neutrophils, monocytes, and mast cells after 24 h. In addition, basophils with distinct morphology were
identified after 48 h in the ileum submucosa. This cellular process coincided with the presence of dead
extracellular parasites. In lymphoid tissues, T. gondii was seen in mast cells, with or without the PV.168 In
intraconjunctival infection with the RH parasites, mast cell influx was detected within 5 and 24 h, with
correlation to the inflammatory process.169
Pregnant Calomys were orally infected with the ME-49 strain resulting in ocular injury and the pres-
ence of cysts in 40% of the fetuses. Ocular lesions were found in 25% of the females and 75% of males,
while 25% of each group had binocular lesions.170 These results indicate that the Calomys model is
promising for studying ocular and congenital toxoplasmosis.
41.2.2.3 Pigs
Knowledge obtained with laboratory animals (especially mice) needs to be validated in medium-sized ani-
mal models before clinical human trials.171 Because pigs and humans have similar anatomy, genetics, and
physiology, this animal is an interesting human infectious disease model172 and has been used as a biomedical
model for preclinical experimentation.173 However, few functional experiments have been carried out in these
animals, including T. gondii infection. Thus, there is great potential concerning this experimental model.
666 Laboratory Models for Foodborne Infections
Besides being an interesting animal model for T. gondii infection, pigs are important in the epidemiol-
ogy of this parasite, since the ingestion of tissue cysts from undercooked pig meat is a relevant source for
T. gondii human infection.174 Most pigs are infected by ingesting oocysts,175 but due to their omnivorous
behavior, the ingestion of birds and rodents with T. gondii cysts is important for the cycle.1,175 Pig infec-
tion has a direct correlation with the environment and animal management.176 Low management, access
of other animals to where food and water is offered to pigs, and low control of rodent access are directly
related to the prevalence of T. gondii infection in pig-breeding farms, especially the noncertified ones.177
In pigs, cysts can be found in organs such as the brain, lungs, liver, and spleen, with the possible occur-
rence of retinopathy.178 In addition, these animals do not usually develop severe pathological clinical
manifestations when infected with T. gondii if the immune system is not compromised, similar to what
occurs in humans with competent immune system.110
Although there is potential use of the pig model for T. gondii infection involving the reproductive
system, only a few groups have been working on this issue. The fact that these animals develop congeni-
tal infection strengthens the advantages of their experimental use. In naturally infected pregnant pigs,
there may be damage to the placenta,179 encephalitis, liver necrosis, and pneumonia in fetuses.180 Similar
changes may occur in human fetuses as a result of parasitemia and placental migration, particularly if the
mother has contact with the parasite for the first time during pregnancy.181 In addition, infective tachyzo-
ites of T. gondii can be found in porcine semen for up to 49 days postinfection, demonstrating a potential
for sexual transmission in this species.182 Surprisingly, the hypothesis of congenital toxoplasmosis as a
consequence of infection through human semen has also been recently proposed.183
An aspect evaluated via experimental infection with T. gondii in swine relates to its presence in blood.
Biological evidence in infected mice demonstrates the viability of this parasite in the pig blood, which
can be observed in neutrophils and monocytes, as well as outside these cells.110 In fact, the parasitemia
is persistent in pigs, and an accurate observation can detect tachyzoites for up to 63 days postinfection in
pigs that show no clinical symptoms.110
Some studies have demonstrated important clinical and hematological alterations in pigs infected with
T. gondii. However, such studies typically use highly infective parasite inoculum or very young ani-
mals,184 factors that possibly enhance the pathology.110 In our study, pigs with developed immune system
were infected with parasite dosages close to a natural infection with the ME-49 strain, and typical clini-
cal manifestations of acute toxoplasmosis were observed, such as general apathy, low fever, and, rarely,
lymphadenopathy. Diarrhea was not observed, although the possibility exists and may depend on the
strain and the infecting dose.184 When orally infected, an increase in the number of band neutrophil fol-
lowed by a tendency to monocytopenia was observed. Under these experimental infection conditions, no
anemia or thrombocytopenia were observed, although liver injury was found as seen by increased serum
alanine aminotransferase readings.110 In addition, a persistent parasitemia without exacerbation of pathol-
ogy was found, which is similar to human toxoplasmosis infections caused by some Brazilian strains.185
This strengthens pig as an animal model to study human toxoplasmosis in immunocompetent patients.
41.2.2.4 Cat
Felids were only discovered as definitive hosts of T. gondii in the last half of the 1960s and in the begin-
ning of the 1970s. Excellent reviews report how the life cycle of T. gondii was discovered.35,186 With the
advent of the serological tests, it was realized that T. gondii infection was disseminated around the globe
in different hosts, including humans. This led to the assumption that transmission through carnivorism
alone could not explain the broad prevalence in the world and how herbivores were being infected; in
addition, transmission by invertebrate vectors was unsuccessful.35,186 Ferguson186 reported that Hutchison
was intrigued by these questions and hypothesized that T. gondii could be fecal–orally transmitted by
domestic animals. By using basic parasitology techniques, cats were fed with brain from infected mice;
the parasite material was purified by flotation from cat feces, stored in water, and then fed to mice that later
became ill with toxoplasmosis.187 This work opened the field to others; the T. gondii oocyst was discov-
ered, the sexual cycle of T. gondii in cats revealed, and the whole life cycle of this parasite solved.34,188–191
The discovery of the T. gondii life cycle explained the broad dissemination of this parasite with rel-
evant importance to the epidemiology of the disease. Oocyst shedding contaminated the environment,
Toxoplasma: Animal and In Vitro Models on Toxoplasmosis 667
ended up infecting herbivores, and was also responsible for human toxoplasma outbreaks by water con-
tamination.192 This led to proper management in farms and reduced the contamination of these ani-
mals.35 In addition, more cats release faster and higher number of oocysts if fed with the bradyzoite form
when compared to the tachyzoites or oocysts.193 This indicates that the natural life cycle of the parasite is
indeed by the transmission to the definitive host by carnivorism of small intermediate hosts (like mice)
containing tissue cysts.
Naturally, the cat became an animal model to better understand the sexual cycle of T. gondii, for oocysts
obtainment for further infection of other hosts, and to answer how the parasite behaves in the definitive
host. Experimental manipulation of the cat with corticosteroids indicated that the immune response is
important to stop oocysts shedding and to control parasite growth in the organs of these animals.194 Young
cats (no older than 12 months) shed more oocysts and also present more parasites in their tissue.195
Simultaneous infection of cats with two distinct strains of T. gondii has been done showing that clas-
sical genetic crosses do occur in this parasite.196 These crosses have been used to create linkage maps of
this parasite,197 identifying 11 chromosomes.198 Later, this technique was used to study virulent factors of
the parasites, and a few have been identified.40 Infected cats have also been used to genetically character-
ize the merozoites forms that multiply in cat enterocytes before gametogenesis. A complete distinct set
of genes was identified in these forms when compared to tachyzoites.37 Thus, the cat is fundamental for
the development of T. gondii biology.
41.2.2.5 Chicken
Chickens are less used in experimental trials compared to the murine model. However, in some specific
cases, chickens are an extremely suitable model for experimental studies.199 The ease in handling, low
maintenance cost, and the prolific characteristics of chickens favor their use in experimentations. In
addition, chickens are the world’s largest population of domestic animals with great economic impor-
tance and, therefore, there is interest in studying them.
Chickens may be 100% infected with T. gondii when raised in backyards, whereas those raised in
free-range organic rearing can present prevalence as high as 50%.200 Because of their feeding habits,
chickens are characterized for being indicators of soil contamination by T. gondii oocysts. Moreover,
these animals may be as important in the biological cycle as mice, due to their greater longevity.200 In
chickens, tissue cysts are more common in muscles than in the brain,1 strengthening the importance of
this animal as a source of human infection.200
Chickens do not normally develop a severe clinical disease in natural infections200 and may not develop
the disease when inoculated with the virulent T. gondii strains for mice.201 However, diarrhea, emacia-
tion, blindness, and sudden death have been reported in chickens naturally infected with T. gondii, with
isolation of the parasite in the heart, liver, brain, and lungs. Interestingly enough, the parasite isolates
were avirulent for mice.202,203
Prior to the discovery of T. gondii life cycle and the importance of oral transmission, studies empha-
sized the parenteral infection route, which was not accompanied by clinical manifestations.202–204 After
this period, oral infections have shown variable results with chickens depending on the strain used.
Chickens inoculated with oocysts of the ME-49 strain (Type II) did not develop any clinical symptoms,
whereas chickens infected with the GT1 strain (Type I) became ill, resulting in 20% (one of five animals)
death.205 In addition, oral infection with oocysts of the E strain caused mild fever only in highly infective
doses.206 Although the relative acute phase of the T. gondii infection in chickens does not result in clinical
manifestations, the chronicity of the disease may cause physiological changes and possible production
losses.207 Chicken infection with the M-7741 strain caused diarrhea and inflammatory cell infiltration in
the intestinal wall, with a strong reduction of the density of myenteric neurons. The chronic phase was
correlated with neuronal death and atrophy of the intestinal wall, causing gastrointestinal disorders.207
The relatively mild acute phase followed by a chronic phase with potential epidemiological impor-
tance indicates that the experimental infection of chickens with T. gondii is indeed an interesting model,
particularly with respect to immunological studies. Therefore, interaction with immune cells from pri-
mary cultures, or with the established cell lines such as the HD11 of the chicken macrophages,70,208 may
generate interesting results.
668 Laboratory Models for Foodborne Infections
41.2.2.6 Fish
T. gondii is known to infect nucleated cells of warm-blooded animals, with mice being the most common
and important intermediate host.73,75 However, some studies have indicated that fish and shellfish may
be involved in the natural infection of different hosts by T. gondii. In China, parasitic DNA was identi-
fied in a small number of crayfish Procambarus clarkii, fish Hypophthalmichthys molitrix, and shrimp
Macrobrachium nipponense.209 The authors indicated that the consumption of raw meat of these animals
may be considered a health threat related to T. gondii infection.209 In Brazil, it was shown that oysters
(Crassostrea rhizophorae) may retain oocysts of T. gondii as these animals filter water, and the parasite
DNA was found in a low percentage of individuals.210 There is a need for more studies on the participa-
tion of these animals as carriers of T. gondii.210 However, the importance of these cold-blooded animals
as an effective intermediate host has not been clearly demonstrated, since the presence of DNA in the
sample does not mean viable parasites. It is necessary to differentiate the participation of a potential host
or parasite carrier from a mechanical vector.
Grizzly bears (Ursus arctos) in Alaska showed a T. gondii serology prevalence up to 37% depending
on the area studied.211 Although this animal feeds on fish, no established relationship with these cold-
blooded animals was established, because toxoplasmosis is also a waterborne disease.6 Thus, one cannot
exclude the infection of these animals through contaminated water.
Experimental studies have demonstrated the inability of T. gondii to persist in tissues of goldfish main-
tained at 37°C.212 However, in appropriate conditions, especially temperature, this can be used as a new
experimental infection model for T. gondii in the zebrafish (Danio rerio).213 Proliferation of T. gondii of
the ME-49 and VEG strains was observed in the cardiac myocytes, endothelium, lumen of peripheral
vessels, liver, spleen, brain, pancreas, ovaries, skeletal musculature, and the eye of this animal infected
after acclimation at 37°C. Two weeks after infection, cystic structures were observed in the brain, sug-
gesting that this model may also be fit for chronic studies. The authors report that the only limitation to
the study of toxoplasmosis in this fish was temperature. This is a promising model because this fish can
be kept at temperatures of 37°C, used in high-throughput screenings for new compound, and be geneti-
cally modified.213
41.3 Concluding Remarks
Toxoplasmosis is a disease with worldwide distribution affecting about 2 × 109 people. First considered a
foodborne disease, it is now also recognized as a waterborne disease. It has been suggested that cultural
customs dictate the way of transmission, especially in regard to the consumption of raw or undercooked
meat. This disease can cause different clinical manifestations, with serious implications to immuno-
compromised patients. The disease also affects a large number of wild and domestic warm-blooded
animals causing great economic losses. The etiological agent of this disease is the obligate intracellular
protozoan Toxoplasma gondii, which infects a vast number of hosts and has an elaborate life cycle that
helps to explain its broad dissemination. With the establishment of T. gondii cell culture, the biological
characteristics of the parasite have been delineated through the infection of primary cells, cell lines, and
transfected cells with distinct strains and mutant parasites. Because of its broad host range, most animal
models are suited to investigate this disease.
With a complex life cycle involving various hosts and a diversity of genotypes made up of multiple
strains, T. gondii is a peculiar parasite. Thus, it is imperative to use different models to better understand
the many distinct steps of infection leading to acute and chronic stages of this disease. In vitro models
are in constant development and yield interesting outcomes, but the obtained results must be confirmed
via in vivo models. To date, no animal model that mimics the human infection by T. gondii exists;
however, recent advances on secreted virulence factors by distinct T. gondii strains and specific antimi-
crobial mechanisms of host cells from different host animals that are strain-dependent have increased
our knowledge and shown how adapted this parasite is. Further research is clearly required to fully elu-
cidate the clinical variations of toxoplasmosis and to unravel if specific microbicidal mechanisms oper-
ate in humans and what parasite genes code for virulence factors that help its evasion of host immune
Toxoplasma: Animal and In Vitro Models on Toxoplasmosis 669
functions. In addition, its genetic diversity also demands that diagnosis and treatment of toxoplasmosis
need to be geographically specific. There is no doubt that the models described here and others to come
will contribute to improved understanding of this important parasite and the disease it causes.
Acknowledgments
The authors would like to thank Andrèa Carvalho César for proofing the manuscript. This study was
supported by the following Brazilian agencies: Conselho Nacional de Desenvolvimento Científico e
Tecnológico (CNPq), Fundação Carlos Chagas Filho de Amparo à Pesquisa do Estado do Rio de Janeiro
(FAPERJ), and Fundação de Coordenação de Pessoal de Nível Superior (CAPES).
REFERENCES
1. Dubey, J. P., and Beattie, C. P., Toxoplasmosis of Animals and Man, Boca Raton, FL: CRC Press,
pp. 1–220, 1988.
2. Robert-Gangneux, F., and Dardé, M, L., Epidemiology of and diagnostic strategies for toxoplasmosis,
Clin. Microbiol. Rev., 25, 264, 2012.
3. Tenter, A. M., Heckeroth, A. R., and Weiss, L. M., Toxoplasma gondii: from animals to humans, Int. J.
Parasitol., 30, 1217, 2000.
4. Dubey, J. P., The history of Toxoplasma gondii—the first 100 years, J. Eukaryot. Microbiol., 55, 467, 2008.
5. Weiss, L. M., and Dubey, J. P., Toxoplasmosis: a history of clinical observations, Int. J. Parasitol., 39,
895, 2009.
6. Jones, J. L., and Dubey, J. P., Waterborne toxoplasmosis: recent developments, Exp. Parasitol., 124,
10, 2010.
7. Bowie, W. R., et al., Outbreak of toxoplasmosis associated with municipal drinking water. The BC
Toxoplasma Investigation Team, Lancet, 350, 173, 1997.
8. Bahia-Oliveira, L. M., et al., Highly endemic, waterborne toxoplasmosis in north Rio de Janeiro state,
Brazil, Emerg. Infect. Dis., 9, 55, 2003.
9. de Moura, L., et al., Waterborne toxoplasmosis, Brazil, from field to gene, Emerg. Infect. Dis., 12, 326, 2006.
10. Vaudaux, J. D., et al., Identification of an atypical strain of Toxoplasma gondii as the cause of a water-
borne outbreak of toxoplasmosis in Santa Isabel do Ivai, Brazil, J. Infect. Dis., 202, 1226, 2010.
11. Diana, J., et al., Migration and maturation of human dendritic cells infected with Toxoplasma gondii
depend on parasite strain type, FEMS Immunol. Med. Microbiol., 42, 321, 2004.
12. Johnson, A. M., Strain-dependent, route of challenge-dependent, murine susceptibility to toxoplasmo-
sis, Z. Parasitenkd., 70, 303, 1984.
13. Dubey, J. P., Lindsay, D. S., and Speer, C. A., Structures of Toxoplasma gondii tachyzoites, bradyzoites,
and sporozoites and biology and development of tissue cysts, Clin. Microbiol. Rev., 11, 267, 1998.
14. Vasconcelos-Santos, D. V., Ocular manifestations of systemic disease: toxoplasmosis, Curr. Opin.
Ophthalmol., 23, 543, 2012.
15. Halonen, S. K., and Weiss, L. M., Toxoplasmosis, Handb. Clin. Neurol., 114, 125, 2013.
16. Demar, M., et al., Acute toxoplasmoses in immunocompetent patients hospitalized in an intensive care
unit in French Guiana, Clin. Microbiol. Infect., 18, E221, 2012.
17. Dubey, J. P., et al., Toxoplasmosis in humans and animals in Brazil: high prevalence, high burden of
disease, and epidemiology, Parasitology, 139, 1375, 2012.
18. Waleska, T. C., et al., Toxoplasmosis and mental retardation. Report of a case control study, Mem. Inst.
Oswaldo Cruz, 88, 253, 1993.
19. Sutterland, A. L., et al., Beyond the association. Toxoplasma gondii in schizophrenia, bipolar disorder,
and addiction: systematic review and meta-analysis, Acta Psychiatr. Scand., 132, 161, 2015.
20. Sibley, L. D., et al., Genetic diversity of Toxoplasma gondii in animals and humans, Philos. Trans. R.
Soc. Lond. B. Biol. Sci., 364, 2749, 2009.
21. Frenkel, J. K., Toxoplasma in and around us, Bioscience, 23, 343, 1973.
22. Torrey, E. F., and Yolken, R. H., Toxoplasma oocysts as a public health problem, Trends Parasitol., 29,
380, 2013.
670 Laboratory Models for Foodborne Infections
23. Levine, N. D., The Protozoan Phylum Apicomplexa, Boca Raton, FL: CRC Press, 1988.
24. Black, M. W., and Boothroyd, J. C., Lytic cycle of Toxoplasma gondii, Microbiol. Mol. Biol. Rev., 64,
607, 2000.
25. Shimada, K., et al., Cyst formation by Toxoplasma gondii (RH strain) in vitro. The role of immunologic
mechanisms, Arch. Ophthalmol., 92, 496, 1974.
26. Jones, T. C., Bienz, K. A., and Erb, P., In vitro cultivation of Toxoplasma gondii cysts in astrocytes in
the presence of γ-interferon, Infect. Immun., 51, 147, 1986.
27. Bohne, W., Holpert, M., and Gross, U., Stage differentiation of the protozoan parasite Toxoplasma gon-
dii, Immunobiology, 201, 248, 1999.
28. Skariah, S., Mcintyre, M. K., and Mordue, D. G., Toxoplasma gondii: determinants of tachyzoite to
bradyzoite conversion, Parasitol. Res., 107, 253, 2010.
29. Fischer, H. G., et al., Host cells of Toxoplasma gondii encystation in infected primary culture from
mouse brain, Parasitol. Res., 83, 637, 1997.
30. Lüder, C. G., et al., Toxoplasma gondii in primary rat CNS cells: differential contribution of neu-
rons, astrocytes, and microglial cells for the intracerebral development and stage differentiation, Exp.
Parasitol., 93, 23, 1999.
31. Ferreira da Silva Mda, F., et al., Stress-related and spontaneous stage differentiation of Toxoplasma
gondii, Mol. Biosyst., 4, 824, 2008.
32. Ferreira da Silva Mda, F., et al., Spontaneous stage differentiation of mouse-virulent Toxoplasma gondii RH
parasites in skeletal muscle cells: an ultrastructural evaluation, Mem. Inst. Oswaldo Cruz, 104, 196, 2009.
33. Dubey, J. P., Tissue cyst tropism in Toxoplasma gondii: a comparison of tissue cyst formation in organs
of cats, and rodents fed oocysts, Parasitology, 115, 15, 1997.
34. Frenkel, J. K., Dubey, J. P., and Miller, N. L., Toxoplasma gondii in cats: fecal stages identified as coc-
cidian oocysts, Science, 167, 893, 1970.
35. Dubey, J. P., History of the discovery of the life cycle of Toxoplasma gondii, Int. J. Parasitol., 39, 877, 2009.
36. Fayer, R., Toxoplasmosis update and public health implications, Can. Vet. J., 22, 344, 1981.
37. Hehl, A. B., et al., Asexual expansion of Toxoplasma gondii merozoites is distinct from tachyzoites and
entails expression of non-overlapping gene families to attach, invade, and replicate within feline entero-
cytes, BMC Genomics, 16, 66, 2015.
38. Speer, C. A., and Dubey, J. P., Ultrastructure of early stages of infections in mice fed Toxoplasma gondii
oocysts, Parasitology, 116, 35, 1998.
39. Dubey, J. P., et al., Genetic diversity of Toxoplasma gondii isolates from chickens from Brazil, Vet.
Parasitol., 157, 299, 2008.
40. Hunter, C. A., and Sibley, L. D., Modulation of innate immunity by Toxoplasma gondii virulence effec-
tors, Nat. Rev. Microbiol., 10, 766, 2012.
41. Howe, D. K., and Sibley, L. D., Toxoplasma gondii comprises three clonal lineages: correlation of para-
site genotype with human diseases, J. Infect. Dis., 172, 1561, 2005.
42. Shwab, E. K., et al., Geographical patterns of Toxoplasma gondii genetic diversity revealed by multilo-
cus PCR-RFLP genotyping, Parasitology, 141, 453, 2014.
43. Cañón-Franco, W. A., et al., An overview of seventy years of research (1944–2014) on toxoplasmosis in
Colombia, South America, Parasite Vectors, 7, 427, 2014.
44. Splendore, A., Um nuovo protozoa parassita dei conigli incontrato nelle lesioni anatomiche duna malat-
tia che ricorda in molti ponti il kala-azar delluomo, Rev. Soc. Sci. São Paulo, 3, 109, 1908.
45. Nicolle, C., and Manceaux, L., Sur une infection a corps de Leishman (ou organisms voisins) du gondi,
C. R. Acad. Sci., 147, 763, 1908.
46. Ferguson, D. J., Toxoplasma gondii: 1908–2008, homage to Nicolle, Manceaux and Splendore, Mem.
Inst. Oswaldo Cruz, 104, 133, 2009.
47. Carrel, A., and Montrose, T. B., Cultivation of tissues in vitro and its technique, J. Exp. Med., 13, 387, 1911.
48. Sabin, A. B., and Olitsky, P. K., Toxoplasma and obligate intracellular parasitism, Science, 85, 336, 1937.
49. Beverley, J. K., and Fry, B. A., Sulphadimidine, pyrimethamine and dapsone in the treatment of toxo-
plasmosis in mice, Br. J. Pharmacol. Chemother., 12, 189, 1957.
50. Maloney, E. D., and Kaufman, H. E., Multiplication and therapy of Toxoplasma gondii in tissue culture,
J. Bacteriol., 88, 319, 1964.
51. Summers, W. A., Antagonism of sulfonamide inhibition by para-aminobenzoic acid and folic acid in
Toxoplasma infected mice, Proc. Soc. Exp. Biol. Med., 66, 509, 1947.
Toxoplasma: Animal and In Vitro Models on Toxoplasmosis 671
52. Wolf, A., Cowen, D., and Paige, B., Human toxoplasmosis: occurrence in infants as an encephalomyeli-
tis verification by transmission to animals, Science, 89, 226, 1939.
53. Wolf, A., Cowen, D., and Paige, B. H., Toxoplasmic encephalomyelitis: IV. Experimental transmission
of the infection to animals from a human infant, J. Exp. Med., 71, 187, 1940.
54. Nakayama, I., and Matsubayashi, H., Experimental transmission of Toxoplasma gondii in mice, Keio J.
Med., 10, 163, 1961.
55. Guimarães, F. N., and Meyer, H. Cultivo de “Toxoplasma” Nicolle & Manceaux, 1909, em cultura de
tecidos, Ver. Bras. Biol., 2, 123, 1942.
56. Jacobs, L., Propagation, morphology, and biology of Toxoplasma, Ann. N. Y. Acad. Sci., 64, 154,
1956.
57. de Souza, W., DaMatta, R. A., and Attias, M., Brazilian contribution for a better knowledge on the biol-
ogy of Toxoplasma gondii, Mem. Inst. Oswaldo Cruz, 104, 149, 2009.
58. Jiménez-Ruiz, E., et al., Advantages and disadvantages of conditional systems for characterization of
essential genes in Toxoplasma gondii, Parasitology, 141, 1390, 2014.
59. Hartl, D. L., and Jones, E. W., Genetics: Principles and Analysis, Boston, MA: Jones and Bartlett
Publishers, 1998.
60. Jones, T. C., and Hirsch, J. G., The interaction between Toxoplasma gondii and mammalian cells: II.
The absence of lysosomal fusion with phagocytic vacuoles containing living parasites, J. Exp. Med.,
136, 1173, 1972.
61. Jones, T. C., Yeh, S., and Hirsch, J. G., The interaction between Toxoplasma gondii and mammalian
cells: I. Mechanism of entry and intracellular fate of the parasite, J. Exp. Med., 136, 1157, 1972.
62. Sibley, L. D., Weidner, E., and Krahenbuhl, J. L., Phagosome acidification blocked by intracellular
Toxoplasma gondii, Nature, 315, 416, 1985.
63. Sinai, A. P., Biogenesis of and activities at the Toxoplasma gondii parasitophorous vacuole membrane,
Subcell. Biochem., 47, 155, 2008.
64. Boyle, J. P., and Radke, J. R., A history of studies that examine the interactions of Toxoplasma with its
host cell: emphasis on in vitro models, Int. J. Parasitol., 39, 903, 2009.
65. Murray, H. W., and Cohn, Z. A., Macrophage oxygen-dependent antimicrobial activity. I. Susceptibility
of Toxoplasma gondii to oxygen intermediates, J. Exp. Med., 150, 938, 1979.
66. Sibley, L. D., Lawson, R., and Weidner, E., Superoxide dismutase and catalase in Toxoplasma gondii,
Mol. Biochem. Parasitol., 19, 83, 1986.
67. Ding, M., et al., The antioxidant systems in Toxoplasma gondii and the role of cytosolic catalase in
defence against oxidative injury, Mol. Microbiol., 51, 47, 2004.
68. Wilson, C. B., Tsai, V., and Remington, J. S., Failure to trigger the oxidative burst by normal macro-
phages. Possible mechanism for survival of intracellular pathogens, J. Exp. Med., 151, 328, 1980.
69. Murray, H. W., and Cohn, Z. A., Macrophage oxygen-dependent antimicrobial activity. III. Enhanced
oxidative metabolism as an expression of macrophage activation, J. Exp. Med., 152, 1596, 1980.
70. Guillermo, L. V., and DaMatta, R. A., Nitric oxide inhibition after Toxoplasma gondii infection of
chicken macrophage cell lines, Poult. Sci., 83, 776, 2004.
71. Shrestha, S. P., et al., Proliferation of Toxoplasma gondii in inflammatory macrophages in vivo is associ-
ated with diminished oxygen radical production in the host cell, Int. J. Parasitol., 36, 433, 2006.
72. Collazo, C. M., et al., Inactivation of LRG-47 and IRG-47 reveals a family of interferon γ-inducible
genes with essential, pathogen-specific roles in resistance to infection, J. Exp. Med., 194, 181, 2001.
73. Howard, J. C., Hunn, J. P., and Steinfeldt, T., The IRG protein-based resistance mechanism in mice and
its relation to virulence in Toxoplasma gondii, Curr. Opin. Microbiol., 14, 414, 2011.
74. Liesenfeld, O., et al., The IFN-γ-inducible GTPase, Irga6, protects mice against Toxoplasma gondii but
not against Plasmodium berghei and some other intracellular pathogens, PLoS One, 6, e20568, 2011.
75. Gazzinelli, R. T., et al., Innate resistance against Toxoplasma gondii: an evolutionary tale of mice, cats,
and men, Cell Host Microbes, 15, 132, 2014.
76. Fentress SJ, and Sibley, L. D., The secreted kinase ROP18 defends Toxoplasma’s border, Bioessays, 33,
693, 2011.
77. Khan, A., et al., Selection at a single locus leads to widespread expansion of Toxoplasma gondii lineages
that are virulent in mice, PLoS Genet., 5, e1000404, 2009.
78. Reese, M. L., et al., Polymorphic family of injected pseudokinases is paramount in Toxoplasma viru-
lence, Proc. Natl. Acad. Sci. USA, 108, 9625, 2011.
672 Laboratory Models for Foodborne Infections
79. Steinfeldt, T., et al., Phosphorylation of mouse immunity-related GTPase (IRG) resistance proteins is an
evasion strategy for virulent Toxoplasma gondii, PLoS Biol., 8, e1000576, 2010.
80. Etheridge, R. D., et al., The Toxoplasma pseudokinase ROP5 forms complexes with ROP18 and ROP17
kinases that synergize to control acute virulence in mice, Cell Host Microbes, 15, 537, 2014.
81. Alaganan, A., et al., Toxoplasma GRA7 effector increases turnover of immunity-related GTPases and
contributes to acute virulence in the mouse, Proc. Natl. Acad. Sci. USA, 111, 1126, 2014.
82. Da Gama, L. M., et al., Reduction in adhesiveness to extracellular matrix components, modulation of
adhesion molecules and in vivo migration of murine macrophages infected with Toxoplasma gondii,
Microbes Infect., 6, 1287, 2004.
83. Lambert, H., et al., The Toxoplasma gondii-shuttling function of dendritic cells is linked to the parasite
genotype, Infect. Immun., 77, 1679, 2009.
84. Seipel, D., et al., Toxoplasma gondii infection positively modulates the macrophages migratory molecu-
lar complex by increasing matrix metalloproteinases, CD44 and alpha v beta 3 integrin, Vet. Parasitol.,
169, 312, 2010.
85. Seipel, D., et al., Monocytes/macrophages infected with Toxoplasma gondii do not increase co-stimulatory
molecules while maintaining their migratory ability, APMIS, 117, 672, 2009.
86. Schuindt, S. H., et al., Secretion of multi-protein migratory complex induced by Toxoplasma gondii
infection in macrophages involves the uPA/uPAR activation system, Vet. Parasitol., 186, 207, 2012.
87. MacMicking, J., Xie, Q. W., and Nathan, C., Nitric oxide and macrophage function, Annu. Rev.
Immunol., 15, 323, 1997.
88. Bogdan, C., Nitric oxide synthase in innate and adaptive immunity: an update, Trends Immunol., 36,
161, 2015.
89. Hayashi, S., et al., Contribution of nitric oxide to the host parasite equilibrium in toxoplasmosis, J.
Immunol., 156, 1476, 1996.
90. Khan, I. A., et al., A dichotomous role for nitric oxide during acute Toxoplasma gondii infection in mice,
Proc. Natl. Acad. Sci. USA, 94, 13955, 1997.
91. Scharton-Kersten, T. M., et al., Inducible nitric oxide is essential for host control of persistent but not
acute infection with the intracellular pathogen Toxoplasma gondii, J. Exp. Med., 185, 1261, 1997.
92. Liesenfeld, O., et al., TNF-α, nitric oxide and IFN-γ are all critical for development of necrosis in the
small intestine and early mortality in genetically susceptible mice infected perorally with Toxoplasma
gondii, Parasite Immunol., 21, 365, 1999.
93. Adams, L. B., et al., Microbiostatic effect of murine-activated macrophages for Toxoplasma gondii.
Role for synthesis of inorganic nitrogen oxides from l-arginine, J. Immunol., 144, 2725, 1990.
94. Nibbering, P. H., et al., Nitrite production by activated murine macrophages correlates with their toxo-
plasmastatic activity, Ia antigen expression, and production of H 2O2, Immunobiology, 184, 93, 1991.
95. DaMatta, R. A., et al., Nitric oxide is not involved in the killing of Trypanosoma cruzi by chicken mac-
rophages, Parasitol. Res., 86, 239, 2000.
96. Seabra, S. H., de Souza, W., and DaMatta, R. A., Toxoplasma gondii partially inhibits nitric oxide pro-
duction of activated murine macrophages, Exp. Parasitol., 100, 62, 2002.
97. Seabra, S. H., de Souza, W., and DaMatta, R. A., Toxoplasma gondii exposes phosphatidylserine induc-
ing a TGF-beta1 autocrine effect orchestrating macrophage evasion, Biochem. Biophys. Res. Commun.,
324, 744, 2004.
98. Santos, T. A., et al., Phosphatidylserine exposure by Toxoplasma gondii is fundamental to balance the
immune response granting survival of the parasite and of the host, PLoS One, 6, e27867, 2011.
99. Padrão, J. C., et al., Toxoplasma gondii infection of activated J774-A1 macrophages causes inducible
nitric oxide synthase degradation by the proteasome pathway, Parasitol. Int., 63, 659, 2014.
100. de Freitas Balanco, J. M., et al., Apoptotic mimicry by an obligate intracellular parasite downregulates
macrophage microbicidal activity, Curr. Biol., 11, 1870, 2001.
101. Wanderley, J. L., and Barcinski, M. A., Apoptosis and apoptotic mimicry: the Leishmania connection,
Cell Mol. Life Sci., 67, 1653, 2010.
102. Guimarães, E. V., de Carvalho, L., and Barbosa, H. S., Primary culture of skeletal muscle cells as a
model for studies of Toxoplasma gondii cystogenesis, J. Parasitol., 94, 72, 2008.
103. Andrade, E. F., et al., Do microtubules around the Toxoplasma gondii-containing parasitophorous vacu-
ole in skeletal muscle cells form a barrier for the phagolysosomal fusion? J. Submicrosc. Cytol. Pathol.,
33, 337, 2001.
Toxoplasma: Animal and In Vitro Models on Toxoplasmosis 673
104. Takács, A. C., Swierzy, I. J., and Lüder, C. G., Interferon-γ restricts Toxoplasma gondii development in
murine skeletal muscle cells via nitric oxide production and immunity-related GTPases, PLoS One, 7,
e45440, 2012.
105. Gomes, A. F., et al., Toxoplasma gondii-skeletal muscle cells interaction increases lipid droplet biogen-
esis and positively modulates the production of IL-12, IFN-g and PGE2, Parasite Vectors, 7, 47, 2014.
106. Mota, L. A., et al., Culture of mouse peritoneal macrophages with mouse serum induces lipid bod-
ies that associate with the parasitophorous vacuole and decrease their microbicidal capacity against
Toxoplasma gondii, Mem. Inst. Oswaldo Cruz, 109, 767, 2014.
107. de Muno, R. M., et al., Spontaneous cystogenesis of Toxoplasma gondii in feline epithelial cells in vitro,
Folia Parasitol. (Praha), 61, 113, 2014.
108. Moura, Mde. A., Amendoeira, M. R., and Barbosa, H. S., Primary culture of intestinal epithelial cells as
a potential model for Toxoplasma gondii enteric cycle studies, Mem. Inst. Oswaldo Cruz, 104, 862, 2009.
109. Lind, P., et al., The time course of the specific antibody response by various ELISAs in pigs experimen-
tally infected with Toxoplasma gondii, Vet. Parasitol., 71, 1, 1997.
110. Miranda, F. J., et al., Experimental infection with the Toxoplasma gondii ME-49 strain in the Brazilian
BR-1 mini pig is a suitable animal model for human toxoplasmosis, Mem. Inst. Oswaldo Cruz, 110, 95,
2015.
111. Dias, R. R., et al., Toxoplasma gondii oral infection induces intestinal inflammation and retinochoroidi-
tis in mice genetically selected for immune oral tolerance resistance, PLoS One, 9(12), e113374, 2014.
112. Lin, M. H., et al., Real-time PCR for quantitative detection of Toxoplasma gondii, J. Clin. Microbiol.,
38, 4121, 2000.
113. Meerburg, B. G., Singleton, G. R., and Kijlstra, A., Rodent-borne diseases and their risks for public
health, Crit. Rev. Microbiol., 35, 221, 2009.
114. Dabritz, H. A., et al., Risk factors for Toxoplasma gondii infection in wild rodents from central coastal
California and a review of T. gondii prevalence in rodents, J. Parasitol., 94, 675, 2008.
115. Hide, G., et al., Evidence for high levels of vertical transmission in Toxoplasma gondii, Parasitology,
136, 1877, 2009.
116. Lilue, J., et al., Reciprocal virulence and resistance polymorphism in the relationship between
Toxoplasma gondii and the house mouse, Elife, 2, e01298, 2013.
117. Lélu, M., et al., When should a trophically and vertically transmitted parasite manipulate its intermedi-
ate host? The case of Toxoplasma gondii, Proc. Biol. Sci., 280, 20131143, 2013.
118. Yap, G. S., and Sher, A., The use of germ line-mutated mice in understanding host-pathogen interac-
tions, Cell Microbiol., 4, 627, 2002.
119. Subauste, C. S., and Remington, J. S., Role of γ interferon in Toxoplasma gondii infection, Eur. J. Clin.
Microbiol. Infect. Dis., 10, 58, 1991.
120. McCabe, R. E., Luft, B. J., and Remington, J. S., Effect of murine interferon-γ on murine toxoplasmosis,
J. Infect. Dis., 150, 961, 1984.
121. Suzuki, Y., et al., Interferon-γ: the major mediator of resistance against Toxoplasma gondii, Science,
240, 516, 1988.
122. Scharton-Kersten, T. M., et al., In the absence of endogenous IFN-γ, mice develop unimpaired IL-12
responses to Toxoplasma gondii while failing to control acute infection, J. Immunol., 157, 4045,
1996.
123. Hunter, C. A., et al., Studies on the role of interleukin-12 in acute murine toxoplasmosis, Immunology,
84, 16, 1995.
124. Hunter, C. A., Chizzonite, R., and Remington, J. S., IL-1 beta is required for IL-12 to induce production
of IFN-γ by NK cells. A role for IL-1 beta in the T cell-independent mechanism of resistance against
intracellular pathogens, J. Immunol., 155, 4347, 1995.
125. Scharton-Kersten, T., et al., Interferon consensus sequence binding protein-deficient mice display
impaired resistance to intracellular infection due to a primary defect in interleukin 12 p40 induction, J.
Exp. Med., 186, 1523, 1997.
126. Gazzinelli, R. T., et al., In the absence of endogenous IL-10, mice acutely infected with Toxoplasma
gondii succumb to a lethal immune response dependent on CD4+ T cells and accompanied by overpro-
duction of IL-12, IFN-γ and TNF-α, J. Immunol., 157, 798, 1996.
127. Scanga, C. A., et al., Cutting edge: MyD88 is required for resistance to Toxoplasma gondii infection and
regulates parasite-induced IL-12 production by dendritic cells, J. Immunol., 168, 5997, 2002.
674 Laboratory Models for Foodborne Infections
128. Yarovinsky, F., and Sher, A., Toll-like receptor recognition of Toxoplasma gondii, Int. J. Parasitol., 36,
255, 2006.
129. Liesenfeld, O., Oral infection of C57BL/6 mice with Toxoplasma gondii: a new model of inflammatory
bowel disease? J. Infect. Dis., 185 Suppl 1, S96, 2002.
130. Schreiner, M., and Liesenfeld, O., Small intestinal inflammation following oral infection with
Toxoplasma gondii does not occur exclusively in C57BL/6 mice: review of 70 reports from the litera-
ture, Mem. Inst. Oswaldo Cruz, 104, 221, 2009.
131. Heimesaat, M. M., et al., Gram-negative bacteria aggravate murine small intestinal Th1-type immuno-
pathology following oral infection with Toxoplasma gondii, J. Immunol., 177, 8785, 2006.
132. Muñoz, M., et al., Interleukin (IL)-23 mediates Toxoplasma gondii-induced immunopathology in the
gut via matrixmetalloproteinase-2 and IL-22 but independent of IL-17, J. Exp. Med., 206, 3047, 2009.
133. Cavalcanti, M. G., et al., MIF participates in Toxoplasma gondii-induced pathology following oral
infection, PLoS One, 6(9), e25259, 2011.
134. Guiton, R., et al., Interleukin 17 receptor signaling is deleterious during Toxoplasma gondii infection in
susceptible BL6 mice, J. Infect. Dis., 202, 427, 2010.
135. Passos, S. T., et al., IL-6 promotes NK cell production of IL-17 during toxoplasmosis, J. Immunol., 184,
1776, 2010.
136. Raetz, M., et al., Parasite-induced TH1 cells and intestinal dysbiosis cooperate in IFN-γ-dependent
elimination of Paneth cells, Nat. Immunol., 14, 136, 2013.
137. Molloy, M. J., et al., Intraluminal containment of commensal outgrowth in the gut during infection-
induced dysbiosis, Cell Host Microbes, 14, 318, 2013.
138. Abi Abdallah, D. S., and Denkers, E. Y., Neutrophils cast extracellular traps in response to protozoan
parasites, Front. Immunol., 3, 382, 2012.
139. Mouse Nomenclature Home Page, http://www.informatics.jax.org/mgihome/nomen/strains.shtml
(accessed September 2015).
140. Dubey, J. P., and Frenkel, J. K., Toxoplasmosis of rats: a review, with considerations of their value as an
animal model and their possible role in epidemiology, Vet. Parasitol., 77, 1, 1998.
141. Chinchilla, M., Alfaro, M., and Guerrero, O. M., Natural adaptation of the white rat to Toxoplasma
gondii, Rev. Biol. Trop., 29, 273, 1981.
142. Chinchilla, M., Guerrero, O. M., and Solano, E., Lack of multiplication of Toxoplasma in macrophages
of rats in vitro, J. Parasitol., 68, 952, 1982.
143. Dubey, J. P., Pathogenicity and infectivity of Toxoplasma gondii oocysts for rats, J. Parasitol., 82, 951, 1996.
144. Freyre, A., et al., Toxoplasma gondii: an improved rat model of congenital infection, Exp. Parasitol.,
120, 142, 2008.
145. Zenner, L., et al., Rat model of congenital toxoplasmosis: rate of transmission of three Toxoplasma
gondii strains to fetuses and protective effect of a chronic infection, Infect. Immun., 61, 360, 1993.
146. Kempf, M. C., et al., Different manifestations of Toxoplasma gondii infection in F344 and LEW rats,
Med. Microbiol. Immunol., 187, 137, 1999.
147. Sergent, V., et al., Innate refractoriness of the Lewis rat to toxoplasmosis is a dominant trait that is intrin-
sic to bone marrow-derived cells, Infect. Immun., 73, 6990, 2005.
148. Cavaillès, P., et al., The rat Toxo1 locus directs toxoplasmosis outcome and controls parasite prolifera-
tion and spreading by macrophage-dependent mechanisms, Proc. Natl. Acad. Sci. USA, 103, 744, 2006.
149. Cavaillès, P., et al., A highly conserved Toxo1 haplotype directs resistance to toxoplasmosis and its asso-
ciated caspase-1 dependent killing of parasite and host macrophage, PLoS Pathog., 10(4), e1004005,
2014.
150. Cirelli, K. M., et al., Inflammasome sensor NLRP1 controls rat macrophage susceptibility to Toxoplasma
gondii, PLoS Pathog., 10(3), e1003927, 2014.
151. Lamkanfi, M., and Dixit, V. M., Mechanisms and functions of inflammasomes, Cell, 157, 1013, 2014.
152. Ewald, S. E., Chavarria-Smith, J., Boothroyd, J. C., NLRP1 is an inflammasome sensor for Toxoplasma
gondii, Infect. Immun., 82, 460, 2014.
153. Gorfu, G., et al., Dual role for inflammasome sensors NLRP1 and NLRP3 in murine resistance to
Toxoplasma gondii, mBio, 5(1), pii, e01117-13, 2014.
154. Witola, W. H., et al., ALOX12 in human toxoplasmosis, Infect. Immun., 82, 2670, 2014.
155. Witola, W. H., et al., NALP1 influences susceptibility to human congenital toxoplasmosis, proinflammatory
cytokine response, and fate of Toxoplasma gondii-infected monocytic cells, Infect. Immun., 79, 756, 2011.
Toxoplasma: Animal and In Vitro Models on Toxoplasmosis 675
156. Gov, L., et al., Human innate immunity to Toxoplasma gondii is mediated by host caspase-1 and ASC
and parasite GRA15, mBio, 4(4) pii, e00255-13, 2013.
157. Bonvicino, C. R., Lima, J. F. S., and Almeida, F. C., A new species of Calomys Waterhouse (Rodentia,
Sigmodontinae) from the cerrado of central Brazil, Rev. Bras. Zool., 20, 301, 2003.
158. Hershkovitz, P., The evolution of mammals of the neotropical region. A zoogeographic and ecological
review, Q. Ver. Biol., 44, 1, 1969.
159. Mello, A. D., Calomys callosus Rengger, 1830 (Rodentia-Cricetidadae): Sua caracterização, distri-
buição, biologia, criação e manejo de uma cepa em laboratório, Mem. Inst. Oswaldo Cruz, 79, 37, 1984.
160. Dubey, J. P., et al., Characterization of Toxoplasma gondii isolates in free-range chickens from Amazon,
Brazil, J. Parasitol., 92, 36, 2006.
161. Franco, P. S., et al., Experimental infection of Calomys callosus with atypical strains of Toxoplasma
gondii shows gender differences in severity of infection, Parasitol. Res., 113, 2655, 2014.
162. Favoreto-Junior, S., et al., Experimental Infection of Calomys callosus (Rodentia, Cricetidae) by
Toxoplasma gondii, Mem. Inst. Oswaldo Cruz, 93, 103, 1998.
163. Ferro, E. A. V., et al., Callomys callosus (Rodentia: Cricetidae) trophoblast cells as host cells to
Toxoplasma gondii in early pregnancy, Parasitol. Res., 85, 647, 1999.
164. Ferro, E. A. V., et al., Effect of Toxoplasma gondii infection kinetics on trophoblast cell population in
Calomys callosus, a model of congenital toxoplasmosis, Infect. Immun., 70, 7089, 2002.
165. Franco, P. S., et al., Evaluation of vertical transmission of Toxoplasma gondii in Calomys callosus
model after reinfection with heterologous and virulent strain, Placenta, 32, 116, 2011.
166. Franco, P. S., et al., Calomys callosus chronically infected by Toxoplasma gondii clonal type II strain and
reinfected by Brazilian strains is not able to prevent vertical transmission, Front. Microbiol., 6, 181, 2015.
167. Barbosa, B. F., et al., Susceptibility to vertical transmission of Toxoplasma gondii is temporally depen-
dent on the preconceptional infection in Calomys callosus, Placenta, 28, 624, 2007.
168. Ferreira, G. L. S, et al., Toxoplasma gondii and mast cell interactions in vivo and in vitro: experimental
infection approaches in Calomys callosus (Rodentia, Cricetidae), Microbes Infect., 6, 172, 2004.
169. Gil, C. D., et al., Mast cells in the eyes of Calomys callosus (Rodentia: Cricetidae) infected by
Toxoplasma gondii, Parasitol. Res., 88, 557, 2002.
170. Pereira, M. F., et al., Acquired and congenital ocular toxoplasmosis experimentally induced in Calomys
callosus (Rodentia, Cricetidae), Mem. Inst. Oswaldo Cruz, 1, 103, 1999.
171. Pearce, A. I., et al., Animal models for implant biomaterial research in bone: a review, Eur. Cell. Mater.,
13, 1, 2007.
172. Meurens, F., et al., The pig: a model for human infectious diseases, Trends Microbiol., 20, 50, 2012.
173. Vodicka, P., et al., The miniature pig as an animal model in biomedical research, Ann. N. Y. Acad. Sci.,
1049, 161, 2005.
174. Dubey, J. P., Toxoplasmosis, J. Am. Vet. Med. Assoc., 189, 166, 1986.
175. Penkert, R. A., Possible spread of toxoplasmosis by feed contaminated for cats, J. Am. Vet. Med. Assoc.,
162, 924, 1973.
176. Kijlstra, A., and Jongert, E., Toxoplasma-safe meat: close to reality? Trends Parasitol., 25, 18, 2009.
177. Piassa, F. R., et al., Prevalence and risk factors for Toxoplasma gondii infection in certified and non-
certified pig breeding farms in the Toledo microregion, PR, Brazil, Rev. Bras. Parasitol. Vet., 19, 152, 2010.
178. Giraldi, N., et al., Estudo da toxoplasmose congênita natural em granjas de suínos em Londrina, PR,
Arq. Bras. Med. Vet. Zootec., 48, 83, 1996.
179. Chang, G. N., et al., Serological survey of swine toxoplasmosis in Taiwan, Southeast Asian J. Trop. Med.
Public Health., 22 Suppl, 111, 1991.
180. Haritani, M., et al., Demonstration of Toxoplasma gondii antigen in stillborn piglets using immunoper-
oxidase technique, Nihon Juigaku Zasshi, 50, 954, 1988.
181. Lopes, F. M. R., et al., Toxoplasma gondii infection in pregnancy, Braz. J. Infect. Dis., 11, 496, 2007.
182. Moura, A. B., et al., Occurrence of anti-Toxoplasma gondii antibodies in swine and ovine slaughtered at
municipality of Guarapuava in the State of Paraná, Brazil, Rev. Bras. Parasitol. Vet., 16, 54, 2007.
183. Flegr, J., Klapilová, K., and Kaňková, S., Toxoplasmosis can be a sexually transmitted infection with
serious clinical consequences. Not all routes of infection are created equal, Med. Hypotheses, 83, 286,
2014.
184. Wingstrand, A., et al., Clinical observations, pathology, bioassay in mice and serological response at
slaughter in pigs experimentally infected with Toxoplasma gondii, Vet. Parasitol., 72, 129, 1997.
676 Laboratory Models for Foodborne Infections
185. Silveira, C., et al., Toxoplasma gondii in the peripheral blood of patients with acute and chronic toxo-
plasmosis, Br. J. Ophthalmol., 95, 396, 2011.
186. Ferguson, D. J., Identification of faecal transmission of Toxoplasma gondii: small science, large charac-
ters, Int. J. Parasitol., 39, 871, 2009.
187. Hutchison, W. M., Experimental transmission of Toxoplasma gondii, Nature, 206, 961, 1965.
188. Hutchison, W. M., et al., Coccidian-like nature of Toxoplasma gondii, Br. Med. J., 1, 142, 1970.
189. Dubey, J. P., Miller, N. L., and Frenkel, J. K., The Toxoplasma gondii oocyst from cat feces, J. Exp.
Med., 132, 636, 1970.
190. Overdulve, J. P., The identity of Toxoplasma Nicolle and Manceaux, 1909 with Isospora Schneider,
1881. I. Proc. K. Ned. Akad. Wet. C., 73, 129, 1970.
191. Sheffield, H. G, and Melton, M. L., Toxoplasma gondii: the oocyst, sporozoite, and infection of cultured
cells, Science, 167, 892, 1970.
192. Dubey, J. P., Advances in the life cycle of Toxoplasma gondii, Int. J. Parasitol., 28, 1019, 1998.
193. Dubey, J. P., and Frenkel, J. K., Feline toxoplasmosis from acutely infected mice and the development of
Toxoplasma cysts, J. Protozool., 23, 537, 1976.
194. Dubey, J. P., and Frenkel, J. K., Immunity to feline toxoplasmosis: modification by administration of
corticosteroids, Vet. Pathol., 11, 350, 1974.
195. Dubey, J. P., Hoover, E. A., and Walls, K. W., Effect of age and sex on the acquisition of immunity to
toxoplasmosis in cats, J. Protozool., 24, 184, 1977.
196. Pfefferkorn, E. R., and Kasper, L. H., Toxoplasma gondii: genetic crosses reveal phenotypic suppression
of hydroxyurea resistance by fluorodeoxyuridine resistance, Exp. Parasitol., 55, 207, 1983.
197. Sibley, L. D., et al., Generation of a restriction fragment length polymorphism linkage map for
Toxoplasma gondii, Genetics, 132, 1003, 1992.
198. Sibley, L. D., and Boothroyd, J. C., Construction of a molecular karyotype for Toxoplasma gondii, Mol.
Biochem. Parasitol., 51, 291, 1992.
199. de Macchi, B. M., et al., Chickens treated with a nitric oxide inhibitor became more resistant to
Plasmodium gallinaceum infection due to reduced anemia, thrombocytopenia and inflammation, Vet.
Res., 44, 8, 2013.
200. Dubey, J. P., Toxoplasma gondii infections in chickens (Gallus domesticus): prevalence, clinical disease,
diagnosis and public health significance, Zoonoses Public Health, 57, 60, 2010.
201. Dubey, J. P., et al., Biological and genetic characterization of Toxoplasma gondii isolates from chickens
(Gallus domesticus) from São Paulo, Brazil: unexpected findings, Int. J. Parasitol., 32, 99, 2002.
202. Erichsen, S., and Harboe, A., Toxoplasmosis in chickens. I. An epidemic outbreak of toxoplasmosis in a
chicken flock in South-Eastern Norway, Acta Pathol. Microbiol. Scand., 33, 381, 1953.
203. Erichsen, S., and Harboe, A., Toxoplasmosis in chickens. II. So-called gliomas observed in chickens
infected with toxoplasms, Acta Pathol. Microbiol. Scand., 33, 381, 1953.
204. Jones, F. E., et al., Experimental toxoplasmosis in chickens, J. Parasitol., 45, 31, 1959.
205. Dubey, J. P., et al., Serologic and parasitologic responses of domestic chickens after oral inoculation
with Toxoplasma gondii oocysts, Am. J. Vet. Res., 54, 1668, 1993.
206. Kaneto, C. N., et al., Experimental toxoplasmosis in broiler chicks, Vet. Parasitol., 69, 203, 1997.
207. Bonapaz, R. S., et al., Effects of infection with Toxoplasma gondii oocysts on the intestinal wall and the
myenteric plexus of chicken (Gallus gallus), Pesq. Vet. Bras., 30, 787, 2010.
208. Ong, Y.C., Boyle, J.P., and Boothroyd, J.C., Strain-dependent host transcriptional responses to
Toxoplasma infection are largely conserved in mammalian and avian hosts, PLoS One, 6(10), e26369,
2011.
209. Zhang, M., et al., Detection of Toxoplasma gondii in shellfish and fish in parts of China, Vet. Parasitol.,
200, 85, 2014.
210. Esmerini, P. O., Gennari, S. M, and Pena, H. F., Analysis of marine bivalve shellfish from the fish market
in Santos City, São Paulo State, Brazil, for Toxoplasma gondii, Vet. Parasitol., 170, 8, 2010.
211. Zarnke, R. L., et al., Serologic survey for Toxoplasma gondii in grizzly bears from Alaska, J. Wildl.
Dis., 33, 267, 1997.
212. Omata, Y., et al., Toxoplasma gondii does not persist in goldfish (Carassius auratus), J. Parasitol., 91,
1496, 2005.
213. Sanders, J. L., et al., The zebrafish, Danio rerio, as a model for Toxoplasma gondii: an initial description
of infection in fish, J. Fish Dis., 38, 675, 2015.
Section VI
Foodborne Infections
due to Helminths
42
Anisakis
CONTENTS
42.1 Introduction................................................................................................................................... 680
42.2 Taxonomy, Life Cycle, and World Distribution of Anisakis Species............................................ 680
42.3 Allergen Nomenclature................................................................................................................. 683
42.4 Pathogenesis, Immunological Response, and Clinical Signs and Symptoms.............................. 683
42.5 Laboratorial Diagnosis.................................................................................................................. 684
42.6 Experimental Models ................................................................................................................... 684
42.6.1 General Considerations.................................................................................................... 684
42.6.2 Guinea Pigs....................................................................................................................... 685
42.6.2.1 Intradermic Route............................................................................................. 685
42.6.2.2 Intraperitoneal Route........................................................................................ 685
42.6.2.3 Intragastric Route.............................................................................................. 685
42.6.3 Pigs................................................................................................................................... 685
42.6.3.1 Oral Route......................................................................................................... 685
42.6.4 Rabbit................................................................................................................................ 686
42.6.4.1 Intragastric Route.............................................................................................. 686
42.6.4.2 Subcutaneous Route.......................................................................................... 686
42.6.4.3 Intramuscular Route......................................................................................... 687
42.6.5 Rats................................................................................................................................... 687
42.6.5.1 Intraperitoneal Larval Implant......................................................................... 687
42.6.5.2 Intragastric Infection........................................................................................ 687
42.6.5.3 In Vivo L3–L4 Transformation Model in Rats................................................. 688
42.6.6 Mice.................................................................................................................................. 688
42.6.6.1 Intraperitoneal Larva Implant........................................................................... 689
42.6.6.2 Intraperitoneal Immunization........................................................................... 689
42.6.6.3 Intragastric Infection........................................................................................ 689
42.6.6.4 Epicutaneous Immunization............................................................................. 690
42.6.6.5 Subcutaneous Immunization............................................................................ 690
42.6.6.6 Intranasal Immunization................................................................................... 690
42.6.6.7 Nematode Molecules as Immunoregulators......................................................691
42.6.7 Fish....................................................................................................................................691
42.6.7.1 Oral Infection.....................................................................................................691
42.6.7.2 Intraperitoneal................................................................................................... 692
42.6.8 In Vitro Cultivation........................................................................................................... 692
42.6.8.1 Culture Media................................................................................................... 693
42.6.8.2 L3–L4 Transformation Model.......................................................................... 693
42.6.9 Larvicidal Models............................................................................................................ 694
42.7 C onclusion..................................................................................................................................... 694
Acknowledgments................................................................................................................................... 695
References............................................................................................................................................... 695
679
680 Laboratory Models for Foodborne Infections
42.1 Introduction
Parasites from the marine environment have historically been overlooked as a risk for human disease
and are thus not in the main stream of basic or clinical investigation, although they can infect humans,
thereby causing anthropozoonosis, and are therefore a public health risk. Within the marine worms
with clinical importance are those pertaining to the Anisakidae family (Anisakis, Pseudoterranova, and
Contracaecum) and Raphidascarididae family (Hysterothylacium) causing anisakidosis.1 Anisakids are
nematodes whose definitive hosts are marine mammals; intermediate hosts are crustaceans (L2), fish,
and cephalopods (L3), and they have a worldwide distribution. Humans become accidental hosts after
ingestion of raw or undercooked infected seafood.2
There is an estimate of 20,000 human cases of anisakidosis, with an annual registration of 2000
new cases. The highest incidence with approximately 90% of all reported cases occurs in Japan,3 prob-
ably owing to the routine habit of eating raw fish in dishes like sushi and sashimi.4 Other countries
that habitually consume raw or undercooked seafood also record an expressive number of cases of the
disease. This is the case of European countries, mainly in the coastal areas of Germany, Netherlands,
and Scandinavian countries that consume salted, pickled, and smoked herring, or Spain, where typical
appetizers are ceviche (fresh seafood marinated in lemon juice) and Boquerón’s en vinegar (pickled
anchovies).1 In the Americas, there has been an increase in the number of reported anisakidosis cases,
probably because of the popularization of oriental cuisine and the consumption of dishes like Lomi-lomi
salmon and ceviche.1,4,5 The improvement of diagnostic methods is probably another explanation for the
increase in the report of new cases.
Since the first descriptions of human cases, the number of researchers who investigate actual and
potential human marine infections has increased, and several animal models have been developed in
order to understand the host–parasite relationship associated with the sensitization of individuals who
accidentally ingest anisakid larvae. In this chapter, we will contextualize several in vitro and in vivo
experimental models that are employed to reproduce and understand the natural history of human dis-
ease and explore the molecular and biological aspects of these parasites.
of the Pseudoterranova genus are more specific having the pinnipeds (seals, walruses, and sea lions) as
definitive hosts. The species belonging to the Contracaecum genus have as definitive hosts fish-eating
birds and pinnipeds, and unlike the other genera, Contracaecum larvae can parasitize both marine and
freshwater fish. Finally, the definite hosts of the species belonging to the Hysterothylacium genus of the
Raphidascarididae family are pinnipeds, fish, and shellfish.7–10 Because the biological cycles of the four
genera are similar, and the aim here is the experimental approach to study these worms, we will only
depict the Anisakis life cycle.
Adult worms release their eggs in the gut of the definite host. Through the feces, the eggs gain access
to the seawater, where they embryonate and form the first larval stage (L1) and progress to the second
stage (L2). The L2 are eaten by small crustaceans such as krill (first intermediate host), where they prog-
ress to the third-stage larvae (L3), the infective stage for the definitive host. Second intermediate hosts
(fish or shellfish) ingest the crustaceans, which in turn are eaten by bigger fish, transferring L3 through
the food chain and resulting in their accumulation in the larger fish until eaten by sea mammals, which
are their definite hosts.11,12 Once L3 have been eaten by their definite hosts, they progress to the fourth
larval stage (L4) and finally become adults13,14 (Figure 42.1).
When fish are captured, soon after their death, L3 migrate to the viscera, peritoneal cavity, and mus-
cles. The degree of migration depends on environmental conditions, the parasite, and fish species. When
humans consume raw or undercooked infected fish or shellfish, they may become accidental hosts. As
the parasites are not adapted to humans, they do not reach sexual maturity although they may cause mild
irritation to anaphylactic shock.5,15,16
At least 200 fish and 25 cephalopods species have been described as being infected with anisakid
larvae.17–19 Within all anisakids, species pertaining to the Anisakis genus are considered the most
7—Humans become
accidental hosts by
ingesting raw or
undercooked sea food
3—Eggs hatch,
ANISAKIS LIFE CYCLE
and L2 larva
swim freely
in water
5—Infected crustaceans
are consumed by fish and
4—Free swimming
squids. These are eaten
larvae are ingested
by larger fish, and so on,
by small crustacean
forming increasingly larger
and develop to L3
numbers of L3 in the gut
pathogenic and cause the largest number of human occurrences.20,21 Although morphologically very
similar, the genus has nine species that have been identified by molecular technologies and have
distinct definitive host distribution worldwide.22–24 As depicted in Figure 42.2, larvae are classified
by their morphology and genetic characteristics in “clades (I and II).” Clade I contains the Anisakis
simplex complex, which includes A. simplex (strict sense), A. pegreffi, and A. simplex (complex); the
other sister species in this clade are A. typica, A. ziphidarum, and A. nascettii. The definitive host of
the Clade I species are mainly distributed in the Atlantic and Pacific oceans. A. simplex (ss) and A.
pegreffi are also found in the Mediterranean, Arctic, and Antarctic Seas.2,12 Anisakis species pertain-
ing to Clade II are classified as A. physeteris complex, which includes A. physeteris, A. brevispiculata,
and A. paggiae species. Although considered to have a cosmopolitan distribution, they are mainly
found in the Atlantic Ocean.2,25
The Pseudoterranova decipiens complex consists of species that include the P. decipiens (sensu
stricto) or P. decipiens B, P. krabbei, P. bulbous, P. azarasi, and P. cattani. They are considered cosmo-
politan and are very abundant in the Atlantic Ocean, occurring from the Arctic to Antarctica.26
The Contracaecum genus has species that are able to parasitize both freshwater and marine organ-
isms. From this genus, the species that most frequently cause anisakidosis pertain to the C. osculatum
complex, which is a set of five members including C. osculatum types A, B, C, D, and E, where C. oscu-
latum sensu stricto corresponds to type C.27 The most frequent geographic distribution of these species
is the Alaskan and Japanese waters, the Baltic Sea, and Antarctic and Atlantic Ocean.7,28
Although the Hysterothylacium genus has a worldwide distribution, it is described as a rarely occur-
ring causative agent of anisakidosis. Apparently, the first human case of H. aduncum was registered in
1996 in Japan.29 Molecular identification of Anisakis and Hysterothylacium larvae from marine fish of
the East China Sea and the Pacific coast of central Japan showed that approximately 10% of the larvae
pertained to the Hysterothylacium genus (H. amoyense—5.0%, H. aduncum—1.6%, H. fabri—3.4%,
and H. spp.—2.9%), while the majority of the remaining Anisakidae nematodes belong to the Anisakis
genus.30 This result correlates well with the clinical finding.
Using classical techniques (morphological taxonomy), human anisakidosis is most frequently described
as being caused by Anisakis and Pseudoterranova genus.29,31–33 Among the Anisakis species, A. simplex
(ss) is reported as responsible for the highest number of human cases. However, after the introduction of
molecular biology in taxonomy, A. pegreffi has been more frequently described as the agent responsible
for anisakiosis in some countries such as Italy.18,34,35
A. physeteris I Physeteridae
A. brevispiculata
Clade 2
II Kogiidae
A. paggiae
A. ziphidarum
Clade 1
A. typica IV Delphinidae
A. simplex complex
Ani s 13
Ani s 14 hemoglobin
Unknown Ani s 3
UniProt - K9USK2
UniProt-?? Tropomyosin
Gonzalez-Fernandez (2015) Ani s 2
Kobayashi et al. UniProt - Q9NAS5
Asturias et al. (2000) Paramyosin
UniProt - L7V1|9
Pérez-Pérez et al. (2000)
Ani s 12
Unknown Ani s 1
UniProt - L7V0K0 Serine protease inhibitor
Kobayashi et al. (2011) (Kunitz type)
UniProt - L7V3Q3
Ani s 11 Moneo et al. (2000)
Unknown
Ani s 4
UniProt - E9RFF3
Cystatin
Kobayashi et al. (2011)
UniProt - Q14QT4
Moneo et al. (2005)
Ani s 10
Unknown Ani s 5
UniProt - D2K835 SXP/RAL-2
Caballero et al. UniProt - A1|KL2
Kobayashi et al. (2007)
Ani s 6
Ani s 9 Ani s 8 Ani s 7 Trypsin inhibitor like cysteine
SXP/RAL-2 SXP/RAL-2 n/a rich domain
UniProt - B2XCP1 UniProt - A7M6S9 UniProt - A9XBJ8 UniProt - A1|KL3
Rodriguez-Perez et al. (2008) Kobayashi et al. (2007) RodrÍguez et al. (2008) Kobayashi et al. (2007)
FIGURE 42.3 Updated Anisakis allergen compiled mainly from data extracted from the Allergome database in combina-
tion with published literature. The colors of the wedges indicate the origin of the antigens: Dark gray—somatic antigens;
medium gray—excretory-secretory antigens; light gray—unknown origin. (http://www.allergen.org/treeview.php.)
42.3 Allergen Nomenclature
The abbreviation of the name of the gender (first three letters) and of the species (first letter) followed by
a number indicating the chronology of the allergen purification was adopted as the systematic nomencla-
ture of allergens, implemented by the Nomenclature Sub-Committee of the World Health Organization
(WHO) and International Union of Immunological Societies. So, the A. simplex allergens are called
“Ani s #” (e.g., Ani S1).36 A. simplex (ss) has 14 allergens characterized by origin and molecular aspects.
The immunoreactivity pattern for these allergens has been studied both with human sera and with
experimental animals. A synthesis of the structural classification of Anisakis allergens is presented in
Figure 42.3 based mainly on the allergen database AllFam,37 which can be accessed on the web at http://
www.meduniwien.ac.at/allergens/allfam. The data was complemented from other published literature.
For example, data from the Conserved Domain Database (CDD) and from the domain of unknown func-
tion (PF; DUF, Pfam) were used.
epigastric pain, nausea, diarrhea, vomiting, and fever.40 Furthermore, when larvae penetrate the submu-
cosa, it may sensitize the host with its excretory-secretory (ES) products by stimulating the development
of a predominantly Th2 immune response, which favors the production of IgE antibodies, responsible for
allergies.41 Persistence of larvae in the tissue can result in direct damage and, in turn, the development
of a eosinophilic granuloma, characterized by an inflammatory infiltrate of eosinophils and neutrophils
associated with a diffuse interstitial edema and proliferation of connective tissue around the body of the
larva.42–46
There is evidence in the literature that the continued exposure to Anisakis antigens by fish factory
workers, anglers, and their families can sensitize them through inhalation or direct contact.47,48 Farmers
are another group of workers who can become sensitized to Anisakis antigens when in direct contact
with the corresponding allergens, e.g., fish meal.47 Gastrointestinal conditions, asthma, conjunctivitis,
and occupational contact dermatitis have been frequently described in Anisakis-sensitized patients.47,49–53
Signs and symptoms can range from discreet allergic symptoms, urticaria up to angioedema, and fatal
anaphylactic reactions with or without gastrointestinal symptoms.54,55
The human immune response to Anisakis sp. antigens is highly heterogeneous, varying both in quan-
tity and in quality between individuals.56 Studies in patients showed that infection with Anisakis larvae
induces a strong immune response with the production of specific antibodies reaching maximum titers
within the first month of infection.57 Infections with low numbers of larvae and continuous exposure
frequently result in the production of high levels of IgE, whereas the exposure to high numbers of lar-
vae frequently results in the production of IgG.58,59 The analysis of the cytokine profile obtained from
the peripheral blood and intestinal biopsy samples of newly infected patients reinforces the concept that
the Th2 response plays an important role in the immunopathogenesis of anisakiosis.38 Further detail
pertaining to the immune response shall be presented during the experimental section.
42.5 Laboratorial Diagnosis
Initially, specific IgG was used to diagnose anisakiosis; however, as the IgG titers persist elevated for a
relatively long period, it is not a good parameter to differentiate current from previous A. simplex infec-
tions. Another observation is that anisakid allergy is frequently associated with high levels of specific
IgG4.60–62
A good diagnostic tool can be the use of the proportion of specific IgE and IgG4 titers. This strategy
has been used to evaluate allergic diseases caused by a variety of other nematodes even if the nematode
is not observed by a gastroscopy.60,63,64 Thus, the serological diagnosis of a gastroallergic anisakiosis can
be a good alternative.57,60,63,65 Chronic urticaria (CU) associated with anisakiosis is another clinical set-
ting in which IgG4 can be used for diagnosis and follow-up. Unlike patients who continue their exposure
to the fish, those that are subjected to a fish-free diet experience a significant reduction in CU symptoms
accompanied by significant reduction of IgG4 levels.65 The comparison of the levels of IgE, IgG, and
IgG4 to A. simplex in CU and GAA patients showed that the latter presented significantly higher levels
of all tested immunoglobulins.66
42.6 Experimental Models
42.6.1 General Considerations
Even if the conditions that are used in animal experimentation do not exactly match those that occur in
the natural history of disease, this is a widely used method for acquiring knowledge of various diseases
in human and veterinary medicine. Through in vivo experimentation, it is possible to answer specific
questions about the pathophysiology of diseases generating information that can then be extrapolated
to the clinical setting, permitting a better understanding of the disease, leading to better prevention and
better treatment.
Anisakis 685
Since the discovery of the first human anisakiosis cases in the 1960s, many animal species have been
used as a model for this disease. The first studies used rabbits and guinea pigs to understand the migra-
tion trajectory of the larvae to the tissues and granuloma formation. However, to study the allergic reac-
tions induced by Anisakis larvae, most researchers prefer to use rats and mice. We chose to present the
animal models by species and route of infection/sensitization.
42.6.2 Guinea Pigs
42.6.2.1 Intradermic Route
In order to evaluate the in vivo chemotactic effect of A. simplex larvae extract, Tanaka and Torisu67
used guinea pigs as experimental animals. These researchers found that a few hours after intrader-
mal injection of crude larvae extract (CE), a dose-dependent accumulation of eosinophils occurred at
the site of injection. To confirm this effect, these authors carried out in vitro chemotaxis assays using
Boyden chambers.68 Using the same concentration of the extract with which eosinophil chemotaxis was
observed, no chemotactic activity was found for neutrophils, supporting the idea that the CE plays an
important role in the development of eosinophilia in anisakiosis.
42.6.2.2 Intraperitoneal Route
Early in the 1980s, in the attempt to determine the etiologic mechanism of the allergic reactions associ-
ated with anisakiosis, guinea pigs were sensitized by implanting live Anisakis sp. larvae in the perito-
neal cavity.69 The Schultz–Dale70,71 reaction was used to determine the presence of type I reactivity. In
short, intestinal fragments of intraperitoneal-sensitized guinea pigs with live Anisakis larvae responded
intensely when stimulated with Anisakis larvae hemoglobin and with less intensity when stimulated with
CE from other anisakids (Contracaecum and Pseudoterranova), whereas no response was observed
when Toxocara canis or Ascaris suum extracts were used. These results confirm the IgE-mediated etiol-
ogy of the allergic reactions associated with anisakiosis.
42.6.2.3 Intragastric Route
To determine the migratory pattern and viability of live Anisakis larvae, these were delivered to the
gastric cavity. Larvae gained different organs and tissues passing the stomach wall through an active
migration mechanism without a preestablished migratory pattern. Live larvae without any morphologi-
cal changes were recovered up to the fifth day after administration. These were able to reinfect another
guinea pig maintaining the same migration capability. However, as of the sixth day post infection, all
larvae disappeared leaving no hint of its presence in any part of the body.
Guinea pigs experimentally infected with A. simplex have also been used to test drugs.72 For example,
oral treatment with ivermectin or albendazole was tested and found to present high in vivo efficacy
against the larvae present in different organs of the guinea pigs.73
42.6.3 Pigs
42.6.3.1 Oral Route
Anisakis larvae infection in pigs was studied by feeding the animals with fish offal contaminated with L3.
In these studies, researchers observed that the severity of injury was proportional to the number of larvae
ingested. Histological alterations due to larvae interaction with the mucosa included primary mechanical
damage accompanied with bleeding, ulceration of the mucosa and submucosa, and intense cellular infil-
tration with connective tissue proliferation around the larva.74 The histological changes of the stomach
mucosa from experimentally infected pigs with Anisakis sp. and Pseudoterranova sp. larvae involved
intense inflammatory reaction around the larva with the presence of numerous eosinophilic cells.75 That
686 Laboratory Models for Foodborne Infections
is, feeding pigs L3 of C. osculatum results in the same histopathological findings that c orresponded to
findings of infections caused by other Anisakis sp. pathogens.76
42.6.4 Rabbit
The histological aspects of intestinal sections of experimentally infected rabbits resemble those of acciden-
tally infected humans, suggesting a similarity of the pathogenesis. Thus, rabbits were successfully intro-
duced as experimental anisakiosis models soon after the publication of the first human anisakiosis cases.77
42.6.4.1 Intragastric Route
In the early 1970s, the experimental determination of the pathogenesis of anisakiosis was performed by
administrating live larvae to the stomach of rabbits and semiquantitatively grading the inflammatory
reaction of the surrounding tissue where larvae penetrated.78 Three days after the oral administration of
40 Anisakis larvae, only a very small number entered the stomach wall, many of which were still alive,
and the degree of the inflammatory reactions of the gastric mucosa surrounding the distinct larvae varied
between mild, moderate, and severe in an individual animal and between individuals.
Necrosis and massive amounts of granulocytes, including eosinophils, were the main findings on day 3
after infection. On day 5, the larval viability declined, and an infiltrate of plasma cells and immunoblasts
was observed along with the granulocytes in the center of the reaction, while fibroblasts were already
present in the periphery. After 7 days, the fibroblast infiltrate became more intense; by 10 days, granula-
tion tissue was observed; and by a month, the necrotic tissue was substituted by new connective tissue
surrounded predominantly by mononuclear cells with moderate amounts of eosinophils.
The serological reactivity in association with the histopathological pattern was also studied in rabbits
infected with 30 live A. simplex larvae through the oral rout. Although most larvae were recovered in the
stomach, some migrated from the gastrointestinal tract and reached extragastric tissues, resulting in the
formation of abscess that contained dead larvae. By 30 days, the reactions progressed to granulomatous
abscesses followed by calcification of the larvae.79
From the serological point of view, IgG peaked by 30 days, coinciding with the granuloma resolution
and calcification of the larva followed by an abrupt decline. Another study that infected rabbits with
10 larvae showed a peak of IgM on the 11th day, whereas IgG peaked approximately a month later.80
Intragastric sensitization of rabbits with Anisakis larvae was also employed to assess the recognition
pattern of somatic and secreted antigens of infective Anisakis larvae comparing possible relationships
with antigens from other nematodes of Ascaroidea family using radioimmunoprecipitation techniques.81
Such as in serum derived from Anisakis infected patients, infected rabbits preferentially respond to
somatic antigens, and the recognition sequence occurs to different components of secreted antigens.
The differences in the recognition of secreted/excreted antigens and somatic components may be due to
the duration of sensitization and the degree of penetration by nematodes in the tissues. Kennedy et al.81
also demonstrated that a 14-kDa component derived from A. simplex cross-reacts with a homologues
component derived from Ascaris suum, Ascaris lumbricoides, and Toxocara canis, species from the
Ascaroidea family.
42.6.4.2 Subcutaneous Route
A chemotactic factor selectively attractive for eosinophils found in the extract from Anisakis larva was
termed eosinophil chemotactic factor of parasites (ECF-P).67 To determine whether the eosinophilic
phlegmonous inflammation typically observed in human anisakiosis could be experimentally repro-
duced, normal and subcutaneously immunized rabbits received intraserosal injection of ECF-P into the
ileum of rabbits. All rabbits developed a significant eosinophilic inflammation at the injection site in a
dose-dependent manner. Although immunized rabbits presented high anti-ECF-P antibody titers while
normal animals had no detectable antibody, there was no significant histological difference between the
lesions observed in either group of rabbits. These results support the argument that, especially in the
Anisakis 687
early phase of primary infection with anisakiosis, ECF-P may contribute to the development of eosino-
philic phlegmonous inflammation without any immunological intervention.67,82
42.6.4.3 Intramuscular Route
One of the experimental protocols involves the intramuscular route to investigate if larval antigens of
A. simplex present molecular similarity to interleukin IL-4. The resulting rabbit anti-mouse IL-4 anti-
bodies were tested against A. simplex ES and CE antigens in ELISA. The anti-IL-4 antibodies showed
a strong cross-reactivity, which was confirmed by western blot analysis. A complementary assay, the
absorption of the anti-IL-4 sera with A. simplex antigen, demonstrated a 70%–80% inhibition of antigen
binding when retested in ELISA. These results support the hypothesis that A. simplex proteins share
several epitopes with IL-4 or conversely that A. simplex larval ES and somatic products present IL-4-
like molecules. This finding implies that the parasite may control and modulate the mucosal Th1-Th2
dichotomy for its own benefit in an attempt to avoid its expelling.83
Currently, experimental Anisakis research has not used rabbits as a model to study allergic reactions
caused by this nematode. However, intramuscular inoculation with Anisakis antigens has been employed
when the aim is to characterize allergens and to produce laboratory reagents.84–88
42.6.5 Rats
Rats have been used extensively to investigate the immune response to Anisakis larvae. Although the
oral route is the natural form of infection, in the experimental scenario, investigators have shown a
limited usefulness of per os administration due to the difficulty in accurately determining the parasite
load, since many larvae are expelled through the anus, hampering the establishment of the relationship
between parasite load and immune response.89 Although the surgical implant may appear to be an inad-
equate route of infection, the argument used to validate this technique and to expect that the antibody
production profile would be the same regardless of the route is that orally administered larvae pass from
the intestinal lumen into the peritoneal cavity after infection.90,91 Another observation that supports this
hypothesis is that extragastrointestinal anisakiosis has also been observed in humans who are infected.92
42.6.5.2 Intragastric Infection
Let us return to the intragastric/intraperitoneal duel. Authors argue that despite the importance of the
live larvae intraperitoneal inoculum studies, the human natural history of gastroallergic anisakiosis is
688 Laboratory Models for Foodborne Infections
given orally; so experiments using this pathway are important.59 Rats were infected with L3 Anisakis by
the oral route twice with an interval of 9 weeks to investigate the kinetics of isotype-specific antibody
expression, and it was found that IgM’s peak with similar titers after primary and reinfection presents
the same antigenic recognition. After reinfection, as expected, IgG1 and IgG2a levels were higher and
showed accelerated kinetics; however, IgG2b level was substantially lower. The biological allergy state
peaked earlier (1 week) than the immunochemical allergy state (2 weeks). Since no meaningful cor-
relation between specific IgE avidity and biological allergy state was found and elevated IgM levels at
reinfection occurred, the hypothesis is that the allergic response induced by oral L3 infection might not
be related to specific IgE avidity.94
A procedure developed recently to deliver live larvae directly to the stomach of mice by an esophageal
catheterization95 was adapted to perform live Anisakis spp. infection in rats.96 The aim of this study was
to understand the histopathological effects of acute (single) and chronic (multiple reinfections—24, 48,
72, and 96 h intervals) Anisakis infection. Live larvae were found anchored to the mucosa at different
locations (whose milieu varied from a very acidic to basic pH gradient), passing through the stomach
wall and in organs out of the gastrointestinal tract. The histopathology showed an acute inflammatory
reaction, with eosinophil predominance and a mild fibrotic reaction. Even though not all larvae were
recovered as previously placed as an obstacle to the oral route, this protocol can be considered a good
experimental model because the histopathological alterations are similar to those described in human
anisakiosis.97
Although there are reported cases of allergic reactions due to the ingestion of cooked and frozen
seafood, there is also evidence that only live larvae trigger the allergic reactions. Consequently, the
debate on the risk of Anisakis-associated hypersensitivity by ingestion of properly cooked and frozen
fish remains. To elucidate this fact, an experimental model was designed to study the antibody produc-
tion kinetics after oral inoculation with live or dead Anisakis L3. The results show that animals produce
specific IgM, IgG, and IgE to ES antigens after primary and secondary inoculation with live L3 but not
after dead L3 (frozen, heated, cut, or homogenized). These results suggest that the ingestion of cooked or
frozen seafood containing Anisakis L3 is safe even for allergic individuals.98
42.6.6 Mice
Because of the accumulated data in the last decades, especially concerning IgE synthesis of the anti-
body associated with allergic reactions, mice are considered better animal models than other species to
investigate allergic reactions.100–102 In food-associated allergies, it is still unclear what conditions make
certain foods strong IgE inductors. There are reports of more than 170 foods causing food allergies, but
only 8 (peanut, tree nuts, milk, egg, wheat, soy, fish, and shellfish) account for 90% of all food-allergic
reactions.103 It is known that the major reaction to food proteins when ingested in physiological condi-
tions usually is a phenomenon called oral tolerance, while the parenteral administration of the same
Anisakis 689
food proteins in experimental models induces sensitization.104–110 This intriguing dichotomy has inter-
ested immunologists.
It is also known that in natural helminthic infections, IgE and eosinophilia are major hallmarks of
the immunological response as the consequence of a Th2 lymphocyte profile activation by helminths.
Among other interleukins, Th2 cells secrete IL-4 and IL-5; the former promotes immunoglobulin class
switching to IgE, and the latter stimulates eosinophil development and activation. Furthermore, in IgE
experimental models, animals are immunized with the antigenic preparations mixed with adjuvants such
as aluminum hydroxide102 or pertussis toxin111,112 and commonly administered by a parenteral route. In
experimental models where the aim is to develop oral sensitization and food allergy, antigens are associ-
ated to cholera toxin.113 Complete or incomplete Freund’s adjuvant is another commonly used adjuvant,
which is considered a good IgG inducer.114
The humoral and cellular immune responses to live A. simplex larvae observed in mice models share
similarities with those observed in human disease. However, due to the difficulty in introducing live
larva into the gastric cavity of mice, the majority of the experiments have been conducted by immuniz-
ing the animals with CE or with ES antigens of cultivated larvae, thus indicating the relevance of the
investigation of the immune mechanisms that control the allergic responses to live and dead Anisakis
spp. larvae.115–118
42.6.6.2 Intraperitoneal Immunization
To help understand some of the unknown immune interactions between helminth infection and allergy,
mice were intraperitoneally sensitized to develop a hypersensitivity reaction with A. simplex proteins,
followed by an intravenous or oral A. simplex challenge. The sensitized mice presented as of the third
week specific IgE, IgG1, and IgG2a to numerous A. simplex allergens, some of which were similar to
those found in human serum. When challenged with intravenous A. simplex antigens but not after an oral
antigen challenge, anaphylaxis and plasma histamine release were observed. The cellular and molecular
profile showed that A. simplex stimulated splenocytes to release IL-10, IFN-γ, IL-4, IL-13, and IL-5 with
a mixed Th1/Th2 pattern.120 This seems to be a good model to investigate the peculiar allergic reactions
to parasitic proteins.
42.6.6.3 Intragastric Infection
Live Anisakis L3 were orally inoculated in C57BL/10 and BALB/c mice to investigate isotype-specific
immune responses to ES and CE products. The C57BL mouse strains typically produce a Th1-Type
cytokine profile, while BALB/c mice produce a Th2-Type cytokine profile. Both ES and CE antigens
stimulated similar antibody patterns; however, CE stimulated the production of higher antibody levels.
690 Laboratory Models for Foodborne Infections
BALB/c mice produced a faster IgM response than C57BL/10 mice, while the latter produced higher
IgG1 and IgG2b antibodies with practically undetectable IgG2a levels.121 Further anisakiosis studies
using BALB/c mice showed that after multiple immunizations using Freund’s adjuvant, mice showed a
single maximum peak of IL-4 between weeks 8 and 14, whereas animals inoculated with a single larva
per os showed two IL-4 peaks—the first with moderate levels between days 6 and 12 p.i. and the second
maintained from week 3 to 9.122 After Perteguer and Cuellar showed the consequences of natural sensi-
tization,121,122 the authors of this chapter proposed a simplified method to introduce live larvae with an
intragastric tube. This technique results in similar data as those published in the literature.95,118
42.6.6.4 Epicutaneous Immunization
As cited before, contact dermatitis is one of the consequences of antigen exposure to Anisakis proteins
in seafood-processing workers. Therefore, to understand the basic mechanisms in the development of
allergic sensitization through the skin, repeated epicutaneous exposure of Anisakis proteins in wild-type
(WT), IL-4-, IL-4Rα-, IL-13-, and IL-4/IL-13-deficient mice was evaluated by following the systemic
signs and symptoms. Epicutaneous sensitization with Anisakis larval antigens induced the WT local-
ized inflammation, epidermal hyperplasia, production of TH2 cytokines, antigen-specific IgE and IgG1,
and anaphylactic shock after intravenous challenge. IL-13-deficient mice failed to develop epidermal
hyperplasia and inflammation, and in IL-4-, IL-4/IL-13-, and IL-4Rα-deficient mice, anaphylaxis was
reduced. These results suggest that interleukin-13 plays a central role in contact dermatitis development,
whereas IL-4 drives the Th2 profile and resultant anaphylactic reactions.48
42.6.6.5 Subcutaneous Immunization
The subcutaneous route is a technique frequently utilized in immunological studies. The footpad is a
frequently used location, since the draining lymph nodes are easily removed making it possible to study
the local immunological response. Taking the advantages of this strategy, the cellular immune response
to A. simplex L3 antigens was compared in mice that were infected either after being pre-sensitized, with
homologous CE antigen or sham sensitized. As the pre-sensitization simulates a primary immune response
the authors addressed the difference between a primary infection and a simulation of a reinfection. This
footpad sensitization protocol induced an increase in the size and weight of the popliteal lymph nodes
(PLN). A high proportion of systemic CD4+ and TCRαβ+ T cells in both groups was observed. A reduc-
tion in B cells accompanied by a decrease of CD8α+ T cells was observed in pre-sensitized and infected
mice, while those only exposed to infection present the greatest increase in CD8α+ and TCRαβ− T cells.
42.6.6.6 Intranasal Immunization
To examine the immunological mechanisms underlying the development of allergic airway inflamma-
tion, WT and interleukin-4 receptor alpha (IL-4Rα)-deficient mice were sensitized to Anisakis anti-
gens through different routes.123 Live or heat-killed Anisakis larvae were administered intraperitoneally,
while Anisakis extract was administered by the intranasal route. Subsequently, all animals were chal-
lenged intranasally with an Anisakis extract. Allergen-specific antibodies developed only in intra-
peritoneally sensitized mice; however, both routes of sensitization induced IL-4Rα-dependent allergic
airway responses in WT mice in an IL-4/IL-13-dependent pathway. Unexpectedly, infection with live
Anisakis larvae induced airway hyperresponsiveness that was abrogated when IFN-γ was neutralized
in vivo. Thus, infection leads to IL-4/IL-13-independent, IFN-γ-dependent airway hyperresponsive-
ness. Together, these results demonstrate that both infection with larvae and inhalational exposure to
Anisakis proteins are potent routes of allergic sensitization, explaining food- and work-related allergies
in humans, which can involve either IL-4/IL-13 or IFN-γ. Importantly for diagnosis, detectable Anisakis-
specific antibodies may not accompany allergic airway inflammation.
In vitro studies demonstrated that a 24-kDa protein (22U homologous; As22U) derived from A. simplex
larva elicits several Th2-related chemokine gene expression, meaning that it may be one of the impor-
tant allergens for the clinical setting. In order to examine their hypothesis, six intranasal applications of
Anisakis 691
ovalbumin (OVA) or recombinant As22U (rAs22U) and OVA were performed. When compared to the
group that only received OVA, the animals challenged with rAs22U associated to OVA presented severe
airway inflammation, immune cell recruitment in special, eosinophils, increased levels of IL-4, IL-5, and
IL-13 in the Bronchoalveolar lavage fluid (BALF), significantly increased airway hyperresponsiveness,
and significantly higher anti-OVA-specific IgE and IgG1. After receiving rAs22U, the GRO-α (CXCL1)
gene expression increased immediately while eotaxin (CCL11) and TARC (CCL17) gene expressions
increased significantly at 6 h. Thus, rAs22U may be responsible for a Th2/Th17-mediated airway allergic
inflammation.124 Using the same experimental protocol, two other Anisakis antigens (Ani s 1 Ani s 9)
were tested, eliciting similar results expressing Th2 (IL-4, IL-5, IL-13, e IL-25) and Th17 (IL-6 e IL-17)
cytokines because of the intranasal exposure.125
42.6.7 Fish
Many marine fish are infected with third-stage larvae of A. simplex (sensu lato). To ensure food safety,
it is important to determine whether these larvae are present in the flesh of commercial fish species.
However, there is little information regarding the tissue specificity of anisakid species. Thus, the ratio-
nale for the use of fish as an experimental model to study Anisakidae nematode is to understand the
infective capacity in commercially relevant fish species, the parasite mechanisms of aggression, and the
host’s immunological response.
42.6.7.1 Oral Infection
Rainbow trout (Oncorhynchus mykiss) and olive flounder (Paralichthys olivaceus) received L3 larvae of two
sibling species of A. simplex per os and were accompanied for 5 weeks. In the rainbow trout, A. simplex s.s.
692 Laboratory Models for Foodborne Infections
predominantly migrated into the body muscle while a small number of freely moving A. pegreffi larvae were
recovered within the body cavity. In the olive flounder, A. simplex s.s. larvae were found both in the body
cavity and the muscle, while A. pegreffi larvae were found only in the body cavity encapsulated in lumps.135
In another set of in vivo investigations, A. simplex was used to experimentally infect rainbow trout
(Oncorhynchus mykiss), Baltic salmon (Salmo salar), and brown trout (Salmo trutta). Of the three spe-
cies, Baltic salmon was the most susceptible, presenting the highest number of successfully established
nematodes, whereas brown and rainbow trout had a higher natural resistance. The preferred A. simplex
larvae microhabitat in the brown trout was the stomach, pyloric caeca, and intestine, while the majority
of larvae found in rainbow trout were located at the pyloric caeca. In the Baltic salmon, the most suscep-
tible fish species, nematodes were dispersed in and on the spleen, head, kidney, liver, swim bladder, and
musculature. CD8+ cells were present while IgM+ -bearing cells were absent in the inflammatory tissue
around the nematodes of all three fish species. MHCII-bearing cells were present in the encapsulated
A. simplex in rainbow and the brown trout, but not in Baltic salmon.136
Yet, another set of recent experiments show that closely related salmonids differ in their susceptibility
toward different anisakid larvae and agree that parasites select different microhabitats in the hosts.137
Orally infected rainbow trout (Oncorhynchus mykiss), brown trout (Salmo trutta), and Atlantic salmon
(Salmo salar) with larval stages of H. aduncum, C. osculatum, or A. simplex were studied to determine
parasite survival and location up to14 days post infection (dpi). Although the most prevalent and numer-
ous nematode in brown trout at 2 dpi was H. aduncum, a large proportion of the worms were already
recovered dead with no tissue penetration. This fish species exhibited the highest natural resistance to
A. simplex. Rainbow trout exhibited the highest susceptibility to C. osculatum larvae at 2, 7, and 14 dpi
with eventual pyloric cecum penetration. A. simplex larvae established a more successful infection in
salmon compared to rainbow trout, although at 2 and 7 dpi this fish showed the highest intensity and
abundance of larvae, but not after 14 days. Although the pyloric ceca was the preferred microhabitat for
Anisakis in both rainbow trout and salmon, larval penetration into muscle and liver was found.
42.6.7.2 Intraperitoneal
Since hydrolytic enzymes play an important role in the nematode host tissue penetration, determination
of what enzymes are present within the ES proteins seems important. Lipase, esterase/lipase, valine and
cysteine arylamidases, naphthol-AS-BI-phosphohydrolase, and α-galactosidase activities were found. To
further elucidate the influence of intraperitoneally injected ES, substances on the immune system of fish-
specific gene expression in spleen and liver of the rainbow trout (Oncorhynchus mykiss) were measured.
The results demonstrate a generalized downregulation of immune-related gene expression, suggesting
a suppressive immunomodulatory role for ES proteins. From the ecological point of view, this makes
biological sense. One can argue that when worm enzymes directly target the host’s immune molecules, a
decreased immune response with an increased worm survival is the consequence.138
For many clinically important parasites, in vitro cultivation is an important diagnosis tool. An array
of commercial systems, which have been developed, such as the Harada-Mori culture technique for
larval-stage nematodes, permit rapid diagnosis. In comparison, although in vitro cultivation techniques
are used more often than in vivo techniques, the in vivo techniques are sometimes used for diagnosing
parasitic infections such as trypanosomiasis and toxoplasmosis. Parasite cultivation continues to be a
challenging diagnostic option. Thus, an overview of intricacies of parasitic culture and an update on
popular methods used for cultivating parasites are presented.
42.6.8.1 Culture Media
The first description of Anisakidae nematode cultivation occurred in the early 1960s. P. decipiens larvae
removed from the flesh of fresh fish were immediately transferred to 199 culture media enriched with
glucose, beef embryo extract, beef liver extract, and antibiotics. In this study, the authors obtained larvae
that reached morphological changes consistent with adult worms.144 Subsequently, with adjustments of
the initial conditions, A. marina developed successfully to adult worms. The first larval developmental
changes were observed within 4 days after the release of cuticles in the medium. The development of
gonadal tissue characterizing the preadult stage occurred between 26 and 98 days. The complete matu-
ration characterized by the worm wall thickening and gonadal maturation can be distinguished in vitro.
The first free larvae were observed after 4–8 days at a temperature of 13°C–18°C and after 20–27 days
at a temperature of 5°C–7°C. The larvae are very active, and their mobility has no fixed direction. In
seawater, they can live for 3–4 weeks at temperatures of 13°C–18°C and for 6–7 weeks at 5°C–7°C; tem-
peratures above 20°C led to increased mortality, and a temperature of 34°C was absolutely inadequate,
indicating that the first intermediate host must be cold-blooded.145
0.2%–7% of cod liver oil. The results demonstrate that although L3 move randomly, they do not stop
randomly. The tendency to move out of a certain area was inversely correlated with lipid concentration.
A second observation indicates that the intentional migration range of larvae is short. In conclusion, L3
prefer high fat content and seek it over short distances. These in vitro data agree with previous observa-
tions that A. simplex L3 randomly tend to migrate out of the fish gut into the flesh.155
42.6.9 Larvicidal Models
With the growing number of human anisakiosis cases, an alternative was the search for active larvicidal
compounds. In vitro and in vivo assays were undertaken to evaluate herbs used to season fish based on
epidemiological observations that prevalence of anisakidosis in the Chinese regions where raw fish is
often seasoned with ginger (rhizome of Zingiber officinale) and/or “perilla mint,” “Chinese basil,” or
“wild basil” (common names for Perilla frutescens [Lamiaceae]) is smaller. 156,157
In vitro studies showed that [6]-Shogaol and [6]-gingerol components derived from ginger rhizome
induced an important reduction in larvae mobility and altered both their cuticle and digestive tract.158
Further studies revealed that [10]-gingerol, [10]-shogoal, and other compounds derived from ginger also
has a very effective larvicidal effect.159
In vivo protocols where rats were infected by delivering larvae directly to the stomach through the
use of a gavage were used to evaluate the action of essential oils on Anisakis L3. Simultaneously or 2 h
after infection, each rat received one of five monoterpenes. To determine the localization and viabil-
ity of the larvae and to determine gastrointestinal histopathological changes, rats were sacrificed at
various time points. The order of in vivo larvicidal activity was peril aldehyde > citral > citronellol >
cuminaldehyde > carvacrol. When peril aldehyde, citral, and citronellol were given together with the
nematodes, no hemorrhages were observed, leading to the conclusion that these monoterpenes somehow
inhibit the fixation and/or penetration capacity of the larvae. The time gap of 2 h between the infesta-
tion and the administration of any of the tested compounds is sufficient for the larvae to develop their
pathogenicity in the rats.160
Three sesquiterpenic derivatives (nerolidol, farnesol, and elemolto) were studied to determine their
in vivo larvicidal activity. The order of in vivo larvicidal activity was nerolidol > farnesol > elemolto; the
first two caused the death of all nematodes, which showed cuticle changes and intestinal wall rupture.
Only 20% of infected rats treated with nerolidol or farnesol showed gastric wall lesions in comparison to
86.6% of control animals, suggesting that nerolidol and farnesol are good candidates for further research
as biocidal agents against L(3) larvae of Anisakis type I.161
The histological parameters to evaluate the effect of potentially larvicidal compounds were the analy-
sis of the cuticle and intestinal wall structure. Fixed formalin A. simplex L3 was assessed by optical
microscopy study of transverse thin sections (0.5–1 μm) stained with hematoxylin eosin, Masson’s tri-
chromic dyes, or toluidine blue.
Knowing that essential oils can irritate the mucosa, gut inflammatory reaction was studied after oral
administration of the tested compounds.161 A marker of neutrophilic infiltration is the titration of myelo-
peroxidase activity (MPO), determined by solubilization of myeloperoxidase with hexadecyltrimethyl-
ammonium bromide and measured with a dianisidine-H2O2 assay.162
42.7 Conclusion
A range of laboratory models are available to investigate foodborne infectious diseases, including those
due to Anisakis nematodes. As presented in the short epidemiological and taxonomical review of the
anisakid family along with the review of laboratory models used to study anisakiosis, there are still many
open questions regarding the life cycle, host–pathogen interaction, pathogenesis, and immune response
of the anisakid family larvae. These questions should be addressed because these nematodes are more
frequently contaminating foods due to the diffusion of oriental and Spanish cuisine, making this an
emerging anthropozoonosis of clinical importance.
Anisakis 695
Acknowledgments
We thank Maira Platais for help in the translation process.
REFERENCES
1. Audicana, M.T. & Kennedy, M.W. Anisakis simplex: from obscure infectious worm to inducer of
immune hypersensitivity. Clin Microbiol Rev 21, 360–79 (2008).
2. Baird, F.J., Gasser, R.B., Jabbar, A. & Lopata, A.L. Foodborne anisakiasis and allergy. Mol Cell Probes
28, 167–74 (2014).
3. Pravettoni, V., Primavesi, L. & Piantanida, M. Anisakis simplex: current knowledge. Eur Ann Allergy
Clin Immunol 44, 150–6 (2012).
4. Chai, J.Y., Darwin Murrell, K. & Lymbery, A.J. Fish-borne parasitic zoonoses: status and issues. Int J
Parasitol 35, 1233–54 (2005).
5. Hochberg, N.S. & Hamer, D.H. Anisakidosis: perils of the deep. Clin Infect Dis 51, 806–12 (2010).
6. Gibbons, L. Keys to the Nematode Parasite of Vertebrates. Supplementary Volume. CAB International,
Wallingford, UK (2010).
7. Koie, M. & Fagerholm, H.P. The life cycle of Contracaecum osculatum (Rudolphi, 1802) sensu stricto
(Nematoda, Ascaridoidea, Anisakidae) in view of experimental infections. Parasitol Res 81, 481–9 (1995).
8. Navone, G.T., Etchegoin, J.A. & Cremonte, F. Contracaecum multipapillatum (Nematoda: Anisakidae)
from Egretta alba (Aves: Ardeidae) and comments on other species of this genus in Argentina. J
Parasitol 86, 807–10 (2000).
9. Navone, G.T., Sardella, N.H. & Timi, J. T. Larvae and adults of Hysterothylacium aduncum (Rudolphi, 1802)
(Nematoda: Anisakidae) in fishes and crustaceans in the South West Atlantic. Parasite 5, 127–36 (1998).
10. Palm, H.W. Ecology of Pseudoterranova decipiens (Krabbe, 1878) (Nematoda: Anisakidae) from
Antarctic waters. Parasitol Res 85, 638–46 (1999).
11. Cipriani, P. et al. Genetic identification and distribution of the parasitic larvae of Anisakis pegreffii and
Anisakis simplex (s. s.) in European hake Merluccius merluccius from the Tyrrhenian Sea and Spanish
Atlantic coast: implications for food safety. Int J Food Microbiol 198, 1–8 (2015).
12. Klimpel, S. & Palm, H.W. Anisakid nematode (Ascaridoidea) life cycles and distribution: increasing
zoonotic potential in the time of climate change? in Progress in Parasitology (ed. Melhorn, H.) 201–22
(Springer, Berlin and Heidelberg, Germany, 2011).
13. Kuhn, T., Hailer, F., Palm, H.W. & Klimpel, S. Global assessment of molecularly identified Anisakis
Dujardin, 1845 (Nematoda: Anisakidae) in their teleost intermediate hosts. Folia Parasitol (Praha) 60,
123–34 (2013).
14. Mattiucci, S. & Nascetti, G. Genetic diversity and infection levels of anisakid nematodes parasitic in fish
and marine mammals from Boreal and Austral hemispheres. Vet Parasitol 148, 43–57 (2007).
15. Ishikura, H. & Kikuchi, Y. Infection by larvae of Anisakis in humans. Nihon Ishikai Zasshi 57, 1649–55
(1967).
16. Ishikura, H. et al. Transition of occurrence of anisakiasis and its paratenic host fishes in Japan, with
pathogenesis of anisakiasis. Hokkaido Igaku Zasshi 63, 376–91 (1988).
17. Abollo, E., Gestal, C. & Pascual, S. Anisakis infestation in marine fish and cephalopods from Galician
waters: an updated perspective. Parasitol Res 87, 492–9 (2001).
18. D’Amico, P. et al. Evolution of the Anisakis risk management in the European and Italian context. Food
Res Int 64, 348–362 (2014).
19. Klimpel, S., Palm, H.W., Ruckert, S. & Piatkowski, U. The life cycle of Anisakis simplex in the
Norwegian Deep (northern North Sea). Parasitol Res 94, 1–9 (2004).
20. Andersen, K. Hysterothylacium aduncum (Rudolphi, 1862) infection in cod from the Oslofjord: seasonal
occurrence of third- and fourth-stage larvae as well as adult worms. Parasitol Res 79, 67–72 (1993).
21. Yorimitsu, N. et al. Colonic intussusception caused by anisakiasis: a case report and review of the litera-
ture. Intern Med 52, 223–6 (2013).
22. Klimpel, S., Kellermanns, E. & Palm, H.W. The role of pelagic swarm fish (Myctophidae: Teleostei) in
the oceanic life cycle of Anisakis sibling species at the Mid-Atlantic Ridge, Central Atlantic. Parasitol
Res 104, 43–53 (2008).
696 Laboratory Models for Foodborne Infections
23. Mattiucci, S. et al. First molecular identification of the zoonotic parasite Anisakis pegreffii (Nematoda:
Anisakidae) in a paraffin-embedded granuloma taken from a case of human intestinal anisakiasis in
Italy. BMC Infect Dis 11, 82 (2011).
24. Valentini, A. et al. Genetic relationships among Anisakis species (Nematoda: Anisakidae) inferred from
mitochondrial cox2 sequences, and comparison with allozyme data. J Parasitol 92, 156–66 (2006).
25. Mattiucci, S. et al. Metazoan parasite infection in the swordfish, Xiphias gladius, from the Mediterranean
Sea and comparison with Atlantic populations: implications for its stock characterization. Parasite 21,
35 (2014).
26. McClelland, G. The trouble with sealworms (Pseudoterranova decipiens species complex, Nematoda): a
review. Parasitology 124 Suppl, S183–203 (2002).
27. Mehrdana, F. et al. Occurrence of zoonotic nematodes Pseudoterranova decipiens, Contracaecum oscu-
latum and Anisakis simplex in cod (Gadus morhua) from the Baltic Sea. Vet Parasitol 205, 581–7 (2014).
28. Koie, M. Experimental infections of copepods and sticklebacks Gasterosteus aculeatus with small
ensheathed and large third-stage larvae of Anisakis simplex (Nematoda, Ascaridoidea, Anisakidae).
Parasitol Res 87, 32–6 (2001).
29. Yagi, K. et al. Femal worm Hysterothylacium aduncum excreted from human: a case report. Jpn J
Parasitol 45, 12–23 (1996).
30. Kong, Q. et al. Molecular identification of Anisakis and Hysterothylacium larvae in marine fishes from
the East China Sea and the Pacific coast of central Japan. Int J Food Microbiol 199, 1–7 (2015).
31. Deardorff, T.L. & Overstreet, R.M. Contracaecum multipapillatum (=C. robustum) from fishes and
birds in the northern Gulf of Mexico. J Parasitol 66, 853–6 (1980).
32. Ishikura, H. et al. Anisakidae and anisakidosis. Prog Clin Parasitol 3, 43–102 (1993).
33. Sakanari, J.A., Staunton, C.E., Eakin, A.E., Craik, C.S. & McKerrow, J.H. Serine proteases from nema-
tode and protozoan parasites: isolation of sequence homologs using generic molecular probes. Proc Natl
Acad Sci USA 86, 4863–7 (1989).
34. Fumarola, L. et al. Anisakis pegreffi etiological agent of gastric infections in two Italian women.
Foodborne Pathog Dis 6, 1157–9 (2009).
35. Lim, H. et al. Molecular diagnosis of cause of anisakiasis in humans, South Korea. Emerg Infect Dis 21,
342–4 (2015).
36. Chapman, M.D., Pomés, A., Breiteneder, H. & Ferreira, F. Nomenclature and structural biology of aller-
gens. J Allergy Clin Immunol 119, 414–420 (2007).
37. Radauer, C., Bublin, M., Wagner, S., Mari, A. & Breiteneder, H. Allergens are distributed into few
protein families and possess a restricted number of biochemical functions. J Allergy Clin Immunol 121,
847–852.e7 (2008).
38. del Pozo, V. et al. Immunopathogenesis of human gastrointestinal infection by Anisakis simplex. J
Allergy Clin Immunol 104, 637–43 (1999).
39. Chung, Y.B. & Lee, J. Clinical characteristics of gastroallergic anisakiasis and diagnostic implications
of immunologic tests. Allergy Asthma Immunol Res 6, 228–33 (2014).
40. Shrestha, S. et al. Intestinal anisakiasis treated successfully with conservative therapy: importance of
clinical diagnosis. World J Gastroenterol 20, 598–602 (2014).
41. Gonzalez-Munoz, M., Rodriguez-Mahillo, A.I. & Moneo, I. Different Th1/Th2 responses to Anisakis sim-
plex are related to distinct clinical manifestations in sensitized patients. Parasite Immunol 32, 67–73 (2010).
42. Hiramatsu, K. et al. A case of acute gastric anisakiasis presenting with malignant tumor-like features:
a large gastric vanishing tumor accompanied by local lymph node swelling. Digest Dis Sci 49, 965–9
(2004).
43. Bouree, P., Paugam, A. & Petithory, J.C. Anisakidosis: report of 25 cases and review of the literature.
Comp Immunol Microbiol Infect Dis 18, 75–84 (1995).
44. Caramello, P. et al. Intestinal localization of anisakiasis manifested as acute abdomen. Clin Microbiol
Infect 9, 734–7 (2003).
45. Choi, S.C., Lee, S.Y., Song, H.O., Ryu, J.S. & Ahn, M.H. Parasitic infections based on 320 clinical
samples submitted to Hanyang University, Korea (2004–2011). Korean J Parasitol 52, 215–20 (2014).
46. Takeuchi, K., Hanai, H., Iida, T., Suzuki, S. & Isobe, S. A bleeding gastric ulcer on a vanishing tumor
caused by anisakiasis. Gastrointest Endosc 52, 549–51 (2000).
47. Anibarro, P.C. et al. Protein contact dermatitis caused by Anisakis simplex. Contact Dermatitis 37, 247
(1997).
Anisakis 697
48. Nieuwenhuizen, N., Herbert, D.R., Brombacher, F. & Lopata, A.L. Differential requirements for inter-
leukin (IL)-4 and IL-13 in protein contact dermatitis induced by Anisakis. Allergy 64, 1309–18 (2009).
49. Anibarro, B. & Seoane, F.J. Occupational conjunctivitis caused by sensitization to Anisakis simplex.
J Allergy Clin Immunol 102, 331–2 (1998).
50. Armentia, A. et al. Occupational asthma by Anisakis simplex. J Allergy Clin Immunol 102, 831–4 (1998).
51. Armentia, A. et al. Anisakis simplex allergy after eating chicken meat. J Investig Allergol Clin Immunol
16, 258–63 (2006).
52. Estrada Rodriguez, J.L. & Gozalo Reques, F. Sensitization to Anisakis simplex: an unusual presentation.
Allergol Immunopathol (Madr) 25, 95–7 (1997).
53. Scala, E. et al. Occupational generalised urticaria and allergic airborne asthma due to Anisakis simplex.
Eur J Dermatol 11, 249–50 (2001).
54. Alonso, A., Daschner, A. & Moreno-Ancillo, A. Anaphylaxis with Anisakis simplex in the gastric
mucosa. N Engl J Med 337, 350–1 (1997).
55. Moreno-Ancillo, A. et al. Allergic reactions to Anisakis simplex parasitizing seafood. Ann Allergy
Asthma Immunol 79, 246–50 (1997).
56. Mattiucci, S. et al. Anisakiasis and gastroallergic reactions associated with Anisakis pegreffii infection,
Italy. Emerg Infect Dis 19, 496–9 (2013).
57. Daschner, A., Cuellar, C., Sanchez-Pastor, S., Pascual, C.Y. & Martin-Esteban, M. Gastro-allergic ani-
sakiasis as a consequence of simultaneous primary and secondary immune response. Parasite Immunol
24, 243–51 (2002).
58. Daschner, A., Alonso-Gomez, A., Cabanas, R., Suarez-de-Parga, J.M. & Lopez-Serrano, M.C.
Gastroallergic anisakiasis: borderline between food allergy and parasitic disease-clinical and allergo-
logic evaluation of 20 patients with confirmed acute parasitism by Anisakis simplex. J Allergy Clin
Immunol 105, 176–81 (2000).
59. Alonso-Gomez, A. et al. Anisakis simplex only provokes allergic symptoms when the worm parasitises
the gastrointestinal tract. Parasitol Res 93, 378–84 (2004).
60. Daschner, A. et al. Specific IgG4: possible role in the pathogenesis and a new marker in the diagnosis of
Anisakis-associated allergic disease. Scand J Immunol 79, 120–6 (2014).
61. del Pozo, M.D. et al. Laboratory determinations in Anisakis simplex allergy. J Allergy Clin Immunol 97,
977–84 (1996).
62. Desowitz, R.S., Raybourne, R.B., Ishikura, H. & Kliks, M.M. The radioallergosorbent test (RAST) for
the serological diagnosis of human anisakiasis. Trans R Soc Trop Med Hyg 79, 256–9 (1985).
63. Gonzalez-Fernandez, J. et al. Haemoglobin, a new major allergen of Anisakis simplex. Int J Parasitol
45, 399–407 (2015).
64. Lorenzo, S. et al. Human immunoglobulin isotype profiles produced in response to antigens recognized
by monoclonal antibodies specific to Anisakis simplex. Clin Exp Allergy 29, 1095–101 (1999).
65. Daschner, A., Vega de la Osada, F. & Pascual, C.Y. Allergy and parasites reevaluated: wide-scale
induction of chronic urticaria by the ubiquitous fish-nematode Anisakis simplex in an endemic region.
Allergol Immunopathol (Madr) 33, 31–7 (2005).
66. Daschner, A., De Frutos, C., Valls, A. & Vega, F. Anisakis simplex sensitization-associated urticaria:
short-lived immediate type or prolonged acute urticaria. Arch Dermatol Res 302, 625–9 (2010).
67. Tanaka, J. & Torisu, M. Anisakis and eosinophil. I. Detection of a soluble factor selectively chemotactic
for eosinophils in the extract from Anisakis larvae. J Immunol 120, 745–9 (1978).
68. Boyden, S. The chemotactic effect of mixtures of antibody and antigen on polymorphonuclear leuco-
cytes. J Exp Med 115, 453–66 (1962).
69. Asaishi, K. et al. Studies on the etiologic mechanism of anisakiasis. 1. Immunological reactions of
digestive tract induced by Anisakis larva. Gastroenterol Jpn 15, 120–7 (1980).
70. Dale, H.H. The anaphylactic reaction of plain muscle in the guinea-pig. J Pharmacol Exp Ther 4, 167–
223 (1913).
71. Schultz, W. Physiological studies in anaphylaxis. II. Reaction of smooth muscle from guinea-pigs ren-
dered tolerant to large doses of serum. J Pharmacol Exp Ther 2, 221–9 (1910).
72. Myers, B.J. The migration of Anisakis-type larvae in experimental animals. Can J Zool 41, 147–8
(1963).
73. Dziekonska-Rynko, J., Rokicki, J. & Jablonowski, Z. Effects of ivermectin and albendazole against
Anisakis simplex in vitro and in guinea pigs. J Parasitol 88, 395–8 (2002).
698 Laboratory Models for Foodborne Infections
74. Smith, J.W. & Wootten, R. Anisakis and anisakiasis. in Advances in Parasitology, Vol. 16 (eds. Lumsden,
W.H.R., Muller, R. & Baker, J.R.) 93–163 (Academic Press, London, 1978).
75. Bier, J.W., Jackson, G.J., Earl, F.L. & Knollenberg, W.G. Gross and microscopic pathology with larval
Anisakis sp. and Phocanema sp. Nematodes from fish. Trans Am Microsc Soc 95, 264–265 (1976).
76. Strom, S.B. et al. Third-stage nematode larvae of Contracaecum osculatum from Baltic cod (Gadus
morhua) elicit eosinophilic granulomatous reactions when penetrating the stomach mucosa of pigs.
Parasitol Res 114, 1217–20 (2015).
77. Kuipers, F.C. Eosinophilic phlegmonous inflammation of the alimentary canal caused by a parasite
from the herring. Pathol Microbiol (Basel) 27, 925–30 (1964).
78. Ruitenberg, E.J., Berkvens, J.M. & Duyzings, M.J. Experimental Anisakis marina infections in rabbits.
J Comp Pathol 81, 157–63 (1971).
79. Hong, S.T. & Lee, S.H. Histopathological and serological observations on experimental anisakiasis of
rabbits. Kisaengchunghak Chapchi 25, 168–180 (1987).
80. Yang, H.J., Cho, Y.J. & Paik, Y.H. Changes of IgM and IgG antibody levels in experimental rabbit
anisakiasis as observed by ELISA and SDS-PAGE/immunoblot. Kisaengchunghak Chapchi 29, 389–96
(1991).
81. Kennedy, M.W. et al. The secreted and somatic antigens of the third stage larva of Anisakis simplex,
and antigenic relationship with Ascaris suum, Ascaris lumbricoides, and Toxocara canis. Mol Biochem
Parasitol 31, 35–46 (1988).
82. Iwasaki, K. & Torisu, M. Anisakis and eosinophil. II. Eosinophilic phlegmon experimentally induced in
normal rabbits by parasite-derived eosinophil chemotactic factor (ECF-P). Clin Immunol Immunopathol
23, 593–605 (1982).
83. Cuellar, C., Perteguer, M.J. & Rodero, M. Presence of IL-4-like molecules in larval excretory-secretory
products and crude extracts from Anisakis simplex. Scand J Immunol 53, 483–8 (2001).
84. Kobayashi, Y., Ishizaki, S., Nagashima, Y. & Shiomi, K. Ani s 1, the major allergen of Anisakis simplex:
purification by affinity chromatography and functional expression in Escherichia coli. Parasitol Int 57,
314–9 (2008).
85. Rodero, M., Jimenez, A., Chivato, T., Laguna, R. & Cuellar, C. Purification of Anisakis simplex antigen
by affinity chromatography. Parasitol Res 87, 736–40 (2001).
86. Rodero, M., Jimenez, A. & Cuellar, C. Evaluation by ELISA of Anisakis simplex larval antigen purified
by affinity chromatography. Mem Inst Oswaldo Cruz 97, 247–52 (2002).
87. Perez-Perez, J. et al. Molecular cloning of paramyosin, a new allergen of Anisakis simplex. Int Arch
Allergy Immunol 123, 120–9 (2000).
88. Iglesias, R., Leiro, J., Santamarina, M.T., Sanmartin, M.L. & Ubeira, F.M. Monoclonal antibodies
against diagnostic Anisakis simplex antigens. Parasitol Res 83, 755–61 (1997).
89. Ishikura, H. Clinical features of intestinal anisakiasis. in Intestinal Anisakiasis in Japan, Vol. III (eds.
Ishikura, H. et al.) 100 (Springer-Verlag, Tokyo, 1990).
90. Asami, K. & Inoshita, Y. Experimental anisakiasis in guinea pigs: factors influencing infection of larvae
in the host. Jpn J Parasitol 16, 415–22 (1967).
91. Oshima, T. Anisakis and anisakiasis in Japan and adjacent area. Progr Med Parasitol Japan 4, 305–93
(1972).
92. Yoshimura, H. Clinical patho-parasitology of extra-gastrointestinal anisakiasis. in Intestinal Anisakiasis
in Japan (eds. Ishikura, H. & Kikuchi, K.) 145–54 (Springer-Verlag, Tokyo, 1990).
93. Amano, T., Nakazawa, M., Sugiyama, H., Secor, W.E. & Oshima, T. Specific antibody patterns of
Wistar rats inoculated with third stage larvae of Anisakis simplex. J Parasitol 81, 536–42 (1995).
94. Cho, T.H. et al. The time course of biological and immunochemical allergy states induced by Anisakis
simplex larvae in rats. Clin Exp Immunol 143, 203–8 (2005).
95. Figueiredo, I., Jr. et al. A technique for the intra-gastric administration of live larvae of Anisakis simplex
in mice. Exp Parasitol 130, 285–7 (2012).
96. Zuloaga, J. et al. A rat model of intragastric infection with Anisakis spp. live larvae: histopathological
study. Parasitol Res 112, 2409–11 (2013).
97. Arizono, N., Yamada, M., Tegoshi, T. & Yoshikawa, M. Anisakis simplex sensu stricto and Anisakis
pegreffii: biological characteristics and pathogenetic potential in human anisakiasis. Foodborne Pathog
Dis 9, 517–21 (2012).
Anisakis 699
98. Abe, N. & Teramoto, I. Oral inoculation of live or dead third-stage larvae of Anisakis simplex in rats
suggests that only live larvae induce production of antibody specific to A. simplex. Acta Parasitol 59,
184–8 (2014).
99. Weerasooriya, M.V., Fujino, T., Ishii, Y. & Kagei, N. The value of external morphology in the identification
of larval anisakid nematodes: a scanning electron microscope study. Z Parasitenkd 72, 765–78 (1986).
100. Bowman, C.C. & Selgrade, M.K. Utility of rodent models for evaluating protein allergenicity. Regulat
Toxicol Pharmacol 54, S58–61 (2009).
101. Katz, D.H. Regulation of IgE antibody production by serum molecules. III. Induction of suppressive
activity by allogeneic lymphoid cell interactions and suppression of IgE synthesis by the allogeneic
effect. J Exp Med 149, 539–44 (1979).
102. Katz, D.H. Recent studies on the regulation of IgE antibody synthesis in experimental animals and man.
Immunology 41, 1–24 (1980).
103. Boyce, J.A. et al. Guidelines for the diagnosis and management of food allergy in the United States:
summary of the NIAID-sponsored expert panel report. J Allergy Clin Immunol 126, 1105–18 (2010).
104. Pabst, O. Trafficking of regulatory T cells in the intestinal immune system. Int Immunol 25, 139–43 (2013).
105. Pabst, O. & Mowat, A.M. Oral tolerance to food protein. Mucosal Immunol 5, 232–9 (2012).
106. Schippers, A. et al. β7 integrin controls immunogenic and tolerogenic mucosal B cell responses. Clin
Immunol 144, 87–97 (2012).
107. Campos, S.M. et al. Maternal immunomodulation of the offspring’s immunological system.
Immunobiology 219, 813–21 (2014).
108. Paschoal, P.O. et al. Food allergy/hypersensitivity: antigenicity or timing? Immunobiology 214, 269–78
(2009).
109. Teixeira, G. et al. Diet selection in immunologically manipulated mice. Immunobiology 213, 1–12
(2008).
110. Cardoso, C.R. et al. Modulation of mucosal immunity in a murine model of food-induced intestinal
inflammation. Clin Exp Allergy 38, 338–49 (2008).
111. Suko, M., Ogita, T., Okudaira, H. & Horiuchi, Y. Preferential enhancement of IgE antibody formation
by Bordetella pertussis. Int Arch Allergy Appl Immunol 54, 329–37 (1977).
112. Tada, T., Okumura, K., Ochiai, T. & Iwasa, S. Effect of lymphocytosis-promoting factor of Bordetella
pertussis on the immune response. II. Adjuvant effect for the production of reaginic antibody in the rat.
Int Arch Allergy Appl Immunol 43, 207–16 (1972).
113. Li, X.M. et al. A murine model of peanut anaphylaxis: T- and B-cell responses to a major peanut aller-
gen mimic human responses. J Allergy Clin Immunol 106, 150–8 (2000).
114. Ishizaka, K. & Ishizaka, T. Mechanisms of reaginic hypersensitivity and IgE antibody response.
Immunol Rev 41, 109–148 (1978).
115. Iglesias, R., Leiro, J., Ubeira, F.M., Santamarina, M.T. & Sanmartin, M.L. Anisakis simplex: antigen
recognition and antibody production in experimentally infected mice. Parasite Immunol 15, 243–50
(1993).
116. Iglesias, R., Leiro, J., Ubeira, F.M., Santamarina, M.T. & Sanmartin, M.L. Anisakis simplex: stage-
specific antigens recognized by mice. J Helminthol 69, 319–24 (1995).
117. Perteguer, M.J., Rodero, M., Flores, J.M., Dorea, R.C. & Cuellar, C. Cellular immune responses in mice
immunized with Anisakis simplex larval antigens. Parasitol Res 87, 396–404 (2001).
118. Vericimo, M.A., Figueiredo, I., Teixeira, G.A. & Clemente, S.C. Experimental anisakid infections in
mice. J Helminthol, 1–5 (2014).
119. Jones, R.E., Deardorff, T.L. & Kayes, S.G. Anisakis simplex: histopathological changes in experimen-
tally infected CBA/J mice. Exp Parasitol 70, 305–13 (1990).
120. Baeza, M.L. et al. Anisakis simplex allergy: a murine model of anaphylaxis induced by parasitic proteins
displays a mixed Th1/Th2 pattern. Clin Exp Immunol 142, 433–40 (2005).
121. Perteguer, M.J. & Cuellar, C. Isotype-specific immune responses in murine experimental anisakiasis.
Zentralbl Veterinarmed B 45, 603–10 (1998).
122. Perteguer, M.J. & Cuellar, C. Interleukin-4 production in BALB/c mice immunized with Anisakis sim-
plex. Mem Inst Oswaldo Cruz 96, 979–82 (2001).
123. Kirstein, F. et al. Anisakis pegreffii-induced airway hyperresponsiveness is mediated by γ-interferon in
the absence of interleukin-4 receptor α responsiveness. Infect Immun 78, 4077–86 (2010).
700 Laboratory Models for Foodborne Infections
124. Park, H.K. et al. A 24 kDa excretory-secretory protein of Anisakis simplex larvae could elicit allergic
airway inflammation in mice. Korean J Parasitol 49, 373–80 (2011).
125. Cho, M.K. et al. Allergenicity of two Anisakis simplex allergens evaluated in vivo using an experimental
mouse model. Exp Parasitol 146, 71–7 (2014).
126. Falcone, F.H., Zang, X., MacDonald, A.S., Maizels, R.M. & Allen, J.E. A Brugia malayi homolog of
macrophage migration inhibitory factor reveals an important link between macrophages and eosinophil
recruitment during nematode infection. J Immunol 167, 5348–54 (2001).
127. Gregory, W.F., Blaxter, M.L. & Maizels, R.M. Differentially expressed, abundant trans-spliced cDNAs
from larval Brugia malayi. Mol Biochem Parasitol 87, 85–95 (1997).
128. Murray, J., Gregory, W.F., Gomez-Escobar, N., Atmadja, A.K. & Maizels, R.M. Expression and immune
recognition of Brugia malayi VAL-1, a homologue of vespid venom allergens and Ancylostoma secreted
proteins. Mol Biochem Parasitol 118, 89–96 (2001).
129. Park, S.K. et al. Macrophage migration inhibitory factor homologs of Anisakis simplex suppress Th2
response in allergic airway inflammation model via CD4 +CD25+Foxp3+ T cell recruitment. J Immunol
182, 6907–14 (2009).
130. Pastrana, D.V. et al. Filarial nematode parasites secrete a homologue of the human cytokine macrophage
migration inhibitory factor. Infect Immun 66, 5955–63 (1998).
131. Yenbutr, P. & Scott, A.L. Molecular cloning of a serine proteinase inhibitor from Brugia malayi. Infect
Immun 63, 1745–53 (1995).
132. Yu, H.S. et al. Anisakis simplex: analysis of expressed sequence tags (ESTs) of third-stage larva. Exp
Parasitol 117, 51–6 (2007).
133. Cho, M.K., Lee, C.H. & Yu, H.S. Amelioration of intestinal colitis by macrophage migration inhibitory
factor isolated from intestinal parasites through toll-like receptor 2. Parasite Immunol 33, 265–75 (2011).
134. Cho, M.K. et al. TLR2-dependent amelioration of allergic airway inflammation by parasitic nematode
type II MIF in mice. Parasite Immunol 37, 180–91 (2015).
135. Quiazon, K.M., Yoshinaga, T. & Ogawa, K. Experimental challenge of Anisakis simplex sensu stricto
and Anisakis pegreffii (Nematoda: Anisakidae) in rainbow trout and olive flounder. Parasitol Int 60,
126–31 (2011).
136. Bahlool, Q.Z., Skovgaard, A., Kania, P., Haarder, S. & Buchmann, K. Microhabitat preference of
Anisakis simplex in three salmonid species: immunological implications. Vet Parasitol 190, 489–95
(2012).
137. Haarder, S., Kania, P.W. & Buchmann, K. Comparative infectivity of three larval nematode species in
three different salmonids. Parasitol Res 112, 2997–3004 (2013).
138. Bahlool, Q.Z., Skovgaard, A., Kania, P.W. & Buchmann, K. Effects of excretory/secretory products from
Anisakis simplex (Nematoda) on immune gene expression in rainbow trout (Oncorhynchus mykiss). Fish
Shellfish Immunol 35, 734–9 (2013).
139. Bass, C.C. & Johns, F.M. The cultivation of malarial plasmodia (Plasmodium vivax and Plasmodium
falciparum) in vitro. J Exp Med 16, 567–79 (1912).
140. Sommerville, R.I. & Davey, K.G. Stimuli for cuticle formation and ecdysis in vitro of the infective larva
of Anisakis sp. (Nematoda: Ascaridoidea). Int J Parasitol 6, 433–9 (1976).
141. Silverman, P.H. In vitro cultivation procedures for parasitic helminths. in Advances Parasitology
(ed. Dawes, B.) 159–222 (Academic Press, London, 1965).
142. Smyth, J.D. In Vitro Cultivation of Parasitic Helminths, (CRC Press, Boca Raton, 1990).
143. Yasuraoka, K. & Hata, H. In vitro cultivation of parasitic helminths. in Progress of Medical Parasitology
Japan, Vol. 7 (eds. Osturu, M., Kamegai, S. & Hayashi, S.) 211–26 (Meguro Parasitological Museum,
Tokyo, 2003).
144. Townsley, P., Wight, H., Scott, M. & Hughes, M. The in-vitro maturation of the parasitic nematode,
Terranova decipiens, from cod muscle. J Fish Board Can 20, 743–7 (1963).
145. Van Banning, P. Some notes on a successful rearing of the herring-worm, Anisakis marina L. (Nematoda:
Heterocheilidae). J Conseil 34, 84–8 (1971).
146. Grabda, J. Studies on survival and development in vitro of Anisakis simplex stage 3 larvae in time
[Worm parasite of herring and other marine fish, Nematoda]. Acta Ichthyol Piscatoria 12, 1 (1982).
147. Davila, C., Malagon, D., Valero, A., Benitez, R. & Adroher, F.J. Anisakis simplex: CO(2)-fixing enzymes
and development throughout the in vitro cultivation from third larval stage to adult. Exp Parasitol 114,
10–5 (2006).
Anisakis 701
148. Adroher, F.J., Malagon, D., Valero, A. & Benitez, R. In vitro development of the fish parasite
Hysterothylacium aduncum from the third larval stage recovered from a host to the third larval stage
hatched from the egg. Dis Aquat Organ 58, 41–5 (2004).
149. Iglesias, L., Valero, A. & Adroher, F.J. Some factors which influence the in vitro maintenance of
Anisakis simplex (Nematoda). Folia Parasitol (Praha) 44, 297–301 (1997).
150. Iglesias, L., Valero, A., Benitez, R. & Adroher, F.J. In vitro cultivation of Anisakis simplex: pepsin
increases survival and moulting from fourth larval to adult stage. Parasitology 123, 285–91 (2001).
151. Iglesias, L., Valero, A., Galvez, L., Benitez, R. & Adroher, F.J. In vitro cultivation of Hysterothylacium
aduncum (Nematoda: Anisakidae) from 3rd-stage larvae to egg-laying adults. Parasitology 125, 467–75
(2002).
152. Malagon, D., Benitez, R., Valero, A. & Adroher, F.J. CO2-fixing enzymes and phosphoenolpyruvate
metabolism in the fish parasite Hysterothylacium aduncum (Ascaridoidea, Anisakidae). Dis Aquat
Organ 85, 217–23 (2009).
153. Mladineo, I., Simat, V., Miletic, J., Beck, R. & Poljak, V. Molecular identification and population
dynamic of Anisakis pegreffii (Nematoda: Anisakidae Dujardin, 1845) isolated from the European
anchovy (Engraulis encrasicolus L.) in the Adriatic Sea. Int J Food Microbiol 157, 224–9 (2012).
154. Stromnes, E. An in vitro study of lipid preference in whaleworm (Anisakis simplex, Nematoda,
Ascaridoidea, Anisakidae) third-stage larvae. Parasitol Res 113, 1113–8 (2014).
155. Young, P. The relationship between the presence of larval anisakine nematodes in cod and marine mam-
mals in British home waters. J Appl Ecol, 459–85 (1972).
156. Goto, C., Kasuya, S., Koga, K., Ohtomo, H. & Kagei, N. Lethal efficacy of extract from Zingiber offici-
nale (traditional Chinese medicine) or [6]-shogaol and [6]-gingerol in Anisakis larvae in vitro. Parasitol
Res 76, 653–6 (1990).
157. Kasuya, S. et al. Lethal efficacy of leaf extract from Perilla frutescens (traditional Chinese medicine) or
perillaldehyde on Anisakis larvae in vitro. Jpn J Parasitol 39, 220–5 (1990).
158. Goto, C., Kasuya, S., Koga, K., Ohtomo, H. & Kagei, N. Lethal efficacy of extract from Zingiber offici-
nale (traditional Chinese medicine) or [6]-shogaol and [6]-gingerol in Anisakis larvae in vitro. Parasitol
Res 76, 653–6 (1990).
159. Lin, R.-J., Chen, C.-Y., Chung, L.-Y. & Yen, C.-M. Larvicidal activities of ginger (Zingiber officinale)
against Angiostrongylus cantonensis. Acta Trop 115, 69–76 (2010).
160. Hierro, I., Valero, A. & Navarro, M.C. In vivo larvicidal activity of monoterpenic derivatives from aro-
matic plants against L3 larvae of Anisakis simplex s.l. Phytomedicine 13, 527–31 (2006).
161. Navarro-Moll, M.C., Romero, M.C., Montilla, M.P. & Valero, A. In vitro and in vivo activity of three
sesquiterpenes against L(3) larvae of Anisakis type I. Exp Parasitol 127, 405–8 (2011).
162. Krawisz, J.E., Sharon, P. & Stenson, W.F. Quantitative assay for acute intestinal inflammation based on
myeloperoxidase activity. Assessment of inflammation in rat and hamster models. Gastroenterology 87,
1344–50 (1984).
43
Clonorchis sinensis
CONTENTS
43.1 Introduction................................................................................................................................... 703
43.1.1 History.............................................................................................................................. 703
43.1.2 Life Cycle and Morphology............................................................................................. 703
43.1.3 Pathogenesis and Pathology............................................................................................. 705
43.1.4 Clinical Manifestations.................................................................................................... 707
43.1.5 Epidemiology.................................................................................................................... 707
43.1.6 Diagnosis.......................................................................................................................... 708
43.1.7 Chemotherapy and Control Measures.............................................................................. 709
43.2 Laboratory Models for C. sinensis Research................................................................................ 709
43.2.1 Rationale on Models......................................................................................................... 709
43.2.2 Laboratory Animals for Host Susceptibility to C. sinensis............................................. 709
43.2.3 Animal Models for Immune Response and Reinfection...................................................710
43.2.4 Animal Models for Metabolism Study..............................................................................711
43.2.5 Animal Models for Chemotherapy....................................................................................711
43.2.6 Animal Models for Histopathology and Carcinogenesis..................................................711
43.2.7 In Vitro Models..................................................................................................................712
43.3 Conclusion......................................................................................................................................713
References................................................................................................................................................713
43.1 Introduction
43.1.1 History
Clonorchis sinensis was first discovered by James McConnell at the autopsy of a Chinese male in
1874. Following a detailed description of the fluke, Spencer Cobbold suggested that the species be
named Distoma sinense in 1875. Afterward, its nomenclature was changed to Opisthorchis sinensis
by Blanchard in 1895 and finally renamed as Clonorchis sinensis by Arthur Looss in 1907. Commonly
called the “Chinese or oriental liver fluke,” C. sinensis belongs to the family Opisthorchiidae. Similar to
those in the families Dicrocoeliidae and Fasciolidae, members of the family Opisthorchiidae infect the
biliary tree of mammals including humans. Currently, C. sinensis is recognized as a major parasite of
the human intrahepatic bile duct causing clonorchiasis [1].
703
704 Laboratory Models for Foodborne Infections
FIGURE 43.2 Gross view of recovered adult worms of Clonorchis sinensis from a man after chemotherapy in Korea. The
uterus of worms that are red in color is full of eggs.
metacercariae excyst in the duodenum after trypsin digestion [2]. The excysted juvenile flukes migrate
into the common bile duct through the ampulla of Vater to reach the intrahepatic biliary duct. One
experimental study using rabbits observed that the juvenile flukes moved to the intrahepatic bile duct
as early as 10–20 min after they excysted and entered into the duodenum [3]. The flukes develop into
adults within 4 weeks and begin to produce eggs, which mix in feces and pass through the intestine [1].
Clonorchis sinensis 705
After their discharge by mammalian hosts, the eggs are ingested by freshwater snail, which is the
intermediate hosts, in the rivers or lakes. The first intermediate snail hosts are Parafossarulus or
Bithynia species (Figure 43.3). In the snail host, the miracidium hatches out in the rectum and pen-
etrates the wall to reach the perirectal tissue. In the perirectal tissue, the miracidium undergoes meta-
morphoses into a sporocyst, which in turn gives rise to rediae and then cercariae. One miracidium may
amplify to 1000–1500 cercariae, which emerge into the water [4]. The emerging cercariae freely swim
and penetrate the skin, gills, fins, or muscles of freshwater fish within 24–48 h. In the fish, the cercariae
encyst to become metacercariae. Numerous species of small-sized freshwater fish such as Pseudorasbora
parva, Pungtungia herzi, Pseudogobio esocinus, Acheilognathus intermedia, Odontobutis interrupta,
and others serve as the second intermediate host of C. sinensis, harboring the metacercariae [5,6].
A study investigating freshwater fish in their infection status with metacercariae of C. sinensis in Korea
reported that the number of metacercariae per fish was found to be 48 (1–1142) in P. parva and 60
(1–412) in P. herzi (Figure 43.4), implying that the two species are able to be the index fish for the estima-
tion of C. sinensis transmission in a certain locality [5]. When humans or mammals consume infected
raw freshwater fish or poorly cooked ones (including smoked or pickled fish), the metacercariae infect
the host and develop into adults.
FIGURE 43.4 Second intermediate host. (A) Pseudorasbora parva. (B) Pungtungia herzi.
FIGURE 43.5 Histopathology of the Clonorchis sinensis-infected intrahepatic bile duct in an experimentally infected
cat. HE stained, original magnification, ×40. The duct is enlarged with adenomatous hyperplasia and periductal fibrosis.
Clonorchis sinensis 707
43.1.4 Clinical Manifestations
Clonorchiasis is rather well adapted to human hosts, and most of the light infected cases show no clinical
findings. When present, clinical features of clonorchiasis are often nonspecific and related to inflamma-
tion and intermittent obstruction of the biliary ducts. Adult worms cause obstruction and blockage of
the bile flow, which may induce epigastric discomfort or pain, swelling, and jaundice. In severe cases
with heavy worm burdens, abdominal pain, nausea, and diarrhea can occur. However, in long-standing
and untreated infections, chronic inflammation of the biliary system can lead to a neoplastic change as
cholangiocarcinoma (CCA) [1].
43.1.5 Epidemiology
Globally, clonorchiasis is a common foodborne zoonosis distributed mainly in East Asia including
China, Korea, Vietnam, the Philippines, and the far eastern part of Russia [1,12]. It may also occur
in other regions where there are immigrants from endemic areas [13]. It is estimated that more than
200 × 106 people are at risk of infection, 15–20 × 106 people are infected, and 1.5–2 × 106 show symp-
toms or complications [1].
Several studies in Korea reported that the infection status of C. sinensis has shown only a little decrease
despite wide use of praziquantel, an effective anthelmintic, for 30 years [14,15]. Cho et al. [14] reported
that the prevalence of C. sinensis was 11.1% among 24,075 inhabitants in the riverside areas of southern
Korea. Recently, several small endemic foci were reported in southern rural communities where eating
raw fish is common [15].
The Korea Centers for Disease Control and Prevention (KCDC) performed the eighth nationwide
survey on prevalence of intestinal parasitic infections in Korea and reported that the overall egg-positive
rate of C. sinensis was 1.9% in 2012 compared to 4.6% in 1971, 1.8% in 1976, 2.6% in 1981, 2.7% in
1986, 2.2% in 1992, 1.4% in 1997, and 2.4% in 2004 [16]. The egg-positive rates, which are regarded as
average prevalence of clonorchiasis in the general population in the surveyed year, have been decreasing
slowly. Hong and Fang [1] explained that the slow decrease is due mainly to frequent treatment failure
or reinfection after treatment. Still, about 1 × 106 residents who live along the river beds are infected in
southern endemic areas in Korea [1,16].
In a study conducted during 2000–2004 in Korea, C. sinensis prevalence (2.1% in Chuncheon, 7.8% in
Chungju, and 31.3% in Haman) was significantly correlated with CCA incidence rates (0.3, 1.8, and 5.5 per
100,000 persons, respectively) [17]. Furthermore, earlier case–control study in Busan, Korea, including 41
patients with CCA and 203 cases of hepatocellular carcinoma (HCC) revealed that CCA was significantly
correlated with C. sinensis eggs in stool, past history of liver flukes, hepatitis history, and a history of heavy
consumption of alcoholic beverages [18]. Shin et al. [19] described that the odds ratio of CCA due to C. sinensis
infection was 4.7, and about 10% of CCA cases in Korea were due to this organism. The overall odds ratio of
CCA is 4.47 for clonorchiasis, and about 5000 CCA cases are estimated annually in the world [20].
In Guangdong, China, where fish farming is common, the prevalence of human clonorchiasis is about
14%, and it may reach 80% in rural endemic areas [8]. A recent analysis of data from three parasitic
disease surveys conducted in Hengxian County, China, in the last 22 years showed substantial increases
in the patterns of clonorchiasis prevalence and infection intensity and decrease in trends of prevalence
of soil-transmitting helminths [21]. Another recent study in Guangdong, China, reported that incidence
rates of clonorchiasis showed a direct increasing trend by years with temperature change [22].
The distribution of clonorchiasis is determined not only by the distribution of snail intermediate hosts
but also by the fish-consuming custom of residents in the endemic community. Fishing and eating of raw
fish is an old popular tradition that has remained habitual and constant for thousands of years in endemic
areas. The eating habit has resulted in persistent transmission of the liver fluke to humans [23,24]. There
are several well-known risk factors that are associated with the transmission of clonorchiasis such as
poor educational level of local residents, lack of sanitation, habit of eating raw or undercooked freshwater
fish, development of freshwater aquaculture, and lack of systematic control activities in many endemic
areas. In particular, freshwater aquaculture has rapidly expanded, with a resulting increase in fish con-
tamination that has resulted from a lack of quarantine measures for fish products [25].
708 Laboratory Models for Foodborne Infections
Regarding accumulated infection, infection rate and burden as measured by egg-positive rate and
number of eggs per gram of feces (EPGs) have been recorded as increasing with age in men [23,26]. The
reason for its increase among adult men reflects behavioral patterns of fishing as an occupation or recre-
ation. Since the lifespan of C. sinensis is estimated to be around 30 years, the infection occurs repeatedly
through the life of humans [23]. That finding suggests that host immunity is not so efficient to prevent
reinfection or superinfection in humans.
43.1.6 Diagnosis
Diagnosis of clonorchiasis is essential in the control programs as well as for treatment. Early diagnosis
and treatment is important to prevent serious complications of clonorchiasis in humans. Detection of
eggs or adult worms in feces, bile, or duodenal fluids is a definite diagnosis. However, since detection of
worms from a human body needs invasive procedures, the gold standard diagnosis of C. sinensis infec-
tion is microscopic identification of eggs in feces. The Kato-Katz (KK) method is reliable for diagnosis
of clonorchiasis by egg detection [27], but the formalin-ether concentration technique is more sensitive
than the KK method in cases of mild infection [28]. Furthermore, Choi et al. [28] reported that is good
correlation between the EPGs determined by KK method and by direct smear. Since the KK method is
far from straightforward for differential diagnosis between eggs of C. sinensis and heterophyid flukes,
well-trained technicians are required for mixed infections [6]. Morphological identification of these eggs
by fecal examination is very tricky and requires much more experience (Figure 43.6).
Methods other than fecal examination include serology tests, detection of DNA, and image diagnosis.
There have been many serological methods used for diagnosis of C. sinensis infection, but ELISA is
more commonly used [29]. The ELISA diagnosis using excretory-secretory antigen (ESA) and crude
antigen (CA) of C. sinensis showed sensitivities of 92.5% and 88.2% and specificities of 93.1% and
87.8%, respectively [30]. The data implied that ESA was a better serodiagnostic antigen than CA for
ELISA. ELISA detects specific IgG antibodies in serum, and the antibodies may last over 2 years after
treatment, which may make it less specific.
Clonorchiasis has been diagnosed using ultrasound scanning, computerized tomography (CT), and
magnetic resonance imaging (MRI) [31,32]. Choi and Hong [31] reported that radiological images
employing CT or MRI of the liver were correlated with worm burdens (EPG counts) frequency and
also severity. The images of the liver and bile ducts can be used as a good practical alternative diag-
nostic method of clonorchiasis. Although most of the positive images are acceptable for diagnosis,
their sensitivity and specificity are lower than those of the fecal examination. This is mainly because
those images recognize the tissue changes caused by the worms but not the worms itself. The images
of cholangiocarcinoma associated with clonorchiasis show both the tumor with obstruction images
FIGURE 43.6 Eggs of C. sinensis by the Kato-Katz smear. Original magnification, ×400.
Clonorchis sinensis 709
and diffuse dilatation of the peripheral intrahepatic bile ducts [31]. The remaining ductal dilatation
with thick wall after chemotherapy further reduces the image specificity [11]. In endemic areas, it
is recommended to make diagnosis of clonorchiasis by any feasible method and to treat positives
more actively.
43.2.1 Rationale on Models
Animal research has provided valuable information about many physiological processes that are relevant
to humans and human pathogens. Rodents (e.g., mice, rats) and rabbits are the most commonly used
laboratory animal models for biomedical studies. Of course, several other mammals have been used. The
animal model for C. sinensis research may depend on host–parasite relationship, feasibility of animal
quarters, research purpose, and cost.
TABLE 43.1
Recovery Rates of C. sinensis in Mammals by Experimental Infection
Recovery Rates (%)
Mammals No. of Heads Mean ± SD
Rats 10 63.9 ± 20.4
Mice 11 17.6 ± 13.0
Hamsters 9 68.0 ± 15.0
Guinea pigs 10 34.7 ± 13.7
Rabbits 7 35.0 ± 7.6
Dogs 3 41.6 ± 14.5
Source: Sohn, W.M., et al., Korean J. Parasitol., 44, 163–166, 2006.
Mice are the most commonly used laboratory animal model in biomedical research, but they are
not susceptible to C. sinensis infection [34,36]. Many strains of mice have been developed for diverse
research purposes, and the susceptibility was different in the strains [36]. ICR, Balb/c, C57BL/6, DDY,
CBA/N, and C3H/HeN mouse strains were not susceptible to C. sinensis infection and showed very low
recovery rates below 10%. Only the FVB strain showed 17.6% recovery rate, which is little higher than
other strains [34]. Since the FVB strain is relatively susceptible compared to other strains, this strain has
been used for in vivo studies of C. sinensis as a mouse model [37,38]. The FVB mice show cystic dilata-
tion of the infected bile duct, which is a unique pathological change [38].
On the other hand, there have been reports that rats are resistant (to some degree) to reinfection
by C. sinensis. The significant increase in the levels of bile IgA antibodies and serum IgE antibodies
in resistant reinfected rats might play a role in the development of resistance to reinfection by C. sinensis [39].
Rabbits, dogs, and cats are susceptible hosts for C. sinensis infection. Rabbits are commonly used to
maintain adult worms in the laboratory. Dogs, cats, and pigs are not used commonly for experimental
infection of C. sinensis, but their natural infection has been reported to be common in China [26,40].
They are good reservoir hosts of C. sinensis in nature.
changes in CBA/N mice as compared to C57BL/6 mice. Based on these results, it has been suggested
that CBA/N could be an appropriate mouse model for studying early immune responses in liver fluke
infection [45].
A few experimental studies on reinfection or superinfection of C. sinensis used mostly rats because
they are susceptible to primary infection but resistant to reinfection [34,39,46]. Meanwhile, hamsters,
dogs, and humans are susceptible to primary and also reinfection or superinfection, and the infection
is accumulated in those hosts. The resistance of the rats was mainly modulated by the local immune
response in the infected tissue [39].
The findings of laboratory studies suggest that C. sinensis infection in mammals may suppress host
immunity, which makes the host susceptible. The host immunity provokes the host resistant to reinfec-
tion or superinfection in rats but not in humans. Humans are repeatedly reinfected.
fibrosis, and CCA [52–54]. In extreme cases, liver enlargement, thickening of the bile ducts, fibrosis, and
some destruction of liver parenchyma are evident. However, unlike Fasciola, Clonorchis does not invade
liver tissues and therefore, does not cause extensive liver necrosis [8].
Several well-documented epidemiological, histopathological, and experimental studies of
C. sinensis have provided convincing evidence of a relationship between this trematode infection and
the tendency for malignant transformation of the biliary epithelium in humans and experimentally
infected animals. The International Agency for Research on Cancer (IARC) of the World Health
Organization recategorized C. sinensis as a Group 1 biological carcinogen with Opisthorchis viver-
rini, as it produces human CCA [55,56].
The neoplastic response that is associated with clonorchiasis is CCA, which is a highly malignant
epithelial cancer of the biliary tract [57]. In humans, primary sclerosing cholangitis, hepatolithiasis,
fibropolycystic diseases of the biliary tract, Caroli’s disease, and liver fluke infection have been con-
sidered as predisposing conditions for development of CCA [18,57,58].
Concerning history of experimental animals for induction of CCA, Thamavit et al. [59] first reported the
possible development of CCA in the hamster model by administering Opisthorchis viverrini and admin-
istration of N-nitrosodimethylamine (NDMA; exogenously) in drinking water. Since then, similar exper-
iments were also performed following C. sinensis infection in combination with 2-acetylaminofluorene
or N-nitroso compounds (NDMA or N-nitrosodihydroxydi-n-propylamine) in hamsters [ 60–62] and rats
[63]. These studies supported the role of C. sinensis as a promotor instead of an initiator. Experimental
infection with C. sinensis alone in animals has never induced bile duct tumors [62]. During the past two
decades, the primary experimental model for CCA induced by C. sinensis [60–62] as well as O. viverrini
is the Syrian golden hamster [64,65]. The study using rat model on the possible modifying potential of
C. sinensis infection and NDMA suggested that C. sinensis infection might facilitate the proliferation of
NDMA-induced preneoplastic lesions of the liver [63].
One experimental animal study using animals other than hamsters or rats reported that intrahe-
patic cholangiocarcinomas (ICCs) could originate from fully differentiated hepatocytes. Using a
mouse model of hepatocyte fate tracing, they found that activated NOTCH and AKT signaling coop-
erated to convert normal hepatocytes into neoplastic biliary cells that acted as precursors of rapidly
progressing, lethal ICCs [66].
For the biliary hyperplasia, several in vitro experiments provided strong evidence that C. sinensis pos-
sessed mitogenic factors in its ESPs and exhibited different levels of cell proliferations. For instance, C.
sinensis ESPs were reported to exhibit antiapoptotic effect in human CCA cells [71]. Likewise, exposure
of human CCA cells (HuCCT1) to C. sinensis ESPs demonstrated increased free radical generation
through activation of NADPH oxidase, inducible nitric oxide synthase (iNOS), and xanthine oxidase,
subsequently leading to nuclear factor-kappa B (NF-κB)-mediated inflammatory processes [72].
There has been a well-established body of knowledge about these highly reactive free radicals that
can damage biologically relevant molecules of cells. Consequently, oxidative stress arising as a result of
an imbalance between free radical production and antioxidant defenses is associated with damage to a
wide range of molecular species including lipids, proteins, and nucleic acids [73,74]. The damage to cells
caused by free radicals, especially the damage to DNA, may trigger the development of neoplasms and
also other health-related disorders.
43.3 Conclusion
C. sinensis, a foodborne trematode infecting intrahepatic bile duct of humans, is considered as one
of the target NTDs by WHO. Since its host specificity is low, most of the laboratory mammals except
for mice are susceptible, and rats or rabbits are commonly used for common experimental infection.
For experimental CCA studies, hamster is the only successful animal model. It is recommended to
develop other animal models such as murine model for experimental studies on immunology and oncol-
ogy of C. sinensis. In vitro cell cultivation is a good option for molecular studies of ESPs and generates
molecular insights into its carcinogenesis.
REFERENCES
1. Hong ST, Fang Y. Clonorchis sinensis and clonorchiasis, an update. Parasitol Int. 2012; 61: 17–24.
2. Li S, Chung YB, Chung BS, Choi MH, Yu JR, Hong ST. The involvement of the cysteine proteases of
Clonorchis sinensis metacercariae in excystment. Parasitol Res. 2004; 93: 36–40.
3. Kim TI, Yoo WG, Kwak BK, Seok JW, Hong SJ. Tracing of the bile-chemotactic migration of juvenile
Clonorchis sinensis in rabbits by PET-CT. PLoS Negl Trop Dis. 2011; 5: e1414.
4. Rim HJ. Current pathobiology and chemotherapy of clonorchiasis. Korean J Parasitol. 1986;
24(Suppl): 1–141.
5. Kim EM, et al. Infection status of freshwater fish with metacercariae of Clonorchis sinensis in Korea.
Korean J Parasitol. 2008; 46: 247–51.
6. Chai JY. Fish-borne parasitic diseases. Hanyang Med Rev. 2010; 30: 223–31.
7. Kim YJ, Choi MH, Hong ST, Bae YM. Proliferative effects of excretory/secretory products from
Clonorchis sinensis on the human epithelial cell line HEK293 via regulation of the transcription factor
E2F1. Parasitol Res. 2008; 102: 411–7.
8. Bogitsh BJ, Carter CE, Oeltmann TN. Human Parasitology. 4th ed. Amsterdam: Elsevier Inc; 2013.
9. Hong ST, Kho WG, Kim WH, Chai JY, Lee SH. Turnover of biliary epithelial cells in Clonorchis
sinensis infected rats. Korean J Parasitol. 1993; 31: 83–9.
10. Rim HJ. Clonorchiasis: an update. J Helminthol. 2005; 79: 269–81.
11. Lee SH, Hong ST, Kim CS, Sohn WM, Chai JY, Lee YS. Histopathological changes of the liver after
praziquantel treatment in Clonorchis sinensis infected rabbits. Korean J Parasitol. 1987; 25: 110–22.
12. Fürst T, Keiser J, Utzinger J. Global burden of human food-borne trematodiasis: a systematic review and
meta-analysis. J Lancet Infect Dis. 2012; 12: 210–21.
13. WHO. Control of foodborne trematode infections. Report of a WHO study group. WHO Tech Rep Ser.
1995; 849: 1–157.
14. Cho SH, et al. Prevalence of clonorchiasis in southern endemic areas of Korea in 2006. Korean
J Parasitol. 2008; 46: 133–7.
15. Park DS, Na SJ, Cho SH, June KJ, Cho YC, Lee YH. Prevalence and risk factors of clonorchiasis among
residents of riverside areas in Muju-gun, Jeollabuk-do, Korea. Korean J Parasitol. 2014; 52: 391–7.
714 Laboratory Models for Foodborne Infections
16. Korea Centers for Disease Control and Prevention. The 8th National surveys on the prevalence of
intestinal parasitic infections. Seoul, Korea; 2013. pp. 35–68. http://www.cdc.go.kr/CDC/contents/
CdcKrContentLink.jsp?fid=31&cid=24152&ctype=6.
17. Lim MK, et al. Clonorchis sinensis infection and increasing risk of cholangiocarcinoma in the Republic
of Korea. Am J Trop Med Hyg. 2006; 75: 93–6.
18. Shin HR, et al. Hepatitis B and C virus, Clonorchis sinensis for the risk of liver cancer: a case-control
study in Pusan, Korea. Int J Epidemiol. 1996; 25: 933–40.
19. Shin HR, et al. Descriptive epidemiology of cholangiocarcinoma and clonorchiasis in Korea. J Korean
Med Sci. 2010; 25: 1011–6.
20. Qian MB, Chen YD, Liang S, Yang GJ, Zhou XN. The global epidemiology of clonorchiasis and its
relation with cholangiocarcinoma. Infect Dis Poverty. 2012; 1: 4.
21. Qian MB, et al. Prevalence and intensity of foodborne clonorchiasis, Hengxian County, China,
1989–2011. Emerg Infect Dis. 2014; 20: 1872–5.
22. Li T, Yang Z, Wang M. Correlation between clonorchiasis incidences and climatic factors in Guangzhou,
China. Parasite Vectors. 2014; 7: 29.
23. Hong ST. Clonorchis sinensis. In: Miliotis MD, Bier JW, editors. International Handbook of Foodborne
Pathogens. New York, Basel: Marcel Dekker, Inc.; 2003. pp. 581–92.
24. Seo M, et al. Paleoparasitological report on the stool from a medieval child mummy in Yangju. Korean
J Parasitol. 2007; 93: 589–92.
25. Fang Y, et al. Investigation and analysis on epidemic status of clonorchiasis in Guangdong province.
J Pathog Biol. 2007; 2: 241–3.
26. Choi MH, et al. Effect of control strategies on prevalence, incidence and re-infection of clonorchiasis in
endemic areas of China. PLoS Negl Trop Dis. 2010; 4: e601.
27. Hong ST, Choi MH, Kim CH, Chung BS, Ji Z. The Kato-Katz method is reliable for diagnosis of
Clonorchis sinensis infection. Diagn Microbiol Inf Dis. 2003; 47: 345–9.
28. Choi MH, Ge T, Yuan S, Hong ST. Correlation of egg counts of Clonorchis sinensis by three methods
of fecal examination. Korean J Parasitol. 2005; 43: 115–7.
29. Lee MK, Hong SJ, Kim HR. Seroprevalence of tissue invading parasitic infections diagnosed by ELISA
in Korea. J Korean Med Sci. 2010; 25: 1272–6.
30. Choi MH, Park IC, Li S, Hong ST. Excretory-secretory antigen is better than crude antigen for the
serodiagnosis of clonorchiasis by ELISA. Korean J Parasitol. 2003; 41: 35–9.
31. Choi D, Hong ST. Imaging diagnosis of clonorchiasis. Korean J Parasitol. 2007; 45: 77–85.
32. Lim JU, Joo KR, Shin HP, Cha JM, Lee JI, Lim SJ. Obstructive jaundice caused by clonorchiasis-
associated duodenal papillitis: a case report. J Korean Med Sci. 2011; 26: 135–7.
33. World Health Organization (WHO). First WHO report on neglected tropical diseases 2010: working
to overcome the global impact of neglected tropical diseases. http://www.who.int/neglected_diseases/
2010report/en/.
34. Sohn WM, Choi MH, Hong ST. Susceptibility of experimental animals to reinfection with Clonorchis
sinensis. Korean J Parasitol. 2006; 44: 163–6.
35. Wang X, et al. Experimental model in rats for study on transmission dynamics and evaluation of
Clonorchis sinensis infection immunologically, morphologically, and pathologically. Parasitol Res.
2009; 106: 15–21.
36. Uddin MH, Li S, Bae YM, Choi MH, Hong ST. Strain variation in the susceptibility and immune
response to Clonorchis sinensis infection in mice. Parasitol Int. 2012; 61: 118–23.
37. Yoon BI, et al. Infectivity and pathological changes in murine clonorchiasis: comparison in immuno-
competent and immunodeficient mice. J Vet Med Sci. 2001; 63: 421–5.
38. Kim EM, Bae YM, Choi MH, Hong ST. Cyst formation, increased anti-inflammatory cytokines and
expression of chemokines support for Clonorchis sinensis infection in FVB mice. Parasitol Int. 2012;
61: 124–9.
39. Zhang H, Chung BS, Li S, Choi MH, Hong ST. Changing patterns of serum and bile antibodies in
re-infected rats with Clonorchis sinensis. Korean J Parasitol. 2008; 46: 17–22.
40. Lin RQ, et al. Prevalence of Clonorchis sinensis infection in dogs and cats in subtropical southern
China. Parasite Vectors. 2011; 4: 180.
Clonorchis sinensis 715
41. Zheng M, Hu K, Liu W, Li H, Chen J, Yu X. Proteomic analysis of different period excretory secretory
products from Clonorchis sinensis adult worms: molecular characterization, immunolocalization, and
serological reactivity of two excretory secretory antigens-methionine aminopeptidase 2 and acid phos-
phatase. Parasitol Res. 2013; 112: 1287–97.
42. Xu Y, et al. Molecular characterization and immune modulation properties of Clonorchis sinensis-
derived RNASET2. Parasite Vectors. 2013; 6: 360.
43. Jin Y, Wi HJ, Choi MH, Hong ST, Bae YM. Regulation of anti-inflammatory cytokines IL-10 and
TGF-β in mouse dendritic cells through treatment with Clonorchis sinensis crude antigen. Exp Mol
Med. 2014; 46: e74.
44. Jeong YI, et al. Identification of anti-allergic effect of Clonorchis sinensis-derived protein venom
allergen-like proteins (CsVAL). Biochem Biophys Res Commun. 2014; 445: 549–55.
45. Jin Y, Wi HJ, Choi MH, Hong ST, Bae YM. Differential regulation of pathogenesis and immune
reactions was performed in C57BL/6 and CBA/N mice infected with Clonorchis sinensis. J Immunol.
2012; 188: 164.22.
46. Chung BS, et al. Development of resistance to reinfection by Clonorchis sinensis in rats. Korean
J Parasitol. 2004; 42: 19–26.
47. Chen T, et al. Sequence analysis and molecular characterization of Clonorchis sinensis hexokinase, an
unusual trimeric 50-kDa glucose-6-phosphate-sensitive allosteric enzyme. PLoS One. 2014; 9: e107940.
48. Chen T, et al. Advanced enzymology, expression profile and immune response of Clonorchis sinensis
hexokinase show its application potential for prevention and control of clonorchiasis. PLoS Negl Trop
Dis. 2015; 9: e0003641.
49. Keiser J, et al. Effect of artesunate and artemether against Clonorchis sinensis and Opisthorchis viver-
rini in rodent models. Int J Antimicrob Agents. 2006; 28: 370–3.
50. Keiser J, Xiao SH, Smith TA. Utzinger J. Combination chemotherapy against Clonorchis sinensis:
experiments with artemether, artesunate, OZ78, praziquantel, and tribendimidine in a rat model.
Antimicrob Agents Chemother. 2009; 53: 93770–6.
51. Qian MB, et al. Efficacy and safety of tribendimidine against Clonorchis sinensis. Clin Infect Dis. 2013;
56: e76–e82.
52. Choi D, et al. Cholangiocarcinoma and Clonorchis sinensis infection: a case-control study in Korea.
J Hepatol. 2006; 44: 1066–73.
53. Sripa B, et al. Liver fluke induces cholangiocarcinoma. PLoS Med. 2007; 4: e201.
54. Fried B, Reddy A, Mayer D. Helminths in human carcinogenesis. Cancer Lett. 2011; 305: 239–49.
55. International Agency for Research on Cancer. A Review of Human Carcinogens Part B: Biological
Agents, Vol. 100 B. IARC Monographs on the Evaluation of Carcinogenic Risks to Humans. Lyon,
France: IARC; 2009.
56. Grosse Y, et al. A review of human carcinogens-part A: pharmaceuticals. Lancet Oncol. 2009; 10: 13–4.
57. Sirica AE. Cholangiocarcinoma: molecular targeting strategies for chemoprevention and therapy.
Hepatology. 2005; 41: 5–15.
58. Khan SA, Thomas HC, Davidson BR, Taylor-Robison SD. Cholangiocarcinoma. Lancet. 2005; 366:
1303–14.
59. Thamavit W, Bhamarapravati N, Sahaphong S, Vajrasthira S, Angsubhakorn S. Effects of dimethylni-
trosamine on induction of cholangiocarcinoma in Opisthorchis viverrini-infected Syrian golden ham-
sters. Cancer Res. 1978; 38: 4634–9.
60. Iida H. Experimental study of the effects of Clonorchis sinensis infection on induction of cholangiocar-
cinoma in Syrian golden hamsters administered 0.03% N-2-fluorenylacetamide (FAA). Jpn J Parasitol.
1985; 34: 7–16.
61. Lee JH, Rim HJ, Bak UB. Effect of Clonorchis sinensis infection and dimethylnitrosamine administration
on the induction of cholangiocarcinoma in Syrian golden hamsters. Korean J Parasitol. 1993; 31: 21–30.
62. Lee JH, Yang HM, Bak UB, Rim HJ. Promoting role of Clonorchis sinensis infection on induction of
cholangiocarcinoma during two-step carcinogenesis. Korean J Parasitol. 1994; 32: 13–8.
63. Jang JJ, Cho KJ, Myong NH, Chai JY. Enhancement of dimethylnitrosamine-induced glutathione
S-transferase P-positive hepatic foci by Clonorchis sinensis infestation in F344 rats. Cancer Lett. 1990;
52: 133–8.
716 Laboratory Models for Foodborne Infections
CONTENTS
44.1 Introduction....................................................................................................................................717
44.2 Life Cycle.......................................................................................................................................719
44.3 Genome, Transcriptome, and Proteome........................................................................................719
44.4 Host–Pathogen Interaction............................................................................................................ 720
44.4.1 Immunity...........................................................................................................................721
44.4.2 Evasion Mechanisms........................................................................................................ 722
44.5 Clinical Manifestations................................................................................................................. 722
44.5.1 Acute Phase...................................................................................................................... 722
44.5.2 Chronic Phase................................................................................................................... 722
44.6 Diagnosis....................................................................................................................................... 723
44.6.1 Parasitological Techniques............................................................................................... 723
44.6.2 Immunological Techniques.............................................................................................. 723
44.6.3 Molecular Techniques...................................................................................................... 723
44.7 Treatment...................................................................................................................................... 723
44.8 Experimental Models in Fasciolosis............................................................................................. 724
44.8.1 Fasciolosis in Mice........................................................................................................... 724
44.8.1.1 Experimental Infection..................................................................................... 724
44.8.1.2 Physicopathological Changes........................................................................... 724
44.8.1.3 Microarray Analysis......................................................................................... 726
44.8.2 Fasciolosis in Rats............................................................................................................ 726
44.8.2.1 Experimental Infection..................................................................................... 726
44.8.2.2 Physicopathological Changes........................................................................... 726
44.8.3 Fasciolosis in Rabbits........................................................................................................ 727
44.8.3.1 Experimental Infection...................................................................................... 727
44.8.3.2 Physicopathological Changes............................................................................ 727
44.9 Experimental Assessment of Vaccine Candidates...................................................................... 728
44.10 Conclusion and Future Perspective............................................................................................. 730
References............................................................................................................................................... 730
44.1 Introduction
Classified in the phylum Platyhelminthes, subphylum Neodermata, class Trematoda, subclass Digenea,
order Echinostomida, and family Fasciolidae, the genus Fasciola consists of two liver fluke species that
cause fasciolosis in human and animal hosts, i.e., Fasciola hepatica and Fasciola gigantica. Jean DeBrie
was the first person who referred fasciolosis to “putrefaction from liver of sheep” in 1379. However, no
association between fasciolosis and the causal agent, the trematode F. hepatica, was mentioned at that
time. On the contrary, there was a belief that the disease could be attributed to toxic substances from
plants that were consumed by animals.1 The discovery of F. hepatica and its life cycle in the intermediate
717
718 Laboratory Models for Foodborne Infections
host (Galba truncatula) was the result of many observations made by several researchers worldwide. The
first observation was by Sir Anthony Fitzherbert (1523) and the Italian physicist Fanensi Gabucin (1549),
who described the presence of worms similar to pumpkin seeds in the blood vessels of sheep and goats.
At this time, it was thought that the onset of the disease was spontaneous. In 1688, the Italian physicist
Francesco Redi demonstrated that the disease was not a spontaneous phenomenon, but was caused by
oviposition of adult worms. In 1698, Govert Bidloo, a professor of anatomy, documented the presence
of worms in the bile ducts of sheep, deer, and calves and also described for the first time the presence of
worms in the liver of human beings.
During the second half of the 18th century, the intermediate phases in the life cycle of this parasite
were described. These observations were made by Johann Swammerdam (1758) while he was dissect-
ing a snail (Paludina vivipara), and he noted living organisms that evidently did not belong to the snail.
Otto Müller (1798) found microscopic living organisms similar to tadpoles in backwaters, and he called
them cercariae, but the life cycle of these worms remained a mystery, and nobody thought that more
than one host will be required for completing its life cycle. In 1803, Johan Zeder noted egg hatching
from different trematode species and also the release of a ciliated embryo in water. In 1807, Christian
Nitzsch described for the first time the encystment of cercariae. He noted that after a period of time,
cercariae stick on the surface of aquatic plants, they lose the tail, and then they were covered with a
gelatinous substance. Guido Wagener (1857) noted miracidia penetration into snail and subsequent
development of rediae. The first person who suggested that larval stages of liver worm were developed
in the G. truncatula snail was the German helminthologist David Weinlad (1875) when he noted cer-
cariae inside the snail. He also described for the first time the encystment of the cercariae on aquatic
plants and the subsequent digestion by sheep, and he argued that these cercariae were the juvenile
stages of liver worm. However, how the encysted cercariae reached the liver and bile ducts remained
unknown. In 1914, the Russian helminthologist Dimitry Sinitsin demonstrated, using rabbits as animal
models, that juvenile stages of worms, once released in the small intestine, penetrate the intestinal wall
and migrate to the liver by the peritoneal cavity. By then, it was clearly evident that F. hepatica involves
the snail as an intermediate host in its life cycle, resulting in the so-called liver-putrefaction disease,
now known as fasciolosis.1–5
During the past decades, the epidemiology of human fasciolosis has markedly changed, and the
number of reported cases has increased since 1990, and currently, it is considered as a reemerging
disease worldwide. The geographic distribution of the two parasitic species causing fasciolosis is
quite different. F. hepatica is cosmopolitan, while F. gigantica is mainly distributed in Africa and
Asia, although its presence has also been reported in some regions of Europe and Russia.6 Although
the European origin of F. hepatica is well accepted, the incidence of human fasciolosis has been
reported in 51 countries worldwide. It is estimated that more than 1 million people are currently
infected, and there are more than 90 million people living in high-risk areas of acquiring the disease.
The distribution of F. hepatica around the world has been mainly caused by cattle exportation from
Europe to other continents and also because F. hepatica has been able to easily adapt into other
mammalian hosts.
The ability of F. hepatica to successfully adapt into other mammalian hosts has been well
described in the literature. For example, F. hepatica has been well adapted to camelids in Africa
and the South of America, in some marsupials in Australia, and also in black rats, otters, and pigs,7–9
which increases fasciolosis transmission rates. F. hepatica has also been able to adapt to different
intermediate hosts. The main species responsible for F. hepatica transmission is the snail G. trun-
catula, with an important role in the distribution of fasciolosis around the world, as these snails
inhabit ponds or waters that are formed, for example, in the rainy season, and its presence has also
been detected in all continents. The snail responsible for the transmission of F. gigantica (Radix)
lives mainly in standing and deep waters rich in aquatic vegetation, and its presence is limited to
certain geographic areas.
The incidence of human fasciolosis could be underestimated, as not all cases are described in inter-
national reports, and they remain as internal reports, thesis, or publications in local journals, without
international diffusion. Also, fasciolosis is not a mandatory notifiable disease in some countries,10 and in
many instances, they are not diagnosed, as they remain asymptomatic.
Fasciola and Fasciolosis 719
Andean (Bolivia, Perú, Chile, and Ecuador), Caribbean (Cuba), north African (Egypt), western
European (Portugal, France, and Spain), and Caspian Sea (Iran) countries have the highest prevalence
of fasciolosis in the world. In Bolivia, the north plateau is the area most affected with human fasciolo-
sis,11 where the prevalence is higher than 72%, and in some individuals even about 5000 eggs have been
found in per gram of feces.12,13 High prevalence has also been noted in the Puno plateau, Cajamarca, and
Mantaro Valley (Perú).14,15
44.2 Life Cycle
An infected mammalian host releases F. hepatica eggs to the environment through the feces. They are
ovoid, are yellow-brown in color, and measure about 130–145 μm long and 70–90 μm wide. Development
of eggs and hatching are closely related to physical and chemical factors such as relative humidity, tem-
perature, oxygen concentration, and pH. It has been demonstrated that the optimal temperature for egg
development is 23°C–26°C; under these conditions, the eggs are embryonated in 2–3 weeks at pH 7.0. The
precise mechanisms involved in egg hatching still remain unknown. Miracidia (hatched from eggs), which
measure about 130 μm long and have great mobility due to their ciliated tegument, swim quickly to find the
intermediate host (snails). And this happens within 24 h; otherwise, they die. Several factors are involved
in the localization and penetration of miracidia into the snail, the most important being the presence of
stimulatory molecules in the snail mucose, such as glucose, amino acids, and lipids. Miracidia penetrate
into snail, lose cilia, and transform into sporocyst. Sporocysts are transformed into rediae, which in turn
transform into cercariae. It has been estimated that one miracidia can develop up to 600 cercariae, through
parthenogenetic proliferation. The cercariae have a propeller tail for swimming actively, which is subse-
quently lost before its encystment and posterior transformation into metacercariae, which attach to aquatic
plants. A gelatinous substance covers the metacercariae to protect from environmental conditions until
they are ingested by the mammalian host. The infection occurs after digesting metacercariae attached
on the surface of aquatic plants. Approximately 1 h after the ingestion of metacercariae, the excystment
occurs in the small intestine. After that, excysted metacercariae perforate the intestinal wall, migrate to
the peritoneal cavity, perforate the Glisson’s capsule, and penetrate into the hepatic parenchyma, reach-
ing the inside of the liver, causing hemorrhage and fibrosis. In this stage, the parasite attains maximum
growth. Finally, the parasites reach the bile ducts (7 weeks after the infection), and they remain here until
full maturity. Occasionally, juvenile parasites can also be found in rare anatomical locations such as lungs,
pancreas, lymph nodes, and thymus. Eight weeks after the infection, eggs can be found in the bile and later
in the feces, thus completing the entire life cycle. Adult worms of F. hepatica can remain alive in the liver
of the mammalian host even for several years and can also produce approximately 20,000 eggs daily.2–5,16
The main phases of the life cycle of F. hepatica are shown in Figure 44.1.
(A) (B)
(C) (D)
FIGURE 44.1 The biology of F. hepatica. (A) Metacercariae. (B) Adolescariae. (C) Adult. (D) Eggs.
44.4 Host–Pathogen Interaction
After ingestion of the metacercariae, the newly excysted juveniles perforate the intestinal wall and
migrate through the hepatic parenchyma to reach the bile ducts. The induced damage to the mammalian
Fasciola and Fasciolosis 721
host during its migration is caused mainly by factors such as mechanical abrasion by its spicules or the
suction caused by their suckers. The hepatic parenchyma suffers a huge destruction with several internal
injuries and bleeding during parasite migration, which are caused not only by the juvenile flukes but also
by the inflammatory and immunological responses that are generated by the mammalian host. Peritonitis
can also be produced as a result of secondary bacterial infections.
Adult worms are located in the bile ducts, but they can also be found in the cystic duct, gall bladder,
Vater blister, and also in the choledochus. Bile ducts are highly dilated with hyperplasia and obstruction
of bile flow.27,28
44.4.1 Immunity
Although one of the biggest challenges in the study and understanding of the immunology of infections
caused by parasites is the vaccine development, the extreme importance of function and regulation of
different types of T-lymphocytes and cytokines they produce help us to gain a better understanding con-
cerning the immunological reactions between the host and the parasite and figure out the mechanisms
evolved by the parasites not only to successfully invade and establish the cells and tissues from the
mammalian host but also to evade the immune response from the host. Helminths are organisms that
can measure from few centimeters to even meters. This is the reason why they do not invade cells but
tissues, and the induced immune response by the mammalian host is also different from the common
immune response generated by other pathogens. Immunomodulation from the host immune response
becomes the most important factor by which helminths successfully survive for long periods of time in
the host. During helminthic infections, the cells of the innate immune system are activated, resulting in
the generation of different cellular phenotypes that promote the differentiation of Th2 cells with secre-
tion of IL-4, IL-5, and IL-13, which stimulate the production of IgG1, IgE, and IgA. At the same time,
the differentiation of Th1 cells that produce high levels of IFN-γ and are supposed to be highly protective
is suppressed.29–31
The chronic infection occurs when the helminths are well established in the mammalian host. At this
time, the Th2 phenotype still remains through the secretion of IL-4, but the T-regulator (Treg) phenotype
is generated with secretion of IL-10 together with other cell types that secrete TGF-β with the expression
of the cytotoxic T-lymphocyte antigen (CTLA).32 Treg cells also play an important role in the suppression
of Th1 cells and also regulate Th2 response that prevent tissue damage in the mammalian host caused
by migration and feeding of parasite.33 It has also been proposed that in helminthic infections, the mam-
malian host suppresses Th1 responses with destructive potential to the parasite and generates a Th2
response instead, which prevents tissue damage.29,34 However, not all the infections caused by helminths
induce the same pattern in the mammalian host.35 Thus, infection caused by Schistosoma spp. induces a
Th1 response at early stages (acute). Only when the helminth has established in the host and egg libera-
tion by adult worms occurs, Th1 response switches to Th2 as a consequence of immunomodulation by
the antigens from the egg parasites.36 A similar behavior has also been observed in the infection caused
by B. malayi using a murine model. Microfilariae induce a Th1 response in the acute phase, and once the
adult worm has been established in the host, a Th2 phenotype differentiation occurs.37
Once F. hepatica enters into the intestinal wall, a complex network of cellular and molecular interac-
tions between the parasite and different cell types from the host, such as dendritic cells, macrophages,
and mastocytes, occurs. The parasite secretes a great number of molecules, which induce both Th2 and
Treg nonprotective immune response in the host, and favors adaptation and survival of the parasite inside
the host. It has been estimated that one adult worm from F. hepatica can remain alive in the host for
2 years and for even 20 years in sheep.1,32
Using a murine model of fasciolosis, it has been reported that the first attack from the immune system
of the host to the parasite occurs during the next 4 h after the infection when the metacercariae have
excysted in the duodenum. It consists of the generation of immunoglobulins that attempt to perform an
effector mechanism, but the parasite releases cysteine proteases that are able to break these immuno-
globulins, thereby preventing its effector action.38–40 It has also been demonstrated that peritoneal mac-
rophages induce the expression of Th2 phenotype cells together with a decrease in the Th1 phenotype
at 24 h after infection. Simultaneously in this early stage of infection, the juvenile parasites also release
722 Laboratory Models for Foodborne Infections
immunomodulatory molecules, which have a direct effect on the function of the innate immune system
such as dendritic cells and macrophages allocated in the intestinal wall and peritoneal cavity.41,42
Once the infection has been well established, there is a secretion of Th2-type cytokines such as IL-4,
IL-5, and IL-13 by the splenocytes; in later stages of infection (3 weeks), there is also generation of Treg
cytokines IL-10 and TGF-β by macrophages and dendritic cells. It has also been demonstrated that in
the peritoneum, most of the T-CD4+ cells secrete only IL-10 without IL-4 or IFN-γ, thus inducing a
suppression of both Th1 and Th2 responses, which are ineffective against the parasite.43 Chronic fas-
ciolosis in cattle has a similar immune response pattern as that of the murine model of fasciolosis, with
predominance of Th2 and suppression of Th1 phenotype. Serological studies have demonstrated high
levels of IgG1 and little or no IgG2a,44–46 and the susceptibility to F. hepatica has been correlated with
an increase in both the IgG1/IgG2a and the IL-4/IFN-γ ratio.47 Owing to such immunoregulation/immu-
nosuppression, the host infected with F. hepatica becomes more susceptible to acquire other infections,
for example, in those infections where a Th1 response is necessary to prevent acquiring a disease. More
specifically, it has been observed that mice infected with F. hepatica and coinfected with Bordetella per-
tussis could not develop a strong Th1 immune response which in turn becomes protective.48–50 However,
not all coinfections with F. hepatica present the same behavior. For example, Toxoplasma gondii induces
a high Th1 response, with high levels of IFN-γ even in the presence of F. hepatica.51
44.4.2 Evasion Mechanisms
F. hepatica has developed several mechanisms to evade the immune response raised by the host. Anatomic
location of adult worms in the bile ducts can be considered as an evasion mechanism as it represents,
immunologically speaking, an inaccessible site. Furthermore, F. hepatica releases eggs and antigens
together with the bile fluid, which also protect them from the immune response of the host. The tegument
of F. hepatica also plays an important role in immune evasion, which is composed by the glycocalyx,
which changes its chemical composition during the phases of migration and maturation of the parasite.
In the adolescariae stage, the tegumental change occurs every 3 h, thus avoiding the contact and attack
of ligated antibody effector cells such as eosinophils and neutrophils. Juvenile flukes are highly resistant
to the action of the complement of the host, and it is supposed to be due to the presence of sialic acid in
surface glycoproteins, which activates the alternative pathway of complement. F. hepatica has mecha-
nisms to prevent the action of nitric oxide, free oxygen radicals, superoxide dismutase (SOD), glutathi-
one peroxidase, and GST. The parasite also secretes molecules that suppress or immunomodulate the
immune response from the host.27,28
44.5 Clinical Manifestations
44.5.1 Acute Phase
The acute or invasive phase from fasciolosis is caused by the migration of the worms through the perito-
neum and the hepatic parenchyma, causing mechanical damage and allergic and toxic reactions within
the first 2–4 months after infection. The more frequent clinical manifestations include fever, abdominal
pain, flatulence, nausea, diarrhea, constipation, urticaria, and respiratory symptoms such as cough, dys-
pnea, and chest pain. A physical exploration of patients with fasciolosis reveals hepatomegaly, ascites,
and jaundice, while analytical data reveal leukocytosis, eosinophilia, anemia, and high activity from
hepatic enzymes.27,28
44.5.2 Chronic Phase
The chronic phase from fasciolosis (also called obstructive phase) could occur within the next
months or years after the infection. Adult worms migrate and establish within the bile ducts, caus-
ing inflammation and hyperplasia of epithelium and also thickening and dilatation of bile ducts and
gallbladder, leading to cholangitis, cholecystitis, and gallbladder obstruction. Analytical data reveal
Fasciola and Fasciolosis 723
leukocytosis, eosinophilia, middle anemia, high activity from hepatic enzymes, hypoalbuminemia,
and hypergammaglobulinemia.27,28
44.6 Diagnosis
The diagnosis of human fasciolosis is difficult, as there are no specific symptoms and also because there
is no egg-parasite detection in the early phases of infection. Two methods are widely used for the diag-
nosis of F. hepatica: parasitological- and immunological-based. There are also other methods based on
the detection of parasite DNA, such as polymerase chain reaction (PCR), which are not commonly used
because of technical difficulties.
44.6.1 Parasitological Techniques
The detection of F. hepatica eggs in feces is performed through sedimentation techniques, and it rep-
resents the most common methodology used for the diagnosis of fasciolosis.27 The eggs released in the
feces have an ovoid morphology, normally with a green-yellow coloration (caused by the direct contact
with bile), and it is 60 μm in width and 130–150 μm in length. Parasitological techniques have several
advantages, as they are not time consuming and are economical. However, these methods cannot be used
in the acute phase of the disease, as eggs are released at 3–4 months after infection. In areas with a high
prevalence of animal fasciolosis, a false-positive detection could occur, which is caused by the ingestion
of bovine-liver-containing eggs from F. hepatica.52
44.6.2 Immunological Techniques
The antibodies produced against the antigens of F. hepatica have high sensitivity, and can be diagnosed
even in the acute phase of the disease. ELISA assay is used, with antigens from F. hepatica, mainly
excretory-secretory antigens,53,54 cathepsin L1,55,56 proteins from the tegument,57 saposin-like proteins,58
leucine aminopeptidase,59 and other recombinant antigens.60 Although there are several antigens for such
detection, cathepsins remain the main source of antigens for the detection of circulating antibodies.61
Monoclonal antibodies have also been used for the detection of antigens in sera samples and also for
the detection of coproantigens. Some examples of monoclonal antibodies currently used are ES78 and
MM3.62,63 A new lateral flow test was constructed with a recombinant cathepsin L1, using protein A and
MM3 monoclonal antibodies as detector reagents.64
44.6.3 Molecular Techniques
Although molecular techniques for the detection of parasitic DNA, such as PCR, have high specificity
and sensibility, because of technical difficulties in areas with high incidence, PCR is not commonly
used for the diagnosis of F. hepatica infection. Also, this is the only technique that identifies the caus-
ative organism both in the intermediate host and in the mammalian one. The molecular techniques
more widely used for the detection of fasciolosis are (1) conventional PCR,65,66 (2) real-time PCR,65,66
(3) LAMP (loop-mediated isothermal amplification),67 and (4) multiplex PCR.68,69
44.7 Treatment
To date, the number of chemotherapeutic agents for the successful treatment of fasciolosis is very scarce,
as most of them are effective only against adult worms and ineffective or partially effective against
immature worms. Triclabendazole (TBZ) is currently the drug of choice for the treatment of fasciolo-
sis, as it is highly active against both mature and immature worms. Furthermore, it is a safe compound
when administered and is well tolerated.70 The recommended dosage is 10 mg/kg/day for 2 consecutive
days, obtaining a curative rate between 92.2% and 93.9%. Adverse effects caused by TBZ are normally
724 Laboratory Models for Foodborne Infections
minimal, the more frequent being abdominal pain and sweating. In some cases, it can also induce
nausea, vomiting, chills, cough, fever, and itching. In animals, the dose regimen may vary depending on
the infected species; thus, bovines receive a single dose of 12 mg/kg, whereas sheep and goats receive a
single dose of 10 mg/kg. For animals that are used for human consumption, such treatment should not be
applied during the 28 days prior to sacrifice. On the other hand, milk from treated animals should not be
consumed for the next 4 days after treatment.71
Structurally, TBZ belongs to the benzimidazole’s family, and its mechanism of action is associated
with the β-tubulin function, attaching to and hampering biological process associated with microtu-
bules.72 The resistance to TBZ was reported for the first time in Australia in the mid-1990s.73 Since then,
resistance to TBZ has also been reported in European countries such as Ireland, Scotland, Wales, Spain,
and Netherlands.74–76 The alternative treatment for the treatment of fasciolosis caused by TBZ-resistant
strains is based on the administration of artemisinin-derivative compounds.
44.8.1 Fasciolosis in Mice
Mice have been used for a long time to study the immunology of the infection and are also used as an
experimental model to assess vaccine efficacy. Furthermore, the use of natural infection models to study
fasciolosis has been hampered because of technical issues such as cost, space, manipulation, and the
availability of specific reagents for immunological studies. The University of Salamanca’s Research
Center for Tropical Diseases (CIETUS) has been working for the last decade on the development of an
appropriate laboratory model to study the infection caused by F. hepatica, including both BALB/c and
CD1 mice. Thus, a hyperinfection model using lethal doses of F. hepatica metacercariae to study both
the immunopathology induced by the infection and the immunoprotective efficacy of several vaccine
candidates was described.77 The current approaches to study fasciolosis in mice are discussed in the fol-
lowing paragraphs.
44.8.1.1 Experimental Infection
Mice are orally infected with five to seven F. hepatica metacercariae, which are suspended in water
or phosphate buffered solution (PBS). This suspension is then administered to each mouse using an
intragastric gavage technique.78,79 Nonlethal dosage in mice is frequently used to study the dynamics
of the infection as mice remain alive during the whole study.80 Other authors have used a large number
of metacercariae to infect mice, including 10 and 15 metacercariae. In both cases, a human endpoint is
established at 3 weeks after experimental infection.49
44.8.1.2 Physicopathological Changes
It is well known that helminth-associated infections induce a strong Th2 immune response in the mam-
malian host.35 Thus, typically the success of the infection is investigated by measuring the level of anti-
bodies raised against the excretory-secretory antigens. It is worth noting that the hyperinfection in mice
with five to seven metacercariae induces death of animals between days 28 and 34.77,79
Fasciola and Fasciolosis 725
44.8.1.2.2 Hepatic Transaminases
Alterations in hepatic transaminase levels are often used to assess the course of infection, and such mea-
sures can be done either in the serum or in the liver of infected mice. Typically, aspartate transaminase
(AST), alanine transaminase (ALT), and γ-glutamyl transpeptidase are measured.
44.8.1.2.4 Cytokines
To assess cellular immune responses induced by F. hepatica, the most representative Th1- and Th2-
Treg and Th17-associated cytokines are measured. For this purpose, infected mice are humanely
euthanized and necropsied for spleen recovery. Later, splenocytes are recovered, in vitro cultured,
and stimulated to quantify the levels of the aforementioned cytokines. The results clearly demon-
strate that F. hepatica induces a Th2-polarized immune response with high levels of IL-4 and IL-5
and little or no IFN-γ.49
44.8.1.2.5 Hepatic Damage
Measuring the hepatic damage induced after infection also assesses the success of the infection by
F. hepatica. At the time of necropsy, the liver is isolated and macroscopically evaluated considering the
size, consistency, color, and dilatation of the bile ducts and repletion of the vessels, and scored as intense
(+++), mild (++), moderate, (+) or without lesions (−).77,79 There is no consensus in the quantitation of
hepatic damage, and most of the times authors use their own scale and parameters. It is worth noting
that not all of the mouse strains are equally susceptible/resistant to the infection with F. hepatica.49 An
example of liver lesions in healthy and infected mice is shown in Figure 44.2.
(A) (B)
FIGURE 44.2 The hepatic damage induced by the experimental infection of F. hepatica in mouse. (A) Liver of one
uninfected healthy mouse. (B) Liver of one mouse infected and necropsied at 21 days after infection. In this stage, bile
duct enlargement is observed together with scars on the hepatic surface and irregular yellowish white area on the liver and
parenchyma.
726 Laboratory Models for Foodborne Infections
44.8.1.2.6 Worm Burden
Although determining worm burden is very useful to evaluate the success of infection with F. hepatica,
the results of this approach are slightly inappropriate when using mouse as an experimental model, as
they could not harbor too many adult worms in the liver, thereby leading to inconclusive statistical infer-
ence. Furthermore, when using lethal doses, mice die before worms reach the liver and bile ducts.
44.8.1.3 Microarray Analysis
Recently, we have investigated the gene expression profile during F. hepatica infection at 7 and 21 days
after experimental infection using a microarray-based methodology. Differential expression of genes occurs
as the infection progresses. For this purpose, we have isolated the RNA from the liver of infected mice at
7 and 21 days postinfection and investigated the gene expression profile compared to untreated mice. Using
functional profiling, we identified several upregulated key genes involved in the induction of hepatic injury.78
44.8.2 Fasciolosis in Rats
Rats have also been widely used to study not only the immunology of the infection but also the protective
efficacy of several vaccine candidates. Rats are more resistant to the infection, and typically they do not die,
thus allowing a more detailed study during advanced stages of the infection. Moreover, previous reports have
demonstrated the important role of black rats, Rattus rattus, in the epidemiology of fasciolosis, mainly in
the Mediterranean island of Corsica. It has been demonstrated that these rats become naturally infected with
the trematode F. hepatica. Although human infections of fasciolosis have also been reported in Corsica, the
prevalence of the disease remains low.82 Thus, the investigation of the immune response and the assessment
of vaccines in rats have attracted interest, and several studies have been developed since then.
44.8.2.1 Experimental Infection
Rats are orally infected with the metacercariae of F. hepatica using the intragastric gavage technique.
However, there is no consensus regarding the number of metacercariae that are needed for a successful
infection, and this decision depends almost exclusively on each researcher’s criteria and experience.83–85
44.8.2.2 Physicopathological Changes
44.8.2.2.1 Antibody Response
Using rats as laboratory experimental model allows a more compressive study of the dynamics of anti-
body production during infection with F. hepatica for a long period of time. Typically, IgG, IgG1, IgG2a,
IgG2b, and IgG2c are measured during infection in rats by the ELISA technique using sera or plasma
samples. IgG antibodies are detected from 1 week after experimental infection, and an increase occurs
during the following 5 weeks. From 7 to 10 weeks after infection, IgG remains unchanged.86 IgG1
and IgG2a are also detected from the first week after the infection, and this increases during the next
10 weeks, with similar patterns. High levels of IgM are induced by 2 weeks after infection, which slowly
decreases as the infection occurs. However, high levels still remain by 10 weeks after infection. IgE has
a biphasic behavior, with peaks being detected at 5 and 9 weeks after infection.87
44.8.2.2.2 Cytokines
Most of the experiments aimed at the study of the production of cytokines after the experimental infec-
tion with F. hepatica are performed in the spleen cells at different times of infection. An increase in the
percentage of T-CD4 cells after the experimental infection is observed while the level of T-CD8 cells
remains unchanged. It has also been demonstrated that the amount of cells producing IFN-γ remains
unchanged during the first week after the infection, but they suffer a decrease at 2 weeks after the infec-
tion,84 thus demonstrating a downregulation of the Th1 phenotype as the infection moves along. This
behavior has also been reported in mice infections.49 An increase in the level of both IL-4 and IL-10 is
observed at 1 and 2 weeks after the infection.83,84
Fasciola and Fasciolosis 727
44.8.2.2.4 Worm Burden
Using rats as an experimental model in the study of fasciolosis allows a more accurate investigation of
worm burden than using mice. From third week after infection until the seventh week, flukes are har-
bored in the hepatic parenchyma. Later, they continue migrating and reach the bile ducts at week 11,
where they remain for a long period of time.89
44.8.2.2.5 Hepatic Damage
As in mice, the hepatic damage induced by experimental infection is assessed considering the size,
consistency, color, dilatation of the bile ducts, and repletion of the vessels, and scored as intense (+++),
mild (++), moderate (+), or without lesions (−).77 However, the pathology in the liver has previously been
described from the first week up to 12 weeks after infection.90
44.8.3 Fasciolosis in Rabbits
The use of rabbits as an experimental model to study the immunology of the infection caused by
F. hepatica has been very limited mainly because immunological assays for this species remain unavail-
able. However, rabbits have been widely used for the production of monoclonal antibodies (as large
quantities of sera could be obtained) and also for the evaluation of the protective efficacy of vaccine
candidates. Currently, there is only one report concerning the cytokine profile in rabbits immunized with
a saposin-like (SAP2) recombinant protein by means of quantification of mRNA by real-time PCR.91
As in rats, rabbits could harbor a large number of adult worms in the liver, making them suitable to
statistically assess worm burden.
44.8.3.1 Experimental Infection
To perform the experimental infection, rabbits are orally infected with the metacercariae of F. hepatica
suspended in either distilled water or PBS. As in the rat model, the number of metacercariae required for
the infection is variable and depends on the researcher’s criteria and experience. Because a large number
of metacercariae are commonly used to infect rabbits, the administration protocol could also be differ-
ent. In some cases, the metacercariae of F. hepatica are contained within a gelatin capsule, thus avoiding
the count of the metacercariae. The number of metacercariae required to infect rabbits is usually in the
range of 25 to 50.91–95
44.8.3.2 Physicopathological Changes
44.8.3.2.1 Antibody Response
After experimental infection, rabbits are weekly bled to investigate the dynamics of antibodies raised
against the parasite. ELISA technique is used to measure antibodies. From the third week after experi-
mental infection, IgG is detectable, reaching a maximum peak at sixth week after infection. From week
6 onward, antibody levels remain unchanged until 10th week.91
44.8.3.2.2 Worm Burden
Rabbits are humanely slaughtered between 12 and 20 weeks after experimental infection, and the flukes
are isolated from bile ducts. Both immature and mature flukes are isolated from the liver. For this pur-
pose, livers are cut, soaked in water or PBS at 37°C, squeezed, and passed through 300-μm mesh sieve,
according to previously described methodologies.91,92
728 Laboratory Models for Foodborne Infections
44.8.3.2.3 Hepatic Damage
At necropsy, independent, experienced pathologists assess hepatic damage. They macroscopically
examine the livers and assign a score according to the intensity of the damage. There is no consensus
regarding the scale used in the quantitation of hepatic damage, but researchers have their own scales,
which are applied independently. Typically, hepatic damage is classified as mild, moderate, and intense.96
729
730 Laboratory Models for Foodborne Infections
REFERENCES
1. Andrews, S.J. The life cycle of Fasciola hepatica. in Fasciolosis. Dalton, J.P., ed. 1–29 (CAB
International, Wallingford, Oxon, 1999).
2. Galaktionov, K.V. & Dobrovolskij, A.A. The Biology and Evolution of Trematodes. An Essay on the
Biology, Morphology, Life Cycles, Transmissions, and Evolution of Digenetic Trematodes. 215–346
(Kluwer Academic Publishers, Dordrecht, the Netherlands, 2003).
3. Gunn, A. & Pitt, S.J. Helminth parasites. in Parasitology: An Integrated Approach (Wiley-Blackwell,
Chichester, 2012).
4. Kendall, S.B. Relationships between the species of Fasciola and their molluscan hosts. Adv Parasitol 8,
251–8 (1970).
5. Olsen, O.W. Animal Parasites: Their Life Cycles and Ecology, 199–267 (General Publishing Company,
Toronto, Canada, 1974).
6. Mas-Coma, S. & Bargues, M.D. Human liver flukes: a review. Res Rev Parasitol 57, 145–218 (1997).
7. Menard, A. et al. Myocastor coypus as a reservoir host of Fasciola hepatica in France. Vet Res 32,
499–508 (2001).
8. Valero, M.A., Marcos, M.D., Fons, R. & Mas-Coma, S. Fasciola hepatica development in the experi-
mentally infected black rat Rattus rattus. Parasitol Res 84, 188–94 (1998).
9. Valero, M.A., Panova, M., Comes, A.M., Fons, R. & Mas-Coma, S. Patterns in size and shedding of Fasciola
hepatica eggs by naturally and experimentally infected murid rodents. J Parasitol 88, 308–13 (2002).
10. Mas-Coma, S. Epidemiology of fascioliasis in human endemic areas. J Helminthol 79, 207–16 (2005).
11. Mas-Coma, S. et al. Human fascioliasis in Bolivia: a general analysis and critical review of existing
data. Res Rev Parasitol 55, 73–79 (1995).
12. Esteban, J.G. et al. A population-based coprological study of human fascioliasis in a hyperendemic area
of the Bolivian Altiplano. Trop Med Int Health 2, 695–9 (1997).
13. Hillyer, G.V. et al. Use of the Falcon assay screening test—enzyme-linked immunosorbent assay
(FAST-ELISA) and the enzyme-linked immunoelectrotransfer blot (EITB) to determine the prevalence
of human fascioliasis in the Bolivian Altiplano. Am J Trop Med Hyg 46, 603–9 (1992).
Fasciola and Fasciolosis 731
14. Esteban, J.G. et al. High fascioliasis infection in children linked to a man-made irrigation zone in Peru.
Trop Med Int Health 7, 339–48 (2002).
15. Marcos-Raymundo, L.A. et al. Hiperendemicidad de fasciolosis humana en el valle del mantaro, perú:
Factores de riesgo de la infección por Fasciola hepatica. Rev Gastroenterol Perú 24, 158–64 (2004).
16. Lopez-Aban, J. et al. Adaptive immune stimulation is required to obtain high protection with fatty acid
binding protein vaccine candidate against Fasciola hepatica in Balb/C mice. J Parasitol 98, 527–35 (2012).
17. Berriman, M. et al. The genome of the blood fluke Schistosoma mansoni. Nature 460, 352–8 (2009).
18. Tsai, I.J. et al. The genomes of four tapeworm species reveal adaptations to parasitism. Nature 496,
57–63 (2013).
19. Zheng, H. et al. The genome of the hydatid tapeworm Echinococcus granulosus. Nat Genet 45, 1168–75
(2013).
20. Cwiklinski, K. et al. The Fasciola hepatica genome: gene duplication and polymorphism reveals
adaptation to the host environment and the capacity for rapid evolution. Genome Biol 16, 71 (2015).
21. Young, N.D., Hall, R.S., Jex, A.R., Cantacessi, C. & Gasser, R.B. Elucidating the transcriptome of
Fasciola hepatica—a key to fundamental and biotechnological discoveries for a neglected parasite.
Biotechnol Adv 28, 222–31 (2010).
22. Margulies, M. et al. Genome sequencing in microfabricated high-density picolitre reactors. Nature 437,
376–80 (2005).
23. Wilson, R.A. et al. Exploring the Fasciola hepatica tegument proteome. Int J Parasitol 41, 1347–59 (2011).
24. Hernandez-Gonzalez, A., Valero, M.L., del Pino, M.S., Oleaga, A. & Siles-Lucas, M. Proteomic analysis
of in vitro newly excysted juveniles from Fasciola hepatica. Mol Biochem Parasitol 172, 121–8 (2010).
25. Morphew, R.M., Wright, H.A., LaCourse, E.J., Woods, D.J. & Brophy, P.M. Comparative proteomics of
excretory-secretory proteins released by the liver fluke Fasciola hepatica in sheep host bile and during
in vitro culture ex host. Mol Cell Proteomics 6, 963–72 (2007).
26. Boukli, N.M., Delgado, B., Ricaurte, M. & Espino, A.M. Fasciola hepatica and Schistosoma mansoni:
identification of common proteins by comparative proteomic analysis. J Parasitol 97, 852–61 (2011).
27. López-Abán, J., Pardo, L.J., Pérez-Arellano, J.L. & Muro, A. Infecciones difícilmente transmisibles en
el inmigrante V: Otras trematodosis. in Manual de Enfermedades Importadas. Muro Álvarez, A. &
Pérez Arellano, J.L., eds (Elsevier, Barcelona, España, 2012).
28. Muro, A., Pérez del Villar, L., Velasco, V. & Pérez-Arellano, J.L. Infecciones por trematodos. Medicine—
Programa de Formación Médica Continuada Acreditado 10, 3717–3728 (2010).
29. Allen, J.E. & Maizels, R.M. Diversity and dialogue in immunity to helminths. Nat Rev Immunol 11,
375–88 (2011).
30. McSorley, H.J., Hewitson, J.P. & Maizels, R.M. Immunomodulation by helminth parasites: defining
mechanisms and mediators. Int J Parasitol 43, 301–10 (2013).
31. van Riet, E., Hartgers, F.C. & Yazdanbakhsh, M. Chronic helminth infections induce immunomodula-
tion: consequences and mechanisms. Immunobiology 212, 475–90 (2007).
32. Dalton, J.P., Robinson, M.W., Mulcahy, G., O’Neill, S.M. & Donnelly, S. Immunomodulatory molecules
of Fasciola hepatica: candidates for both vaccine and immunotherapeutic development. Vet Parasitol
195, 272–85 (2013).
33. Hill, J.A., Benoist, C. & Mathis, D. Treg cells: guardians for life. Nat Immunol 8, 124–5 (2007).
34. Allen, J.E. & Wynn, T.A. Evolution of Th2 immunity: a rapid repair response to tissue destructive
pathogens. PLoS Pathog 7, e1002003 (2011).
35. Maizels, R.M. & Yazdanbakhsh, M. Immune regulation by helminth parasites: cellular and molecular
mechanisms. Nat Rev Immunol 3, 733–44 (2003).
36. Pearce, E.J. & MacDonald, A.S. The immunobiology of schistosomiasis. Nat Rev Immunol 2, 499–511
(2002).
37. Lawrence, R.A., Allen, J.E., Osborne, J. & Maizels, R.M. Adult and microfilarial stages of the filar-
ial parasite Brugia malayi stimulate contrasting cytokine and Ig isotype responses in BALB/c mice.
J Immunol 153, 1216–24 (1994).
38. Berasain, P., Carmona, C., Frangione, B., Dalton, J.P. & Goni, F. Fasciola hepatica: parasite-secreted
proteinases degrade all human IgG subclasses: determination of the specific cleavage sites and identifi-
cation of the immunoglobulin fragments produced. Exp Parasitol 94, 99–110 (2000).
39. Berasain, P. et al. Proteinases secreted by Fasciola hepatica degrade extracellular matrix and basement
membrane components. J Parasitol 83, 1–5 (1997).
732 Laboratory Models for Foodborne Infections
40. Carmona, C., Dowd, A.J., Smith, A.M. & Dalton, J.P. Cathepsin L proteinase secreted by Fasciola
hepatica in vitro prevents antibody-mediated eosinophil attachment to newly excysted juveniles. Mol
Biochem Parasitol 62, 9–17 (1993).
41. Donnelly, S., O’Neill, S.M., Sekiya, M., Mulcahy, G. & Dalton, J.P. Thioredoxin peroxidase secreted by
Fasciola hepatica induces the alternative activation of macrophages. Infect Immun 73, 166–73 (2005).
42. Donnelly, S. et al. Helminth 2-Cys peroxiredoxin drives Th2 responses through a mechanism involving
alternatively activated macrophages. FASEB J 22, 4022–32 (2008).
43. Walsh, K.P., Brady, M.T., Finlay, C.M., Boon, L. & Mills, K.H. Infection with a helminth parasite
attenuates autoimmunity through TGF-β-mediated suppression of Th17 and Th1 responses. J Immunol
183, 1577–86 (2009).
44. Hoyle, D.V. & Taylor, D.W. The immune response of regional lymph nodes during the early stages of
Fasciola hepatica infection in cattle. Parasite Immunol 25, 221–9 (2003).
45. Mulcahy, G. et al. Immune responses of cattle to experimental anti-Fasciola hepatica vaccines. Res Vet
Sci 67, 27–33 (1999).
46. Mulcahy, G. et al. Correlation of specific antibody titre and avidity with protection in cattle immunized
against Fasciola hepatica. Vaccine 16, 932–9 (1998).
47. Pleasance, J., Wiedosari, E., Raadsma, H.W., Meeusen, E. & Piedrafita, D. Resistance to liver fluke
infection in the natural sheep host is correlated with a type-1 cytokine response. Parasite Immunol 33,
495–505 (2011).
48. Brady, M.T., O’Neill, S.M., Dalton, J.P. & Mills, K.H. Fasciola hepatica suppresses a protective Th1
response against Bordetella pertussis. Infect Immun 67, 5372–8 (1999).
49. O’Neill, S.M. et al. Fasciola hepatica infection downregulates Th1 responses in mice. Parasite Immunol
22, 147–55 (2000).
50. Vukman, K.V. et al. The effects of Fasciola hepatica tegumental antigens on mast cell function. Int
J Parasitol 43, 531–9 (2013).
51. Miller, C.M. et al. Immunological interactions between 2 common pathogens, Th1-inducing protozoan
Toxoplasma gondii and the Th2-inducing helminth Fasciola hepatica. PLoS One 4, e5692 (2009).
52. Taira, N., Yoshifuji, H. & Boray, J.C. Zoonotic potential of infection with Fasciola spp. by consumption
of freshly prepared raw liver containing immature flukes. Int J Parasitol 27, 775–9 (1997).
53. El-Aziz, M.M., Ghazy, A.A. & Effat, M.M. Immunodiagnosis of bovine fasciolosis using Fasciola
hepatica excretory-secretory antigens ELISA. J Egypt Soc Parasitol 31, 327–34 (2001).
54. Espino, A.M., Dumenigo, B.E., Fernandez, R. & Finlay, C.M. Immunodiagnosis of human fascioliasis
by enzyme-linked immunosorbent assay using excretory-secretory products. Am J Trop Med Hyg 37,
605–8 (1987).
55. O’Neill, S.M., Parkinson, M., Strauss, W., Angles, R. & Dalton, J.P. Immunodiagnosis of Fasciola
hepatica infection (fascioliasis) in a human population in the Bolivian Altiplano using purified cathep-
sin L cysteine proteinase. Am J Trop Med Hyg 58, 417–23 (1998).
56. Rokni, M.B., Massoud, J., O’Neill, S.M., Parkinson, M. & Dalton, J.P. Diagnosis of human fasciolosis
in the Gilan province of Northern Iran: application of cathepsin L-ELISA. Diagn Microbiol Infect Dis
44, 175–9 (2002).
57. Morales, A. & Espino, A.M. Evaluation and characterization of Fasciola hepatica tegument protein
extract for serodiagnosis of human fascioliasis. Clin Vaccine Immunol 19, 1870–8 (2012).
58. Figueroa-Santiago, O., Delgado, B. & Espino, A.M. Fasciola hepatica saposin-like protein-2-based
ELISA for the serodiagnosis of chronic human fascioliasis. Diagn Microbiol Infect Dis 70, 355–61 (2011).
59. Marcilla, A. et al. Leucine aminopeptidase is an immunodominant antigen of Fasciola hepatica excre-
tory and secretory products in human infections. Clin Vaccine Immunol 15, 95–100 (2008).
60. Caban-Hernandez, K., Gaudier, J.F., Ruiz-Jimenez, C. & Espino, A.M. Development of two antibody
detection enzyme-linked immunosorbent assays for serodiagnosis of human chronic fascioliasis. J Clin
Microbiol 52, 766–72 (2014).
61. Cordova, M. et al. Fasciola hepatica cysteine proteinases: immunodominant antigens in human fascio-
liasis. Am J Trop Med Hyg 57, 660–6 (1997).
62. Espino, A.M. & Finlay, C.M. Sandwich enzyme-linked immunosorbent assay for detection of excretory
secretory antigens in humans with fascioliasis. J Clin Microbiol 32, 190–3 (1994).
63. Ubeira, F.M. et al. MM3-ELISA detection of Fasciola hepatica coproantigens in preserved human stool
samples. Am J Trop Med Hyg 81, 156–62 (2009).
Fasciola and Fasciolosis 733
64. Martinez-Sernandez, V. et al. Development and evaluation of a new lateral flow immunoassay for sero-
diagnosis of human fasciolosis. PLoS Negl Trop Dis 5, e1376 (2011).
65. Ai, L. et al. Specific PCR-based assays for the identification of Fasciola species: their development,
evaluation and potential usefulness in prevalence surveys. Ann Trop Med Parasitol 104, 65–72 (2010).
66. Rokni, M.B. et al. Identification and differentiation of Fasciola hepatica and Fasciola gigantica using
a simple PCR-restriction enzyme method. Exp Parasitol 124, 209–13 (2010).
67. Ai, L. et al. Rapid identification and differentiation of Fasciola hepatica and Fasciola gigantica by a
loop-mediated isothermal amplification (LAMP) assay. Vet Parasitol 174, 228–33 (2010).
68. Magalhaes, K.G. et al. Isolation and detection of Fasciola hepatica DNA in Lymnaea viatrix from
formalin-fixed and paraffin-embedded tissues through multiplex-PCR. Vet Parasitol 152, 333–8 (2008).
69. Magalhaes, K.G., Passos, L.K. & Carvalho Odos, S. Detection of Lymnaea columella infection by
Fasciola hepatica through multiplex-PCR. Mem Inst Oswaldo Cruz 99, 421–4 (2004).
70. Boray, J.C., Jackson, R. & Strong, M.B. Chemoprophylaxis of fascioliasis with triclabendazole. N Z Vet
J 33, 182–5 (1985).
71. Ibarra-Velarde, F., Vera-Montenegro, Y., Najera-Fuentes, R. & Sanchez-Albarran, A. Efficacy of com-
bined chemotherapy against gastrointestinal nematodes and Fasciola hepatica in cattle. Vet Parasitol
99, 199–204 (2001).
72. Brennan, G.P. et al. Understanding triclabendazole resistance. Exp Mol Pathol 82, 104–9 (2007).
73. Overend, D.J. & Bowen, F.L. Resistance of Fasciola hepatica to triclabendazole. Aust Vet J 72, 275–6 (1995).
74. Brockwell, Y.M. et al. Confirmation of Fasciola hepatica resistant to triclabendazole in naturally
infected Australian beef and dairy cattle. Int J Parasitol Drugs Drug Resist 4, 48–54 (2014).
75. Fairweather, I. Triclabendazole: new skills to unravel an old(ish) enigma. J Helminthol 79, 227–34 (2005).
76. Martinez-Valladares, M., Cordero-Perez, C. & Rojo-Vazquez, F.A. Efficacy of an anthelmintic combi-
nation in sheep infected with Fasciola hepatica resistant to albendazole and clorsulon. Exp Parasitol
136, 59–62 (2014).
77. López-Abán, J., Nogal-Ruíz, J.J., Muñoz-Pera, B.C., Martínez-Fernández, A.R. & Muro, A. Evaluation
of a fasciolosis hyper-infection model in mice for vaccination trials. Rev Ibérica Parasitol 65, 71–78
(2005).
78. Rojas-Caraballo, J. et al. Gene expression profile in the liver of BALB/c mice infected with Fasciola
hepatica. PLoS One 10, e0134910 (2015).
79. Rojas-Caraballo, J. et al. In vitro and in vivo studies for assessing the immune response and protection-
inducing ability conferred by Fasciola hepatica-derived synthetic peptides containing B- and T-cell
epitopes. PLoS One 9, e105323 (2014).
80. Bundesen, P.G. & Janssens, P.A. Biochemical tracing of parasitic infections. I. Fasciola hepatica L. in
mice—a qualitative study. Int J Parasitol 1, 7–14 (1971).
81. Milbourne, E.A. & Howell, M.J. Eosinophil responses to Fasciola hepatica in rodents. Int J Parasitol
20, 705–8 (1990).
82. Mas-Coma, S. et al. Small mammals as natural definitive hosts of the liver fluke, Fasciola hepatica
Linnaeus, 1758 (Trematoda: Fasciolidae): a review and two new records of epidemiologic interest on the
island of Corsica. Riv Parassitol 5, 73–78 (1987).
83. Cervi, L., Cejas, H. & Masih, D.T. Cytokines involved in the immunosuppressor period in experimental
fasciolosis in rats. Int J Parasitol 31, 1467–73 (2001).
84. Tliba, O. et al. Early hepatic immune response in rats infected with Fasciola hepatica. Vet Res 33,
261–70 (2002).
85. Tliba, O., Sibille, P., Boulard, C. & Chauvin, A. Local hepatic immune response in rats during primary
infection with Fasciola hepatica. Parasite 7, 9–18 (2000).
86. Poitou, I., Baeza, E. & Boulard, C. Humoral and cellular immune responses in rats during a primary
infestation with Fasciola hepatica. Vet Parasitol 45, 59–71 (1992).
87. Poitou, I., Baeza, E. & Boulard, C. Kinetic responses of parasite-specific antibody isotypes, blood
leucocyte pattern and lymphocyte subsets in rats during primary infestation with Fasciola hepatica. Vet
Parasitol 49, 179–90 (1993).
88. Kolodziejczyk, L., Siemieniuk, E. & Skrzydlewska, E. Fasciola hepatica: effects on the antioxidative
properties and lipid peroxidation of rat serum. Exp Parasitol 113, 43–8 (2006).
89. Keegan, P.S. & Trudgett, A. Fasciola hepatica in the rat: immune responses associated with the devel-
opment of resistance to infection. Parasite Immunol 14, 657–69 (1992).
734 Laboratory Models for Foodborne Infections
90. Thorpe, E. The pathology of experimental fascioliasis in the albino rat. J Comp Path 75, 39–44 (1965).
91. Espino, A.M. & Rivera, F. Quantitation of cytokine mRNA by real-time RT-PCR during a vaccination
trial in a rabbit model of fascioliasis. Vet Parasitol 169, 82–92 (2010).
92. Acosta, D. et al. Fasciola hepatica leucine aminopeptidase, a promising candidate for vaccination
against ruminant fasciolosis. Mol Biochem Parasitol 158, 52–64 (2008).
93. Espino, A.M. & Hillyer, G.V. A novel Fasciola hepatica saposinlike recombinant protein with immuno-
prophylactic potential. J Parasitol 90, 876–9 (2004).
94. Maggioli, G. et al. A recombinant thioredoxin-glutathione reductase from Fasciola hepatica induces a
protective response in rabbits. Exp Parasitol 129, 323–30 (2011).
95. Muro, A., Ramajo, V., Lopez, J., Simon, F. & Hillyer, G.V. Fasciola hepatica: vaccination of rabbits with
native and recombinant antigens related to fatty acid binding proteins. Vet Parasitol 69, 219–29 (1997).
96. Casanueva, R. et al. Immunoprophylaxis against Fasciola hepatica in rabbits using a recombinant Fh15
fatty acid-binding protein. J Parasitol 87, 697–700 (2001).
97. Hillyer, G.V. Fasciola antigens as vaccines against fascioliasis and schistosomiasis. J Helminthol 79,
241–7 (2005).
98. Martinez-Fernandez, A.R. et al. Vaccination of mice and sheep with Fh12 FABP from Fasciola hepatica
using the new adjuvant/immunomodulator system ADAD. Vet Parasitol 126, 287–98 (2004).
99. Lopez-Aban, J. et al. Progress in the development of Fasciola hepatica vaccine using recombinant fatty
acid binding protein with the adjuvant adaptation system ADAD. Vet Parasitol 145, 287–96 (2007).
100. Muro, A. et al. Identification of Fasciola hepatica recombinant 15-kDa fatty acid-binding protein T-cell
epitopes that protect against experimental fascioliasis in rabbits and mice. J Parasitol 93, 817–23 (2007).
101. Lopez-Aban, J. et al. The addition of a new immunomodulator with the adjuvant adaptation ADAD sys-
tem using fatty acid binding proteins increases the protection against Fasciola hepatica. Vet Parasitol
153, 176–81 (2008).
45
Haplorchis
Dongyou Liu
CONTENTS
45.1 Introduction................................................................................................................................... 735
45.1.1 Classification and Morphology......................................................................................... 736
45.1.1.1 Classification..................................................................................................... 736
45.1.1.2 Morphology....................................................................................................... 736
45.1.2 Life Cycle and Epidemiology........................................................................................... 737
45.1.3 Clinical Features.............................................................................................................. 737
45.1.4 Diagnosis.......................................................................................................................... 738
45.1.5 Treatment and Prevention................................................................................................. 738
45.2 Laboratory Models........................................................................................................................ 738
45.2.1 Animal Models................................................................................................................. 738
45.2.1.1 Rodents............................................................................................................. 738
45.2.1.2 Dogs.................................................................................................................. 739
45.2.1.3 Foxes................................................................................................................. 739
45.2.1.4 Chicks............................................................................................................... 739
45.2.2 In Vitro Models................................................................................................................. 739
45.3 Conclusion..................................................................................................................................... 739
References............................................................................................................................................... 739
45.1 Introduction
The genus Haplorchis contains several small intestinal flukes that require mollusks (first intermediate
host), fish (second intermediate host), humans, other mammals, or birds (definitive hosts) for complet-
ing life cycles. In comparison to the liver flukes Opisthorchis viverrini and Clonorchis sinensis, which
are well known and extensively investigated (as shown by 1320 and 1166 entries in recent PubMed
searches using the terms Opisthorchis and Clonorchis, respectively), Haplorchis species are relatively
understudied (as shown by 149 entries in PubMed search using the term Haplorchis). Nevertheless,
this does not make Haplorchis a less significant pathogen. In fact, intestinal flukes Haplorchis tai-
chui and Metagonimus yokogawai (see Chapter 46) of the family Heterophyidae are listed by WHO
as foodborne trematodes of medical importance and public health concern in Asia, along with liver
flukes O. viverrini (see Chapter 47) and C. sinensis (see Chapter 43) of the family Opisthorchiidae.
Given the medical prominence of H. taichui, this chapter will focus on the genus Haplorchis, despite
the presence of several other human-infecting genera (e.g., Heterophyes, Centrocestus, Pygidiopsis,
Stellantchasmus, and Procerovum) in the family Heterophyidae.
735
736 Laboratory Models for Foodborne Infections
45.1.1.2 Morphology
Similar to other members of the family Heterophyidae (often referred to as heterophyids), Haplorchis
species are small flukes (of 0.3–1 mm in length and 0.14–0.2 mm in width), with tegument covered by
spines. In addition, they possess a small oral sucker (which is armed with spines) and a ventrogenital
sucker complex (also with spines). Other organs present in Haplorchis species include pharynx, simple
intestinal system, ceca, one testis (compared to two testes seen in some other heterophyids), and vitellaria
(both located in posterior part of the body); however, cirrus and bursa are absent.
Within the Haplorchis genus, H. pleurolophocerca adult worms (of 0.32–0.42 × 0.14–0.17 mm in size) are
characterized by having only one testis and a ventrogenital sucker complex armed with spines. Freshwater
snails Melanoides tuberculata and Cleopatra bulimoides are the first intermediate host (harboring cer-
cariae); freshwater fish Gambusia affinis is the second intermediate host (harboring metacercariae); and
cats are the natural definitive hosts. H. pleurolophocerca infection in humans has been reported in Egypt.
H. pumilio adult worms (of 0.45–0.89 × 0.2–0.4 in size) are characterized by the presence of only
one testis and a ventrogenital sucker complex armed with 27–39 (average 32) gonotyl and chitinous
spines. The freshwater snail Melania reiniana var. hitachiens appears to act as the first intermediate
host; freshwater fish belonging to the families Cyprinidae, Siluridae, and Cobitidae are the second
intermediate host; and dogs and cats serve as the natural definitive hosts. This parasite is present in the
Philippines, Thailand, Laos, Vietnam, South China, Taiwan, Malaysia, India, Sri Lanka, Iraq and Egypt;
and H. pumilio infection in humans was first documented in Thailand in 1983 [4].
H. taichui adult worms (of 0.47–0.64 × 0.18–0.22 mm in size) are characterized by the presence of only
one testis and 14–20 (av. 15) large chitinous, fan-shaped spines (hooklets) on the ventrogenital sucker
complex. Under scanning electron microscope (SEM), the whole body surface of newly excysted juvenile
H. taichui shows numerous transverse rows of scale-like spines and two types of sensory papillae (type I,
ciliated knob-like swellings and type II, round swellings of the tegument) [5]. The first intermediate hosts
are freshwater snails Melania obliquegranosa, Melania juncea, or M. tuberculata [6]; the second inter-
mediate hosts are freshwater fish, including Cyclocheilichthys repasson, Cyprinus auratus, Cyprinus
carpio, Gambusia affinis, Hampala dispar, Labiobarbus leptocheila, Puntius binotatus, Puntius bre-
vis, Puntius gonionotus, Puntius leicanthus, Puntius orphoides, Puntius palata, Pseudorasbora parva,
Rhodeus ocellatus, and Zacco platypus in addition to Raiamas guttatus, Mystacoleucus marginatus,
and Henichoryhnchus siamensis; and the natural definitive hosts are dogs, cats, and birds. This parasite
Haplorchis 737
is found in Taiwan, the Philippines, Bangladesh, India, Sri Lanka, Malaysia, Thailand, Laos, Vietnam,
South China, Iraq, Palestine and Egypt. The first human case of H. taichui infection was reported in the
Philippines [4,7].
H. vanissimus adult worms (of 0.38–0.51 × 0.25–0.51 mm in size) utilize freshwater fish as the second
intermediate host, and pelicans and wild mammals as the definitive hosts. Human infection with H.
vanissimus was described in the Philippines [4].
H. yokogawai adult worms (of 0.47–0.64 × 0.18–0.22 mm in size) are characterized by having
only one testis and numerous (uncountable) minute chitinous spines on the ventrogenital sucker com-
plex. The first intermediate host is freshwater snail (M. tuberculata or Stenomelania newcombi); the
second intermediate host is freshwater fish, belonging to Cyclocheilichthys armatus, Hampala dispar,
Labiobarbus leptocheila, Misgurnus sp., Mugil spp., Onychostoma elongatum, Ophiocephalus striatus,
and Puntius spp.; and the natural definitive hosts are dogs, cats, cattle, and other mammals. The fluke is
known to occur in the Philippines, South China, Malaysia, Indonesia, Thailand, Laos, Vietnam, India,
Australia, and Egypt; and human infection with H. yokogawai was first noted in the Philippines [4].
The complete mitochondrial genome of H. taichui measures 15,130 bp in length and contains 12
protein-coding genes, 2 ribosomal RNAs (rRNAs, a small and a large subunit), and 22 transfer RNAs
(tRNAs). Similar to other trematodes, the atp8 gene is absent, although a single long noncoding region
is present between trnE and trnG in H. taichui mitochondrial genome. On the whole, H. taichui appears
to be more closely related to Opisthorchiidae than other trematode groups [8].
45.1.3 Clinical Features
With an incubation period of between 1 and 15 days, Haplorchis infections in humans are generally
asymptomatic or mild and transient in immunocompetent individuals. However, in heavily infected or
immunocompromised individuals, Haplorchis infestations may cause weakness, discomfort, abdominal
pain, diarrhea, loss of appetite, nausea, and vomiting [16].
In case that Haplorchis eggs released in the intestine by adult worms gain entry into the blood and
lymph vascular systems via mucosa, and migrate to the valves and myocardium, the heart, the brain, or
spinal cord, they may induce cardiac failure, neurological disorders, and occasional fatalities.
738 Laboratory Models for Foodborne Infections
Microscopic examination of the small intestine from patients with H. taichui infection may reveal
mucosal ulceration, mucosal and submucosal hemorrhages, fusion and shortening of villi, chronic
inflammation, and fibrosis of the submucosa [17].
45.1.4 Diagnosis
Diagnosis of Haplorchis infections has traditionally relied on microscopic observation of adult worms
or eggs in feces. This is sometimes problematic as adult worms may not always be present and because
Haplorchis eggs are indistinguishable from those of O. viverrini and C. sinensis [18].
The development of molecular methods based on nucleic acid amplification has offered new oppor-
tunities to improve the identification of Haplorchis adult worms, metacercariae, and eggs. By target-
ing ribosomal RNA gene and mitochondrial cytochrome c oxidase subunit I (mCOI) gene, polymerase
chain reaction (PCR) and its derivatives [e.g., PCR-restriction fragment length polymorphism (RFLP),
multiplex PCR, nested PCR, real-time PCR, and PCR pyrosequencing] have made rapid, sensitive, and
precise determination and phylogenetic analysis of heterophyid trematodes possible [19–23].
Thaenkham et al. [24] showed that using primers (COI-OV-Hap F&R primers) from mitochondrial
cytochrome c oxidase subunit I (COI) gene, two common fish-borne trematodes in Thailand, O. viverrini
and H. taichui, can be rapidly and precisely differentiated. In addition, Thaenkham et al. [25] described a
PCR-RFLP based on 28S ribosomal RNA gene and restriction enzyme MboII digestion for identification
of metacercaria of C. sinensis, O. viverrini, H. taichui, H. pumilio, and H. yogokawai.
Similarly, Sato et al. [19] designed primers from ITS 1 and ITS 2 regions in ribosomal DNA that
enabled amplification of 800, 820, 1250, and 930 bp products (ITS1) and 380, 390, 380, and 530 bp prod-
ucts (ITS2) from the eggs of O. viverrini, C. sinensis, H. pumilio, and H. taichui, respectively, allowing
their effective discrimination.
Furthermore, Wongsawad et al. [26,27] developed species-specific primers for H. taichui (Hapt_F
5′-GGCCAACGCAATCGTCATCC-3′ and Hapt_R1 5′-CTCTCGACCTCCTCTAGAAT-3′, which
yield a 170 bp PCR product) and O. viverrini (OpV-1F: 5′-AATCGGGCTGCATATTGACCGAT-3′ and
OpV-1R: 5′-CGGTGTTGCTTATTTTGCAGACAA-3′, which generate a 319 bp PCR product). Use of
these primers facilitated the specific confirmation of both parasites in fish and snail intermediate hosts.
45.2 Laboratory Models
45.2.1 Animal Models
45.2.1.1 Rodents
Mice (Mus musculus) are a suitable experimental definitive host for adult H. taichui. Sukontason et al.
[29] noted that mice infected orally with 200 active H. taichui metacercaria developed mature adult
worms in a few days, with egg production as early as 3 days postinfection, and increasing daily to about
50–60 eggs. Kay et al. [30] also found mice highly susceptible to H. pumilio infection, with peak adult
worm recovery 7 days after oral inoculation with metacercariae.
Rats may be used as definitive host for H. taichui infection. Saenphet et al. [31] observed that
male rats orally fed with 300 H. taichui metacercariae produced adult worms as early as 3 days
postinfection.
Haplorchis 739
Syrian golden hamster (Mesocricetus auratus) is also capable of supporting growth of Haplorchis
parasites, producing adult worms in the intestine 14 days after being fed metacercariae via a stomach
tube.
45.2.1.2 Dogs
Nissen et al. [32] showed that dogs (3–6 months old) infected with 500 H. pumilio metacercariae yielded
adult flukes without clinical symptoms. The infection lasted for at least 2 months, and up to 2 eggs/g feces
were excreted. The predilection site of the flukes appeared to be the lower part of jejunum (93% of total
worm burden).
45.2.1.3 Foxes
Nissen et al. [33] reported that foxes orally infected with 2000 H. pumilio metacercariae displayed
anorexia at 12 days postinfection (which last for approximately a week), produced between 116 and 2070
adult flukes per animal mostly in the lower part of the jejunum, and excreted H. pumilio eggs (initially
about 16–20 with a maximum of 1443 ± 1176 eggs/g of feces).
45.2.1.4 Chicks
Kumchoo et al. [34] demonstrated the suitability of chicks (Gallus gallus domesticus) as definitive host
for infection with H. taichui, since chick infected orally with 200 H. taichui metacercariae generated
mature adult worms as early as days 3 postinfection. Kay et al. [30] also confirmed that chicks are useful
host for H. pumilio metacercariae infection, although mice appear to be significantly more susceptible
to infection than chicks.
45.3 Conclusion
The genus Haplorchis consists of more than 20 intestinal trematode species whose complex life cycles
involve two intermediate hosts (snail and fish) and a definitive host (mammals and birds). Of the five
Haplorchis species (H. pleurolophocerca, H. pumilio, H. taichui, H. vagabundi, and H. yokogawai)
implicated in human infections, H. taichui is particularly common and poses a major public health
threat in Asia, parts of Africa, and the Americas. In order to decipher the pathogenic mechanisms and
immunobiology of haplorchiasis, use of laboratory models is crucial. Fortunately, a number of laboratory
animals (e.g., mice, rats, hamsters, dogs, foxes, and chicks) have been shown to support the growth and
maturation of Haplorchis adult worms from metacercaria recovered from fish. Further experimentation
will undoubtedly contribute to an improved understanding of human haplorchiasis and to the develop-
ment of novel intervention strategies against Haplorchis parasites.
REFERENCES
1. Thaenkham U, Blair D, Nawa Y, Waikagul J. Families Opisthorchiidae and Heterophyidae: are they
distinct? Parasitol Int. 2012;61(1):90–3.
2. Pearson JC, OwYang CK. New species of Haplorchis from Southeast Asia, together with keys to the
Haplorchis-group of heterophyid trematodes of the region. Southeast Asian J Trop Med Public Health.
1982;13:35–60.
3. ITIS report. Available at http://www.itis.gov/servlet/SingleRpt/SingleRpt?search_topic=TSN&search_
value=57114. Accessed on February 12, 2016.
740 Laboratory Models for Foodborne Infections
4. Chai JY, Shin EH, Lee SH, Rim HJ. Foodborne intestinal flukes in Southeast Asia. Korean J Parasitol.
2009;47(Suppl):S69–102.
5. Sukontason KL, et al. Surface ultrastructure of excysted metacercariae of Haplorchis taichui
(Trematoda: Heterophyidae). Southeast Asian J Trop Med Public Health. 2000;31(4):747–54.
6. Chontananarth T, Wongsawad C. Prevalence of Haplorchis taichui in field-collected snails: a molecular
approach. Korean J Parasitol. 2010;48(4):343–6.
7. Sommerville C. The life-history of Haplorchis pumilio (Looss, 1896) from cultured tilapias. J Fish Dis.
1982;5:233–41.
8. Lee D, et al. Complete mitochondrial genome of Haplorchis taichui and comparative analysis with other
trematodes. Korean J Parasitol. 2013;51(6):719–26.
9. Kumchoo K, Wongsawad C, Chai JY, Vanittanakom P, Rojanapaibul A. High prevalence of Haplorchis
taichui metacercariae in cyprinoid fish from Chiang Mai Province, Thailand. Southeast Asian J Trop Med
Public Health. 2005;36(2):451–5.
10. Skov J, Kania PW, Dalsgaard A, Jorgensen TR, Buchmann K. Life cycle stages of heterophyid trema-
todes in Vietnamese freshwater fishes traced by molecular and morphometric methods. Vet Parasitol.
2009;160:66–75.
11. Nithikathkul C, Wongsawad C. Prevalence of Haplorchis taichui and Haplorchoides sp. meta-
cercariae in freshwater fish from water reservoirs, Chiang Mai, Thailand. Korean J Parasitol.
2008;46(2):109–12.
12. Chai JY, et al. Prevalence of the intestinal flukes Haplorchis taichui and H. yokogawai in a moun-
tainous area of Phongsaly Province, Lao PDR. Korean J Parasitol. 2010;48(4):339–42.
13. Wijit A, Morakote N, Klinchid J. High prevalence of haplorchiasis in Nan and Lampang provinces,
Thailand, proven by adult worm recovery from suspected opisthorchiasis cases. Korean J Parasitol.
2013;51(6):767–9.
14. Krailas D, Veeravechsukij N, Chuanprasit C, Boonmekam D, Namchote S. Prevalence of fish-borne trema-
todes of the family Heterophyidae at Pasak Cholasid Reservoir, Thailand. Acta Trop. 2016;156:79–86.
15. Sohn WM, et al. Prevalence of Haplorchis taichui among humans and fish in Luang Prabang
Province, Lao PDR. Acta Trop. 2014;136:74–80.
16. Watthanakulpanich D, et al. Haplorchis taichui as a possible etiologic agent of irritable bowel
syndrome-like symptoms. Korean J Parasitol. 2010;48:225–9.
17. Sukontason K, Unpunyo P, Sukontason KL, Piangjai S. Evidence of Haplorchis taichui infection as
pathogenic parasite: three case reports. Scand J Infect Dis. 2005;37:388–90.
18. Sato M, Sanguankiat S, Pubampen S, Kusolsuk T, Maipanich W, Waikagul J. Egg laying capacity of
Haplorchis taichui (Digenea: Heterophyidae) in humans. Korean J Parasitol. 2009;47:315–8.
19. Sato M, Thaenkham U, Dekumyoy P, Waikagul J. Discrimination of O. viverrini, C. sinensis,
H. pumilio and H. taichui using nuclear DNA-based PCR targeting ribosomal DNA ITS regions. Acta
Trop. 2009;109:81–3.
20. Sato M, et al. Copro-DNA diagnosis of Opisthorchis viverrini and Haplorchis taichui infection in
an endemic area of Lao PDR. Southeast Asian J Trop Med Public Health. 2010;41:28–35.
21. Dung DT, Hop NT, Thaenkham U, Waikagul J. Genetic differences among Vietnamese Haplorchis
taichui populations using the COI genetic marker. J Helminthol. 2013;87(1):66–70.
22. Chontananarth T, Wongsawad C, Chomdej S, Krailas D, Chai JY. Molecular phylogeny of trema-
todes in Family Heterophyidae based on mitochondrial cytochrome c oxidase subunit I (mCOI). Asian
Pac J Trop Med. 2014;7(6):446–50.
23. Tantrawatpan C, et al. Development of a PCR assay and pyrosequencing for identification of impor-
tant human fish-borne trematodes and its potential use for detection in fecal specimens. Parasites
Vectors. 2014;7:88.
24. Thaenkham U, Visetsuk K, Dung Do T, Waikagul J. Discrimination of Opisthorchis viverrini from
Haplorchis taichui using COI sequence marker. Acta Trop. 2007;103:26–32.
25. Thaenkham U, et al. Rapid and simple identification of human pathogenic heterophyid intestinal
fluke metacercariae by PCR-RFLP. Parasitol Int. 2011;60:503–6.
26. Wongsawad C, Wongsawad P, Chai JY, Anuntalabhochai S. Development of a HAT-RAPD marker
for the detection of minute intestinal fluke infection. Exp Parasitol. 2009;123(2):158–61.
Haplorchis 741
Jong-Yil Chai
CONTENTS
46.1 Introduction................................................................................................................................... 744
46.1.1 Classification and Morphology......................................................................................... 744
46.1.1.1 Metagonimus yokogawai Katsurada, 1912....................................................... 744
46.1.1.2 Metagonimus takahashii Suzuki, 1930.............................................................745
46.1.1.3 Metagonimus miyatai Saito, Chai, Kim, Lee, and Rim, 1997..........................745
46.1.1.4 Metagonimus minutus Katsuta, 1932................................................................ 746
46.1.1.5 Metagonimus katsuradai Izumi, 1935.............................................................. 746
46.1.1.6 Metagonimus otsurui Saito and Shimizu, 1968............................................... 746
46.1.1.7 Metagonimus hakubaensis Shimazu, 1999...................................................... 746
46.1.2 Biology and Life Cycle......................................................................................................747
46.1.3 Epidemiology and Distribution........................................................................................ 748
46.1.4 Pathogenesis and Clinical Features...................................................................................749
46.1.5 Diagnosis.......................................................................................................................... 750
46.1.6 Treatment and Control.......................................................................................................751
46.2 Laboratory Models.........................................................................................................................751
46.2.1 General Considerations.....................................................................................................751
46.2.2 Animal Models Used for Studies on Morphology and Taxonomy...................................752
46.2.2.1 Dogs...................................................................................................................752
46.2.2.2 Cats....................................................................................................................752
46.2.2.3 Hamsters............................................................................................................752
46.2.2.4 Mice and Rats....................................................................................................753
46.2.2.5 Rabbits, Guinea Pigs, and Other Mammals......................................................753
46.2.2.6 Ducks and Chickens...........................................................................................753
46.2.3 Animal Models Used for Studies on Worm Development and Host Susceptibility.........753
46.2.3.1 Dogs...................................................................................................................753
46.2.3.2 Cats................................................................................................................... 754
46.2.3.3 Hamsters........................................................................................................... 754
46.2.3.4 Mice and Rats................................................................................................... 754
46.2.3.5 Rabbits, Guinea Pigs, and Other Mammals......................................................755
46.2.3.6 Ducks and Chickens...........................................................................................755
46.2.4 Animal Models Used for Studies on Pathogenesis and Pathology...................................756
46.2.4.1 Dogs, Cats, Rats, and Mice................................................................................756
46.2.5 Animal Models Used for Studies on Immunity and Host–Parasite Interaction...............756
46.2.5.1 Dogs, Cats, Rats, Mice, and Hamsters..............................................................756
46.2.6 Animal Models Used for Studies on Chemotherapy and Control....................................758
46.2.6.1 Dogs and Rats....................................................................................................758
743
744 Laboratory Models for Foodborne Infections
46.1 Introduction
Twelve genera (29 species) of the family Heterophyidae are so far known to infect humans around the
world: Apophallus, Ascocotyle, Centrocestus, Cryptocotyle, Haplorchis, Heterophyes, Heterophyopsis,
Metagonimus, Procerovum, Pygidiopsis, Stellantchasmus, and Stictodora.1 Among them, Haplorchis,
Heterophyes, and Metagonimus are the three most important genera in public health significance. The
genus Metagonimus is distinct morphologically from Haplorchis and Heterophyes and also differs in its
life cycle and geographical distribution.1,2
The genus Metagonimus was established by F. Katsurada in 1912 with M. yokogawai as the type
species.3,4 Later, six more species of Metagonimus have been described in the literature: M. takahashii
Suzuki, 19305; M. minutus Katsuta, 19326; M. katsuradai Izumi, 19357; M. otsurui Saito and Shimizu,
19688; M. miyatai Saito et al., 19979; and M. hakubaensis Shimazu, 1999.10 From public health points
of view, M. yokogawai, M. takahashii, and M. miyatai are the three major species frequently causing
human infections.2,11 M. minutus was also listed as a human-infecting species12; however, no literature
background is available to verify it. With regard to M. katsuradai, an experimental human infection was
reported7; however, no natural human infections have been documented yet. M. otsurui was originally
described from experimental hamsters infected with metacercariae from freshwater fishes,8 and later,
natural infection of the Japanese water shrew was discovered13 but never from humans. M. hakubaensis
was first found from experimental rats that were fed the metacercariae in lampreys in Japan,10 but human
infection is yet unknown.
Human Metagonimus spp. infections have been found exclusively in the Far East, including the
Republic of Korea (=Korea), China, Japan, and the Far Eastern Russia.1,2,11 However, the life cycle of
M. yokogawai was also detected in Taiwan and eastern Europe. The potential for human infections with
other species of Metagonimus remains to be elucidated.
The pathogenicity of Metagonimus spp. to humans and animals has been reported to be generally mild.1
The habitat of the adult flukes in immunocompetent mice was confined to the mucosal layer, mainly
in the intervillous space and crypt of the small intestine, causing mucosal inflammations, but did not
extend beyond the submucosa and underneath.14 However, in immunosuppressed mice, the adult flukes
could invade into a deeper layer of the submucosa where mesenteric vessels are available.15 This suggests
a potential deep invasion of Metagonimus flukes to elicit significant pathogenesis and pathology in the
human host. To verify this suggestion, experiments with proper laboratory animal models are strongly
needed. In this chapter, laboratory models so far used to study the life cycle, host–parasite relationships,
pathogenesis, immunity, and other related aspects of Metagonimus infection are briefly reviewed.
46.1.1.1 M
etagonimus yokogawai Katsurada, 1912
(syn. Loxotrema ovatum Kobayashi, 1912; Metagonimus ovatus Yokogawa, 1913; Loossia romanica
Ciurea, 1915; Loossia parva Ciurea, 1915; Loossia dobrogensis Ciurea, 1915)16
Metagonimus 745
This species was originally reported from an experimental dog fed with metacercariae from the sweet-
fish (Plecoglossus altivelis) in 1912 from Taiwan and named as Heterophyes yokogawai.3,30 However, the
adult worm morphology was significantly different from Heterophyes, and subsequently it was renamed
as M. yokogawai in the same year.3,4 It is the most highly prevalent of all Metagonimus spp. and has a
most wide geographical distribution, including the Republic of Korea, Japan, China, Taiwan, Russia,
Ukraine, India, Romania, Serbia, Bulgaria, Israel, Egypt, the Balkan States, and Spain.2,3,12,17,18–22 Some
old literature on M. yokogawai were actually referring to M. takahashii or M. miyatai, and caution is
required when reviewing M. yokogawai sensu stricto.3
The characteristic morphologies of M. yokogawai include the presence of two testes, which are closely
adjacent to each other near the posterior end of the body.2,16 However, in M. miyatai and M. takahashii,
the two testes are more or less separated from each other (see below).2,3,23 Another differential character
of M. yokogawai is the distribution of vitelline follicles; they extend in lateral fields from the level of the
ovary down to the posterior end of the posterior testis, but not beyond the posterior testis.16 By contrast,
in M. takahashii, vitelline follicles are abundant from the level of the ovary down to the posterior testis
and usually extend beyond the posterior testis level.9,23 In M. miyatai, vitelline follicles distribute from
the level of the ovary down to the anterior level of the posterior testis; there are no vitellaria distribution
beyond the posterior testis.9,23 In addition, M. yokogawai has the uterine tubule, which does not overlap
or cross over the middle portion of the anterior testis, whereas M. takahashii and M. miyatai have the
uterine tubules that overlap the whole anterior testis.9,23
The adult flukes of M. yokogawai are slightly smaller (0.80–1.32 × 0.42–0.54 mm) than those of
M. takahashii (0.86–1.19 × 0.44–0.57 mm) and M. miyatai (1.00–1.30 × 0.46–0.63 mm).3,9 The eggs
of M. yokogawai are also smaller (0.026–0.030 × 0.015–0.018 mm) than those of M. takahashii
(0.030–0.036 × 0.017–0.020 mm) and M. miyatai (0.029–0.032 × 0.017–0.020 mm), although there are
some overlaps in the range.3,23 The adult flukes of M. minutus differ from those of M. yokogawai,
M. takahashii, and M. miyatai by having a smaller body (0.46 × 0.28 mm) and smaller egg size (0.023 ×
0.013 mm).6,23 The adult flukes of M. katsuradai, M. otsurui, and M. hakubaensis differ from those of
M. yokogawai, M. takahashii, M. miyatai, and M. minutus in that the formers have a larger ventral
sucker than the oral sucker.7,10,23
46.1.1.3 M etagonimus miyatai Saito, Chai, Kim, Lee, and Rim, 1997
This species was first described by F. Katsurada9,30 (only by a figure drawing) together with
M. yokogawai in 1912 in Taiwan and then by I. Miyata in 1941 in Japan.31 However, its specific status
746 Laboratory Models for Foodborne Infections
was not acknowledged, and no special name was given until 1984 when S. Saito began to call it as the
“Metagonimus Miyata type.”9 It was in 1997 when the specific level of this fluke became acknowledged
and described as a new species, M. miyatai.9 The new species description was based on adult flukes col-
lected from dogs and hamsters experimentally fed with the metacercariae from the sweetfish, dace, com-
mon fat-minnow Morocco steindachneri, pale chub Zacco platypus, and dark chub Zacco temmincki in
Korea and Japan.3,9 Human infections have been reported from Korea and Japan.9,23
The specific morphological characters of M. miyatai include the two markedly separated testes from
each other, the posterior one being just touching the terminal end of the worm, its vitelline follicles
never distributing beyond the posterior testis, and the egg size, which is intermediate between those
of M. yokogawai and M. takahashi.2,9,16 M. miyatai is also genetically distinct from M. yokogawai and
M. takahashii.32–34 M. miyatai differs from M. minutus in its larger body and egg size.6,23 It also dif-
fers from M. katsuradai, M. otsurui, and M. hakubaensis in that the latter three species have a smaller
ventral sucker compared with the oral sucker, whereas in M. miyatai, the ventral sucker is larger than
its oral sucker.10,23
46.1.1.4 M
etagonimus minutus Katsuta, 1932
This species was originally described in Taiwan in 1932 based on adult flukes from cats and mice experi-
mentally fed with the metacercariae in the brackish water mullet.6,16 It has body and egg sizes smaller
than those of M. yokogawai, M. takahashii, and M. miyatai.2,3 The body size of M. minutus is similar to
that of M. katsuradai, but its egg size is smaller than that of M. katsuradai.3,6,7 The relative size of the
oral and ventral suckers is also a characteristic feature; the oral sucker is bigger than the ventral sucker
in M. katsuradai, and the oral sucker is smaller than the ventral sucker in M. minutus.3,6,7 M. minutus is
listed as a human-infecting species; however, no relevant background literature is available.12,16
46.1.1.7 M
etagonimus hakubaensis Shimazu, 1999
This species was described in Japan in 1999 based on adult flukes from experimental rats fed with the
metacercariae in a lamprey, Lethenteron reissneri, a fish species collected in Hakuba Village, Nagano
Metagonimus 747
In Russia, human M. yokogawai infection is highly endemic in the Amur and Ussuri valleys of the
Khabarovsk Territory.12 The prevalence among the ethnic minority was reported between 20% and
70%.12 In the north of Sakhalin Island, the infection rate was 1.5% among Russians and 10% among eth-
nic minorities, and sporadic cases were also reported in the Amur district and the Primorye Territory.12,16
The population at risk is estimated to be about 859,000, which is 14.7% of the total population in these
territories.12
In eastern Europe, its existence in fish hosts and wild animals has been reported in Ukraine,21
Serbia,18,22 Bulgaria,19,75 and Czech Republic,20 although no human infections have been confirmed so far.
Regarding M. takahashii infection, in the Republic of Korea, the adult flukes were first recovered from
rabbits experimentally fed with the metacercariae in 1960.76 Thereafter, the presence of human infec-
tions was first demonstrated from riparian people along the Hongcheon River, Gangwon-do, in 1988.77
Subsequently, in 1993, an endemic focus was discovered from Umsong-gun, Chunchungnam-do, along
the upper reaches of the Namhan River.23 The riparian people along this river were mixed-infected with
M. miyatai, with an egg-positive rate of 9.7% for both species.23 A year later, in 1994, M. takahashii adult
flukes (8–402 in number) were recovered in six residents living in Kochang-gun, Gyeongsangnam-do.78
The major sources of human infections were the crucian carp C. carassius.26 The perch, L. japonicus,
was also identified as the fish host in Gyeongsangnam-do.79
In Japan, several articles were published on M. takahashii after this species was first recorded in 1930.3,4
However, before the studies of S. Saito in the 1970s–1990s,13,35,36 its precise epidemiological character-
istics, including the prevalence and geographical distribution of human infections, have not been clearly
defined. However, it is worthwhile to refer to Takahashi,24 who originally discovered M. takahashii;
in 1929 in Okayama City, 43 (0.64%) of 6680 residents were infected with M. takahashii, whereas 54
(0.81%) were infected with M. yokogawai.3 Later, in 1957 in Fuchu City, Hiroshima Prefecture, 11 (4.8%)
of 231 residents examined were infected with M. takahashii, whereas 81 (35.1%) were infected with
M. yokogawai.3 Quite recently, in 2003, Semisulcospira snails collected around the Lake Biwa were
found infected with the cercariae of M. takahashii, together with those of M. yokogawai, M. hakubaensis,
M. otsurui, and M. katsuradai.80
In the Republic of Korea, before M. miyatai was designated as a distinct species, its human infec-
tions were noticed by detecting eggs in the feces, which were slightly larger in size than those of
M. yokogawai.52 Later, adult flukes (under the name Metagonimus Miyata type) were recovered from
32 riverside people living along the Namhan River in Umsong-gun (those people were concurrently
infected with M. takahashii) and Yongwol-gun (infected only with M. miyatai).23 In Yongwol-gun, the
egg-positive rate of M. miyatai among 77 riparian residents was 48.1%.23 Adult flukes were also recov-
ered from residents along the Hantan River, Chorwon-gun81 and Talchon River, Chungwon-gun.82
In Japan, epidemiological studies, particularly on human infections, are scarce. Saito et al.9 enlisted
dogs, foxes, raccoon dogs, and black-eared kites for animal definitive hosts in Shimane, Kochi, and
Yamagata Prefectures. In Hiroi River basin, Nagano Prefecture, cercariae and rediae were found in
Semisulcospira snails, and metacercarial infection was detected in P. lagowski steindachneri fish.37 In
Shizuoka Prefecture, many small rivers were found to have M. miyatai metacercariae in fish.70
The geographical distribution of M. katsuradai is confined to Japan.3,7 In Hyogo Prefecture, the larval
infection rate of Semisulcospira snails was 2.0% for M. yokogawai, 8.6% for M. takahashii, and 1.9% for
M. katsuradai in 1939.3 In Kobe City, dogs were reported as a natural definitive host.3 Around the Lake
Biwa, cercariae of M. katsuradai (having six oral spines) were discovered in 2003.80 Natural human
infections have never been documented.4
decreased villus/crypt height ratio.2,14,15,84 Almost the same intestinal histopathology seems to occur in
human M. yokogawai infection.2,16,85
In immunocompetent hosts, the location of M. yokogawai worms is confined to the intestinal mucosa,
not beyond the submucosa or muscularis mucosa.14 However, immunosuppression could allow a deeper
invasion of worms into the submucosa or underneath.15 Immunosuppression can also enhance survival
of the worms and prolong their life spans.15,86 Poor absorption of intestinal secretions from the secretory
crypt cells seems to induce watery diarrhea in humans or animal hosts.2,16 In M. miyatai infection in
mice, the intestinal histopathology was similar to that in M. yokogawai infection, although the degree of
mucosal damage was less severe than in M. yokogawai infection.87
A big question regarding the pathogenicity of M. yokogawai is whether this fluke can cause extrain-
testinal infections in humans and animal hosts.16 It was reported in 1940 in the Philippines that several
species of heterophyid flukes, including Stellantchasmus falcatus, Haplorchis spp., and Procerovum
spp., caused erratic parasitism in the heart, brain, and spinal cord in humans, which was often fatal.2,17,88
Such erratic parasitism may have occurred in immunocompromised patients.2,16 In M. yokogawai-
infected human patients, no direct evidence of extraintestinal infections has been reported.16 However,
it is worth noting that intracerebral hemorrhage and diabetes mellitus occurred in a metagonimiasis
patient, and the intracerebral hemorrhage may be an acute complication, whereas diabetes mellitus may
be a chronic sign.89
The clinical symptoms in human metagonimiasis, unless complicated by other diseases and there
are no signs of immunosuppression, include mild to moderate degrees of abdominal pain, diarrhea,
lethargy, anorexia, and weight loss.2,16 However, the severity of symptoms may vary depending on vari-
ous host-side factors.4,16 One factor is the intensity of infection—the number of worms infected in each
patient; heavier infection results in more severe illness.2,16,17 The second factor is the immune status of
the host.16,17 In immunocompromised patients, M. yokogawai infection can possibly cause severe clinical
manifestations, including erratic parasitism in vital organs.4,16A severe clinical case of M. yokogawai
infection complicated with multiple intracerebral hemorrhages and diabetes mellitus may have been
immunosuppressed.89 A third factor is the history of previous infections that might confer some degree
of acquired immunity in individual patients.16 New visitors to an endemic area are likely to suffer from
a severe illness from a primary infection.2,16 On the other hand, individuals living in an endemic area for
a long time, who inevitably are infected repeatedly, may be protected from severe pathology and symp-
toms caused by M. yokogawai worms.4,16
46.1.5 Diagnosis
Human Metagonimus infection can be diagnosed by detecting eggs in fecal samples.16 The direct
smear, cellophane thick smear, and Kato-Katz thick smear techniques can be applied in field con-
ditions, whereas in the laboratory equipped with centrifuges, concentration techniques, for exam-
ple, formalin-ether sedimentation technique and brine (or zinc sulfate) floatation techniques, can be
performed.4,16
However, morphological differentiation of eggs in fecal samples may be problematic in areas hav-
ing coprevalence of Clonorchis sinensis and/or heterophyid fluke (Metagonimus spp., Heterophyes
spp., Pygidiopsis summa, and Haplorchis spp.) infections.4 Eggs of M. yokogawai can be differed
from eggs of other heterophyids by their length of 26.9–31.6 µm, elliptical shape with length/width
ratio of 1.5–2.1, clean shell surface (without muskmelon patterns), less prominent operculum, lack
of shoulder rims, and dark yellow or brown color.90,91 Eggs of M. takahashii and M. miyatai are
very similar to those of M. yokogawai, except their sizes being larger in the former and smaller in
M. yokogawai.23,90,91 In patients infected with less than 100 worms, the probability of detecting eggs
in one Kato-Katz smear is almost zero,4,16 assuming that the daily number of eggs produced per
M. yokogawai worm in the human host is 14–64.92 In such cases, serological tests, including ELISA,
are helpful.16
Molecular techniques can be applied to detect Metagonimus spp. infections in the feces71 or food
materials including fish.93
Metagonimus 751
46.2 Laboratory Models
46.2.1 General Considerations
Metagonimus spp. are zoonotic, and various animals are known to serve as natural definitive hosts.
In endemic areas of metagonimiasis due to M. yokogawai or M. miyatai, for example, humans are
the principal host and the most important epidemiological element. The infected humans may suffer
from mild to moderate degrees of abdominal discomfort, diarrhea, indigestion, and weight loss.1,2,4,17
However, the mechanisms of diarrhea and other clinical symptoms as well as host immune responses
against Metagonimus spp. infection are poorly understood. Therefore, for proper understanding of
human metagonimiasis, laboratory models, in particular, experimental metagonimiasis in laboratory
animals (Table 46.1), or in vitro culture models, are indispensable. For this purpose, mammalian ani-
mals like mice, rats, dogs, and hamsters have been most commonly used, and rarely cats, rabbits,
chickens, and ducks were used. The study subjects using laboratory animals have been the morphol-
ogy, taxonomy, growth and sexual development of worms, their longevity and fecundity, comparative
susceptibility of different animal species, pathogenesis and pathology, host–parasite interactions, host
immune responses, chemotherapy, and control.
TABLE 46.1
Species of Metagonimus Reported in the Literature and Laboratory Animals Used
Laboratory Animals
Species Human Infection Source of Infection Models
Metagonimus yokogawai Natural Sweetfish, dace, perch Dog, cat, hamster, rat,
mouse, rabbit, guinea pig,
gerbil, mink, raccoon dog,
fox, duck, chicken
(unsuccessful)
Metagonimus takahashii Natural Crussian carp, common carp, Dog, cat, hamster, rat,
dace, perch mouse, rabbit
Metagonimus miyatai Natural Sweetfish, dace, minnow, pale Dog, hamster, rat, mouse,
chub, dark chub duck (unsuccessful)
Metagonimus minutus Uncertaina Mullet Cat, mouse
Metagonimus katsuradai Experimental Carp, pale chub, Acheilognathus Dog, cat, hamster, rat,
lanceolata mouse, rabbit, duck
Metagonimus otsurui None Freshwater fish Dog, hamster, chicken
(unsuccessful)
Metagonimus hakubaensis None Freshwater fish Dog, hamster, rat
a Enlisted among the species infecting humans,12 but without literature background.
752 Laboratory Models for Foodborne Infections
46.2.2.2 Cats
Cats have been used rarely as a laboratory host for Metagonimus spp.7,97 Kobayashi97 infected cats
and dogs with the metacercariae of M. yokogawai isolated from sweetfish muscle to obtain the adult
flukes. Kim et al.79 infected a cat with metacercariae from perches and obtained M. takahashii adult
flukes. Katsuta6 also infected cats with metacercariae of M. minutus and obtained adult flukes. Izumi7
used cats and several other mammalian animals to recover the adult flukes of M. katsuradai. Cats
were also used for pathology103,104 and immunology studies105,106 in Metagonimus spp. infection (see
46.2.4 and 46.2.5).
46.2.2.3 Hamsters
Hamsters were occasionally used as an experimental definitive host for Metagonimus spp. Yokogawa
and Sano107 infected hamsters, mice, rats (cotton rats and Wistar rats), and guinea pigs with the metacer-
cariae of M. yokogawai to obtain the adult flukes and to observe the development of worms in different
hosts. Kagei and Kihata99 also studied the morphology, recovery, and the life span of M. yokogawai in
hamsters. Miyamoto and Kutsumi108 infected hamsters and dogs to recover M. yokogawai adult flukes. A
similar study was performed recently by Li et al.46 in Taiwan. Hamsters were also used to differentiate
morphological characters of M. yokogawai from those of M. miyatai9,29,37,70,109 and M. takahashii.26,29,109
Moreover, unique morphological features of M. katuradai54, M. otsurui,8,13 and M. hakubaensis55 were
also studied using hamsters.
The merits of using hamsters for an experimental definitive host include their high susceptibility to
Metagonimus spp. infection with high worm recovery and longer worm survival.46,107 Suitability as a
laboratory animal and easy handling, compared with dogs and cats, are also among the merits.
Metagonimus 753
of M. yokogawai was 35–45 eggs at day 7 postinfection in the dog host. Miyamoto and Kutsumi108
confirmed that the dace Tribolodon spp. is a second intermediate host of M. yokogawai by recovery of
adult flukes from experimentally infected dogs from day 8 to 129 postinfection. Kang et al.84 infected
dogs with the metacercariae of M. yokogawai and obtained the worm recovery rate of about 20% even at
week 6 postinfection; a repeated infection led to displacement of worms from the duodenum and jejunum
down to the ileum.
With regard to M. miyatai, dogs were used for studies on the worm development and/or fecundity.9,38,50,53
Ahn and Ryang53 reported that the worm development of M. miyatai and susceptibility was better in dogs
than in rats; the worm maturity was the best at day 35 after infection in dogs. In M. takahashii, few stud-
ies have been performed using dogs as the definitive host. Takahashi24 observed better development of
M. takahashii worms in dogs compared to those developed in mice. Similarly, Koga28 studied the satis-
factory worm development of M. takahashii (under the name, large egg-type Metagonimus) in dogs and
cats. Dogs were further used to study the worm development and host susceptibility of M. katsuradai,7,39
M. otsurui,8 and M. hakubaensis.55 Kudo et al.55 reported that dogs and hamsters were more susceptible
to M. hakubaensis infection than cats, rats, mice, chickens, and quails.
Regardless of the species of Metagonimus, dogs were found to be a highly susceptible host to experi-
mental Metagonimus infection. The worms grow quickly (within 3–6 days) in the small intestine of dogs
compared with other laboratory hosts including mice and rats, and survive remarkably a longer time.
Their fecundity in dogs was fairly good.
46.2.3.2 Cats
Kobayashi97 first used cats to infect the metacercariae of M. yokogawai isolated from the sweetfish.
Thereafter, only a few researchers73,103–106 used cats to study M. yokogawai. These studies were mainly
to recover the adult flukes or to observe the intestinal pathology and host immune responses103–106 rather
than to observe the life cycle, development, and sexual maturity of worms. Sohn et al.73 infected cats and
dogs to recover adult M. yokogawai after an experimental infection with the metacercariae in freshwater
fish from China.
Studies using cats were also scarce for other Metagonimus spp. As for M. takahashii, Kim et al.79
infected a cat with the metacercariae in the perch Lateolabrax japonicus from Korea and obtained
adult flukes 21 days later. Similarly, Katsuta6 obtained M. minutus adult flukes from cats experimentally
infected with the metacercariae in Taiwan, and Izumi7 recovered adult flukes of M. katsuradai from
experimental cats.
46.2.3.3 Hamsters
Hamsters were popularly used to study the development and longevity of Metagonimus spp. worms.
Yokogawa and Sano107 reported that hamsters were a highly suitable animal host for M. yokogawai infec-
tion compared with mice, rats, and guinea pigs. However, Kagei and Kihata99 reported that although some
M. yokogawai worms survived up to a year in dogs, no worms were retained in the intestine of hamsters
after 5 months. Miyamoto and Kutsumi108 reported similar findings; in dogs, a considerable number of
worms survived at day 129 postinfection, but in hamsters, only a few worms survived at day 29 postinfec-
tion. However, Li et al.46 obtained a high worm recovery rate (75.3%) of M. yokogawai from hamsters at day
14 postinfection. Compared with hamsters or dogs, mice were not a suitable host for M. yokogawai infec-
tion.99,107 Therefore, it can be said that the suitability of laboratory animals to infection with M. yokogawai
is the highest for dogs, followed by hamsters, rats, mice, and other rodent animals. Hamsters were also used
for studies on the development and host susceptibility of M. miyatai37 and M. otsurui.13
survived until day 26. Yokogawa and Sano107 compared the recovery and growth of M. yokogawai in
mice, Wistar rats, cotton rats, hamsters, and guinea pigs. They reported that the worms obtained full
maturation by week 1 after infection in all of these animals except guinea pigs, which showed no worms
even at 1 week. Hamsters and cotton rats retained more worms until week 6 after infection, whereas
mice and Wistar rats retained smaller number of worms.107 Hong and Seo113 observed the full growth
and development of M. yokogawai in mice by day 7 postinfection, although the rate of growth varied
individually. Kagei and Kihata99 reported that M. yokogawai developed well in mice, hamsters, and
dogs; however, the worm survived shorter than 1 month in mice, whereas it survived until 4 months in
hamsters and up to a year in dogs. On the other hand, Li et al.46 obtained a fairly high worm recovery
rate (70.0%) of M. yokogawai from ICR mice at day 14 postinfection, comparable with that (75.3%)
obtained in hamsters. Chai et al.86 infected M. yokogawai to various strains of mice (CBH, A, C57BL,
DBA, and KK) and observed the susceptibility of each mouse strain judged by the worm develop-
ment and worm recovery. KK mice (diabetic mice) showed the highest worm recovery at day 7 after
infection.86
Mice and rats were also used for studies on the development and maturation of M. miyatai and/or
M. takahashii.29,38,50,51–53,77 Kim et al.38 observed the development and site distribution of M. miyatai
worms (under the name Metagonimus Miyata type) in the intestine of mice. Ahn and Ryang53 reported
that M. miyatai (under the name Metagonimus Miyata type) continuously increased the body size in
rats until day 30 postinfection when the number of uterine eggs also peaked. Some retarded worms
were recovered until day 55 postinfection in rats.53
Guk et al.51 studied on the comparative growth and development of M. yokogawai, M. miyatai, and
M. takahashii in different strains of mice (BALB/c, ddY, C57BL/6J, C3H/HeN, and A/J). They found
that ddY mice were the most suitable host for M. yokogawai with the highest worm recovery, that
BALB/c and ddY were equally suitable for M. miyatai infection, and that BALB/c, ddY, and C3H/HeN
mice were equally suitable for M. takahashii infection.51 Generally, mice were fairly susceptible to M.
yokogawai infection with high worm recovery, good worm dimension, and high worm fecundity but
less susceptible to M. miyatai and M. takahashii infection.51
Mice were also used to study the growth and development of M. minutus; the sexual maturation was
completed by day 7 postinfection.6 By comparison, the development of M. katsuradai was completed
earlier in mice, by day 5 postinfection; moreover, eggs were seen in the feces of mice from day 4 (90 h)
postinfection.7 Fully mature specimens of M. hakubaensis were recovered in experimentally infected
rats at days 7–22 postinfection.10
succeeded in recovery of two adult flukes at day 7 after infection of a duck with M. katsuradai meta-
cercariae. Chicks were used for infection with M. yokogawai73 and M. otsurui,8 but no worms were
recovered.
He performed repeated infections of rats (2–3 times) and mice (2–6 times) with M. yokogawai at 4- to
30-day intervals and reported the following results: Acquired resistance was obtained in the reinfected
animals, and the resistance became stronger in accordance with the frequency of reinfection and the
lapse of the days after reinfection. Worm development was retarded in reinfected animals, particularly in
genital organs, and animals possessing immature worms got reinfected more easily, whereas those pos-
sessing fully developed worms got reinfected with a markedly lower incidence rate.110 He also notified a
tendency of changing parasitic locations from the upper part of the small intestine to the lower part of the
intestine.110 He further notified that the resistance could be acquired by an injection of emulsified worms
after refrigeration, drying, and powdering.110 The shifting of the parasitic locations from the upper part
of the small intestine to the lower part of the intestine was also observed in 1983 in dogs reinfected with
M. yokogawai.84
Chai et al.86 reported that the susceptibility of mice to M. yokogawai infection was highly dependent
on the immune status of the host. In immunocompetent ICR mice experimentally infected with M. yok-
ogawai, most of the worms were expelled spontaneously before day 7 after infection.86 However, ICR
mice immunosuppressed with prednisolone injection allowed a longer survival (until day 21 postinfec-
tion) of a greater number of worms than in immunocompetent control mice.86 Eosinophils were sug-
gested to have an important role to induce such protective immunity against M. yokogawai infection.86,126
Other effector mechanisms conferring protective immune responses of the host may include intestinal
intraepithelial lymphocytes (IEL), lamina propria lymphocytes (LPL), mucosal mast cells (MMC), and
goblet cells.1,2,4,11 The kinetic of intestinal IEL was studied by Chai et al.83; the IEL number was mark-
edly increased at day 5 postinfection, which decreased thereafter until day 24 postinfection and then
normalized. The day 5 postinfection was well corresponded with the time of active worm expulsion
from the host intestine.86 Moreover, the location of the IEL was moved to the apical portion of the villi
from the original basal or intermediate portion around the day 5 after infection.83 Mucosal mastocytosis
was observed throughout the infection period, from week 1 to 4, in rats,120 and, from week 1 to 3, in
mice,125 experimentally infected with M. yokogawai.120 Thus, MMC seemed also a significant factor
for inducing immunopathological effects and damages on the mucosal regions and finally, expulsion
of worms. Increased mucosal permeability was also suggested to be a factor related to the expulsion of
worms from mice.125
Antigenicity of body portions of M. yokogawai was studied using adult worms recovered from experi-
mental cats.105,106 Tegumental cells and tegumental syncytium as well as the intestinal content of the
worms were suggested to be the major antigens of M. yokogawai.105,106 However, a purified 100-kDa anti-
gen reacting to the tegumental and subtegumental layers of M. yokogawai appeared to be not a species-
specific antigen but a common antigen crossly recognizing even the liver fluke (Clonorchis sinensis),
lung fluke (Paragonimus westermani), and another intestinal fluke species (Gymnophalloides seoi).124
The antigenicity of M. yokogawai was generally stronger than that of M. miyatai as assessed by the lower
levels of the villus/crypt height ratio and the lowered expression patterns of the proliferating cell nuclear
antigen in the former.87 In addition, the parasitic location of M. yokogawai was generally at the upper
level of the small intestine in experimental hamsters, whereas that of M. miyatai was generally at the
middle level of the small intestine.70
An interesting observation of the worm posture within the ecological niche of M. yokogawai in the
dog’s small intestine was that the worms, during the early stage of infection (around day 3 postinfec-
tion), invade the slit of the Lieberkhün’s crypts by making their anterolateral bodies as one or more
reversible tube-like structures.122 Thereafter, until week 10 of observation, worms with such protruded
anterior bodies decreased continuously in number when most of the worms were floating on the top of
the villi.122 Another point of interest in host–parasite interactions includes the mechanisms of diarrhea
in metagonimiasis. It has been suggested that diarrhea is caused by excessive water content in the small
intestine of M. yokogawai-infected hosts.14,103,127 The excessive water content is considered to be due
to poor absorption of intestinal secretions resulting from mucosal inflammation in the affected small
intestine.103 The poor absorption of intestinal secretions is likely resulting from the reduced absorptive
surface due to the decreased villous height; however, this may be a reversible phenomenon.123 It was also
reported that the activities of brush border membrane-bound enzymes decreased in the small intestine of
M. yokogawai-infected mice and participated in the generation of diarrhea and malabsorption.127
758 Laboratory Models for Foodborne Infections
REFERENCES
1. Chai, J.Y., Intestinal flukes, in Food-Borne Parasitic Zoonoses: Fish and Plant-Borne Parasites,
Murrell, K.D. and Fried, B., Eds., Springer, New York, 2007.
2. Chai, J.Y., Shin, E.H., Lee, S.H., and Rim, H.J., Foodborne intestinal flukes in Southeast Asia, Korean
J. Parasitol., 47, S103, 2009.
3. Ito, J., Metagonimus and other human heterophyid trematodes, Progr. Med. Parasitol. Jpn., 1, 314, 1964.
4. Chai, J.Y., Metagonimus, in Biology of Foodborne Parasites, Xiao, L., Ryan, U. and Feng, Y., Eds., CRC
Press, Taylor & Francis Group, Boca Raton, FL, 427–443, 2007.
5. Suzuki, S., Yokogawa’s Metagonimus. List of publications on special animals in Okayama prefecture,
Okayama Prefectural Rep., 1, 146, 1930.
6. Katsuta, I., Studies on the metacercariae of Formosan brackish water fishes (2) On a new species,
Metagonimus minutus n. sp. parasitic in Mugil cephalus, Taiwan Iggakai Zasshi, 31, 26, 1932 (in Japanese).
7. Izumi, M., Studies on a new species of Metagonimus and its life cycle, Tokyo Iji Shinshi, 2929, 1224,
1935 (in Japanese).
8. Saito, S. and Shimizu, T., A new trematode, Metagonimus otsurui sp. nov. from the fresh-water fishes
(Trematoda: Heterophyidae), Jpn. J. Parasitol., 17, 167, 1968.
9. Saito, S., et al., Metagonimus miyatai sp. nov. (Digenea: Heterophyidae), a new intestinal trematode
transmitted by freshwater fishes in Japan and Korea, Korean J. Parasitol., 35, 223, 1997.
10. Shimazu, T. Metagonimus hakubaensis sp. n. (Digenea, heterophyidae) from Nagano, Japan: morphol-
ogy and life cycle, Bull. Nat. Sci. Mus., Tokyo Ser. A, 25, 87, 1999.
11. Chai, J.Y., Murrell, K.D., and Lymbery, A.J., Fish-borne parasitic zoonoses: status and issues, Int. J.
Parasitol., 35, 1233, 2005.
12. Yu, S.H. and Mott, K.E., Epidemiology and morbidity of food-borne intestinal trematode infections,
Trop. Dis. Bull., 91, R125, 1994.
13. Shimazu, T. and Urabe, M. Morphology and life cycle of Metagonimus otsurui (Digenea: Heterophyidae)
from Nara, Japan, Bull. Nat. Sci. Mus., Tokyo Ser. A, 28, 21, 2002.
14. Chai, J.Y., Study on Metagonimus yokogawai (Katsurada, 1912) in Korea V. Intestinal pathology in
experimentally infected albino rats, Seoul J. Med., 20, 104, 1979.
15. Chai, J.Y., Kim, J., and Lee, S.H., Invasion of Metagonimus yokogawai into the submucosal layer of the
small intestine of immunosuppressed mice, Korean J. Parasitol., 33, 313, 1995.
16. Yu, J.R. and Chai, J.Y., Metagonimus. In Molecular Detection of Human Parasitic Pathogens, Liu, D.
(Ed.), CRC Press, Boca Raton, FL, 2013.
17. Chai, J.Y. and Lee, S.H., Food-borne intestinal trematode infections in the Republic of Korea, Parasitol.
Int., 51, 129, 2002.
18. Cakiç, P., et al., Metagonimus yokogawai, a new parasitic trematoda species in ichthyoparasitofauna of
the Serbia, Acta Vet. (Beograd), 57, 537, 2007.
19. Nachev, N. and Sures, B., The endohelminth fauna of barbel (Barbus barbus) correlates with water qual-
ity of the Danube River in Bugaria, Parasitology, 136, 545, 2009.
20. Francová, K., et al., Parasite fauna of native and non-native populations of Neogobius melanosto-
mus (Pallas, 1814) (Gobiidae) in the longitudinal profile of the Danube River, J. Appl. Ichthyol., 27,
879, 2011.
21. Davydov, O.N., Lysenko, V.N., and Kurovskaya, L.Y., Species diversity of carp, Cyprinus carpio
(Cypriniformes, Cyprinidae), parasites in some cultivation regions, Vestn. Zool., 45, e9, 2011.
22. Djikanovic, V., et al. Parasitofauna of freshwater fishes in the Serbian open waters: a checklist of para-
sites of freshwater fishes in Serbian open waters, Rev. Fish Biol. Fish. 22, 297, doi:10.1007/s11160-011-
9226-6, 2012.
23. Chai, J.Y., et al., An epidemiological study of metagonimiasis along the upper reaches of the Namhan
River, Korean J. Parasitol., 31, 99, 1993.
24. Takahashi, S., On the life history of Metagonimus yokogawai, a new species of Metagonimus, and
Exorchis major, Okayama Igakkai Zasshi, 41, 2687, 1929 (in Japanese).
25. Asada, S., On Metagonimus yokogawai and its related species, Rinsho Igaku (Clin. Med.), 22, 43, 1934
(in Japanese).
26. Saito, S., On the differences between Metagonimus yokogawai and Metagonimus takahashii I. The
morphological comparisons, Jpn. J. Parasitol., 21, 449, 1972 (in Japanese).
760 Laboratory Models for Foodborne Infections
27. Saito, S., On the differences between Metagonimus yokogawai and Metagonimus takahashii II. The
experimental infections to the second intermediate hosts, Jpn. J. Parasitol., 22, 39, 1973 (in Japanese).
28. Koga, G., Studies on the genus Metagonimus, Igaku Kenkyu, 12, 3471, 1938 (in Japanese).
29. Chai, J.Y., et al., Three morphological types of the genus Metagonimus encysted in the dace, Tribolodon
taczanowskii, caught from the Sumjin River, Korean J. Parasitol., 29, 217, 1991.
30. Katsurada, F., On the genus Heterophyes in Japan, Okayama Igakkai Zasshi (no. 268), 373, 1912
(in Japanese).
31. Miyata, I., Comments on the classification of trematodes of the genus Metagonimus, Dobutsugaku
Zasshi, 56, 16, 1944 (in Japanese).
32. Yang, H.J., et al., Molecular differentiation of three species of Metagonimus by simple sequence repeat
anchored polymerase chain reaction (SSR-PCR) amplification, J. Parasitol., 86, 1170, 2000.
33. Yu, J.R., et al., PCR-RFLP pattern of three kinds of Metagonimus in Korea, Korean J. Parasitol., 35,
271, 1997.
34. Lee, S.U., et al., A cytogenetic study on human intestinal trematodes of the genus Metagonimus
(Digenea: Heterophyidae) in Korea, Korean J. Parasitol., 37, 237, 1999.
35. Oyamada, T., et al., Metagonimus otsurui metacercarial infection in a gobiid fish (Tridentiger brevispi-
nis) collected from Lake Ogawara in Aomori Prefecture, Japan, Jpn. J. Parasitol., 45, 275, 1996.
36. Cho, S.Y., Kang, S.Y., and Lee, J.B., Metagonimiasis in Korea, Arzneimittelforschung, 34(9B), 1211,
1984.
37. Shimazu, T., Life cycle and morphology of Metagonimus miyatai (Digenea: Heterophyidae) from
Nagano, Japan, Parasitol. Int., 51, 271, 2002.
38. Kim, C.H., et al., Studies on the Metagonimus fluke in the Daecheong reservoir and the upper stream of
Geum River, Korea, Korean J. Parasitol., 25, 69, 1987 (in Korean).
39. Kurokawa, T., Studies on the genus Metagonimus, especially on the life history of M. katsuradai Izumi,
1935 and determination of its first intermediate host, Tokyo Iji Shinshi, 3161, 2877, 1939 (in Japanese).
40. Besprozvannykh, V.V., et al., Occurrence of Metagonimus katsuradai Izumi, 1935 (Trematoda:
Heterophyidae) in the southern Primor’e region. In Gel’minty I vyzyvaemye imi zabollevaniya, Yu, L.
(Ed.), Mamaev, Vladivostock, Russia, 1987.
41. Ahn, Y.K., Lateolaborax japonicus, a role of the second intermediate host of Metagonimus yokogawai,
New Med. J., 26, 135, 1983 (in Korean).
42. Cho, S.Y., Kang, S.Y., and Ryang, Y.S., Helminthes infections in the small intestine of stray dogs in
Ejungbu City, Kyunggi Do, Korea, Korean J. Parasitol., 19, 55, 1981.
43. Seo, B.S., et al., Studies on parasitic helminths of Korea 5. Survey on intestinal trematodes of house rats,
Korean J. Parasitol., 19, 131, 1981.
44. Huh, S., Sohn, W.M., and Chai, J.Y., Intestinal parasites of cats purchased in Seoul, Korean J. Parasitol.,
31, 371, 1993.
45. Miyamoto, K., Studies on zoonoses in Hokkaido 7. Survey of natural definitive hosts of Metagonimus
yokogawai, Jpn. J. Parasitol., 34, 371, 1985 (in Japanese).
46. Li, M.H., Metagonimus yokogawai: metacercariae survey in fishes and its development to adult worms
in various rodents, Parasitol. Res., 112, 1647, 2013.
47. Li, M.H., et al., Infectivity and development of Metagonimus yokogawai in experimentally infected
domestic ducks, Vet. Parasitol., 168, 45, 2010.
48. Yamaguti, S., Digenetic trematodes of vertebrates, Systema Helminthum, Volume 1, Interscience
Publishers Inc., New York, 1958.
49. Kamiya, H., et al., Helminths of stray dogs in Sapporo. II, Jpn. J. Parasitol., 24, 41, 1975 (in Japanese).
50. Ahn, Y.K., Intestinal flukes of genus Metagonimus and their second intermediate hosts in Kangwon-do,
Korean J. Parasitol., 31, 331, 1993 (in Korean).
51. Guk, S.M., et al., Susceptibility of inbred mouse strains to infection with three species of Metagonimus
prevalent in the Republic of Korea, J. Parasitol., 91, 12, 2005.
52. Kim, C.H., Study on the Metagonimus sp. in Gum River basin, Chungchung-nam Do, Korea, Korean
J. Parasitol., 18, 215, 1980 (in Korean).
53. Ahn, Y.K. and Ryang, Y.S., Growth, egg-laying and life span of Miyata type of genus Metagonimus
(Trematoda: Heterophyidae) in the final hosts, J. Wonju Med. Coll., 8, 1, 1995 (in Korean).
54. Shimazu, T., Morphology of metacercariae and adults of Metagonimus katsuradai Izumi (Digenea,
Heterophyidae) from Shiga, Japan, Bull. Nat. Sci. Mus. Tokyo Ser. A, 29, 47, 2003.
Metagonimus 761
55. Kudo, N., et al., Experimental final hosts of Metagonimus hakubaensis (Trematoda: Heterophyidae) and
their susceptibility to the fluke, J. Vet. Med. Sci., 76, 1651, 2014.
56. Kudo, N., et al., Discovery of natural infection by Metagonimus hakubaensis Shimizu, 1999 (Trematoda:
Heterophyidae) in Japanese water shrews (Chimarrogalus platycephala) J. Vet. Med. Sci., 76, 1531, 2014.
57. Chai, J.Y., et al., High endemicity of Metagonimus yokogawai infection along residents of Samcheok-
shi, Kangwon-do, Korean J. Parasitol., 38, 33, 2000.
58. Chai, J.Y., et al., Heterophyid trematodes recovered from people residing along the Boseong River,
South Korea, Acta Trop., 148, 142, 2015.
59. Lee, J.J., et al., Decrease of Metagonimus yokogawai endemicity along the Tamjin River basin, Korean
J. Parasitol., 46, 289, 2008.
60. Center for Diseases Prevention and Control, Ministry of Health and Welfare (Korea), Statistics of intes-
tinal helminths nationwide (the 8th Report), Osong, Korea, pp. 1–210, 2013.
61. Cho, S.H., et al., Prevalence of Metagonimus metacercariae in sweetfish, Plecoglossus altivelis, from
eastern and southern coastal areas of Korea, Korean J. Parasitol., 49, 161, 2011.
62. Sohn, W.M. and Chai, J.Y., Infection status with helminthes in feral cats purchased from a market in
Busan, Republic of Korea, Korean J. Parasitol., 43, 93, 2005.
63. Shin, S.S., et al., Zoonotic intestinal trematodes in stray cats (Felis catus) from riverside areas of the
Republic of Korea, Korean J. Parasitol., 53, 209, 2015.
64. Seo, M., et al., Gymnophalloides seoi eggs from the stool of a 17th century female mummy found in
hadong, Republic of Korea, J. Parasitol., 94, 467, 2008.
65. Shin, D.H., et al., Scanning electron microscope study of ancient parasite eggs recovered from Korean
mummies of the Joseon Dynastry, J. Parasitol., 95, 137, 2009.
66. Kagei, N., Epidemiological studies of metagonimiasis in Japan. I. Epidemological survey of metagoni-
miasis among the Takatsu River, Shimane Prefecture, Bull. Inst. Public Health, Japan, 14, 213, 1965 (in
Japanese).
67. Yoshimura, S., et al., Epidemological studies of metagonmiasis in Chokai village, Akita Prefecture, Jpn.
J. Parasitol., 21, 400, 1972 (in Japanese).
68. Ito, J., et al., On the prevalence of Metagonimus sp. among the inhabitants at Hamamatsu basin in
Shizuoka Prefecture, Japan, Jpn. J. Parasitol., 40, 274, 1991.
69. Kagei, N. and Oshima, T., Nationwide epidemiological surveys on the metacercariae of Metagonimus
yokogawai in “ayu”, Plecoglossus altivelis, in Japan, Jpn. J. Parasitol., 17, 461, 1968 (in Japanese).
70. Kino, H. et al., Geographical distribution of Metagonimus yokogawai and M. miyatai in Shizuoka
Prefecture, Japan, and their site preferences in the sweetfish, Plecoglossus altivelis, and hamsters,
Parasitol. Int., 55, 201, 2006.
71. Jeon, H.K., et al., Human infections with liver and minute intestinal flukes in Guangxi, Chia: analysis
by DNA sequencing, ultrasonography, and immunoaffinity chromatography, Korean J. Parasitol., 50,
391, 2012.
72. Wang, C.R., et al., Prevalence of helminthes in adult dogs in Heilongjiang Province, the People’s
Republic of China, Parasitol. Res., 99, 627, 2006.
73. Sohn, W.M., et al., Fishborne trematode metacercariae in freshwater fish from Guangxi Zhuang
Autonomous Region, China, Korean J. Parasitol., 47, 249, 2009.
74. Chen, Y., et al., Investigation of snails transmitting parasitic diseases in Yunnan Province, J. Pathog.
Biol. (China), 4, 211, 2009 (in Chinese).
75. Ondračkova, M., et al., Shoreline distribution and parasitic infection of black-striped pipefish Syngnathus
abaster Risso, 1827 in the low River Danube, J. Appl. Ichthyol., 28, 590, 2012.
76. Chun, S.K., A study on the metacercaria of Metagonimus takahashii and Exorchis oviformis from
Carassius carassius, Bull. Pusan Fish Coll., 3, 31, 1960 (in Korean).
77. Ahn, Y.K. and Ryang, Y.S., Epidemiological studies on Metagonimus infection along the Hongcheon
river, Kangwon Province, Korean J. Parasitol., 26, 207, 1988 (in Korean).
78. Son, W.Y., Intestinal trematode infections in the villagers in Koje-myon, Kochang-gun, Kyongsangnam-do,
Korea, Korean J. Parasitol., 32, 149, 1994.
79. Kim, D.G., et al., Heterophyid metacercarial infections in brackish water fishes from Jinju-man (Bay),
Kyongsangnam-do, Korea, Korean J. Parasitol., 44, 7, 2006.
80. Urabe, M., Trematode fauna of prosobranch snails of the genus Semisulcospira in Lake Biwa and the
connected drainage system, Parasitol. Int., 52, 21, 2003.
762 Laboratory Models for Foodborne Infections
81. Park, M.S., et al., Intestinal parasite infections in the inhabitants along the Hantan River, Chorwon-gun,
Korean J. Parasitol., 31, 375, 1993.
82. Yu, J.R., Kwon, S.O., and Lee, S.H., Clonorchiasis and metagonimiasis in the inhabitants along
Talchongang (River), Chungwon-gun, Korean J. Parasitol., 32, 267, 1994.
83. Chai, J.Y., et al., Chronological observation on intestinal histopathology and intraepithelial lymphocytes
in the intestine of rats infected with Metagonimus yokogawai, Korean J. Parasitol., 32, 215, 1994 (in
Korean).
84. Kang, S.Y., et al., A study on intestinal lesions of experimentally reinfected dogs with Metagonimus
yokogawai. Korean J. Parasitol., 21, 58, 1983.
85. Chi, J.G., et al., Intestinal histopathology in human metagonimiasis with ultrastructural observations of
parasites, J. Korean Med. Sci., 3, 171, 1988.
86. Chai, J.Y., Seo, B.S., and Lee, S.H., Study on Metagonimus yokogawai (Katsurada, 1912) in Korea VII.
Susceptibility of various strains of mice to Metagonimus infection and effect of prednisolone, Korean J.
Parasitol., 22, 153, 1984.
87. Yu, J.R., Myong, N., and Chai, J.Y., Expression patterns of proliferating cell nuclear antigen in the small
intestine of mice infected with Metagonimus yokogawai and Metagonimus Miyata type, Korean J.
Parasitol., 35, 239, 1997.
88. Africa, C.M., de Leon, W., and Garcia, E.Y., Visceral complications in intestinal heterophyidiasis of
man. Acta Med. Philipp. Monogr. Ser., 1, 1–132, 1940.
89. Yamada, S.M., et al., A case of metagonimiasis complicated with multiple intracerebral hemorrhages
and diabetes mellitus, J. Nippon Med. Sch., 75, 32–35, 2008.
90. Lee, J.J., et al. Comparative morphology of minute intestinal fluke eggs that can occur in human stools
in the Republic of Korea, Korean J. Parasitol., 50, 207–213, 2012.
91. Lee, S.H., et al., Comparative morphology of eggs of heterophyids and Clonorchis sinensis causing
human infections in Korea, Korean J. Parasitol., 22, 171, 1984.
92. Ahn, Y.K., Epidemiological studies on Metagonimus yokogawai infection in Samcheok-gun,
Kangwon-do, Korea, Korean J. Parasitol., 22, 161, 1984 (in Korean).
93. Pyo, K.H., et al., Species identification of medically important trematodes in aquatic food samples using
PCR-RFLP targeting 18S rRNA, Foodborne Pathog. Dis., 10, 1, 2013.
94. Chai, J.Y., Praziquantel in treatment of trematode and cestode infections, Infect. Chemother., 45, 1,
2013.
95. Chai, J.Y., et al., Effects of gamma-irradiation on the survival and development of Metagonimus yok-
ogawai metacercariae in rats, Korean J. Parasitol., 33, 297, 1995.
96. Yokogawa, S., On a new trematode which takes Plecoglossus altivelis as second intermediate host-
Metagonimus, Tokyo Igakkai Zashii, 27(9), 685, 1913 (in Japanese).
97. Kobayashi, H., On a new genus of trematode (a preliminary report), Dobutsugaku Zasshi, 24(289), 1,
1912 (in Japanese).
98. Muto, S., Studies on human trematodes caused by their intermediate host, freshwater fish in Biwa-Lake,
especially on Metagonimus yokogawai from Cyprinidae, Nippon Shokakibyo Gakkai Zasshi, 16(2), 135,
1917 (in Japanese).
99. Kagei, N. and Kihata, M., On the development of Metagonimus yokogawai (Katsurada, 1912)
(Heterophyidae, Trematoda) in laboratory animals, Rep Natl Inst Public Hyg (Japan), 19(1), 48, 1970
(in Japanese).
100. Ahn, Y.K., Soh, C.T., and Lee, S.K., Egg laying capacity of Metagonimus yokogawai, Yonsei Rep. Trop.
Med., 12, 11, 1981.
101. Saito, S., On the synonymy of the trematodes of the genus Metagonimus, Report of Parasite Classification
and Morphology Seminar (Japan), 2, 1, 1984 (in Japanese).
102. Inatomi, S., et al., The ultrastructure of helminth 2. The body wall of Metagonimus takahashii Suzuki,
1930, Jpn. J. Parasitol., 17, 455, 1968 (in Japanese).
103. Lee, J.B., et al., Study on pathology of metagonimiasis in experimentally infected cat intestine, Korean
J. Parasitol., 19, 109, 1981.
104. Rho, I.H., et al., Observation on the pathogenesis of villous changes in early phase of experimental
metagonimiasis, Chung-Ang J. Med., 9, 67, 1984 (in Korean).
105. Ahn, H., Rim, H.J., and Kim, S.J., Antigenic localities in the tissues of Metagonimus yokogawai
observed by immunogold labeling method, Korean J. Parasitol., 29, 245, 1991 (in Korean).
Metagonimus 763
106. Rim, H.J., Kim, S.J., and Yang, M.G., Antigenic localities in the tissues of Metagonimus yokogawai in
the period of growth, Korean J. Parasitol., 30, 309, 1992 (in Korean).
107. Yokogawa, M. and Sano, M., Studies on the intestinal flukes IV. On the development of the worm in the
experimentally infected animals with metacercariae of Metagonimus yokogawai, Jpn. J. Parasitol., 17,
540, 1968 (in Japanese).
108. Miyamoto, K. and Kutsumi, H., Studies on zoonoses in Hokkaido, Japan 2. On the second intermediate
hosts of Metagonimus yokogawai in Asahikawa City, Kamikawa District, Jpn. J. Parasitol., 27, 445,
1978 (in Japanese).
109. Rim, H.J., Kim, K.H., and Joo, K.H., Classification of host specificity of Metagonimus spp. from Korean
freshwater fish, Korean J. Parasitol., 34, 7, 1996.
110. Gushima, M., Studies on the immunity of Metagonimus yokogawai, Igaku Kenkyu, 13, 1713, 1939 (in
Japanese).
111. Komiya, Y., Ito, J., and Yamamoto, S., Studies on Metagonimus yokogawai infected in a brackish water
fish Salangichthys microdon from the lake Kasumigaura, Jpn. J. Parasitol., 7, 7, 1958 (in Japanese).
112. Chun, S.K., A study on Metagonimus yokogawai from Plecoglossus altivelis in the Miryang River, Bull.
Pusan Fish Coll., 3, 24, 1960 (in Korean).
113. Hong, N. T. and Seo, B.S., Study on Metagonimus yokogawai (Katsurada, 1912) in Korea, Korean J.
Parasitol., 7, 129, 1969.
114. Lee, S.U., et al., Sequence comparisons of 28S ribosomal DNA and mitochondrial cytochrome c oxi-
dase subunit I of Metagonimus yokogawai, M. takahashii and M. miyatai, Korean J. Parasitol., 42, 129,
2004.
115. Lee, S.H., et al., Study on Metagonimus yokogawai (Katsurada, 1912) in Korea VII. Electron micro-
scopic observation on the tegumental structure, Korean J. Parasitol., 22, 1, 1984 (in Korean).
116. Chai, J.Y., et al., Surface ultrastructure of Metagonimus miyatai metacercariae and adults, Korean J.
Parasitol., 36, 217, 1998.
117. Chai, J.Y., et al., Surface ultrastructure of Metagonimus takahashii metacercariae and adults, Korean J.
Parasitol., 38, 9, 2000.
118. Yu, J.R., Chung, J.S., and Chai, J.Y., Different RAPD patterns between Metagonimus yokogawai and
Metagonimus Miyata type, Korean J. Parasitol., 35, 295, 1997.
119. Kogame, Y., On Metagonimus yokogawai ovatus (Kobayashi, 1913) found from Tribolodon hakonensis
in Daiseiji River, Ishikawa Prefecture, Japan, Tokyo Iji Shinshi, (3127), 793, 1939 (in Japanese).
120. Chai, J.Y., et al., Mucosal mast cell responses to experimental Metagonimus yokogawai infection in rats,
Korean J. Parasitol., 31, 129, 1993.
121. Hong, S.J., et al., Worm recovery and small intestinal lesions of albino rats coinfected with Fibricola
seoulensis and Metagonimus yokogawai, Korean J. Parasitol., 31, 109, 1993.
122. Jang, Y.K., et al., In situ posture of anterior body of Metagonimus yokogawai in experimentally infected
dog, Korean J. Parasitol., 23, 203, 1985.
123. Cho, S.Y., et al., A preliminary observation on water content of small intestine in Metagonimus
yokogawai infected dog, Korean J. Parasitol., 23, 175, 1985.
124. Han, E.T., et al., Metagonimus yokogawai: a 100-kDa somatic antigen commonly reacting with other
trematodes, Korean J. Parasitol., 52, 201, 2014.
125. Ohnishi, Y. and Taufen, M., Increases in the intestinal mucosa of mice infected with Metagonimus
yokogawai, Jpn. J. Vet. Sci., 46, 885, 1984.
126. Ohnishi, Y., Eosinophil responses in the mice infected with Metagonimus yokogawai, Jpn. J. Parasitol.,
36, 271, 1987.
127. Hong, S.T., et al., Activities of brush border membrane bound enzymes of the small intestine in
Metagonimus yokogawai infection in mice, Korean J. Parasitol., 29, 1, 1991.
128. Mehlhorn, H., et al., Ultrastructural investigations on the effects of praziquantel on human trematodes
from Asia: Clonorchis sinensis, Metagonimus yokogawai, Opisthorchis viverrini, Paragonimus wester-
mani and Schistosoma japonicum, Arzneimit-Forsch/Drug Res., 33, 91, 1983.
129. Hamajima, F., et al., Studies on the in vitro effects of bithionol and menichlopholan on flukes of
Clonorchis sinensis, Metagonimus takahashii and Paragonimus miyazakii, Int. J. Parasitol., 9, 241,
1979.
130. Yasuraoka, K. and Kojima, K., In vitro cultivation of the heterophyid trematode, Metagonimus
yokogawai, from the metacercaria to adult, Jpn. J. Parasitol., 23, 199, 1970.
47
Opisthorchis viverrini
Thidarut Boonmars
CONTENTS
47.1 Introduction................................................................................................................................... 765
47.2 Morphological Characteristics of O. viverrini.............................................................................. 765
47.3 Animal Models for O. viverrini.................................................................................................... 766
47.3.1 Establishment of O. viverrini Infection in Animal Models..............................................767
47.3.2 Gross Pathology of Hamster and Gerbil Liver..................................................................767
47.3.3 Pathology of Liver Fluke Infection in Hamster and Gerbil..............................................767
47.3.4 The Level of White Blood Cells in the Liver Fluke Infection in Hamsters..................... 768
47.3.5 Opisthorchiasis-Associated Cholangiocarcinoma............................................................ 769
47.4 Conclusion..................................................................................................................................... 770
References............................................................................................................................................... 771
47.1 Introduction
Opisthorchis viverrini is a trematode (commonly called liver fluke) that is classified in the family
Opisthorchiidae, order Opisthorchiata, subclass Digenea, class Trematoda, phylum Platyhelminthes,
kingdom Animalia (Figure 47.1). With a high prevalence in Southeast Asia, particularly Thailand, Lao
PDR, Cambodia, and Vietnam,1 this parasite lives in the hepatobiliary tract of humans, causing hepato-
biliary diseases such as cholangitis, lithiasis, gallstone, hepatitis, and also cholangiocarcinoma. Humans
normally acquire infection with this liver fluke by ingestion of uncooked cyprinoid fish that contains or
is contaminated by the infective stage (namely “metacercaria”) of O. viverrini. After ingestion, the cyst
wall of metacercaria is digested by gastric juice and duodenum juice, and the excysted juvenile migrates
to the biliary tract and grows to adult in the hepatobiliary tract where it dwells (Figure 47.2). This infec-
tion (often called “opisthorchiasis” or “opisthorchiosis”) induces many changes in hepatobiliary tract,
such as dilatation of the bile ducts with hyperplasia, desquamation, proliferation of the bile duct epithe-
lial cells, glandular formation, and fibrous connective tissue infiltration of the walls, and other complica-
tion of hepatobiliary diseases (e.g., cholangitis, cystitis, cholecystitis including cholangiocarcinoma).2–4
Besides O. viverrini, which mainly occurs in Thailand, Lao PDR, Vietnam, and Cambodia,1 Clonorchis
sinensis and Opisthorchis felineus within the family Opisthorchiidae also cause liver fluke infections in
humans, dogs, and cats. While C. sinensis infection appears in China, Korea, Japan, and Vietnam,5
O. felineus infection is present in Russia and European countries.6
765
766 Laboratory Models for Foodborne Infections
Oral sucker
Ventral sucker
Vitelline gland
Ovary
Testes
1.Ingestion of metacercaria
Adult
2.Metacercaria is
excysted and then
4.Cercaria invade to junvenile migrate to
cyprinod fish and bile ducts and gallbladder
develop to
metacercaria
columns (Figure 47.1). In vivo study showed that O. viverrini in different hosts show varied shape and
size.7 Rat and mouse are resistant to O. viverrini, whereas hamster and gerbil are susceptible to this
parasite. In general, worm recovery from hamster is higher than that from the gerbil, and the recovered
worm from gerbil is larger than that from hamster. The worm size correlates to the size of the reproduc-
tive organs (testes, uterus, and vitelline gland), eggs per worm, and worms per gram of feces as shown
in Figure 47.3 and Table 47.1.8
(A) (B)
FIGURE 47.3 The representative adult of O. viverrini from hamster (A) and gerbil (B) at 1.5 month.
TABLE 47.1
The O. viverrini Infection in Animal Models
Animal Number of Size of Adult Worm
Model Metacercariae (Length × Height) in mm % Infection References
Hamster 50 1.43 ± 0.08 × 4.4 ± 0.3 28 ± 9.8 7
Gerbil 50 1.81 ± 0.06 × 5 ± 0.12 60.5 ± 14.44
Mouse 50 — 0 8
Rat 50 — 0
Cyprinoid fishes Fishes digestion OV-Metacercariae collection Hamster were orally infected
50 OV-metacercariae
FIGURE 47.5 Representative gross liver pathology of the infected hamster (A–C) and gerbil (D–F) at 30 days (A,D),
60 days (B,E), and 90 days (C,F). Gall bladder (G); common bile duct (arrow). Note: Liver obstruction and cirrhosis could
be observed at all time points.
FIGURE 47.6 Representative liver pathology of the infected hamster (A–C) and gerbil (D–F) at 30 days (A,D), 60 days
(B,E), and 90 days (C,F). Parasite (P). Note: Fibrosis could be observed in infected gerbil at all time points.
bile ducts appears and thickens in line with the duration of the infection, which corresponds to gross
pathological bile ducts (Figure 47.7).
47.3.4 The Level of White Blood Cells in the Liver Fluke Infection in Hamsters
The levels of white blood cells in infected hamster in the first period (<2 months postinfection) are signif-
icantly increased in comparison with that in the uninfected hamster (Table 47.2). Among various types
of white blood cells, lymphocytes are higher in all phases of the infection. Neutrophils significantly
increase at 6 months postinfection. Eosinophils peak at the first period of ~1 month and decrease at
Opisthorchis viverrini 769
TABLE 47.2
Blood Cell Counts in Uninfected/Infected Hamster
Estimated White Blood Cell Count (Months Postinfection)
1 2 3 6
Experimental
Group Mean ± SEM Mean ± SEM Mean ± SEM Mean ± SEM
Total white blood Normal 2040.0 ± 160.0 2012.5 ± 173.7 1800 ± 115.5 1266.7 ± 176.4
cell count OV 2600.0 ± 739.4 2960.0 ± 865.8 2850.0 ± 895.8 2250.0 ± 639.7
Neutrophil Normal 32.0 ± 3.3 26.7 ± 2.1 25.7 ± 5.4 23.9 ± 2.0
OV 42.2 ± 4.7 20.9 ± 1.8 31.5 ± 6.7 33.5 ± 2.8*
Monocyte Normal 2.8 ± 1.0 4.5 ± 1.0 3.8 ± 1.9 0.0 ± 0.0
OV 17.3 ± 1.6* 21.0 ± 3.5* 14.0 ± 2.4* 0.5 ± 0.5
Eosinophil Normal 0.0 ± 0.0 0.0 ± 0.0 0.0 ± 0.0 0.0 ± 0.0
OV 3.8 ± 1.6* 1.2 ± 0.5* 1.0 ± 0.6 0.0 ± 0.0
Lymphocyte Normal 65.2 ± 4.0 68.8 ± 1.6 72.0 ± 4.0 76.1 ± 2.0
OV 35.0 ± 5.0* 56.9 ± 4.4* 54.0 ± 5.1* 66.0 ± 2.9*
*p = 0.05. OV, O. viverrini.
2 months postinfection. Monocytes are significantly higher during the first 1–3 months and s ignificantly
decrease at 6 months postinfection.
47.3.5 Opisthorchiasis-Associated Cholangiocarcinoma
Cholangiocarcinoma (CCA) is defined as a carcinoma arising from bile duct epithelium within the
liver, from the large hilar duct and the extrahepatic duct. CCA may arise anywhere in the biliary tree,
but the most frequent sites are the ampulla of Vater, lower end of the common bile duct, hepatic duct, and
the junction of hepatic duct to the common bile duct. CCA is usually small, extending for 1–2 cm along
the duct, thickening the of the duct.12 CCA is a relatively rare cancer in the western world, with the inci-
dence of 1–2 cases per 100,000 people13 but rising worldwide over the past several decades. Recent epi-
demiological studies revealed that the incidence of CCA has increased in the United States, the United
Kingdom, and Australia.14,15 The prevalence of CCA is higher in Thailand, Loa PDR, and Cambodia
770 Laboratory Models for Foodborne Infections
where opisthorchiasis is endemic.3,16 CCA is a crucial public health problem in the Northeastern part of
Thailand. The incidence of CCA is approximately >50% of all liver cancer in Thailand and ~90% of all
liver cancer in the northeastern part of Thailand.17 This incidence rate is the highest in the world.18 It is
well known that both liver flukes O. viverrini and C. sinensis have been classified as class I carcinogens
by the International Agency for Research on Cancer owing to their association with CCA and hepato-
carcinoma in humans.3,19,20
The risk factors for CCA in northeastern part of Thailand was a clear association with past or pres-
ent infection with O. viverrini, as indicated by raised serum antibodies, and at least two-thirds of cases
can be attributed to this cause, and males may be at a higher risk than females.21,22 CCA patients who
have been infected with a liver fluke develop this cancer, and patients who had been infected and treated
for opisthorchiasis have increased risk of developing CCA than the noninfected population.23 Several
experiments and clinical reports revealed the correlation between O. viverrini and CCA development,
including clinical studies3,19,20,21,24 and experimental models.25–28
The mechanism of O. viverrini-associated CCA relates to chronic inflammation resulting in a
combination of mechanical damage, parasite secretions, and immunopathology.28 The primary
pathological change in epithelial desquamation may cause mechanical irritation by the liver fluke
and/or produce metabolic products from the parasite. The mechanical damage is caused by oral
sucker and ventral sucker of fluke during activities of feeding and migrating in the bile duct. The
fluke eggs are trapped in the lumen of the bile duct, leading to ulcerates, granulomatous inflamma-
tion, and cholelithiasis.29 The excretory-secretory (ES) products released by O. viverrini cause cell
proliferation.30,31 After infection for 2 weeks, the bile duct epithelium hyperplasia and periductal
inflammatory cells, predominantly eosinophils and lymphocytes, are observed. Furthermore, immu-
nopathological processes lead to the long-standing hepatobiliary damage. Host immune response can
induce the inflammatory cell infiltrations surrounding the hepatic bile ducts for killing the parasite
by reactive oxygen species (ROS) and reactive nitrogen species (RNS).32 ROS not only destroy para-
site, but also destroy host tissue and DNA leading to DNA fragmentation, which is well known during
apoptosis. Moreover, RNS, which are produced during inflammation, may play a role in initiation
and subsequent modulation stage of CCA development through DNA mutation leading to cancer
development.33,34 In O. viverrini-associated CCA, Sripa and Kaewkes found O. viverrini antigens
located in epithelium bile ducts in hamster model, and Thuwajit et al.30,31 reported that the ES prod-
ucts of O. viverrini increased NIH3T3 cell proliferations, which may enhance bile duct proliferation
and account for nitrative and oxidative DNA damage in hamsters infected with O. viverrini.34 The
development of cholangiocarcinogenesis involves many changes of cancer-relevant genes. Several
studies on the molecular mechanisms of O. viverrini-associated CCA have highlighted the potential
influences of KRAS and TP53 mutations in a large number of biological processes, including hamster
CCA,35 interleukin 6 (IL-6),36 transforming growth factor-β (TGF-β), IL-8, tumor necrosis factor-α
(TNF-α) and platelet-derived growth factor (PDGF),37 proteomic profiling of CCA,38 Rb1 and related
genes in CCA,39 comparative protein expression profiles (MUC5AC, Akt2, CK8, annexin II, and
VEGF A) of hilar and peripheral hepatic CCA,40 MMP-7 and MMP-9 in CCA,41 p38delta/MAPK13
as a diagnostic marker for CCA,42 and c-ski and related genes in CCA.43 However, infection with O.
viverrini alone may not produce CCA, which requires a combination of factors such as O. viverrini
infection, nitrosamine, host immune response, telomerase, alcohol, and hepatobiliary disease.
47.4 Conclusion
Opisthorchis viverrini is a liver fluke living in the hepatobiliary tract that causes opisthorchiasis
in human and animal reservoir host. The highest prevalence of this disease was found in Southeast
Asia, especially in Thailand, Cambodia, and Lao PDR. Chronic infection with O. viverrini enhances
the hepatobiliary diseases including CCA, which is a rare cancer but a highly fatal disease in the
world. However, the highest prevalence of CCA is in northeastern Thailand, which correlates to
the O. viverrini infection and other combination factors. To date, one effective anthelmintic drug
Opisthorchis viverrini 771
is praziquantel, but the CCA remains high. Therefore, use of animal models for the investigation
of O. viverrini pathogenesis is important. Syrian hamster is the best animal model for the study of
O. viverrini infection and CCA.
REFERENCES
1. WHO. Control of food borne trematode infections. WHO Tech Rep Ser 849, 1–157 (1995).
2. Srivatanakul, P. et al. Opisthorchis viverrini infestation and endogenous nitrosamines as risk factors for
cholangiocarcinoma in Thailand. Int J Cancer 48, 821–5 (1991).
3. IARC. Infection with liver flukes (Opisthorchis viverrini, Opisthorchis felineus and Clonorchis
sinensis). IARC Monogr Eval Carcinog Risks Hum 61, 121–75 (1994).
4. Sriamporn, S., Parkin, D.M., Pisani, P., Suwanrungruang, K. & Pengsaa, P. Behavioural risk factors for
cervical cancer from a prospective study in Khon Kaen, Northeast Thailand. Cancer Detect Prev 28,
334–9 (2004).
5. Rim, H.J. et al. Fishborne trematode metacercariae detected in freshwater fish from Vientiane
Municipality and Savannakhet Province, Lao PDR. Korean J Parasitol 46, 253–60 (2008).
6. Pozio, E., Armignacco, O., Ferri, F. & Gomez Morales, M.A. Opisthorchis felineus, an emerging
infection in Italy and its implication for the European Union. Acta Trop 126, 54–62 (2013).
7. Boonmars, T., Boonjaraspinyo, S. & Kaewsamut, B. Animal models for Opisthorchis viverrini infection.
Parasitol Res 104, 701–3 (2009).
8. Wonkchalee, O. et al. Comparative studies on animal models for Opisthorchis viverrini infection: host
interaction through susceptibility and pathology. Parasitol Res 110, 1213–23 (2012).
9. Wonkchalee, O. et al. Opisthorchis viverrini infection causes liver and biliary cirrhosis in gerbils.
Parasitol Res 109, 545–51 (2011).
10. Juasook, A. et al. Immunosuppressive prednisolone enhances early cholangiocarcinoma in Syrian ham-
sters with liver fluke infection and administration of N-nitrosodimethylamine. Pathol Oncol Res 19,
55–62 (2013).
11. Sriraj, P. et al. Does a combination of opisthorchiasis and ethyl alcohol consumption enhance early
cholangiofibrosis, the risk of cholangiocarcinoma? Parasitol Res 112, 2971–81 (2013).
12. Gatto, M. & Alvaro, D. Cholangiocarcinoma: risk factors and clinical presentation. Eur Rev Med
Pharmacol Sci 14, 363–7 (2010).
13. Landis, S.H., Murray, T., Bolden, S. & Wingo, P.A. Cancer statistics, 1998. CA Cancer J Clin 48, 6–29
(1998).
14. Patel, T. Increasing incidence and mortality of primary intrahepatic cholangiocarcinoma in the United
States. Hepatology 33, 1353–7 (2001).
15. Gores, G.J. A spotlight on cholangiocarcinoma. Gastroenterology 125, 1536–8 (2003).
16. Sithithaworn, P. & Haswell-Elkins, M. Epidemiology of Opisthorchis viverrini. Acta Trop 88, 187–94
(2003).
17. Srivatanakul, P., Sriplung, H. & Deerasamee, S. Epidemiology of liver cancer: an overview. Asian Pac
J Cancer Prev 5, 118–25 (2004).
18. Vatanasapt, V. et al. Cancer incidence in Thailand, 1988–1991. Cancer Epidemiol Biomarkers Prev 4,
475–83 (1995).
19. de Martel, C., Plummer, M. & Franceschi, S. Cholangiocarcinoma: descriptive epidemiology and risk
factors. Gastroenterol Clin Biol 34, 173–80 (2010).
20. Fried, B., Reddy, A. & Mayer, D. Helminths in human carcinogenesis. Cancer Lett 305, 239–49
(2011).
21. Parkin, D.M. et al. Liver cancer in Thailand. I. A case-control study of cholangiocarcinoma. Int J Cancer
48, 323–8 (1991).
22. Srivatanakul, P. et al. Endogenous nitrosamines and liver fluke as risk factors for cholangiocarcinoma in
Thailand. IARC Sci Publ 105, 88–95 (1991).
23. Haswell-Elkins, M.R., Satarug, S. & Elkins, D.B. Opisthorchis viverrini infection in northeast Thailand
and its relationship to cholangiocarcinoma. J Gastroenterol Hepatol 7, 538–48 (1992).
24. Kurathong, S. et al. Opisthorchis viverrini infection and cholangiocarcinoma. A prospective,
case-controlled study. Gastroenterology 89, 151–6 (1985).
772 Laboratory Models for Foodborne Infections
25. Thamavit, W., Kongkanuntn, R., Tiwawech, D. & Moore, M.A. Level of Opisthorchis infestation and
carcinogen dose-dependence of cholangiocarcinoma induction in Syrian golden hamsters. Virchows
Arch B Cell Pathol Incl Mol Pathol 54, 52–8 (1987).
26. Thamavit, W. et al. Promotion of cholangiocarcinogenesis in the hamster liver by bile duct ligation after
dimethylnitrosamine initiation. Carcinogenesis 14, 2415–7 (1993).
27. Thamavit, W., Pairojkul, C., Tiwawech, D., Shirai, T. & Ito, N. Strong promoting effect of Opisthorchis
viverrini infection on dimethylnitrosamine-initiated hamster liver. Cancer Lett 78, 121–5 (1994).
28. Sripa, B. et al. Liver fluke induces cholangiocarcinoma. PLoS Med 4, e201 (2007).
29. Sripa, B. Pathobiology of opisthorchiasis: an update. Acta Trop 88, 209–20 (2003).
30. Thuwajit, C. et al. Increased cell proliferation of mouse fibroblast NIH-3T3 in vitro induced by excretory/
secretory product(s) from Opisthorchis viverrini. Parasitology 129, 455–64 (2004).
31. Thuwajit, C. et al. Gene expression profiling defined pathways correlated with fibroblast cell proliferation
induced by Opisthorchis viverrini excretory/secretory product. World J Gastroenterol 12, 3585–92
(2006).
32. Coussens, L.M. & Werb, Z. Inflammation and cancer. Nature 420, 860–7 (2002).
33. Ohshima, H. et al. Increased nitrosamine and nitrate biosynthesis mediated by nitric oxide synthase
induced in hamsters infected with liver fluke (Opisthorchis viverrini). Carcinogenesis 15, 271–5 (1994).
34. Pinlaor, S. et al. Mechanism of NO-mediated oxidative and nitrative DNA damage in hamsters infected
with Opisthorchis viverrini: a model of inflammation-mediated carcinogenesis. Nitric Oxide 11, 175–83
(2004).
35. Tangkawattana, S., Kaewkes, S., Pairojkul, C., Tangkawattana, P. & Sripa, B. Mutations of KRAS and
TP53 in a minor proportion of Opisthorchis viverrini-associated cholangiocarcinomas in a hamster
model. Asian Pac J Cancer Prev 9, 101–6 (2008).
36. Sripa, B. et al. Advanced periductal fibrosis from infection with the carcinogenic human liver fluke
Opisthorchis viverrini correlates with elevated levels of interleukin-6. Hepatology 50, 1273–81 (2009).
37. Berthiaume, E.P. & Wands, J. The molecular pathogenesis of cholangiocarcinoma. Semin Liver Dis 24,
127–37 (2004).
38. Scarlett, C.J. et al. Proteomic classification of pancreatic adenocarcinoma tissue using protein chip
technology. Gastroenterology 130, 1670–8 (2006).
39. Boonmars, T. et al. Alterations of gene expression of RB pathway in Opisthorchis viverrini infection-
induced cholangiocarcinoma. Parasitol Res 105, 1273–81 (2009).
40. Guedj, N. et al. Comparative protein expression profiles of hilar and peripheral hepatic cholangiocarci-
nomas. J Hepatol 51, 93–101 (2009).
41. Leelawat, K., Sakchinabut, S., Narong, S. & Wannaprasert, J. Detection of serum MMP-7 and MMP-9
in cholangiocarcinoma patients: evaluation of diagnostic accuracy. BMC Gastroenterol 9, 30 (2009).
42. Tan, F.L. et al. p38delta/MAPK13 as a diagnostic marker for cholangiocarcinoma and its involvement
in cell motility and invasion. Int J Cancer 126, 2353–61 (2010).
43. Boonmars, T. et al. Involvement of c-Ski oncoprotein in carcinogenesis of cholangiocacinoma induced
by Opisthorchis viverrini and N-nitrosodimethylamine. Pathol Oncol Res 17, 219–27 (2011).
48
Paragonimus
Dongyou Liu
CONTENTS
48.1 Introduction................................................................................................................................... 773
48.1.1 Classification and Morphology..........................................................................................774
48.1.1.1 Classification......................................................................................................774
48.1.1.2 Morphology........................................................................................................774
48.1.2 Life Cycle and Epidemiology............................................................................................774
48.1.3 Clinical Features...............................................................................................................775
48.1.3.1 Acute Paragonimiasis (Early-Stage Disease)....................................................775
48.1.3.2 Chronic Pleuropulmonary Paragonimiasis (Late-Stage Disease).................... 776
48.1.3.3 Ectopic Paragonimiasis..................................................................................... 776
48.1.4 Diagnosis.......................................................................................................................... 776
48.1.5 Treatment and Prevention................................................................................................. 777
48.2 Laboratory Models........................................................................................................................ 777
48.2.1 Animal Models................................................................................................................. 777
48.2.1.1 Rodents............................................................................................................. 778
48.2.1.2 Cats................................................................................................................... 778
48.2.1.3 Dogs.................................................................................................................. 778
48.2.2 In Vitro Cultures............................................................................................................... 778
48.3 Conclusion..................................................................................................................................... 779
References............................................................................................................................................... 779
48.1 Introduction
The genus Paragonimus covers a large group of flatworms that are associated with parenchymal and/or
pleural lung infections (thus collectively known as lung flukes). Although Naterer made initial obser-
vation of lung flukes in 1828, it was Coenraad Kerbert who first described Paragonimus westermani
(originally named Distoma westermani) infection in a Bengal tiger suffering from pneumonia at the
Amsterdam zoological gardens in 1878. In the subsequent year, B. S. Ringer identified P. wester-
mani in the lungs of a human patient who died of an aortic aneurysm, thus confirming the first case
of human paragonimiasis. Following the establishment of the genus Paragonimus (“para”—Greek
word meaning “on the side of” and “gonimos”—Greek word meaning “gonads or genitalia”) by
Max Braun in 1899, in which P. westermani was the type species, much work has been done toward
the identification of many other species in this genus, including the independent, near-simultaneous
discovery of the only North American Paragonimus species (P. kellicotti) by Henry Ward and D. S.
Kellicott in 1894 [1,2].
773
774 Laboratory Models for Foodborne Infections
48.1.1.2 Morphology
Paragonimus adult worm has an oval- shaped body (of 7.5–12 mm in length, 4–6 mm in width, and 3.5–5 mm
in thickness), composed of smooth muscle that is protected by a thick tegument, which is in turn covered
with variably scattered spines. Both the oral terminal sucker (0.19 mm in diameter) and ventral sucker
(or acetabulum, 0.12 mm in diameter) are round and muscular. The lobed ovary is located behind the
acetabulum and anterior to the two branched testes. Between the ovary and the acetabulum are the semi-
nal receptacle, the tightly coiled uterus, and the thick-walled terminal part (metraterm). The digestive
system comprises a truncated pharynx and esophagus that bifurcates early into paired ceca [17].
Paragonimus eggs (80–120 × 45–70 μm) are ovoid, thick-shelled (measuring 2–4 μm in thickness),
golden brown, and operculate (showing a shoulder- or ridge-like structure). The eggs are produced by
self-fertilization (hermaphrodites), and more often by cross-fertilization in lung or pleura-encysted pairs.
Following release into bronchioles, the eggs are coughed up along with sputum, and then either expecto-
rated or swallowed for later discharge in feces.
Morphological features useful for the identification of Paragonimus adult worm include size and
shape, the patterns of lobation of the ovary and testes, and the appearance of the tegumental spines.
For example, the P. kellicotti group has tegumental spines arranged in groups, whereas the P. africanus
group shows individual spines. Microscopic observation of the thick shell-walled, unembryonated, oper-
culated eggs within sputum or feces is a clear indication of paragonimiasis, since other helminthic para-
sites that produce operculate eggs (e.g., trematodes Clonorchis sinensis, Opisthorchis species, Fasciola
hepatica, and Fasciolopsis buski, as well as cestode Diphyllobothrium latum) do not cause pulmonary
infections. In addition, the morphological features of Paragonimus adult, cercariae, metacercariae, and
metecercarial cyst may also be exploited for differentiation from other trematodes [17].
of the snail) to become infective cercariae (with anterior stylets and short tails). The infective cercariae
released by snails into freshwater (or infected snails eaten by freshwater crustaceans) penetrate the gills
and other soft tissue sites on the exoskeletons of crustaceans (the second intermediate host) and develop
into metacercariae [18].
Following consumption of raw, undercooked, or alcohol-pickled freshwater crustaceans (crabs or cray-
fish or undercooked infected tissues from a paratenic/nonpermissive mammalian host such as rats, pigs,
and possibly water birds), the permissive mammalian definitive host acquires the metacercariae, which
move from the intestine into the abdomen and the lungs [19]. In the lungs, the metacercariae mature into
adult worm, encyst (being spherical to ovoid in shape and up to 2 cm in diameter, each containing two or
more adult flukes or a diploid or triploid variant as in the exceptional case of P. westermani), and cross-
fertilize each other. With the eventual rupture of the cyst in the lungs, the eggs are released and coughed
up or swallowed and excreted in the feces for the next cycle of infection.
The first molluscan intermediate hosts of Paragonimus species include 54 species of freshwa-
ter snails from the families Pleuroceridae and Thiaridae of the superfamilies Cerithioidea and
Rissooidea; the second crustacean intermediate hosts consist of freshwater crabs or crayfish (belong-
ing to 53 species of 21 genera); and the definitive hosts are omnivorous and carnivorous crustacean-
eating mammals such as dogs, cats, tigers, and humans [20]. In the case of P. kellicotti, besides
domestic dogs and cats, wild animals [including skunks (Melphitis mephitis), red foxes (Vulpes
vulpes), coyotes (Canis latrans), mink (Mustela vison), and bobcats (Felis rufus)] also function as
competent definitive hosts.
Reflecting the natural distribution of permissive definitive and intermediate hosts that support infec-
tion and the customs of consuming raw or undercooked seafood or wild boar or using raw seafood
preparation as folk medicine, Paragonimus species have an endemic presence in East and South Asia
(e.g., China, Korea, Japan, Laos, Thailand, Vietnam, Malaysia, and the Philippines), sub-Saharan Africa
(e.g., Cameroon), and in the Americas (from Peru to Canada). It is of interest to note that P. westermani
and other Asian species have been traditionally referred to as Asian lung flukes, and P. kellicotti and
Latin American species have been called American lung flukes [11,21]. Worldwide, about 293 million
people are at risk of Paragonimus infection, and nearly 20 million people (of both sexes and all age) are
being infected [2].
48.1.3 Clinical Features
In comparison with other taxonomically related flukes in the class Trematoda (intestinal fluks, liver
flukes, and blood flukes or schistosomes), lung flukes (formerly oriental lung fluke, pulmonary disto-
matosis, and benign endemic hemoptysis) of the genus Paragonimus demonstrate a unique tropism in
their definitive hosts for encysting within the parenchyma or on the pleural surfaces of the lungs, and
occasionally in ectopic sites such as the brain (cerebral paragonimiasis), mesentery and visceral organs,
abdominal wall, muscles, and subcutaneous tissues (trematode larva migrans). Thus, human paragoni-
miasis may be categorized as acute paragonimiasis (early-stage disease), chronic pleuropulmonary para-
gonimiasis (late-stage disease), and ectopic paragonimiasis (with the parasite in a location other than the
lungs).
48.1.3.3 Ectopic Paragonimiasis
Besides the pleurae and lungs, Paragonimus parasites may migrate to other locations (e.g., the brain,
skin, breast, adrenal gland, heart, mediastinum, and genital organs), especially during heavy infections.
More common in children than in adults, cerebral paragonimiasis often produces two manifestations:
an expansive, space-occupying lesion in the brain (particularly the cerebral cortex) and a meningitis or
meningoencephalitis due to the migration of the worm, with common clinical signs ranging from head-
ache, vomiting, seizures, personality changes, decline of cognitive function, coma, to death (through
herniation caused by increased intracranial pressure). The next common site for ectopic lesions of para-
gonimiasis is the skin, resulting in multifocal skin lesions (e.g., subcutaneous nodules).
The pathogenic mechanisms of paragonimiasis largely reflect the host’s immune responses to
Paragonimus worms and eggs that enter and migrate from the intestines to the lungs, causing edema,
effusion, and subsequently fibrosis that entraps and restricts the lungs, contributing to the associated
signs and symptoms (e.g., shortness of breath). While nodules most likely contain immature cysts,
the mature cysts harboring the adult worms may gradually become fibrotic and die. The cysts with
dead worms degenerate, form scars that hold residual eggs entrapped in the fibrous tissue, and become
calcified.
48.1.4 Diagnosis
Given its largely nonspecific symptoms, clinical diagnosis of paragonimiasis based on clinical observa-
tions, radiological findings, dietary history, and travel history is insufficient and inconclusive [23,24].
Therefore, several conventional laboratory procedures have been routinely used to confirm the pres-
ence of Paragonimus eggs spp. in sputum (through alkaline decontamination and centrifuge sedimenta-
tion) and/or feces (through formalin–ether concentration). Considering that microscopic examination of
respiratory and/or stool specimens has limited sensitivity (30%–40% for a single sputum specimen and
54%–89% for multiple sputum specimens in comparison to 11%–15% for a single stool specimen and
about 25% for three stool specimens) and relatively low specificity, serological tests (e.g., ELISA for IgG
in serum and/or pleural effusion fluid) are helpful when clinical suspicion for paragonimiasis is high
Paragonimus 777
but Paragonimus eggs are not detected [14,25]. In addition, molecular methods based on nucleic acid
amplification have been applied in recent years to further enhance the sensitivity, specificity, and speed
of laboratory diagnosis of paragonimiasis [26–30].
Among various nucleic acid amplification techniques that have been developed to date, polymerase
chain reaction (PCR) has been a standout due to its unprecedented sensitivity, specificity, speed, and
robustness. In a relatively short period since its inception, PCR (and its variants such as conventional
PCR, random amplified polymorphic DNA-PCR, multiplex PCR, and real-time PCR) has proven to be
highly valuable for improved detection and/or species discrimination of Paragonimus parasites directly
from clinical samples (e.g., lung biopsy specimens, bronchoalveolar lavage fluid, sputum, pleural fluid,
or feces) [31]. The commonly used genetic targets for Paragonimus diagnosis include the internal tran-
scribed spacer (ITS) regions of ribosomal genetic complexes, particularly ITS2, and the mitochondrial
cytochrome c oxidase gene (CO1) [32–36].
Sugiyama et al. [37] established PCR-restriction fragment length polymorphism (RFLP) and spe-
cific PCR with primers from the second internal transcribed spacer (ITS2) region of ribosomal DNA
to discriminate the metacercariae of the lung flukes, P. westermani and P. miyazaki, both of which
are found in the same freshwater crab species and are morphologically similar. In addition, Sugiyama
et al. [38] successfully applied primers from the second internal transcribed spacer (ITS2) region of the
nuclear ribosomal DNA in PCR-RFLP and multiplex PCR for precise differentiation between individual
metacercariae of P. heterotremus and P. westermani that occur in Thailand. Using a similar approach,
Sugiyama et al. [39] developed multiplex PCR and PCR-RFLP (with ScrFI) that enabled differentia-
tion of metacercaria from five Paragoniumus species occurring in Thailand, including P. heterotremus
(ca. 310 and 520 bp), P. westermani (ca. 140 and 520 bp), P. siamensis (520 bp), P. bangkokensis (520 bp),
and P. harinasutai (520 bp). Digestion of the 520 bp products with restriction enzyme ScrFI yielded three
bands (ca. 60, 210, and 250 bp) for P. harinasutai, two bands (ca. 250 and 270 bp) for P. bangkokensis,
and an uncut band (520 bp) for P. siamensis. In a more recent study, Tantrawatpan et al. [40] described a
real-time fluorescence resonance energy transfer PCR (real-time FRET PCR) with melting curve analy-
sis for specific and sensitive detection of P. heterotremus eggs in the feces of infected cats, with the
detection limit of 10(–3) ng of P. heterotremus genomic DNA or 10 eggs of P. heterotremus per gram of
cat feces.
48.2 Laboratory Models
48.2.1 Animal Models
A number of laboratory animals have been attempted as experimental models for Paragonimus spe-
cies, including rodents (rats, mice, Syrian hamsters, and Mongolian gerbils), dogs, and cats, with varied
success [43]. While dogs and cats, especially puppies and kittens, are susceptible to most Paragonimus
species prevalent in Asia, rats and mice serve well as paratenic hosts when infected naturally or experi-
mentally with some Paragonimus species.
778 Laboratory Models for Foodborne Infections
48.2.1.1 Rodents
Rats have been found to be susceptible to P. westermani and P. ohirai infection, with metacercaria
d eveloping to adult worms that are capable of producing eggs [44,45].
Narain et al. [46] demonstrated that outbred Wistar rats infected with P. heterotremus metacercariae
produced adult worms in the lungs and pleural cavity, with some immature flukes being present in the
skeletal muscles, highlighting the utility of outbred Wistar rats as a model for pulmonary P. heterotremus
infection.
Weina et al. [47] showed that Syrian hamsters infected with 3–16 metacercariae of P. kellicotti
displayed acute pleuritis, reactive mesothelial hyperplasia, subpleural accumulations of reactive and
mature plasma cells, neovascularization, fibrohistiocytic thickening with and without giant cells,
raised fibroconnective tissue lesions, and granulomatous inflammation with hemorrhage within
35 days post infection (dpi). In addition, perivascular plasmacytic (lymphocytic) infiltrate, multifocal
bronchopneumonia, and parenchymal necrotizing suppurative granulomatous inflammation, hemor-
rhagic pneumonia, and diffuse sprinkling of eosinophils, neutrophils, and intraalveolar macrophages
were also observed. These results confirmed the suitability of Syrian hamsters as a small animal
mortality model for P. kellicotti.
Fischer et al. [48] reported that Mongolian gerbils (Meriones unguiculatus) infected intraperitone-
ally or by oral gavage with three to eight metacercariae of P. kellicotti, developed signs of apathy,
weight loss, dehydration as early as 14 dpi, and yielded mature, gravid lung flukes as early as 39 dpi,
with 69% of infected gerbils succumbing to the infection by 49 dpi. Necropsies revealed pulmonary
hemorrhage with necrosis, and flukes as long as 8 mm were recovered from intrathoracic tissues.
Thus, gerbils appear to be a suitable small animal for producing P. kellicotti parasite material and
for studying parasite migration, immunobiology, and pathogenesis. Sanpool et al. [49] also described
that Mongolian gerbils injected intraperitoneally with P. macrorchis metacercariae formed adult
worms in the lungs 45 dpi.
48.2.1.2 Cats
Weina et al. [50] noted that cats infected orally with 2–30 P. kellicotti metacercariae underwent changes
in the lungs of intense eosinophilic pneumonia, granulomatous pneumonitis, squamous epithelial-lined
cyst formation of bronchogenic origin, and partial resolution of the host response. Fan et al. [51] showed
that cats fed with P. pulmonalis metacercariae developed mature adult worms in the lungs and the pleu-
ral cavity. Interestingly, while eggs from worms encased in a cyst were passed in the feces, those from
worms in the pleural cavity were only found on the lung surface and pleural cavity.
48.2.1.3 Dogs
Xue et al. [41,42] observed that dogs infected with 100 P. westermani metacercariae developed adult
worms in the lungs. In early study, Yokogama et al. [44] demonstrated dogs’ susceptibility to pulmonary
P. ohirai infection after animals fed with metacercariae developed adult worms in lungs. Tsubokawa et al.
[45] also noted that dogs infected with P. westermani metacercaria yielded egg-producing adult worms.
Furthermore, Singh et al. [43] found that only puppies appeared suitable for pulmonary P. heterotremus
infection whereas rats, mice, guinea pigs, and rabbits were largely refractory to oral infection with
metacercariae.
48.3 Conclusion
The genus Paragonimus comprises more than 50 trematode species whose adult worms inhabit the lungs
of various mammalian hosts (so called lung flukes), and whose eggs and larval stages are found in snail
and crustacean hosts. Of the nine human-infecting Paragonimus species, five occur mainly in Asia
(P. westermani, P. miyazaki, P. skrjabini, P. heterotremus, and P. hueitungensis), two in Africa (P. uter-
obilateralis and P. africanus), and two in the Americas (P. kellicotti and P. mexicanus). Humans usually
acquire Paragonimus infection through consumption of raw or undercooked crustaceans or meat from
a paratenic host, with serious consequence. Clearly, continued application of rapid, sensitive, and spe-
cific laboratory diagnostic procedures and prompt implementation of appropriate treatment regimens are
critical for limiting the harmful effects of human paragonimiasis. However, to keep an upper hand over
Paragonimus parasites, it is essential to use appropriate laboratory models that will help e lucidate the
molecular mechanisms of host–parasite interactions and pathogenesis, and contribute to the development
of innovative control strategies against paragonimiasis.
REFERENCES
1. Procop GW. North American paragonimiasis (caused by Paragonimus kellicotti) in the context of
global paragonimiasis. Clin Microbiol Rev. 2009;22(3):415–46.
2. Blair D. Paragonimiasis. Adv Exp Med Biol. 2014;766:115–52.
3. Blair D, et al. A molecular perspective on the genera Paragonimus Braun, Euparagonimus Chen and
Pagumogonimus Chen. J Helminthol. 1999;73:295–9.
4. Waikagul J. A new species of Paragonimus (Trematoda: Troglotrematidae) from a cat infected with
metacercariae from mountain crabs Larnaudia larnaudii. J Parasitol. 2007;93:1496–500.
5. Iwagami M, Rajapakse RP, Paranagama W, Okada T, Kano S, Agatsuma T. Ancient divergence of
Paragonimus westermani in Sri Lanka. Parasitol Res. 2008;102:845–52.
6. Uniprot. www.uniprot.org/taxonomy (accessed June 30, 2016).
7. Iwagami M, et al. A molecular phylogeographic study based on DNA sequences from individual
metacercariae of Paragonimus mexicanus from Guatemala and Ecuador. J Helminthol. 2003;77(1):33–8.
8. Aka NA, Adoubryn K, Rondelaud D, Dreyfuss G. Human paragonimiasis in Africa. Ann Afr Med.
2008;7(4):153–62.
9. Liu Q, Wei F, Liu W, Yang S, Zhang X. Paragonimiasis: an important food-borne zoonosis in China.
Trends Parasitol. 2008;24(7):318–23.
10. Singh TS, Sugiyama H, Rangsiruji A. Paragonimus & paragonimiasis in India. Indian J Med Res.
2012;136(2):192–204.
11. Diaz JH. Paragonimiasis acquired in the United States: native and nonnative species. Clin Microbiol
Rev. 2013;26(3):493–504.
12. Doanh PN, Horii Y, Nawa Y. Paragonimus and paragonimiasis in Vietnam: an update. Korean
J Parasitol. 2013;51(6):621–7.
13. Calvopiña M, Romero D, Castañeda B, Hashiguchi Y, Sugiyama H. Current status of Paragonimus and
paragonimiasis in Ecuador. Mem Inst Oswaldo Cruz. 2014;109(7):849–55.
14. Fischer PU, Weil GJ. North American paragonimiasis: epidemiology and diagnostic strategies. Expert
Rev Anti Infect Ther. 2015;13(6):779–86.
15. Nagayasu E, Yoshida A, Hombu A, Horii Y, Maruyama H. Paragonimiasis in Japan: a twelve-year
retrospective case review (2001–2012). Intern Med. 2015;54(2):179–86.
16. Yatera K, et al. A rare case of paragonimiasis miyazaki with lung involvement diagnosed 7 years after
infection: a case report and literature review. Parasitol Int. 2015;64(5):274–80.
17. Devi KR, et al. Morphological and molecular characterization of Paragonimus westermani in
northeastern India. Acta Trop. 2010;116:31–8.
18. Chai JY. Paragonimiasis. Handb Clin Neurol. 2013;114:283–96.
19. Lane MA, Barsanti MC, Santos CA, Yeung M, Lubner SJ, Weil GJ. Human paragonimiasis in North
America following ingestion of raw crayfish. Clin Infect Dis. 2009;49(6):e55–61.
20. Vargas-Arzola J, et al. Detection of Paragonimus mexicanus (Trematoda) metacercariae in crabs from
Oaxaca, Mexico. Acta Trop. 2014;137:95–8.
780 Laboratory Models for Foodborne Infections
21. Rekha Devi K, et al. Presence of three distinct genotypes within the Paragonimus westermani complex
in northeastern India. Parasitology 2013;140:76–86.
22. Boland JM, et al. Pleuropulmonary infection by Paragonimus westermani in the United States: a rare
cause of eosinophilic pneumonia after ingestion of live crabs. Am J Surg Pathol. 2011;35(5):707–13.
23. Shim SS, Kim Y, Lee JK, Lee JH, Song DE. Pleuropulmonary and abdominal paragonimiasis: CT and
ultrasound findings. Br J Radiol. 2012;85(1012):403–10.
24. Henry TS, Lane MA, Weil GJ, Bailey TC, Bhalla S. Chest CT features of North American paragonimia-
sis. AJR Am J Roentgenol. 2012;198(5):1076–83. Erratum in: AJR Am J Roentgenol. 2013;201(5):1165.
25. McNulty SN, Fischer PU, Townsend RR, Curtis KC, Weil GJ, Mitreva M. Systems biology studies of
adult Paragonimus lung flukes facilitate the identification of immunodominant parasite antigens. PLoS
Negl Trop Dis. 2014;8(10):e3242.
26. Le TH, Van De N, Blair D, McManus DP, Kino H, Agatsuma T. Paragonimus heterotremus Chen and
Hsia (1964), in Vietnam: a molecular identification and relationships of isolates from different hosts and
geographical origins. Acta Trop. 2006;98:25–33.
27. Tandon V, Prasad PK, Chatterjee A, Bhutia PT. Surface fine topography and PCR-based determination
of metacercaria of Paragonimus sp. from edible crabs in Arunachal Pradesh, Northeast India. Parasitol
Res. 2007;102(1):21–8.
28. Chen MX, et al. Sensitive and rapid detection of Paragonimus westermani infection in humans and
animals by loop-mediated isothermal amplification (LAMP). Parasitol Res. 2011;108(5):1193–8.
29. Intapan PM, et al. Molecular identification of a case of Paragonimus pseudoheterotremus infection in
Thailand. Am J Trop Med Hyg. 2012;87(4):706–9.
30. Sanpool O, et al. Molecular variation in the Paragonimus heterotremus complex in Thailand and
Myanmar. Korean J Parasitol. 2013;51(6):677–81.
31. Iwagami I, et al. Molecular phylogeographic studies on Paragonimus westermani in Asia. J Helminthol.
2000;74:315–22.
32. Park GM, Im KI, Yong TS. Phylogenetic relationship of ribosomal ITS2 and mitochondrial CO1 among
diploid and triploid Paragonimus westermani isolates. Korean J Parasitol. 2003;41:47–55.
33. Doanh PN, Shinohara A, Horii Y, Habe S, Nawa Y. Discovery of Paragonimus westermani in Vietnam
and its molecular phylogenetic status in P. westermani complex. Parasitol Res. 2009;104:1149–55.
34. Doanh PN, Dung DT, Thach DT, Horii Y, Shinohara A, Nawa Y. Human paragonimiasis in Viet Nam:
epidemiological survey and identification of the responsible species by DNA sequencing of eggs in
patients’ sputum. Parasitol Int. 2011;60:534–7.
35. Prasad PK, Goswami LM, Tandon V, Chatterjee A. PCR-based molecular characterization and insilico
analysis of food-borne trematode parasites Paragonimus westermani, Fasciolopsis buski and Fasciola
gigantica from Northeast India using ITS2 rDNA. Bioinformation 2011;6(2):64–8.
36. Biswal DK, Chatterjee A, Bhattacharya A, Tandon V. The mitochondrial genome of Paragonimus
westermani (Kerbert, 1878), the Indian isolate of the lung fluke representative of the family
Paragonimidae (Trematoda). Peer J. 2014;2:e484.
37. Sugiyama H, Morishima Y, Kameoka Y, Kawanaka M. Polymerase chain reaction (PCR)-based
molecular discrimination between Paragonimus westermani and P. miyazaki at the metacercarial stage.
Mol Cell Probes 2002;16(3):231–6.
38. Sugiyama H, et al. Molecular discrimination between individual metacercariae of Paragonimus
heterotremus and P. westermani occurring in Thailand. Southeast Asian J Trop Med Public Health
2005;36(Suppl 4):102–6.
39. Sugiyama H, Morishima Y, Rangsiruji A, Binchai S, Ketudat P, Kawanaka M. Application of multiplex
pcr for species discrimination using individual metacercariae of Paragonimus occurring in Thailand.
Southeast Asian J Trop Med Public Health 2006;37(Suppl 3):48–52.
40. Tantrawatpan C, et al. Application of a real-time fluorescence resonance energy transfer polymerase
chain reaction assay with melting curve analysis for the detection of Paragonimus heterotremus eggs in
the feces of experimentally infected cats. J Vet Diagn Invest. 2013;25(5):620–6.
41. Xue J, Utzinger J, Zhang YN, Tanner M, Keiser J, Xiao SH. Artemether and tribendimidine lack activity
in experimental treatment of Paragonimus westermani in the dog. Parasitol Res. 2008;102(3):537–40.
42. Xiao SH, Xue J, Li-li X, Zhang YN, Qiang HQ. Effectiveness of mefloquine against Clonorchis sinensis
in rats and Paragonimus westermani in dogs. Parasitol Res. 2010;107(6):1391–7.
Paragonimus 781
43. Singh TS, Sugiyama H, Devi KR, Singh LD, Binchai S, Rangsiruji A. Experimental infection with
Paragonimus heterotremus metacercariae in laboratory animals in Manipur, India. Southeast Asian
J Trop Med Public Health 2011;42(1):34–8.
44. Yokogama M, et al. Chemotherapy of paragonimiasis with bithionol. I. Experimental chemotherapy on
the animals infected with Paragonimus westermani or P. ohirai. Jpn J Parasitol. 1961;10(2):302–16.
45. Tsubokawa D, et al. Collection methods of trematode eggs using experimental animal models. Parasitol
Int. 2016;65:584–87.
46. Narain K, Rekha Devi K, Mahanta J. A rodent model for pulmonary paragonimiasis. Parasitol Res.
2003;91(6):517–9.
47. Weina PJ, Burns WC. Mortality in Syrian hamsters infected with Paragonimus kellicotti. J Parasitol.
1992;78(2):378–80.
48. Fischer PU, Curtis KC, Marcos LA, Weil GJ. Molecular characterization of the North American lung
fluke Paragonimus kellicotti in Missouri and its development in Mongolian gerbils. Am J Trop Med Hyg.
2011;84(6):1005–11.
49. Sanpool O, et al. Morphological and molecular identification of a lung fluke, Paragonimus macrorchis
(Trematoda, Paragonimidae), found in central Lao PDR and its molecular phylogenetic status in the
genus Paragonimus. Parasitol Int. 2015;64(6):513–8.
50. Weina PJ, England DM. The American lung fluke, Paragonimus kellicotti, in a cat model. J Parasitol.
1990;76(4):568–72.
51. Fan PC, Lu H, Lin LH. Egg production capacity of Paragonimus pulmonalis in cats. J Parasitol.
1998;84(6):1282–5.
52. Yokogama M, Oshima T, Kihata M. Studies to maintain excysted metacercariae of Paragonimus
westermani in vitro. J Parasitol. 1955;41(6):28.
49
Taenia
Dongyou Liu
CONTENTS
49.1 Introduction................................................................................................................................... 783
49.1.1 Classification and Morphology......................................................................................... 784
49.1.1.1 Classification..................................................................................................... 784
49.1.1.2 Morphology....................................................................................................... 784
49.1.2 Life Cycle and Epidemiology........................................................................................... 786
49.1.2.1 Life Cycle.......................................................................................................... 786
49.1.2.2 Epidemiology.................................................................................................... 786
49.1.3 Clinical Features.............................................................................................................. 786
49.1.4 Diagnosis.......................................................................................................................... 787
49.1.5 Treatment and Prevention................................................................................................. 787
49.1.5.1 Treatment.......................................................................................................... 787
49.1.5.2 Prevention......................................................................................................... 788
49.2 Laboratory Models........................................................................................................................ 788
49.3 Conclusion..................................................................................................................................... 788
References............................................................................................................................................... 789
49.1 Introduction
The genus Taenia encompasses a large group of parasitic tapeworms that undergo three distinct stages
of development (i.e., adult worm, eggs, and larva/metacestode) during their life cycles and require two
mammalian hosts for transmission. The ribbon-like adult worm (so called tapeworm) resides in the intes-
tines of carnivores or omnivores, causing intestinal taeniasis, whereas the cyst-like larva (metacestode)
occurs in the internal organs of herbivores or omnivores, causing cysticercosis or neurocysticercosis
(representing different forms of taeniasis).
Of the 45 described Taenia species, three [i.e., Taenia solium (pork tapeworm), Taenia saginata (beef
tapeworm), and Taenia asiatica (Asian tapeworm)] are known to infect humans. Indeed, as the only
definitive host for these three species, human infection with the adult worms leads to intestinal parasit-
ism, which is largely asymptomatic. However, the larva (metacestode) of T. solium causes cysticercosis
(cysticercus cellulosae) in pigs and humans, that of T. saginata causes cysticercosis (cysticercus bovis) in
cattle, and that of T. asiatica causes cysticercosis (cysticercus viscerotropica) in pigs (Table 49.1).
Although tapeworms were mentioned in ancient Egyptian works dated back to 2000 BC, as well as
in the History of Animals written by Greek author Aristotle (384–322 BC), DNA analyses point out the
likelihood that ancestors of modern humans in Africa had acquired parasite through consumption of
infected antelope and bovid meats more than 10,000 years ago and subsequently passed it to other ani-
mals such as domestic pigs. A more recent description of cysticercosis was made by Johannes Udalric
Rumler in 1555; however, the connection between tapeworm and cysticercosis was not established until
the middle of the 19th century.
783
784 Laboratory Models for Foodborne Infections
49.1.1.2 Morphology
Taenia adult worms (Taenia is derived from Greek ταίvɩα, tainia meaning ribbon, bandage, or stripe)
are flat, ribbon-like in appearance, and white or yellowish-white in color. and consist of a knob-like
head called scolex, a short neck, and a ribbon-like body (also known as strobila). The scolex (hold-
fast organ) often possesses suckers (acetabula), rostellum, and spiny hooks. It is noteworthy that the
scolex of T. solium is spheroidal, with a rostellum (of 1 mm in diameter), which is armed with two
rows of 22–32 hooks; that of T. saginata is cuboidal (of 2 mm in width) without rostellum or hooks;
and that of T. asiatica is spheroidal, with a cuspidal rostellum (of 0.8 mm in width), which has no
hooks (Table 49.1). The strobila (up to 22 m long depending on the species) is composed of a chain
of segments called proglottids, each of which contains a set of male and female reproductive organs
(thus hermaphroditic). The reproductive organs are made up of tubular unbranched uterus (filled with
eggs), ovary, genital pore, testes, and vitelline gland, with testes and ovary opening into a common
genital pore located on the side. Being an acoelomate animal, the adult worm has no body cavity or
digestive system, and relies entirely on its penetrable tegument to absorb nutrients. As body growth
starts from the neck region, immature proglottids are found near the neck, mature proglottids in the
middle, and gravid (oldest) proglottids at the posterior end. On average, T. solium adult worm has
about 1000 proglottids, each producing 50,000 eggs; T. saginata adult worm has about 1000–2000
proglottids, each producing 100,000 eggs; T. asiatica adult worm has 700–900 proglottids, each
producing 80,000 eggs [5].
Taenia eggs are spherical, of about 40–48 µm in diameter, and surrounded by a thick striated wall,
which encases an embryo (oncosphere). After gravid proglottids detach from the strobila or discharge
Taenia 785
TABLE 49.1
Morphological and Biological Characteristics of Human-Infecting Taenia Species
Taenia solium (Pork Taenia saginata (Beef Taenia asiatica (Asian
Tapeworm) Tapeworm) Tapeworm)
Adult worm Adult worm of 2–8 m in length Adult worm of 4–12 m in length Adult worm of about 3.5 m in
resides in human small (as long as 25 m) resides in length resides in human small
intestine; its scolex has four human small intestine; its intestine; its scolex has four
suckers that surround the scolex has four suckers, and an suckers surrounding the
rostellum, with 22–32 hooks apical pit instead of a rostellum rostellum, without hooks; its
arranged in two rows; its and hooks; its strobila contains strobila contains 700–900
strobila contains 1000 1000–2000 proglottids; its proglottids; its gravid proglottid
proglottids; its gravid gravid proglottid has no has posterior protuberances
proglottid has no posterior posterior protuberances
protuberances
Metacestode Cysticercus cellulosae (5–8 × Cysticercus bovis Cysticercus viscerotropica
(Cysticercus) 3–6 mm) occurs mostly in pig (6–10 × 4–6 mm) occurs inside (2 × 2 mm) occurs in liver and
muscle, as fluid-filled cyst muscle, liver, and lungs of lungs in pigs, wild boars, and
(0.5–1.5 cm in diameter) cattle, as fluid-filled cyst occasionally cattle, as fluid-filled
containing single scolex with containing single scolex cyst containing single scolex
hooks; its external surface without hooks; its external with two rows of rudimentary
lacks wart-like formations surface lacks wart-like hooks; its external surface has
formations (or not prominent if wart-like formations
present)
Genome 122–131 Mb in size, with 169 Mb in size, with 13,161 168 Mb in size, with 13,323
11,902–12,481 coding genes coding genes coding genes
Clinical features Humans infected with adult Humans infected with adult Humans infected with adult
worms show minimal worms are usually worms are largely asymptomatic
symptoms; accidental ingestion asymptomatic, although heavy
of embryonated eggs or infection may lead to weight
proglottids (via autoinfection loss, dizziness, abdominal pain,
or contaminated food) by diarrhea, headaches, nausea,
humans leads to cysticercosis, constipation, chronic
which shows three indigestion, and loss of
morphologically distinct forms appetite; cysticercosis is not
[the ordinary “cellulose” observed in humans
cysticercus, with a fluid-filled
cyst and an invaginated scolex;
the intermediate form with a
scolex; the “racemose” form
(20 cm in length and 60 mL of
fluid) with no evident scolex]
Distribution Cosmopolitan, and highly Prevalent in Africa, Europe, Prevalent in Taiwan, South Korea,
prevalent in Mexico, Latin Southeast Asia, South Asia, Indonesia, the Philippines,
America, West Africa, Russia, and Latin America Thailand, China, Vietnam, Japan,
India, Pakistan, China, and and Nepal
Southeast Asia
Sources: Parija, S.C. and Ponnambath, D.K., Trop. Parasitol., 3, 120–124, 2013; Wang, S. et al., Nature Commun., 7, 12845, 2016.
along with feces, eggs are released. Being highly resistant to desiccation and sewage treatment, these
eggs can survive on pastures for weeks. Upon ingestion by intermediate host, the eggs hatch and develop
into infective cysticerci in selective organs (e.g., muscle, liver, and lungs).
Taenia larvae (metacestodes) such as those of T. solium, T. saginata, and T. asiatica form small,
pearly-white, fluid-filled cysts (cysticerci) (hence the common name bladder worms) within which a
single invaginated protoscolex (infective stage) is located [5]. The larvae of other Taenia spp. may show
varied appearance, with some forming a strobilocercus (containing noninvaginated protoscolex) and oth-
ers forming a large coenurus (containing several invaginated protoscoleces).
786 Laboratory Models for Foodborne Infections
49.1.2.2 Epidemiology
With humans as the definitive host and pigs as the intermediate host, T. solium is present in many parts
of the world where humans live in close proximity with pigs and eat undercooked pork [7]. In addi-
tion, humans may function as aberrant intermediate host after accidental ingestion of embryonated
eggs, either through autoinfection or consumption of contaminated food, leading to neurocysticercosis,
which demonstrates a predilection for brain tissue and other soft muscle tissues [8]. Geographically,
T. solium infection is particularly common in Mexico, Latin America, West Africa, India, Pakistan,
Southeast Asia, China, Russia, and Slavic countries of Europe. Previous reports indicate that the sero
prevalence of T. solium in areas of Guatemala, Bolivia, and Peru may be as high as 20% in humans and
37% in pigs.
In the case of T. saginata, humans and cattle act as the definitive and intermediate hosts, respectively.
This parasite is found in places where beef is eaten, and is relatively common in Africa, the Middle East,
the Philippines, Eastern Europe, and Latin America.
T. asiatica utilizes humans as the definitive host, and pigs, wild boar, and possibly cattle as the inter-
mediate host. While the adult worm is found in the small intestine of humans, pea-sized, fluid-filled cyst
(cysticercus) is present in liver, serosa, and lungs of pigs and in the liver of cattle. Geographically, the
parasite is essentially restricted to Taiwan, Korea, Indonesia, Nepal, Thailand, and China, in addition to
Japan, the Philippines, and Vietnam [9,10].
49.1.3 Clinical Features
Infection with T. solium adult worm in humans generally involves one to two worms and is often
asymptomatic, although heavy infection may lead to mild intermittent diarrhea or constipation, ane-
mia, indigestion, inappetite, and emaciation, together with urticaria, anal pruritus, and eosinophilia.
However, accidental consumption of T. solium eggs from contaminated vegetables/water or ingestion
of T. solium eggs or proglottids, which rupture within the host intestines, may result in cysticercosis
or neurocysticercosis [11]. In the case of cysticercosis, solid lumps of 1–2 cm in size may develop
under the skin near the trunk and extremities, become painful and swollen, and then resolve. In the
case of neurocysticercosis, cysts of 5–20 mm in diameter may form in the parenchyma of the brain.
In more severe cases (racemose neurocysticercosis), lesions as large as 6 cm in diameter, lobulated,
and may occur in subarachnoid space and fissures. Clinical presentations may include severe head-
aches, dizziness, epilepsy, seizures, dementia, hypertension, lesions in the brain, blindness, tumor-
like growths, hydrocephalus, paraplegy, meningitis, convulsions, and even death.
Taenia 787
Similarly, human infection with T. saginata adult worm is asymptomatic, but heavy infection causes
dizziness, abdominal pain, diarrhea, headaches, nausea, constipation, chronic indigestion, inappetite,
and weight loss. Other rare clinical signs include ileus, pancreatitis, cholecystitis, and cholangitis.
Further, human infection with T. asiatica adult worm is usually asymptomatic. Nevertheless, in severe
cases, damage and bleeding in the stomach and intestine may be observed.
49.1.4 Diagnosis
Identification of Taenia species (T. saginata, T. asiatica, and T. solium) in the definitive host has tradi-
tionally relied on microscopic detection of eggs or proglottids in feces. However, this approach lacks
desired sensitivity and specificity given that the eggs from different Taenia species are morphologically
indistinguishable [12].
Use of computed tomography (CT) or magnetic resonance imaging (MRI) enables detection of cysti-
cerci in the brain, as in the case of human neurocysticercosis. In addition, application of, X-rays facili-
tates detection of calcified cysticerci in the subcutaneous and muscle tissues.
Serological methods (ELISA and IET) based on adult worm excretion-secretion antigens (ES Ag)
(ES33 and ES38) of T. solium have proven useful for confirmation of T. solium taeniasis, with sensitivity
and specificity of >97% and 91%, respectively [13]. Furthermore, serological detection of coproantigens
in fecal specimens allows genus-specific discrimination. Enzyme-linked immunoelectrotransfer blot
(EITB) detects an immunoblot band of 21.5 kDa from T. asiatica, permitting effective differentiation of
asiatica from other taeniid infections.
Utilization of molecular methods targeting various gene regions of Taenia species has greatly
enhanced the diagnosis of taeniasis and cysticercosis [14–25]. Polymerase chain reaction (PCR) amplifi-
cation of ribosomal 5.8S and HDP2 gene sequences enables differentiation among T. solium, T. saginata,
and T. asiatica [26,27]. Sequencing analysis of the variable regions (V1–V5) within nuclear 18S rRNA
gene reveals pertinent phylogenetic details within the genus Taenia. The mitochondrial gene sequence
(mtDNA) provides a valuable molecular marker for uncovering evolutionary relationships among dis-
tantly related taxa, and also for investigating the phylobiogeography of closely related species. A mul-
tiplex PCR based on the primers Ta4978F, Ts5058F, Tso7421F, and Rev7915 from the mitochondrial
gene sequence has streamlined differential diagnosis, molecular characterization, and epidemiological
surveys of Taenia species [28]. Moreover, use of primers (Ta7216F, Ts7313F, Tso7466F, and Rev7915)
from valine transfer RNA and NADH dehydrogenase subunit two genes in multiplex PCR generates
species-specific products of 706, 629, and 474 bp for T. asiatica, T. saginata, and T. solium in a sensitive,
specific, and speedy manner [29].
49.1.5.2 Prevention
The key for preventing Taenia infections in humans is to break their transmission cycle through stringent
meat inspection, condemnation of infected carcasses for human consumption, proper cooking (80°C) or
freezing (–10°C for 9 days) of meat, sanitary disposal of feces, prohibiting the use of sewage for fertil-
izing pastures, washing salad vegetables, strict personal hygiene, and improved access to clean water.
To prevent Taenia infections in pigs and cattle, it is important to avoid the exposure of pigs and cattle
to environments contaminated with human feces; to eliminate the use of untreated sewage effluent to
irrigate land that is later used by pigs and cattle for forage and food crops; and to vaccinate pigs with
genetically engineered 45W-4B antigens or S3PVAC consisting of three protective peptides (KETc12,
KETc1, and GK1) against T. solium infection [31,32].
49.2 Laboratory Models
As humans act as the only definitive host for T. solium, T. saginata, and T. asiatica, use of laboratory
models is not only necessary but also important in helping elucidate the pathogenesis, host–parasite
relationship, and other related issues concerning taeniasis and cysticercosis [33,34]. From published
data, it is clear that pigs, dogs, cats, and rabbits, even in immunosuppressed status, are relatively poor
hosts for experimental investigation of taeniid infections, On the other hand, various rodent hosts (rats,
mice, hamsters, gerbils, and chinchillas) support the maintenance, growth, and maturation of taeniid
tapeworms, contributing to our improved understanding of taeniasis and cysticercosis [35,36].
In an early study, Wang et al. [37] showed that immunosuppressed Syrian hamsters are susceptible to
T. solium infection, with adult worm developing in the intestine after oral ingestion of cysticerci contained
in severe combined immunodeficiency (SCID) mouse muscles. Separately, Avila et al. [38] demonstrated
that hamsters, gerbils, and chinchillas are adequate experimental models for investigating the growth and
immune responses of T. solium adult worm infection. Furthermore, chinchillas and, to a lesser extent,
hamsters appear to be more permissive than gerbils, allowing longer-term survival and development of
T. solium parasites. In a more recent report, Verastegui et al. [39] described a useful neurocysticercosis
model based on Holtzman rats. It was noted that rats injected extraparenchymally as well as intraparen-
chymally with activated T. solium oncospheres developed cysticerci in the brain tissue after 4 months; that
the cysticerci were present in the parenchymal, ventricle, or submeningeal brain tissue; and that epilepsy
was observed in 9% of rats with neurocysticercosis. The age of rats appeared to be crucial for the success
of infection, although the sites of injection and T. solium oncosphere dosages were largely irrelevant.
Several reports showed that nonobese diabetic SCID (NOD/Shi-scid) mice of both sexes are suscep-
tible to infection with in vitro hatched oncospheres of human-infecting Taenia species (T. solium, T. saginata,
and T. asiatica), producing cysticerci comparable to those developed in their known intermediate host
animals [40–44]. On the other hand, only female SCID mice of BALB/c, C57BL, or C.B-17 inbred
strains allow the development of cysticerci from oncospheres of human taeniid species [44].
Additionally, normal C3H/HeN female mice were showed to initiate Th1 cells immune response
against T. asiatica during the early stages of oncosphere infection [45]. Similarly, hamsters are also
capable of eliciting cytokine expression at the anchor site of T. solium [46–48].
Chowdhury et al. [49] successfully induced neurocysticercosis in rhesus monkey with 12,000 T. solium
eggs, resulting in development of hyperexcitability, epileptic seizures, muscular tremors, digital cramps at
10 days postinfection, and paralysis of limbs and death days post infection. Necropsy of the infected mon-
key revealed numerous cysticerci in the brain, and histopathological examination uncovered liquefactive
necrosis and formation of irregular cystic cavities lined by atrophied parenchymal septa with remnants of
neuropil of the cerebrum.
49.3 Conclusion
Members of the genus Taenia are parasitic tapeworms that involve two mammalian hosts (a carnivorous/
omnivorous definitive host and a herbivorous/omnivorous intermediate host) during their life cycles.
Taenia 789
Taenia infection usually begins when Taenia eggs and proglottids produced by adult worm that reside in
the small intestine of definitive hosts are discharged in stools, and subsequently ingested by intermediate
host. Inside intermediate host, Taenia eggs hatch to release oncospheres, which then penetrate through
intestinal wall, migrate via blood circulation to various internal organs and muscles, and mature into
cyst-like larvae (metacestodes). Once taken up by definitive host, Taenia larvae (metacestodes) release
protoscoleces, which utilize their hooks to anchor onto the intestinal wall and grow into adult worms that
produce eggs for the next cycle of infection.
Of 45 Taenia species identified to date, three (T. solium, T. saginata, and T. asiatica) cause taeniasis
in humans and cysticercosis in pigs and cattle as well as humans. Given that the adult worms of these
human-infecting Taenia species only occur in humans, and the larvae are present in pigs and cattle,
detailed examination of taeniid pathogenesis and host–parasite relationship have been hampered by
ethical and cost concerns. For this reason, use of laboratory models is critically important in helping
to improve our understanding of taeniasis and cysticercosis, and leading to innovative control measures
against these parasites. Fortunately, of various types of laboratory animals available, rodents (e.g., rats,
mice, hamsters, gerbils, and chinchillas) have been shown to support the maintenance, growth, and
maturation of human taeniid tapeworms (in both taeniasis and cysticercosis/neurocysticercosis forms)
and to elicit competent immune responses against these parasites. It is envisaged that in our ongoing
effects to elucidate the molecular basis of host–parasite interactions and to develop novel, tailor-made
control strategies against Taenia parasites, rodents will continue to play an indispensable role [35,50].
REFERENCES
1. Romig T, Ebi D, Wassermann M. Taxonomy and molecular epidemiology of Echinococcus granulosus
sensu lato. Vet Parasitol. 2015;213(3–4):76–84.
2. Hoberg EP. Phylogeny of Taenia: species definitions and origins of human parasites. Parasitol Int.
2006;55(Suppl):S23–30.
3. Haukisalmi V, Lavikainen A, Laaksonen S, Meri S. Taenia arctos n. sp. (Cestoda: Cyclophyllidea:
Taeniidae) from its definitive (brown bear Ursus arctos Linnaeus) and intermediate (moose/elk Alces
spp.) hosts. Syst Parasitol. 2011;80(3):217–30.
4. Nakao M, et al. Molecular phylogeny of the genus Taenia (Cestoda: Taeniidae): proposals for the res-
urrection of Hydatigera Lamarck, 1816 and the creation of a new genus Versteria. Int J Parasitol.
2013;43(6):427–37.
5. Eom KS, Rim HJ. Morphologic descriptions of Taenia asiatica sp. n. Korean J Parasitol. 1993;31:1–6.
6. Parija SC, Ponnambath DK. Laboratory diagnosis of Taenia asiatica in humans and animals. Trop
Parasitol. 2013;3(2):120–4.
7. Singh AK, Prasad KN, Prasad A, Tripathi M, Gupta RK, Husain N. Immune responses to via-
ble and degenerative metacestodes of Taenia solium in naturally infected swine. Int J Parasitol.
2013;43(14):1101–7.
8. Bobes RJ, et al. Evolution, molecular epidemiology and perspectives on the research of taeniid parasites
with special emphasis on Taenia solium. Infect Genet Evol. 2014;23:150–60.
9. Eom KS, Jeon HK, Rim HJ. Geographical distribution of Taenia asiatica and related species. Korean J
Parasitol. 2009;47:S115–24.
10. Ooi HK, Ho CM, Chung WC. Historical overview of Taenia asiatica in Taiwan. Korean J Parasitol.
2013;51(1):31–6.
11. Garcia HH, Gonzalez AE, Gilman RH. Cysticercosis of the central nervous system: how should it be
managed? Curr Opin Infect Dis. 2011;24(5):423–7.
12. Rodrignez-Hidalgo R, Geysen D, Benitez-Ortiz W, Geerts S, Brandt J. Comparison of conventional
techniques to differentiate between Taenia solium and Taenia saginata and an improved polymerase
chain reaction-restriction fragment length polymorphism assay using a mitochondrial 12S rDNA frag-
ment. J Parasitol. 2002;88:1007–11.
13. Ochoa-Sánchez A, Jiménez L, Landa A. The hamster model for identification of specific antigens of
Taenia solium tapeworms. J Biomed Biotechnol. 2011;2011:504959.
14. Mayata H, et al. Differentiating Taenia solium and Taenia saginata infections by simple hematoxylin-
eosin staining and PCR-restriction enzyme analysis. J Clin Microbiol. 2000;38:133–7.
790 Laboratory Models for Foodborne Infections
15. Jeon HK, Kim KH, Eom KS. Complete sequence of the mitochondrial genome of Taenia saginata:
comparison with T. solium and T. asiatica. Parasitol Int. 2007;56:243–6.
16. Jeon HK, Kim KH, Eom KS. Molecular identification of Taenia specimens after long-term preservation
in formalin. Parasitol Int. 2011;60:203–5.
17. Gonzales LM, et al. Differential diagnosis of Taenia saginata and Taenia saginata asiatica taeniasis
through PCR. Diagn Microbiol Infect Dis. 2004;49:183–8.
18. Yamasaki H, et al. DNA differential diagnosis of taeniasis and cysticercosis by multiplex PCR. J Clin
Microbiol. 2004;42:548–53.
19. Yamasaki H, Nakaya K, Nakao M, Sako Y, Ito A. Significance of molecular diagnosis using histopatho-
logical specimens in cestode zoonoses. Trop Med Health. 2007;35:307–21.
20. McManus DP. Molecular discrimination of taeniid cestodes. Parasitol Int. 2006;55(Suppl):31–7.
21. Lavikainen A, Laaksonen S, Beckmen K, Oksanen A, Isomursu M, Meri S. Molecular identification
of Taenia spp. in wolves (Canis lupus), brown bears (Ursus arctos) and cervids from North Europe and
Alaska. Parasitol Int. 2011;60(3):289–95.
22. Tsai IJ, et al. The genomes of four tapeworm species reveal adaptations to parasitism. Nature.
2013;496(7443):57–63.
23. Zhang G, et al. Utility of DNA barcoding in distinguishing species of the family Taeniidae. J Parasitol.
2014;100(4):542–6.
24. Pajuelo MJ, et al. Identification and characterization of microsatellite markers derived from the whole
genome analysis of Taenia solium. PLoS Negl Trop Dis. 2015;9(12):e0004316.
25. Roelfsema JH, Nozari N, Pinelli E, Kortbeek LM. Novel PCRs for differential diagnosis of cestodes.
Exp Parasitol. 2016;161:20–6.
26. González LM, et al. Characterization of the Taenia spp HDP2 sequence and development of a novel
PCR-based assay for discrimination of Taenia saginata from Taenia asiatica. Parasites Vectors.
2010;3:51.
27. Yan H, et al. The nuclear 18S ribosomal RNA gene as a source of phylogenetic information in the genus
Taenia. Parasitol Res. 2013;112(3):1343–7.
28. Jeon HK, Eom KS. Molecular approaches to Taenia asiatica. Korean J Parasitol. 2013;51(1):1–8.
29. Jeon HK, et al. Differential diagnosis of Taenia asiatica using multiplex PCR. Exp Parasitol.
2009;121:151–6.
30. Palomares-Alonso F, Palencia Hernández G, Rojas-Tomé IS, Jung-Cook H, Pinzón-Estrada E. Murine
cysticercosis model: influence of the infection time and the time of treatment on the cysticidal efficacy
of albendazole and praziquantel. Exp Parasitol. 2015;149:1–6.
31. Lightowlers MW. Fact or hypothesis: Taenia crassiceps as a model for Taenia solium, and the S3Pvac
vaccine. Parasite Immunol. 2010;32(11–12):701–9.
32. Salazar AM, et al. Genotoxicity induced by Taenia solium and its reduction by immunization with cal-
reticulin in a hamster model of taeniosis. Environ Mol Mutagen. 2013;54(5):347–53.
33. Siles-Lucas M, Hemphill A. Cestode parasites: application of in vivo and in vitro models for studies on
the host-parasite relationship. Adv Parasitol. 2002;51:133–230.
34. Nash TE, et al. Neurocysticercosis: a natural human model of epileptogenesis. Epilepsia.
2015;56(2):177–83.
35. Flisser A, et al. Taenia solium: current understanding of laboratory animal models of taeniosis.
Parasitology. 2010;137(3):347–57.
36. Hamamoto Filho PT, et al. Development of an experimental model of neurocysticercosis-induced
hydrocephalus. Pilot study. Acta Cir Bras. 2015;30(12):819–23.
37. Wang IC, Guo JX, Ma YX, Chung WC, Lu SC, Fan PC. Sexual development of Taenia solium in ham-
sters from rodent-derived cysticerci. J Helminthol. 1999;73(4):347–50.
38. Avila G, et al. Laboratory animal models for human Taenia solium. Parasitol Int. 2006;55
(Suppl):S99–103.
39. Verastegui MR, et al, Novel rat model for neurocysticercosis using Taenia solium. Am J Pathol.
2015;185(8):2259–68.
40. Ito A, et al. Human Taenia eggs develop into cysticerci in SCID mice. Parasitology. 1997;114 (1):85–8.
41. Ito A, Ito M. Human Taenia in severe combined immunodeficiency (SCID) mice. Parasitol Today.
1999;15(2):64–7.
Taenia 791
42. Ito A, Nakaya K, Sako Y, Nakao M, Ito M. NOD-SCID mouse as an experimental animal model for
cysticercosis. Southeast Asian J Trop Med Public Health. 2001;32(Suppl 2):85–9.
43. Chang SL, et al. Development of Taenia saginata asiatica metacestodes in SCID mice and its infectivity
in human and alternative definitive hosts. Parasitol Res. 2005;96(2):95–101.
44. Nakaya K, et al. Usefulness of severe combined immunodeficiency (SCID) and inbred mice for studies
of cysticercosis and echinococcosis. Parasitol Int. 2006;55(Suppl):S91–7.
45. Peng SY, et al. Infection of normal C3H/HeN mice with Taenia saginata asiatica oncospheres. Res Vet
Sci. 2009;86(2):261–6.
46. Cruz-Rivera M, et al. Cytokine expression at the anchor site in experimental Taenia solium infection in
hamsters. Vet Parasitol. 2014;200(3–4):299–302.
47. Garcia HH, Rodriguez S, Friedland JS; Cysticercosis Working Group in Peru. Immunology of Taenia
solium taeniasis and human cysticercosis. Parasite Immunol. 2014;36(8):388–96.
48. Peón AN, Ledesma-Soto Y, Terrazas LI. Regulation of immunity by taeniids: lessons from animal mod-
els and in vitro studies. Parasite Immunol. 2016;38(3):124–35.
49. Chowdhury N, Saleque A, Sood NK, Singla LD. Induced neurocysticercosis in rhesus monkeys
(Macaca mulatta) produces clinical signs and lesions similar to natural disease in man. Sci World J.
2014;2014:248049.
50. Ito A, et al. Recent advances in basic and applied science for the control of taeniasis/cysticercosis in
Asia. Southeast Asian J Trop Med Public Health. 2002;33(Suppl 3):79–82.
50
Trichinella
CONTENTS
50.1 Trichinella and Trichinellosis....................................................................................................... 793
50.2 Mouse............................................................................................................................................ 794
50.2.1 Contribution of MHC-Linked and Background Genes to the Control of
the Response..................................................................................................................... 794
50.2.2 Response of the Host during Intestinal and Muscle Phase of the Infection.................... 795
50.2.3 Applied Studies on Mouse Model for Trichinellosis........................................................ 797
50.3 Rat................................................................................................................................................. 797
50.4 Swine............................................................................................................................................. 798
50.4.1 Applied Studies on Swine Model for Trichinella Infection............................................. 799
50.5 Wild Boar...................................................................................................................................... 800
50.6 Horse............................................................................................................................................. 800
50.7 Other Animal Species as Models for Trichinella Infection.......................................................... 801
50.8 Conclusion..................................................................................................................................... 802
References............................................................................................................................................... 803
50.1 T
richinella and Trichinellosis
Trichinellosis is a foodborne disease caused by the consumption of raw or undercooked meat of dif-
ferent animal origins, e.g., pork, horse, or game, infected with helminth of the Trichinella genus [1,2].
The genus consists of two clades that inhabit mainly striated muscles of the host. Parasites from the first
clade encapsulate, and those from the second do not [3]. Among the so-far identified 12 taxa of the genus,
the species and genotypes of the first clade parasitize only mammals and include T. spiralis, T. britovi
with its closely related genotype T8, T. nativa and related T6, T. murrelli and related T9, T. nelsoni, and
T. patagoniensis. The second clade comprises three species: T. pseudospiralis, which infects mammals
and birds, and T. papuae and T. zimbabwensis, which parasitize mammals and reptiles, respectively [3].
Although Trichinella has cosmopolitan distribution and infects more than 2500 people annually [4],
its infection results in minimal health burden and low mortality rate in human population [5]. Of all
Trichinella taxa, T. spiralis accounts for most human infections [2].
Trichinella spp. are unique among various helminthes in that all three life cycle stages of the parasite-
infective muscle larvae, adult, and newborn larvae develop in one host. Infection is acquired by con-
sumption of infected raw or undercooked meat. Under the influence of gastric juice, larvae are released
from infected meat in the stomach, molt, and develop into the adult stage inside the enterocytes of small
intestine. After mating, newborn larvae move into the circulation and spread throughout the tissues and
organs, and only those that penetrate striated muscles mature into muscle larvae [6]. This parasite has
ability to transform the infected muscle cell into a new type of cell in the host body, the so-called nurse
cell (NC) [7,8]. From this immunologically privileged place, parasite achieves long-lasting communica-
tion with the host through muscle larvae excretory-secretory products (ES).
793
794 Laboratory Models for Foodborne Infections
Unlike what happens in humans, T. spiralis can reach a high worm burden in animals without induc-
ing clinical symptoms [2]. Although infectivity for laboratory animals differs among various Trichinella
taxa, infection provides valuable in vivo models for basic research.
This chapter reviews the most significant observations gained in biological, pathological, and immu-
nological studies performed on rodent models (inbred mice and rats, or genetically manipulated), which
have the benefit of being easy to handle and relatively cost-effective. It also covers data from some larger
animal models using domestic and wild swine, horses, and other extremely costly animals that host
Trichinella species (including even marine mammals and reptiles).
50.2 Mouse
Mice are the most frequently used mammalian model system due to their close genetic (over 95% of the
mouse genome is identical to human genome) and physiological similarities to humans. Creation of vari-
ous inbred strains by genetic manipulation, as well as knock-out and transgenic animals, enabled studies
of complex phenomenon like susceptibility to the infection and immune responses elicited.
Trichinella is not a picky nematode, as it invades and completes its life cycle in the variety of host
organisms, including mice. Among Trichinella species, the most extensively studied is T. spiralis.
Immune response to primary infection with this intestinal and tissue-dwelling parasite starts with recog-
nition of the antigens and proceeds to the inflammation of the intestinal mucosa, leading to the reduction
of female fecundity, cytopathological damage, and expulsion of adults from intestine during primary
infection, whereas in challenge infection, it induces accelerated expulsion of larvae and adults from the
intestine and reduction of growth and fecundity [9,10]. Differences in the resistance to T. spiralis infec-
tion between various strains have a genetic background. Some strains possess alleles that enable rapid
(strong) response, which means that they expel worms more quickly, reduce female worm fecundity, and
as a consequence have lower muscle larvae burden than slow (weak) responders, which possess another
set of alleles [9,11]. Although Wakelin [9] used time of expulsion as a parameter for determining the
response phenotypes of inbred strains of mice infected with T. spiralis, the number of recovered larvae
from the muscles should also be considered for distinguishing between susceptible and resistant strains,
since it takes into account all variables that affect the response to infection.
phenotypic generations were analyzed [16]. The results pointed out that there are at least two dominant
non-H-2 genes that control rejection of adult worms. The presence of both of them generates strong
responding phenotype, while each gene on its own is responsible for intermediate phenotype.
50.2.2 Response of the Host during Intestinal and Muscle Phase of the Infection
Response of the host implies immunological and nonimmunological mechanisms at the level of intestine
and muscles, engaged in the process of defense from T. spiralis invasion. Using genetically defined strains
of mice or mice with targeted deletions in cytokine or cytokine receptor genes, it is possible to identify
particular components and mechanisms of the host response important for the outcome of T. spiralis
infection. Effective immunity in intestinal phase of T. spiralis infection is crucial for the establishment
of parasitism, since it controls elimination of adult worms from the gut, fecundity of female parasites,
and therefore the number of newborn larvae that migrate toward muscle cells. Parasite provokes T-cell-
mediated immune response at intestinal level, which is accelerated upon reinfection [17]. Investigation
encompassing four strains of inbred mice that exhibit a range of responsiveness to T. spiralis infection—
NIH (high), CBA (intermediate), C57BL/10 (intermediate to low), and B10.BR (low)—revealed that
the resistance is correlated with the intensity of intestinal inflammatory responses. NIH and CBA mice
were the most resistant to T. spiralis and have the most intense intestinal inflammatory responses [18].
Intestinal inflammation is absent in congenitally athymic mice infected with T. spiralis [19], indicating
an important role of T cells, especially CD4+ T cells [20] in host-protective immunity. T. spiralis induce
inflammatory responses by distorting the gut epithelial cell layer. This is a signal that generates release
of Th1 cytokines such as IFN-γ within 2 days [21], followed by predominant Th2 response that occurs
between days 2 and 8 in all the aforementioned strains [18].
Th1 type of response leads to susceptibility to worm infection. Mice deficient in cytokine IFN-γ expel
T. spiralis more rapidly than wild-type mice [22]. Th1 type responses are under control of proinflam-
matory cytokine IL-12, produced by dendritic cells and macrophages. Transgenic mice overexpressing
IL-12 show delayed worm expulsion, accompanied with increased IFN-γ and decreased IL-4 and IL-13
production [23].
Resistance to T. spiralis infection is mediated by Th2 type of response [24], primarily IL-4, IL-9, and
IL-13 cytokines [17]. The presence of type-2 cytokines causes eosinophilia, intestinal mastocytosis,
and elevated IgE production. Primary infection of C57BL/6 IL-4 knock-out (KO) mice with T. spiralis
pointed out the importance of IL-4 in the expulsion of parasites [25], and treatment with anti-IL-4
receptor monoclonal antibody caused prolonged adult infections and higher muscle larvae burdens [26].
IL-4 deficiency also leads to impaired Th2 antibody response, both IgE and IgG1. However, BALB/c
IL-4 KO mice showed no difference in the time of expulsion or in enteropathy from wild-type mice.
Moreover, expulsion was delayed in wild-type C57BL/6 mice compared to BALB/c mice with no sig-
nificant differences in Th2 responses, which indicates that the function of cytokines depends on genetic
background [27]. Studies with IL-4-receptor-α deficient mice proved that expression of this receptor on
bone-marrow- and non-bone-marrow-derived cells has a crucial role in worm expulsion and thereby in
host protection [28]. Another important cytokine in Th2 response—IL-13—which shares the same IL-4
receptor α chain (IL-4Rα), stimulates B cell proliferation and antibody class switching to IgE, eosino-
philia, and mastocytosis [29,30]. Binding of either of these cytokines to this receptor results in the phos-
phorylation and activation of signal transducer and activator of transcription factor 6 (Stat6), which leads
to eosinophilia, mastocytosis, and expulsion of T. spiralis [22,31]. Degranulation of mast cells that occur
in response to T. spiralis antigens increases the permeability of intestinal epithelial cells, thus promoting
parasite expulsion [32]. Signaling via Stat6 also induces intestinal smooth muscle cell hypercontractility,
another mechanism necessary for efficient worm expulsion [33]. In T. spiralis infection, intestinal muscle
hypercontractility is under control of Th2 responses [34]. Hypercontractility does not occur in athymic
mice and in animals with major histocompatibility class II or CD4+ cell deficiencies, implicating T cell
control [35,36]. A shift to Th1 response via an overexpression of IL-12 significantly inhibits T. spiralis-
induced muscle hypercontractility and delays worm expulsion [23].
Th2 cytokines IL-3 and IL-5 are also produced during T. spiralis infection [24]; however, treat-
ment with anti-IL-3 and IL-5 antibodies, or using IL-3 KO mice, revealed that these cytokines are not
796 Laboratory Models for Foodborne Infections
significant for host protection. Since IL-5 controls eosinophilia provoked by T. spiralis [37], it could be
concluded that eosinophils are not essential for the expulsion of the parasites and thus do not affect host
protection, although they exert larval cytotoxicity in T. spiralis infection [38]. However, experiments
with IL-5-deficient mice revealed that IL-5 and eosinophils are involved in rapid expulsion during chal-
lenge (secondary) T. spiralis infections [39,40]. Another role for IL-5 and eosinophils was suggested in
a model of T. spiralis infection in mouse strains that constitutively overexpress IL-5. It appears that high
levels of IL-5 and eosinophilia may be advantageous for the parasite, since the number of muscle larvae
is much higher in transgenic mice than in wild-type [41]. This was confirmed by the work of Huang
et al. [42], who pointed out immunoregulatory role of eosinophils in the early muscle phase of the infec-
tion. They have found that eosinophils produce IL-10 necessary for the expansion of IL-10-producing
DCs and CD4+ T cells, which then inhibit inducible nitric oxide synthase (iNOS) expression and the
production of NO, thus protecting intracellular larvae.
Using IL-9 transgenic mice that constitutively overexpress this cytokine, the role of IL-9 in resistance
to T. spiralis was revealed for the first time [43]. Enhanced level of IL-9 in vivo caused high levels of par-
asite-specific IgG1, marked intestinal mastocytosis, and accelerated worm expulsion. IL-9 might medi-
ate effective expulsion of T. spiralis by enhancing the release of mouse mast cell protease-1 (mMCPT-1).
The involvement of IL-9 in mucosal immunity, i.e., recruitment and/or survival of mast cells, was sug-
gested [44]. These data imply that IL-9 is a specific effector molecule involved in host defense against
T. spiralis infection.
Mast cells appear to be involved in the expulsion in nematode infections [45]. Mice in which mast cell
depletion was induced either by genetic manipulation [46] or by application of specific antibodies [47]
exhibit a significant delay in expulsion of T. spiralis. Mast cells contribute to intestinal inflammation
through the production of proinflammatory cytokines, including tumor necrosis factor, proteinases,
and other inflammatory mediators such as histamine, leukotrienes, and prostaglandins [48]. It appears
that mastocytosis in the infected mucosa may represent an immunopathological rather than a protective
response. Mice deficient in TNF receptor, infected with T. spiralis, exhibit minimal pathology in the
gut and the absence of intestinal mastocytosis, and yet they are able to expel adult worms. TNF may
mediate pathological effects in gastrointestinal helminth infections via the production of iNOS [49].
iNOS-deficient mice infected with T. spiralis show reduced pathogenic but not protective response to
T. spiralis [50].
When newborn larvae reach striated muscle tissue, they invade muscle cells and rearrange them to
form a completely new entity, the NC [7]. NC is also composed of satellite cells that undergo cell divi-
sion and join the invaded muscle cell [8,51,52]. Tissue damage provoked initially by parasite penetration
accounts also to the presence of inflammatory cells, which produce high levels of reactive oxygen species
and other free radicals, after activation [53]. These actions result in an inflammatory response, which
causes transient myositis.
Inflammatory response to various encapsulated and nonencapsulated Trichinella species differs [54].
T. pseudospiralis, a nonencapsulated species, induce a lower inflammatory reaction around the parasites
than encapsulated species (T. spiralis and T. britovi). There is also difference in the level of inflammation
between encapsulated species; that is, T. spiralis is accompanied by the higher inflammatory response,
compared to that induced by T. britovi [55].
The immunological response to T. spiralis muscle invasion in mice is mixed, although mainly Th2
type, characterized by the production of IL-5, IL-10, IL-13, and IFN-γ [56] and by the presence of
parasite-specific IgG1 and IgE during the chronic infection [57]. Eighty percent of IgG1 specific for
larval antigens recognize a single shared epitope, highly immunogenic sugar, tyvelose [58]. Although
chronic, lifelong infection in mice is characterized by persistent B cell response, local inflammatory
response to the T. spiralis–NC complex is limited, suggesting the existence of suppressive parasite
and/or host factors.
The cellular infiltrate surrounding the NC is composed of macrophages, able to invade the cytoplasm
of the NC [57]. Among other cells (CD4+ T cells, CD8+ T cells, Treg, B cells), eosinophils enter sites
of infection immediately after tissue invasion by T. spiralis larvae, and by producing IL-10, control
the activation of proinflammatory macrophages and neutrophils that otherwise kill parasite larvae by
releasing NO [42]. Inflammatory infiltrates fail to form around NCs in T-lymphocyte-deficient mice,
Trichinella 797
identifying these cells as the coordinators of the cellular response to muscle infection [59]. In IL-10
KO mice, the extent of the inflammatory infiltrate around the NC was markedly increased compared
to control mice during the acute phase of the infection, although the cellular composition remained the
same [57,60]. The other source of IL-10 that participates in reduction of inflammation is CD4+ CD25-T
effector cells [56]. Another cytokine with immunosuppressive effect on T cells is TGF-β, produced by
regulatory T cells (Treg) [61], which acts together with IL-10 to control local inflammation. When IL-10
KO mice were treated with anti-TGF-β, they develop a strong inflammation around the NC–parasite
complex with a worm burden significantly reduced compared to control mice [56]. Control of inflam-
mation in the chronic phase of the infection is IL-10-independent and could be ascribed to strong Th2
response [57].
50.3 Rat
While carnivores are considered as particular and permissive species for Trichinella infections, rats are
considered as selective hosts in which only a restricted number of species other than T. spiralis reproduce
through either natural [69,70] or experimental infection [71]. As experimental model, rats are used for the
investigation of Trichinella life cycle, host–parasite relationship, and immune response provoked by this
parasite as well as for investigation of parasite’s potential to manipulate the immune response of the host
and to suppress some severe diseases.
Since they are characterized as selective host species for Trichinella infection, rats have been used as
a model for investigation of Trichinella species-specific defense. These experiments were performed by
comparing T. spiralis and T. nativa infections in Wistar rats [71,72]. In T. spiralis-infected rats, muscle
larvae burden and in vitro newborn larvae production were many fold higher compared to T. nativa-
infected rats, which indicates that the infectivity of T. nativa is lower compared to T. spiralis. No differ-
ences between two Trichinella species were observed in intestine histology, serological response, and
weight dynamics of infected rats. This helped understanding that defense against different Trichinella
species not only is solely enteral, but also happens in the parenteral phase of the parasite’s life cycle.
Investigations of the influence of host genetics on the susceptibility to primary T. spiralis infection
on rat model system gave valuable contribution to understanding of parasitological and parasite-induced
immunological differences between strains. Rat strain variation in response to T. spiralis infection
examined on five inbred strains (AO, LOU, PVG, WKA/H, and LEWIS) was evaluated through differ-
ent rate of worm elimination in each strain [73]. LOU and LEWIS rats were the most efficient in expel-
ling T. spiralis. In these two rat strains, the lowest level of serum IgE was found, while the intestinal IgE
response was the most pronounced. Variations in rat response to T. spiralis were amended by investiga-
tion of interstrain differences with respect to muscle larvae burden between AO and DA rats [74], and it
was shown that DA rats express higher susceptibility in comparison with AO rats.
798 Laboratory Models for Foodborne Infections
Immune responses of rats to Trichinella infection during the intestinal phase involve both Th1 and
Th2 cell subsets [75]. During the muscle stage of infection, with the emphasis on rat strain differences,
T. spiralis provoked the increased level of anti-inflammatory cytokines (IL-4 and IL-10), indicating a
shift toward Th2 and regulatory response, without the restriction of Th1 type of response in DA rats,
while the response in AO rats remained in favor of Th1 type [74]. In vivo application of dendritic cells
(key cells in the initiation and regulation of immune response) primed with T. spiralis ES products
creates an immune status that resembles the one observed during Trichinella infection [76].
Research on effector mechanisms involved in protection during the intestinal phase of the T. spiralis
infection indicated that the rapid worm expulsion is IgE-mediated and T-cell-mediated [10]. Extensive
investigation of the relationship between intestinal immune responses and worm expulsion, which occur
upon the infection with T. spiralis, performed on several rat strains (BN strain—mast cell and IgE high
responders; F344—low responders on the level of mast cells and IgE activation; DA strain—intermedi-
ate responders comparing to BN and F344; Ws/Ws strain—mast cell deficient rats), revealed that mast
cells in addition to IgE could contribute to the worm expulsion following T. spiralis infection in rats [77].
Studies of host–parasite relationships in Trichinella-infected animals with respect to different media-
tors involved in infected muscle cell transformation also used rat model system. Potential differences in
signals derived or provoked by different Trichinella species (encapsulated and nonencapsulated) were
studied regarding the production of NO, a known cell-to-cell mediator. It was found that host macro-
phages differ in NO production between encapsulated and nonencapsulated Trichinella species [78].
Macrophages stimulated with T. pseudospiralis muscle larvae antigens produced increased levels of NO
only at the highest antigen concentrations, while T. spiralis antigens induced increased production of
NO even when applied in much lower concentrations compared to doses of nonencapsulated Trichinella
antigens.
Investigation of the dynamics of antibody response was performed on Wistar rats using different doses
of T. spiralis muscle larvae. It was demonstrated that rats survive the high infection doses (up to 16,000
muscle larvae) and that low doses (10 muscle larvae) can be infective and can cause detectable levels of
specific antibodies [79]. Time of seroconversion and antibody titers showed to be dose-dependent in rats.
Significant contribution to the body of evidence considering biochemical parameters of the host
and their changes during the infection with Trichinella was given by the study of lipid status, levels of
enzyme with antiatherogenic properties (paraoxonase-1), and oxidative stress in rats infected with this
parasite [80]. It was demonstrated that during the infection, serum activity of cardioprotective enzyme
paraoxanase-1 was significantly reduced accompanied with the increased levels of triglycerides and LDL
cholesterol and decreased levels of HDL.
Very important information regarding the potential of Trichinella infection or its products to manipu-
late the host immune response and to suppress some severe diseases like experimental autoimmune
encephalomyelitis (EAE), as experimental model of multiple sclerosis, were obtained in experiments
performed on DA rat model system. It was demonstrated that chronic infection with this parasite as well
as the application of Trichinella ES products, or dendritic cells stimulated with these products, has a
strong impact on the course of EAE, i.e., on the reduction of the disease severity by shifting the immune
response toward Th2 and regulatory type [81,82].
50.4 Swine
Although human Trichinella infections can be a consequence of the consumption of infected meat origi-
nating from different animal species including feral animals, T. spiralis-infected pork meat still remains
the main source of the infection [2,83].
Owing to the model of experimentally infected pigs, predilection sites for T. spiralis and other
Trichinella species were discovered in these animals [84,85]. It was established that all Trichinella
spp. accumulate preferentially in diaphragm, tongue, and masseter of infected pigs. Investigation of
infectivity of the Trichinella genotypes to pigs showed that T. spiralis is highly infective and T. britovi
and T. nelsoni are moderately infective but as persistent as T. spiralis, while T. nativa, T. murrelli,
T. pseudospiralis, and Trichinella (T6) are low infective and nonpersistent [86].
Trichinella 799
Several methods such as trichinelloscopy, artificial digestion, immunofluorescence assay (IFA), and
ELISA were compared for establishing reliable test for detection of experimental porcine trichinellosis.
It was concluded that protection of public health from Trichinella in pork and prevention of the infec-
tion spreading can be ensured only by application of parasitological methods and, in particular, artificial
digestion [87,88]. This was due to the finding that digestion detects infection as early as the larvae enter
and encapsulate in muscle tissue i.e., 17–21 days post infection (p.i.), while serological methods cannot
detect infection before seroconversion occurs, i.e., mean times were 5–6 weeks p.i. [88]. Although serol-
ogy cannot be used as a procedure to certify the safety of pork meat, it could be a useful method for
surveillance and epidemiological studies of swine Trichinella infection [89–91].
Trichinella-specific antibodies in swine could be detected by 1.5 years p.i. [92], and specific anti-
bodies raised by the infection with any of 12 different taxa of the genus Trichinella could be reliably
detected by one common antigen since all taxa share similar antigenic pattern [2,86,90,93]. The inves-
tigations performed on experimentally infected pigs provided significant impact on the development
of serological tests, and defining their performances and the identification of highly immunogenic
antigens enabled the improvement of diagnostic tests. Diagnostic tools for antibody detection in swine
evolved from application of the whole larvae in IFA, via total larvae extract and up to ES or its com-
ponents that belong to TSL-1 in ELISA and western blot [87,94,95]. The specificity of anti-Trichinella
antibodies relies on recognition of antigens from TSL-1 group, which consists of protein and unique
sugar component (tyvelose) [96]. Experimentally infected pigs are currently used for development of
international biological standards or reference materials in an aim to validate in-house tests or com-
mercial kits and to improve the interlaboratory comparability for the serological detection of anti-
Trichinella IgG in pigs [97].
Trichinella larvae recovered from experimentally infected pigs were used during development and
evaluation of specificity and sensitivity of polymerase chain reaction methods for the identification of
species or genotypes of Trichinella single muscle larvae [98,99].
Besides the use of the aforementioned outbred swine, few studies that used SLA-inbred miniature
swine proved to be an important experimental model for mapping of genetic resistance to Trichinella
infection. The influence of genes within the MHC (SLA) versus background (non-MHC) genes on the
initiation, maturation, and effector arm of host antiparasite defense mechanisms was investigated. Swine
selectively bred so that they are homozygous at their SLA complex for three SLA haplotypes, aa, cc, or
dd, were used to determine whether those genes regulate swine immune response to T. spiralis [100]. The
obtained results indicated that swine with SLAc/c haplotype have lower worm burden correlating with
the earlier development of humoral antibody response. This and work of other authors implied that this
“resistance” to infection relies not only on SLA locus (expression of one allele from SLAa haplotype) but
also on other non-SLA-encoded genes [101].
Some extension of this work performed on outbred swine indicated that besides the role of SLA
class II molecules, SLA class I phenotype also influences mechanisms that mediate the presentation of
Trichinella antigens and consequently the host immune response. Swine that expressed particular SLA I
phenotype (2–12–3 positive) found to be more resistant to infection (with lower larvae burden), compared
to negative animals [102].
Among the trials that aimed to develop vaccine against establishment of Trichinella infection in
swine, Marti et al. [106] showed that vaccine based on newborn larvae (NBL) antigen was considerably
more protective than the one based on muscle larvae stichosomal fractions or ES proteins [107,108].
Experimentally infected swine are currently in use for determination of specific epitopes on NBL [109],
adult and muscle larvae antigens [110], targeted by host antibodies, which could potentially be used for
new approach to vaccine development as well as for early diagnosis of Trichinella infection.
50.5 Wild Boar
Wild boar infections with both domestic and sylvatic Trichinella spp. occur quite frequently in Europe,
North America, Asia, and Africa, and infected meat of this animal species presents an important risk fac-
tor for human Trichinella infection [111,112]. First experimental Trichinella infection was performed to
determine infectivity, muscle larvae distribution, and ability of investigated species to generate immune
response in wild boars [113]. This study revealed that T. spiralis is highly infective for wild boars;
T. britovi, T. nelsoni, and T. pseudospiralis are moderately infective, while T. nativa, T. murrelli, and
Trichinella T6 are not well adapted for this host species. Predilection sites, independently of Trichinella
genotype and infective dose, are diaphragm and tongue [113,114]. Antibody response increased rapidly
from week 3 to 5 p.i. for all investigated Trichinella genotypes except for T. pseudospiralis where it
increased gradually. For encapsulated species of Trichinella, high antibody level remained until the end
of examined period (10 weeks), whereas for nonencapsulated species, antibody levels declined during the
postinfection period. Data concerning wild boars correlate with those observed in domestic pig model
system [86].
A long-lasting experiment was designed using wild boars to investigate the freezing tolerance of
Trichinella-encapsulated muscle larvae T. spiralis and T. britovi [115]. It was demonstrated that muscle
larvae of both Trichinella species could be inactivated after 1 week on −21 ± 2°C. In contrast to this find-
ing, Gari-Toussaint et al. [116] reported six cases of human trichinellosis originating from the consump-
tion of T. britovi-infected wild boar meat that had been frozen for 1 week at −35°C. It has been suggested
that wild animals’ muscles contain substances that protect Trichinella muscle larvae from freezing [117].
Hence, although the study on experimentally infected wild boars provided some new data on Trichinella
freezing tolerance, freezing method is not recommended as a control measure to inactivate this parasite
in game meat [89].
50.6 Horse
Infected horse meat has been recognized as a source of human trichinellosis since 1975 when the out-
breaks were reported for the first time in Italy and France [118,119]. Because horse has been considered
as an atypical host for Trichinella spp., many aspects of the biology and epidemiology of Trichinella
infection in the horse were poorly investigated and understood during the previous century. Still, being
recognized as a serious food safety risk for consumers of fresh horse meat, the infection was started to be
investigated in experimental horse model from the 1970s. Those very early studies were aimed to detect
muscle larvae and specific antibodies presence during the course of the infection, but latter investigations
included the route of infection, tissue distribution of muscle larvae, parasite life span, and assessment
of reliability of parasitological and serological diagnostic methods for the detection of infected animals.
The first report on distribution of Trichinella muscle larvae (L1) in individual muscles was published
by Pampiglione et al. [120] referring to the examination performed on 6-month-old horses infected
with 10,000 L1 in which the highest muscle larvae burden was detected in tongue, masseter, and dia-
phragm. In this millennium, all three aforementioned muscles were confirmed to be predilection sites
for Trichinella muscle larvae [114]. The study included 30 horses experimentally infected with three
Trichinella genotypes: T. spiralis, T. britovi, and T. pseudospiralis. Limited study on three ponies, [121]
Trichinella 801
as well as studies of other authors [92,122,123], corroborated that the tongue is the main predilection
site. Results obtained in experimentally infected horses were in accordance with those observed in natu-
rally infected animals [124], and they supported the contention that food safety inspection of the horse
meat should target the tongue as a tissue most promising to yield positive results in horses infected with
Trichinella.
Several studies have been undertaken to describe the infection in this atypical host [123,125–127].
Horses mostly do not develop the disease during Trichinella infection, except the rare transient muscular
disturbance observed between 2 and 6 weeks p.i. in case of animals infected with a high dose (50,000)
of L1 larvae. Moreover, hematological parameters were not significantly changed, except for the level of
eosinophils, which was slightly increased (up to 15%) independently of infective larvae dose. Levels of
enzymes that indicate the muscle damage (lactate-dehydrogenase, aldolase, and creatine phosphokinase)
were increased from 2 to 17 weeks p.i. [125].
Experimental infection of horses with Trichinella spp. was used for the examination of kinetics of
antibody production and dependence of antibody response on different Trichinella spp. that cause the
infection in horses. Detection of specific, anti-Trichinella antibodies depend on Trichinella species
(T. spiralis induced higher level of anti-Trichinella IgG than T. murrelli due to its antigenicity), the infec-
tive dose applied for horse infection, as well as on the antigen used for antibody detection [124]. A num-
ber of studies demonstrated that in high-dose infection (more than 20,000 L1), anti-Trichinella-specific
IgG appears between 2 and 5 weeks p.i. and disappears between 16 and 40 weeks p.i. [120,125,128],
judging by indirect IFA and ELISA tests. Low doses of infective larvae (1100 L1) also induce antibody
production detectable by ELISA for up to 18 weeks p.i. [129]. The short period of time (in average up to
26 weeks p.i.) during which anti-Trichinella antibodies in infected horses could be detected by ELISA
makes this kind of test unreliable for detection of Trichinella infection in horses [119,125,130]. On the
other hand, other serological methods were proven to be effective in specific antibody detection for a
longer period (IFA of paraffin sections of L1 and western blot analysis of ES and tyvelose-BSA, at least
8 months) [129]. It was supposed that for diagnosis of horse trichinellosis, additional tests including
antigen detection should be performed. One such test was a dot-blot test for the detection of Trichinella
ES muscle larvae antigens in circulation, which allowed the detection between weeks 4 and 32 p.i. in the
sera of all experimentally infected horses [131].
The most extensive and long-term experimental study was performed on 35 horses infected with 1000,
5000, or 10,000 T. spiralis muscle larvae in which the course of infection was followed for 1 year [92].
It was shown that the persistence of infective muscle larvae is at least 1 year, but serological response
could be detected only by 26 weeks p.i. (ELISA with ES). This study offered the conclusion that parasite
recovery methods are the only suitable detection assays for both meat inspection and epidemiological
studies of Trichinella infection in horses.
The question considering the enigma of early disappearance of detectable antibody response in horses
versus the existence of live muscle larvae raised by this research could partly find the answer in data
showing the presence of seven IgG subclasses in horse and five other isotypes [132,133] and the presump-
tion that anti-horse IgG used in previous studies may be inappropriate for detecting subclass-specific
anti-Trichinella reactivity in the horse [92].
Since horses are herbivores and nevertheless, natural infections with Trichinella occur from time to
time, an intriguing question considering the source of infection emerged. Eating behavior of these ani-
mals studied by intentional feeding of horse with meat products [134] revealed that 31% of horses readily
consumed it and that this feeding practice could be a source of Trichinella infection.
Studies comparing the muscle distribution of different Trichinella genotypes were performed, among
other species, on foxes. In these animals, where all Trichinella species were established in high numbers,
the encapsulating species were found primarily in the tongue, extremities, and diaphragm, whereas the
nonencapsulating species were found primarily in the diaphragm [114]. Foxes were one of the experi-
mental models used for studies of Trichinella larvae freeze tolerance. Since T. nativa is the most frequent
Trichinella species in arctic wildlife and well adapted to the cold climate, it was useful for testing the
tolerance on repeated freezing and thawing. Results obtained in experimentally infected foxes showed
that T. nativa isolates originating from carnivores from higher northern latitudes expressed highest toler-
ance to freezing and that temperature fluctuation around freezing point did not influence larval infectiv-
ity [135]. Foxes were also experimentally infected with T. nativa for the purpose of serological survey,
which showed that western blot is a more reliable method compared to ELISA for antibody detection
in this animal species [136]. Potential of Trichinella to cross the placental barrier was investigated on
experimentally infected foxes, and it was found that, unlike some other animal species (ferrets, guinea
pigs, and mice), vertical transmission, measured as recovery of muscle larvae in the offspring, does not
happen in foxes [137].
Since marine mammals may acquire Trichinella infection in nature by scavenging even small amounts
of infected tissue left by hunters or predators, the important data considering infection in this group of
animals were obtained on seals experimentally infected with T. nativa in order to determine the suscepti-
bility of seals to this parasite infection and freeze tolerance of larvae in the seal meat [138]. Seals showed
high susceptibility to Trichinella infection and measurable antibody response unrelated to the infection
dose. Experiments investigating freeze tolerance of muscle larvae in the meat indicated that there was
correlation between resistance to freezing and the age of host–parasite tissue complex; that is, freeze
tolerance increases with time post infection. The results obtained in this study were complemented with
research in which the meat of seals experimentally infected with T. nativa was used for the preparation of
traditional northern foods in an aim to assess the infectivity of larvae present in those foods [139]. Data
indicated significant food safety risk associated with the food containing infected seal meat.
One of the most unusual experimental models for trichinellosis is caiman. Notification of Trichinella
zimbabwensis (nonencapsulated species) infection in farmed crocodiles in Zimbabwe due to the feeding
practice [140] raised a question of susceptibility of reptiles to infection with other Trichinella spp. The
experimental infection of caimans with different Trichinella isolates showed that these animals can be
infected with nonencapsulated T. pseudospiralis, which is, from evolutionary point of view, likely to
infect reptiles since it can be infective for both birds and mammals, while the infection with encapsu-
lated species was impossible [141].
50.8 Conclusion
Animal models for Trichinella and trichinellosis are a tool that allows detailed investigation of host–
parasite relationships. The host’s susceptibility to different Trichinella genotypes indicates the impact of
genetic background of the host and the parasite on the parasitism establishment. The vast range of exper-
imental animal models reveals insight into parasite muscle distribution and predilection sites, which
form the basis of meat sample properties for accurate parasitological detection and diagnosis. Studies
using experimental Trichinella infection yield valuable information on the course of innate and adaptive
immune response. Knowledge about dynamic of specific antibody synthesis provides a guide for design-
ing and implementing specific and sensitive serological tests in surveillance, monitoring, and control
programs. The rodent models highlight the importance of antigen-presenting cells to drive T-helper
type 2 and T-regulatory responses in Trichinella infection that are responsible for parasite long-lasting
survival and creation of tolerogenic environment in the host body. Animal models have been, and will
continue to be, vital in helping elucidate the mechanisms that underline the host–parasite coevolution,
which is, among others, responsible for the training of host immune systems for accurate discrimination
between foreign and self-antigens.
Trichinella 803
REFERENCES
1. Gottstein B, Pozio E, Nöckler K. Epidemiology, diagnosis, treatment, and control of trichinellosis. Clin
Microbiol Rev. 2009; 22:127–45.
2. Pozio E, Zarlenga DS. New pieces of the Trichinella puzzle. Int J Parasitol. 2013; 43:983–97.
3. Pozio E, Hoberg E, La Rosa G, Zarlenga DS. Molecular taxonomy, phylogeny and biogeography of
nematodes belonging to the Trichinella genus. Infect Genet Evol. 2009; 9:606–16.
4. Murrell KD, Pozio E. Worldwide occurrence and impact of human trichinellosis, 1986–2009. Emerg
Infect Dis. 2011; 17:2194–202.
5. Devleesschauwer B, et al. The low global burden of trichinellosis: evidence and implications. Int J
Parasitol. 2015; 45:95–9.
6. Bruschi F, Gómez-Morales MA. The translational immunology of trichinellosis: from rodents to humans.
In: Jirillo E, Magrone T, Miragliotta G. (eds) Immune Response to Parasitic Infections: Immunity to
Helminths and Novel Therapeutic Approaches. Bantham E-Books, Beijing, 2014; 2:125–61.
7. Despommier DD. How does Trichinella spiralis make itself at home. Parasitol Today. 1998; 14:318–23.
8. Wu Z, Sofronic-Milosavljevic L, Nagano I, Takahashi Y. Trichinella spiralis: nurse cell formation with
emphasis on analogy to muscle cell repair. Parasites Vectors. 2008; 1:27–41.
9. Wakelin D. Helminth infections. In: Wakelin D, Blackwell JM. (eds) Genetics of Resistance to Bacterial
and Parasitic Infection. Taylor & Francis, London, 1988; 153–224.
10. Bell, RG. The generation and expression of immunity to Trichinella spiralis in laboratory rodents. In:
Baker JR. (ed) Advance in Parasitology. Academic press, London, 1998; 149–217.
11. Bell RG, Adams LS, Ogden RW. Trichinella spiralis: genetics of worm expulsion in inbred and F1 mice
infected with different worm doses. Exp Parasitol. 1984; 58:345–55.
12. Wassom DL, David CS, Gleich GJ. Genes within the major histocompatibility complex influence sus-
ceptibility to Trichinella spiralis in the mouse. Immunogenetics. 1979; 9:491–6.
13. Wakelin D, Donachie AM. Genetic control of immunity to parasites: adoptive transfer of immunity
between inbred strains of mice characterized by rapid and slow immune expulsion of Trichinella spira-
lis. Parasite Immunol. 1980; 2:249–60.
14. Wassom DL, et al. Genetic control of immunity to Trichinella spiralis infection of mice. Hypothesis to
explain the role of H-2 genes in primary and challenge infections. Immunology. 1984; 51:625–31.
15. Wassom DL, Brooks BO, Cypess RH. Trichinella spiralis: role of Non-H-2 genes in resistance to pri-
mary infection in mice. Exp Parasitol. 1983; 55:153–8.
16. Bell RG. Genetic analysis of expulsion of adult Trichinella spiralis in NFS, C3H/He, and 810 l BR mice.
Exp Parasitol. 1988; 66:57–65.
17. Artis D, Grencis RK. T helper cell cytokine responses during intestinal nematode infection: induction,
regulation and effector function. In: Kennedy MW, Harnett W. (eds) Parasitic Nematodes. Molecular
Biology, Biochemistry and Immunology. CABI Publishing, Wallingford, UK, 2001; 331–71.
18. Dehlawi MS, Goyal PK. Responses of inbred mouse strains to infection with intestinal nema-
todes. J Helminthol. 2003; 77:119–24.
19. Ruitenberg EJ, Elgersma A. Absence of intestinal mast cell response in congenitally athymic mice dur-
ing Trichinella spiralis infection. Nature. 1976; 264(5583):258–60.
20. Grencis RK, Riedlinger J, Wakelin D. L3T4-positive T lymphoblasts are responsible for transfer of
immunity to Trichinella spiralis in mice. Immunology. 1985; 56:213–18.
21. Ishikawa N, et al. Early cytokine responses during intestinal parasitic infections. Immunology. 1998;
93:257–63.
22. Urban JF, Jr., et al. Stat6 signaling promotes protective immunity against Trichinella spiralis through a
mast cell- and T cell-dependent mechanism. J Immunol. 2000; 164:2046–52.
23. Khan WI, et al. IL-12 gene transfer alters gut physiology and host immunity in nematode-infected mice.
Am J Physiol Gastrointest Liver Physiol. 2001; 81:G102–10.
24. Grencis RK, Hültner L, Else KJ. Host protective immunity to Trichinella spiralis in mice: activa-
tion of Th cell subsets and lymphokine secretion in mice expressing different response phenotypes.
Immunology. 1991; 74:329–32.
25. Lawrence CE, et al. IL-4 regulated enteropathy in an intestinal nematode infection. Eur J Immunol.
1998; 28:2672–84.
804 Laboratory Models for Foodborne Infections
26. Finkelman FD, et al. Cytokine regulation of host defense against parasitic gastrointestinal helminths:
lessons from studies with rodent models. Ann Rev Immunol. 1997; 15:505–33.
27. Scales HE, Ierna MX, Lawrence CE. The role of IL-4, IL-13 and IL-4Rα in the development of protec-
tive and pathological responses to Trichinella spiralis. Parasite Immunol. 2007; 29:81–91.
28. Urban JF, Jr, et al. Cutting edge: IL-4 receptor expression by non-bone marrow derived cells is required
to expel gastrointestinal nematode parasites. J Immunol. 2001; 167:6078–81.
29. Barner M, Mohrs M, Brombacher F, Kopf M. Differences between IL-4R α-deficient and
IL-4-deficient mice reveal a role for IL-13 in the regulation of Th2 responses. Curr Biol. 1998;
8:669–72.
30. Emson CL, et al. Interleukin (IL)-4-independent induction of immunoglobulin (Ig) E, and perturbation
of T cell development in transgenic mice expressing IL-13. J Exp Med. 1998; 188:399–404.
31. Khan WI, et al. Stat6 dependent goblet cell hyperplasia during intestinal nematode infection. Parasite
Immunol. 2001; 23:39–42.
32. McDermott JR, et al. Mast cells disrupt epithelial barrier function during enteric nematode infection.
Proc Natl Acad Sci USA. 2003; 100:7761–6.
33. Khan WI, et al. Critical role for signal transducer and activator of transcription factor 6 in mediating
intestinal muscle hypercontractility and worm expulsion in Trichinella spiralis-infected mice. Infect
Immun. 2001; 69:838–44.
34. Khan WI, et al. Disruption of CD40-CD40 ligand pathway inhibits the development of intestinal muscle
hypercontractility and protective immunity in nematode infection. Am J Physiol Gastrointest Liver
Physiol. 2005; 288:G15–22.
35. Vallance BA, Croitoru K, Collins SM. T lymphocyte-dependent and -independent intestinal smooth
muscle dysfunction in the T. spiralis-infected mouse. Am J Physiol. 1998; 275:G1157–65.
36. Vallance BA, Galeazzi F, Collins SM, Snider DP. CD4 T cells and major histocompatibility com-
plex class II expression influence worm expulsion and increased intestinal muscle contraction during
Trichinella spiralis infection. Infect Immun. 1999; 67:6090–97.
37. Herndon FJ, Kayes SG. Depletion of eosinophils by anti-IL-5 monoclonal antibody treatment of mice
infected with Trichinella spiralis does not alter parasite burden or immunologic resistance to reinfec-
tion. J Immunol. 1992; 149:3642–47.
38. Gurish MF, et al. CCR3 is required for tissue eosinophilia and larval cytotoxicity after infection with
Trichinella spiralis. J Immunol. 2002; 168:5730–6.
39. Vallance BA, Matthaei KI, Sanovic S, Young IG, Collins SM. Interleukin-5 deficient mice exhibit
impaired host defence against challenge Trichinella spiralis infections. Parasite Immunol. 2000;
22:487–92.
40. Huang L, et al. Eosinophils mediate protective immunity against secondary nematode infection.
J Immunol. 2015; 194:283–90.
41. Dent LA, et al. Immune responses of IL-5 transgenic mice to parasites and aeroallergins. Mem Inst
Oswaldo Cruz. 1997; 92:45–54.
42. Huang L, et al. Eosinophil-derived IL-10 supports chronic nematode infection. J Immunol. 2014;
193(8):4178–87.
43. Faulkner HF, et al. Interleukin-9 is involved in host-protective immunity to intestinal nematode infec-
tion. Eur J Immunol. 1997; 27:2536–40.
44. Veldhoen M, et al. Transforming growth factor-β “reprograms” the differentiation of T helper 2 cells
and promotes an interleukin 9-producing subset. Nat Immunol. 2008; 9:1341–6.
45. Brown JK, Donaldson DS, Wright SH, Miller HR. Mucosal mast cells and nematode infection: strain
specific differences in mast cell precursor frequency revisited. J Helminthol. 2003; 77:155–61.
46. Ha TY, Reed ND, Crowle PK. Delayed expulsion of adult Trichinella spiralis by mast cell-deficient
W/Wv mice. Infect Immun. 1983; 41:445–7.
47. Donaldson LE, et al. A critical role for stem cell factor and c-kit in host protective immunity to an intes-
tinal helminth. Int Immunol. 1996; 8:559–67.
48. Garside P, Kennedy MW, Wakelin D, Lawrence CE. Immunopathology of intestinal helminth infection.
Parasite Immunol. 2000; 22:605–12.
49. James SL. Role of nitric oxide in parasitic infections. Microbiol Rev. 1995; 59:533–47.
50. Lawrence CE, et al. Nitric oxide mediates intestinal pathology but not immune expulsion during
Trichinella spiralis infection in mice. J Immunol. 2000; 164:4229–34.
Trichinella 805
51. Matsuo A, Wu Z, Nagano I, Takahashi Y. Five types of nuclei present in the capsule of Trichinella spi-
ralis. Parasitology. 2000; 121:203–10.
52. Wu Z, et al. Different response of satellite cells in the kinetics of myogenic regulatory factors and
ultrastructural pathology after Trichinella spiralis and T. pseudospiralis infection. Parasitology. 2001;
123:85–94.
53. Chiumiento L, Bruschi F. Enzymatic antioxidant systems in helminth parasites. Parasitol Res. 2009;
105:593–603.
54. Boonmars T, Wu Z, Nagano I, Takahashi Y. Trichinella pseudospiralis infection is characterized by
more continuous and diffuse myopathy than T. spiralis infection. Parasitol Res. 2005; 97:13–20.
55. Bruschi F, Marucci G, Pozio E, Masetti M. Evaluation of inflammatory responses against muscle larvae
of different Trichinella species by an image analysis system. Vet Parasitol. 2009; 159:258–62.
56. Beiting BP, et al. Coordinated control of immunity to muscle stage Trichinella spiralis by IL-10, regula-
tory T cells, and TGF-β. J Immunol. 2007; 178:1039–47.
57. Beiting BP, et al. Interleukin-10 limits local and body cavity inflammation during infection with muscle-
stage Trichinella spiralis. Infect Immun. 2004; 72:3129–37.
58. Reason AJ, et al. Novel tyvelose-containing tri- and tetra-antennary N-glycans in the immunodominant
antigens of the intracellular parasite Trichinella spiralis. Glycobiology. 1994; 4:593–603.
59. Walls RS, Carter RL, Leuchars E, Davies AJ. The immunopathology of trichiniasis in T-cell deficient
mice. Clin Exp Immunol. 1973; 13:231–42.
60. Fabre MV, Beiting DP, Bliss SK, Appleton JA. Immunity to Trichinella spiralis muscle infection. Vet
Parasitol. 2009; 159:245–48.
61. Nakamura K, Kitani A, Strober W. Cell contact–dependent immunosuppression by CD4 + CD25+ reg-
ulatory T cells is mediated by cell surface–bound transforming growth factor β. J Exp Med. 2001;
194:629–44.
62. García A, et al. Novel albendazole formulations given during the intestinal phase of Trichinella spiralis
infection reduce effectively parasitic muscle burden in mice. Parasitol Int. 2013; 62:568–70.
63. Basyoni MM, El-Sabaa AA. Therapeutic potential of myrrh and ivermectin against experimental
Trichinella spiralis infection in mice. Korean J Parasitol. 2013; 51:297–304.
64. Codina AV, et al. Efficacy of albendazole:β-cyclodextrin citrate in the parenteral stage of Trichinella
spiralis infection. Int J Biol Macromol. 2015; 77:203–6.
65. Sofronic-Milosavljevic L, Ilic N, Pinelli E, Gruden-Movsesijan A. Secretory products of Trichinella
spiralis muscle larvae and immunomodulation: implication for autoimmune diseases, allergies, and
malignancies. J Immunol Res. 2015; 2015:523875.
66. Dea-Ayuela MA, Rama-Iniguez S, Torrado-Santiago S, Bolas-Fernandez F. Microcapsules formulated
in the enteric coating copolymer Eudragit L100 as delivery systems for oral vaccination against infec-
tions by gastrointestinal nematode parasites. J Drug Target. 2006; 14:567–75.
67. Ortega-Pierres G, Muniz E, Coral-Vazquez R, Parkhouse RM. Protection against Trichinella spi-
ralis induced by purified stage-specific surface antigens of infective larvae. Parasitol Res. 1989;
75:563–7.
68. Ortega-Pierres G, et al. Induction of protection in murine experimental models against Trichinella spi-
ralis: an up-to-date review. J Helminthol. 2015; 89(5):526–39.
69. Oivanen L, et al. Associations between Trichinella species and host species in Finland. J Parasitol.
2002; 88:84–8.
70. Dick TA, Pozio E. Trichinella spp. and trichinellosis. In: Samuel WM, Pybus MJ, Kocan AA. (eds)
Parasitic Diseases of Wild Mammals, 2nd edn. Iowa State University Press, Ames, IA, 2008; 380–96.
71. Pozio E, La Rosa G, Rossi P, Murrell KD. Biological characterization of Trichinella isolates from vari-
ous host species and geographical regions. J Parasitol. 1992; 78:647–53.
72. Airas N, et al. The different infectivity of Trichinella spiralis and Trichinella nativa in rat does not
solely localize to enteral or parenteral phase. Parasitol Res. 2012; 111:2281–8.
73. Negrao-Correa D, Adams LS, Bell RG. Variability of the intestinal immunoglobulin E response of
rats to infection with Trichinella spiralis, Heligmosomoides polygyrus or Nippostrongylus brasiliensis.
Parasite Immunol. 1999; 21:287–97.
74. Vasilev S, Gruden-Movsesijan A, Ilic N, Sofronic-Milosavljevic L. Strain difference in susceptibil-
ity to Trichinella spiralis infection between dark agouti and albino oxford rats. Vet Parasitol. 2009;
159:229–31.
806 Laboratory Models for Foodborne Infections
75. Stewart GL, Na H, Smart L, Seelig Jr. LL. The temporal relationship among anti-parasite immune
elements expressed during the early phase of infection of the rat with Trichinella spiralis. Parasitol Res.
1999; 85:672–7.
76. Gruden-Movsesijan A, et al. The impact of Trichinella spiralis excretory-secretory products on den-
dritic cells. Comp Immunol Microbiol Infect Dis. 2011; 34:429–39.
77. Suzuki T, et al. The effectors responsible for gastrointestinal nematode parasites, Trichinella spiralis,
expulsion in rats. Parasitol Res. 2008; 103:1289–95.
78. Andrade AM, et al. Trichinella: differing effects of antigens from encapsulated and non-encapsulated
species on in vitro nitric oxide production. Vet Parasitol. 2007; 143:86–90.
79. Franssen FJ, et al. Antibody response against Trichinella spiralis in experimentally infected rats is dose
dependent. Vet Res. 2011; 42:113–22.
80. Mido S, et al. Trichinella spiralis: infection changes serum paraoxonase-1 levels, lipid profile, and
oxidative status in rats. Exp Parasitol. 2012; 131:190–4.
81. Gruden-Movsesijan A, et al. Trichinella spiralis: modulation of experimental autoimmune
encephalomyelitis in DA rats. Exp Parasitol. 2008; 118:641–7.
82. Radovic I, et al. Immunomodulatory effects of Trichinella spiralis-derived excretory—secretory
antigens. Immunol Res. 2015; 61:312–25.
83. Murrell KD. Strategies for the control of human trichinosis transmitted by pork. Food Technol. 1985;
39:65–8.
84. Zimmermann WJ. Reproductive potential and muscle distribution of Trichinella spiralis in swine. J Am
Vet Med Assoc. 1970; 156:770–4.
85. Nöckler K, et al. Experimental studies in pigs on Trichinella detection in different diagnostic matrices.
Vet Parasitol. 2005; 132:85–90.
86. Kapel CM, Gamble HR. Infectivity, persistence, and antibody response to domestic and sylvatic
Trichinella spp. in experimentally infected pigs. Int J Parasitol. 2000; 30:215–21.
87. Van Knapen F, et al. Comparison of four methods for early detection of experimental Trichinella spira-
lis infection in pigs. Vet Parasitol. 1981; 9:117–23.
88. Gamble HR. Sensitivity of artificial digestion and enzyme immunoassay methods of inspection for
trichinae in pigs. J Food Prot. 1998; 61:339–343.
89. Gamble HR, et al. International Commission on Trichinellosis: recommendations on methods for the
control of Trichinella in domestic and wild animals intended for human consumption. Vet Parasitol.
2000; 93:393–408.
90. Gamble HR, et al. 2004. International Commission on Trichinellosis: recommendations on the use of
serological tests for the detection of Trichinella infection in animals and man. Parasite. 2004; 11:3–13.
91. European Commission. Commission Regulation (EC) No. 2075/2005 of the European Parliament and
of the Council of 5 December 2005 Laying down specific rules on official controls for Trichinella in
meat. Off J Eur Comm L. 2005; 338:60–82.
92. Hill DE, et al. Larval viability and serological response in horses with long-term Trichinella spiralis
infection. Vet Parasitol. 2007; 146:107–16.
93. Ortega-Pierres G, et al. Workshop on a detailed characterization of Trichinella spiralis antigens: a plat-
form for future studies on antigens and antibodies to this parasite. Parasite Immunol. 1996; 18:273–84.
94. Smith HJ. Evaluation of the ELISA for serological diagnosis of trichinosis in Canadian swine. Can J Vet
Res. 1987; 51:194–7.
95. Ilic N, Gruden-Movsesijan A, Zivojinovic M, Sofronic-Milosavljevic L. Characteristic band pattern in
western blots for specific detection of anti-Trichinella spiralis antibodies in different host species. Acta
Vet (Beograd). 2014; 64:33–43.
96. Yepez-Mullia L, et al. Contributions to the study of Trichinella spiralis TSL-1 antigens in host immunity.
Parasite Immunol. 2007; 29:661–70.
97. Gómez-Morales MA, Ludovisi A, Amati M, Pozio E. Candidates for reference swine serum with
anti-Trichinella antibodies. Vet Parasitol. 2015; 208:218–24.
98. Pozio E, Kapel CMO, Gamble HR. Specificity and sensitivity of amplified polymorphic DNA analyses
for the identification of single muscle larvae of Trichinella after experimental infection of pigs. Parasitol
Res. 1999; 85:504–6.
99. Guenther S, et al. Detection of Trichinella spiralis, T. britovi and T. pseudospiralis in muscle tissue with
real-time PCR. J Microbiol Methods. 2008; 75:287–92.
Trichinella 807
100. Lunney JK, Murrell KD. Immunogenetic analysis of Trichinella spiralis infection in swine. Vet
Parasitol. 1988; 29:179–93.
101. Madden KB, et al. Trichinella spiralis: genetic basis and kinetics of the anti-encysted muscle larval
response in miniature swine. Exp Parasitol. 1993; 77:23–35.
102. Bugarski D, Sofronic-Milosavljevic L, Popovic L, Cuperlovic K. Changes in peripheral blood mono-
nuclear cell subsets during Trichinella spiralis infection in pigs. In: Cambell V, Pozio E, Bruschi F. (eds)
Proceedings of 8th ICT on Trichinellosis. Istituto Superiore di Sanita Press, Rome, 1993; 413–6.
103. Frontera E, et al. Concurrent infection with Trichinella spiralis and other helminths in pigs. Vet
Parasitol. 2007; 146:50–7.
104. Bokken G, Bergwerff A, van Knapen F. A novel bead-based assay to detect specific antibody responses
against Toxoplasma gondii and Trichinella spiralis simultaneously in sera of experimentally infected
swine. BMC Vet Res. 2012; 8:36.
105. Liu P, et al. Screening of early antigen genes of adult-stage Trichinella spiralis using pig serum from
different stages of early infection. Vet Parasitol. 2013; 194:222–5.
106. Marti HP, Murrell KD, Gamble HR. Trichinella spiralis: immunization of pigs with newborn larval
antigens. Exp Parasitol. 1987; 63:68–73.
107. Murrell KD, Despommier DD. Immunization of swine against Trichinella spiralis. Vet Parasitol. 1984;
15:263–70.
108. Gamble HR, Murrell KD, Marti HP. Immunization of pigs against Trichinella spiralis using excretory-
secretory antigens. Am J Vet Res. 1986; 47:2396–9.
109. Yang Y, et al. Identification and characterization of immunodominant linear epitopes on the antigenic
region of a serine protease in newborn Trichinella larvae. J Helminthol. 2015; 90(2):232–7. doi:10.1017/
S0022149X15000267.
110. Bien J, Cabaj W, Moskwa B. Proteomic analysis of potential immunoreactive proteins from muscle
larvae and adult worms of Trichinella spiralis in experimentally infected pigs. Folia Parasitol (Praha).
2015; 62. pii:2015.022.
111. Jongwutiwes S, et al. First outbreak of human trichinellosis caused by Trichinella pseudospiralis. Clin
Infect Dis. 1998; 26:111–5.
112. Pozio E, Kapel CMO. Trichinella nativa in sylvatic wild boar. J Helminthol. 1999; 73:87–9.
113. Kapel CM. Sylvatic and domestic Trichinella spp. in wild boars; infectivity, muscle larvae distribution,
and antibody response. J Parasitol. 2001; 87:309–14.
114. Kapel CM, Webster P, Gamble HR. Muscle distribution of sylvatic and domestic Trichinella larvae in
production animals and wildlife. Vet Parasitol. 2005; 132:101–5.
115. Lacour SA, et al. Freezing tolerance of Trichinella muscle larvae in experimentally infected wild boars.
Vet Parasitol. 2013; 194:175–8.
116. Gari-Toussaint M, et al. Human trichinellosis due to Trichinella britovi in southern France after con-
sumption of frozen wild boar meat. Euro Surveill. 2005; 10:117–18.
117. Dick TA. Infectivity of isolates of Trichinella and the ability of an arctic isolate to survive freezing
temperatures in the raccoon, Procyon lotor, under experimental conditions. J Wildl Dis. 1983; 19:333–6.
118. Mantovani A, Filippini I, Bergomi S. Indagini su un’epidemia di trichinellosi umana verificatasi in
Italia. Parassitologia (Rome). 1980; 22:107–32.
119. Boireau P, et al. Trichinella in horses: a low frequency infection with high human risk. Vet Parasitol.
2000; 93:309–20.
120. Pampiglione S, et al. Infezione sperimentale del cavallo con larve di trichina. Parassitologia (Rome).
1978; 20:183–93.
121. Smith HJ, Snowdon KE. Detection of Trichinella spiralis native antibodies in porcine sera by ELISA
using T. spiralis spiralis excretory-secretory antigen. Can J Vet Res. 1987; 51:413–4.
122. Gamble HR, Gajadhar AA, Solomon MB. Methods for the detection of trichinellosis in horses. J Food
Prot. 1996; 59:420–5.
123. Pozio E, et al. Predilection sites of Trichinella spiralis larvae in naturally infected horses. J Helminthol.
1999; 73:233–7.
124. Pozio E, et al. Evaluation of ELISA and western blot analysis using three antigens to detect anti-
Trichinella IgG in horses. Vet Parasitol. 2002; 108:163–78.
125. Soule C, et al. Experimental trichinellosis in horses: biological and parasitological evaluation. Vet
Parasitol. 1989; 31:19–36.
808 Laboratory Models for Foodborne Infections
126. Soule C, et al. Biological and parasitic variations in horses infested and reinfected by Trichinella
spiralis. Vet Res. 1993; 24:21–31.
127. Pozio E, et al. Distribution of Trichinella spiralis larvae in muscles from a naturally infected horse. Vet
Parasitol. 1998; 74:19–27.
128. Van Knapen F, Franchimont JH, Hendriks WML, Eysker M. Experimental Trichinella spiralis infection
in two horses. In: Geerts S, Kumar V, Brandt J. (eds) Helminth Zoonoses. Martinus Nijhoff Publisher,
Dordrecht, 1987; 192–201.
129. Sofronic-Milosavljevic L, et al. Anti-Trichinella antibodies detected in chronically infected horses by
IFA and western blot, but not by ELISA. Vet Parasitol. 2005; 132:107–11.
130. Yepez-Mullia L, et al. Detection of Trichinella infection in slaughter horses by ELISA and western blot
analysis. Vet Parasitol. 1999; 81:57–68.
131. Ilic N, Petrovic M, Djordjevic M, Sofronic-Milosavljevic L. A novel dot blot test for Trichinella spiralis
antigen detection. Acta Veterinaria (Beograd). 2004; 54:301–10.
132. Sugiura T, Kondo T, Imagawa H, Kamada M. Production of monoclonal antibodies to six isotypes of
horse immunoglobulin. Vet Immunol Immunopathol. 1998; 62:145–51.
133. Keggan A, Freer H, Rollins A, Wagner B. Production of seven monoclonal equine immunoglobulins
isotyped by multiplex analysis. Vet Immunol Immunopathol. 2013; 153:187–93.
134. Murrell KD, et al. Epidemiology of Trichinella infection in the horse: the risk from animal product feed-
ing practices. Vet Parasitol. 2004; 123:223–33.
135. Davidson RK, Handeland K, Kapel CM. High tolerance to repeated cycles of freezing and thawing in
different Trichinella nativa isolates. Parasitol Res. 2008; 103:1005–10.
136. Davidson RK, Opertveit I, Moller L, Kapel CM. Serological detection of anti-Trichinella antibodies in
wild foxes and experimentally infected farmed foxes in Norway. Vet Parasitol. 2009; 163:93–100.
137. Webster P, Kapel CM. Studies on vertical transmission of Trichinella spp. in experimentally infected
ferrets (Mustela putorius furo), foxes (Vulpes vulpes), pigs, guinea pigs and mice. Vet Parasitol. 2005;
130:255–62.
138. Kapel CM, et al. Experimental Trichinella infection in seals. Int J Parasitol. 2003; 33:1463–70.
139. Forbes LB, Measurel L, Gajadhar A, Kapel CM. Infectivity of Trichinella nativa in traditional northern
(country) foods prepared with meat from experimentally infected seals. J Food Prot. 2003; 66:1857–63.
140. Pozio E, et al. Trichinella zimbabwensis n.sp. (Nematoda), a new non-encapsulated species from croco-
diles (Crocodylus niloticus) in Zimbabwe also infecting mammals. Int J Parasitol. 2002; 32:1787–99.
141. Kapel CM, et al. Trichinella spiralis, Trichinella britovi, and Trichinella nativa: infectivity, larval dis-
tribution in muscle, and antibody response after experimental infection of pigs. Parasitol Res. 1998;
84:264–71.
Index
A E4 proteins, 19
intermediate genes and, 19
ABC transport, 357 L4 genes, 19–20
A2B toxin, 133 pathogenesis, 21–22
Acanthamoeba, 579 structural proteins
animal models for, 585–586 capsid core proteins, 17–18
classification, 580–582 hexon, 16
clinical features and pathogenesis, 583–584 hexon-associated proteins, 16
amebic (acanthamoebic) keratitis, 583 minor coat protein pIIIa, 16
cutaneous acanthamoebiasis and sinusitis, 583 penton base and fiber proteins, 17
granulomatous amoebic encephalitis, 583 pVI, 16–17
diagnosis, 584–585 pVIII and pIX, 17
epidemiology, 582–583 virions, TEM of, 15
genomics, 580–582 Adherent invasive E. coli. See AIEC
life cycle, 582–583 Adhesion, 640, 641
morphology, 580–582 assays, 252
pathogenesis, 583 Adolescariae, 720
precautions, 585 AdV infections, 14
treatment and prevention, 585 cytokine response induced by, 22–23
in vitro models, 586 gastroenteric, 23
Acanthamoebic keratitis, 582 Aerobic endospore-forming rods, 132
Acanthamoebidae, 580, 582 Aeromonas, 2, 244
Acute liver failure animal models for
HEV-induced, 47 C. elegans, 243
Acute lung infection mouse models rodents, 242–243
of P. aeruginosa infection, 377 Tetrahymena spp., 243
Acute-on-chronic liver failure (ACLF), 48 zebrafish, 243
Acute poststreptococcal glomerulonephritis bacteremia, 241
(APSGN), 3, 228 biliary tract infections, 241
Acute rheumatic fever (ARF), 228 biology of, 240
Adaptive immune response classification of, 238
to astrovirus infection, 34 diagnosis of, 241–242
to HEV infections, 47 epidemiology of, 240
to urinary tract infections, 365 extraintestinal infections, 240–241
Adenovirus (AdV), 13 gastroenteritis, 240
cell entry, 20 genomics of, 239
cell lines as laboratory models for, 14 history of, 237
classification of, 14 isolates, 241–242
clinical diseasess caused by, 13 morphology of, 238–239
conserved features, 15–16 risk factors for, 240
DNA packaging and virion assembly, 21 treatment and prevention of, 242
DNA replication, 20–21 in vitro models for, 243
enteric. See Enteric AdVs Aflatoxins (AFs), 456–457
genome organization, 15–16 A-hemolytic species, 224
genus-common proteins, 16 A. hydrophila, 241
host–pathogen interaction, 22 AIEC, 323
life cycle of, 21 Airborne GAS epidemics, 227
as model systems, 23 A/J mice, 145
morphology of, 14–15 Alanine aminotransferase (ALT) levels
nonstructural proteins HEV infection and, 44–45
E1A proteins, 18 Albendazole, 516
E1B gene products, 18 Alcmaeon of Croton, 4
E2 gene products, 18–19 Alfalfa seedling infection model, 277
E3 proteins, 19 Alkaline peptone water (APW), 241
809
810 Index
Gastrointestinal anthrax, murine model of, 145 Group A Streptococcus (GAS) infections
Gastrointestinal infections, 240 aminal models for
pathognomonic signs of, 144 C. elegans, 232
Gastrointestinal injuries associated with SEs, 216 chinchilla otitis media model, 232
Gastrointestinal models for foodborne Candida insects, 232
infections, 502 nonhuman primate, 232
Gastrointestinal virus surrogate, bacteriophage MS2 as, rodents, 230–232
87–88 zebrafish, 232
Genbank information, 201 clinical features and pathogenesis, 227–229
Gene polymorphism, 665 APSGN, 228
Gene regulation, 646 ARF, 228
Genomic rearrangements, 203 GAS pharyngitis, 227
Genotypes, 668 impetigo, 228
Genotype 3 strain (3c), 45 PANDAS, 228–229
Genotype 3 virus culture systems, 55 scarlet fever, 228
Genotype 4 virus culture systems, 56 complications of, 225
Gerbils, 188–189, 643 diagnosis, 229
Germfree pig model. See Gnotobiotic (Gn) pig model foodborne or airborne, 227
Giardia intestinalis, 606 of skin, 228
Giardia lamblia, 635 treatment and prevention, 229
antigenic variation in, 645 in vitro models for, 232–233
biology of, 635 Group B Streptococcus (GBS), 225
drug resistance in, 646 Group C Streptococci, 225
encystation methods, 637 Group D streptococci, 175, 225
epidemiology, 639 Group G streptococci, 225
history of, 635 Group H streptococci, 225
immunology, 643 Guinea pig models
life cycle of, 636, 637 for Brucella infections, 265
models used for studies of, 641 for listeriosis, 188
morphology of, 636 of Salmonella infection, 398
trophozoites, electron micrographs of, 638 of shigellosis, 402–403
Giardia muris, 641 Gut-derived sepsis mouse models of P. aeruginosa
Giardiasis, 644, 646 infection, 377–378
diagnosis, 636 Gut epithelium, 186
drug resistance in, 646 Gut microbiota and Gn pig’s response to
epidemiology, 639 HRV, 105
nitroheterocyclic resistance in, 646 Gut, model organisms recapitulating aspects of, 374
pathology, 641
GiardiaVax, 644 H
Glanders, 271, 272
laboratory infection models of, 272–273 Hamster models
Glycopeptide, 179 for Acanthamoeba, 586
Gnotobiotic (Gn) pig model for HRV research of C. difficile infection, 164
advancements made using, 102 of glanders infection, 272
features of, 103–104 for melioidosis, 274
HGM and non-HGM-transplanted, 105 for studying prion infections, 122–123
history of, 97, 99 Haplorchis infections
HRV vaccines and therapeutics, 104 animal models for
probiotics and commensal microbiota role in, chicks, 739
104–105 dogs, 739
protective immunity mechanisms, 103 foxes, 739
Wa HRV infection, 104 rodents, 738–739
Gnotobiotic pigs, 86 clinical features of, 737–738
Gram-negative Fe(III) acquisition systems, 357 diagnosis of, 738
Granulomatous amoebic encephalitis, 583 treatment and prevention of, 738
Group A rotaviruses, 96 in vitro models of, 739
Group A Streptococcus (GAS). See also S. pyogenes Haplorchis species, 735
immune dysfunction, 3 classification of, 736
local diseases, 3 genome of, 737
localized inflammatory lesions, 3 life cycle and epidemiology of, 737
pharyngitis, 227 morphology of, 736–737
820 Index