Lisosoma Nature
Lisosoma Nature
Lysosomal storage diseases (LSDs) are heritable (inborn) specific genetic defect and the biochemical nature of
1
Department of
errors of metabolism that affect the function of the lyso- the stored macromolecules, LSDs can also cause skeletal
Pharmacology, University of
Oxford, Oxford, UK. some. LSDs comprise a group of 70 monogenic disorders dysmorphia due to bone pathology and can be associ-
2
Department of Genetics, St.
of lysosomal catabolism, most of which are inherited as ated with developmental delay or other central nervous
Jude Children’s Research autosomal recessive traits, but three are X-linked. These system (CNS) deficits, in addition to symptoms affect-
Hospital, Memphis, TN, USA. disorders are caused by mutations in genes encoding ing other organ systems (Fig. 1; Table 1). Patients present
3
The Raymond G. Perelman lysosomal proteins, such as lysosomal glycosidases, with a continuum of disease severity that loosely corre-
Center for Cellular and proteases, integral membrane proteins, transporters, lates with the type of mutation and residual activity of
Molecular Therapeutics,
enzyme modifiers or activators. Mutations in lysosomal the mutant protein, but they are generally classified
Children’s Hospital of
Philadelphia, Philadelphia,
genes affect the function of the encoded protein, result- on the basis of the type of disorder and the age of onset of
PA, USA. ing in lysosomal malfunction and the gradual accumu- the clinical signs as congenital or infantile (which usually
4
Department of Pathology lation of substrates inside the lysosome (that is, ‘storage’), have the most severe presentation), late-infantile, juve-
and Laboratory Medicine, which ultimately leads to cell dysfunction and cell death. nile and adult types. Diagnosis of LSDs is based on clin-
University of Pennsylvania, Of the ~1,300 genes involved in lysosomal function, ical symptoms and confirmation of increased storage or
Philadelphia, PA, USA.
many monogenic disorders have been described, includ- genetic alterations using several diagnostic tests, includ-
5
Department of Biological ing 50 enzyme deficiencies, which can be subclassified ing enzymatic analysis and single gene sequencing7.
Chemistry, David Geffen
School of Medicine, University
according to the biochemical type of stored material However, diagnosis, especially of milder cases with
of California–Los Angeles, Los (for example, the sphingolipidoses, mucopolysacchari- longer survival, is often delayed owing to clinical symp-
Angeles, CA, USA. doses and glycoproteinoses) into 7 disorders involving toms that are characteristic of other, more common
6
Office of the Clinical Director integral membrane proteins, 12 disorders of lysosome- conditions. More recently, next-generation sequenc-
and Medical Genetics Branch, related organelles (LROs) and 14 disorders involving the ing, particularly whole-exome sequencing, is becoming
National Human Genome production of lipofuscin1–6 (Table 1). routine and might reduce the time from presentation
Research Institute, NIH,
Bethesda, MD, USA.
LSDs are genetically and clinically heterogeneous dis- to diagnosis.
orders (Fig. 1; Table 1) but frequently present as paediatric Our understanding of the pathophysiology of LSDs
*e-mail: frances.platt@
pharm.ox.ac.uk neurodegenerative diseases that are often accompanied has undergone major advances that have identified
https://doi.org/10.1038/ by visceromegaly (the enlargement of abdominal organs multiple potential clinical intervention points. Through
s41572-018-0025-4 such as the liver and spleen)7. However, depending on the the use of animal models, various therapies have been
NATUre RevIewS | DISeASe PRImeRS | Article citation ID: (2018) 4:27 1
Primer
NATUre RevIewS | DISeASe PRImeRS | Article citation ID: (2018) 4:27 3
Primer
NATUre RevIewS | DISeASe PRImeRS | Article citation ID: (2018) 4:27 5
Primer
evaluated in vivo and have progressed to clinical trials example, CLN5 and CLN8) and other genetic isolates
and regulatory approval. Enzyme replacement therapy owing to founder effects10. These values are undoubt-
(ERT) to restore defective enzymes is the cornerstone of edly underestimates, as calculations of incidence pre-
current treatment paradigms for some LSDs, but small- sume that all cases are ascertained or that accurate
molecule therapies that reduce storage by inhibiting the carrier frequencies are known. Although these factors
production of the stored substrates are also approved and are known for the more severe subtypes of LSDs with
are an expanding area of drug development. Nucleic- infantile presentations, such as Tay–Sachs disease in the
acid-based medicines (such as gene replacement, anti- Ashkenazi Jewish population, it might not be the case
sense oligonucleotide (ASO) therapies or gene editing) for LSDs that occur in individuals of all ethnicities, or
are also emerging. As more treatments for LSDs become juvenile-onset and adult-onset LSDs, whereby many
available, adding these conditions to newborn screening individuals are diagnosed up to decades after symptom
panels should speed the diagnosis, improve the clinical onset and some individuals never receive a diagnosis.
care of patients and aid in the prevention of LSDs in Despite a low incidence of individual LSDs in general,
subsequent pregnancies. the incidence can be higher in specific ethnicities, often
This Primer provides an overview of the LSDs and owing to a founder effect (Table 2). For example, aspar-
the epidemiology and genetics of these disorders as well tylglucosaminuria (one of the glycoproteinoses) is a rare
as current insights into their pathophysiology and the condition worldwide (the exact incidence is estimated as
present status of therapies. <0.2 cases per 100,000 individuals) that is common in
the Finnish population (estimated carrier frequency of
Epidemiology 2.3–3%) owing to a founder effect and genetic isolation11.
Incidence and prevalence Similarly, Hermansky–Pudlak syndrome (a LRO disor-
As a group, LSDs are common, with an estimated inci- der) is found in 1 in 500,000 individuals worldwide but
dence of 1 in 5,000 to 1 in 5,500 for LSDs that involve in ~1 in 1,800 residents of northwestern Puerto Rico12,13.
lysosomal enzyme or integral membrane protein defects;
however, individual LSDs are rare, with estimated inci- Effect of survival and treatment
dences ranging from 1 in 50,000 to 1 in 250,000 live The prevalence of an LSD is determined by the incidence
births8. The most common LSDs are Fabry disease and the average survival for that disorder. The advent
(up to 2.5 cases per 100,000 males), Gaucher disease (up to of approved therapies for some LSDs and/or more-
2 cases per 100,000 individuals), metachromatic leu- effective supportive care for patients, most notably the
kodystrophy (up to 2.5 cases per 100,000 individuals) use of feeding tubes and better pulmonary hygiene,
and Pompe disease (up to 2.5 cases per 100,000 indivi have increased survival in resource-rich countries and
duals)9. In addition, the neuronal ceroid lipofuscinoses might increase disease prevalence. Before the approval
(NCLs) are relatively common, with an incidence of of ERT, 12.3% of infants with Pompe disease survived
1 case per 100,000 individuals in the general popu- to 18 months of age, but in the initial ERT trial, all
lation, but have a higher incidence in Finland (for 18 children treated with ERT before 6 months of age were
NATUre RevIewS | DISeASe PRImeRS | Article citation ID: (2018) 4:27 7
Primer
Fig. 1 | Organs affected in disorders of lysosomes and LROs. Mutations in genes causing lysosomal storage diseases
(LSDs) can be associated with symptoms in specific organs. Despite profound neurological involvement in many LSDs,
involvement of multiple organ systems is frequent. Blue-shaded boxes relate to specific organs as indicated, whereas
yellow-shaded boxes reflect symptoms that are not specific to one organ system. The specific LSDs associated with each
genetic mutation can be found in Table 1; note this figure is not exhaustive. LRO, lysosome-related organelle.
alive at 18 months14,15. ERT, although life-saving, was not As treatments become available for more LSDs,
definitive, and secondary complications of the disease clinicians should be alert to ‘secondary phenotypes’ that
have occurred in most patients receiving this therapy might require additional therapeutic interventions.
alone16. In addition, even in the absence of approved
therapy, infants with Tay–Sachs disease who historically Mechanisms/pathophysiology
died at 1.5 years of age now survive to an average of General mechanisms
5–6 years of age owing to better supportive therapy17. Lysosomes are responsible for the breakdown and
In general, LSDs affect multiple organ systems but recycling of macromolecules (including carbohy-
have one defining system predominating, such as hyper- drates, lipids, nucleic acids and proteins) and function
trophic cardiomyopathy in patients with infantile Pompe as metabolic hubs that control nutrient sensing, amino
disease, CNS deficits in children with CNL2 or hepato acid and ion homeostasis and calcium signalling22–24.
splenomegaly in children with Gaucher disease type I. These organelles are constantly in a dynamic state;
For Pompe disease and Gaucher disease, the major com- they fuse with autophagosomes, phagosomes and the
plications have been mitigated with approved therapy, plasma membrane and therefore have a pivotal role
and the CNS deficits can be slowed in CNL2 (ref.18). in cell–cell and cell–extracellular matrix communi-
However, additional complications have been reported cation, response to infection and maintenance of cell
owing to this increased survival, such as oro-motor defi- homeostasis22,23 (Fig. 2). In addition, late endosomes and
ciencies and diaphragm weakness in Pompe disease19–21. lysosomes tether with other intracellular organelles
without fusing with them, such as mitochondria and are spared in associated LSDs, whereas other neurons
the endoplasmic reticulum, forming functional mem- will store these macromolecules, leading to neuronal
brane contact sites. These membrane structures are damage and death. Circulating monocytes and tissue
signalling microdomains that allow the transfer of macrophages (forming the mononuclear phagocytic
lipids and the exchange of metabolites and calcium system) are the primary cells affected in most LSDs
ions between the organelles24–26. The lipid and protein owing to their role in the phagocytic clearance of cellular
composition of these contact sites drives their func- debris, apoptotic or necrotic cells and microorganisms,
tional characteristics and influences each of the teth- and the impaired catabolism of products they themselves
ered organelles. For instance, mitochondria–lysosome synthesize. These cells have a central role in immune
contact sites modulate mitochondrial fission and lyso- responses and inflammatory processes that can be elic-
somal dynamics through GTP hydrolysis of Ras-related ited by loss of cell integrity and homeostasis in LSDs28;
protein Rab7a (RAB7; also known as RAB7A)27. Thus, indeed, the storage of macromolecules by macrophages
it is understandable that in an LSD, the biochemical in LSDs triggers inflammation that actively contributes
properties and in turn the functions of these micro- to disease progression29.
domains might be altered by changes in composition Substrate accumulation inside lysosomes can initi-
of the endosomal and lysosomal membranes due to ate a cascade of secondary effects, ultimately leading to
impaired lysosomal activity25,26. irreversible cellular damage, cell death as well as organ
As previously mentioned, mutations in genes encod- dysfunction and degeneration. Given the complexity of
ing lysosomal proteins, including lysosomal hydrolases, the clinical presentations and the broad range of accu-
lysosomal membrane proteins, lipid and ion transport- mulated macromolecules in LSDs, many of which are
ers, enzyme modifiers or activators, are the cause of still unknown, defining pathogenetic pathways that
LSDs. Mutations in these genes lead to the aberrant pro- are potentially common to all disorders is challenging.
cessing and degradation of substrates, impaired trans- However, in general terms, several common pathways
port of lipids and metabolites and progressive primary are dysregulated, including deficits in cellular trans-
accumulation of non-degraded or partially degraded port and degradation (such as autophagy, endocytosis,
macromolecules inside lysosomes. The secondary stor- phagocytosis and lysosomal exocytosis), calcium
age of substrates can also occur in LSDs resulting from homeostasis, oxidative stress, inflammatory and innate
defects in non-enzymatic lysosomal proteins (for exam- immune responses and cell death pathways (includ-
ple, transporters); although the mechanisms leading to ing apoptosis and in some instances necroptosis)30,31.
secondary storage are not fully understood, it might be In addition, general mechanisms of disease progres-
the result of trafficking defects. The biochemical type sion include chronic inflammation, including neuro–
of the stored macromolecules differentially affects lyso inflammation, which is linked to cellular dysfunction
some function as different macromolecules have roles in and death, and autoimmune responses to specific self-
specific cellular processes, which underlies the variable antigens or accumulated metabolites28. Several stud-
clinical pathology of LSDs. Cells can store substrates ies have shown that the activation of innate immune
and/or metabolites only if the cell in question synthe- cells, such as microglia in the brain and macrophages
sizes or ingests these molecules. For example, glyco- in the periphery, actively contributes to LSD progres-
sphingolipid expression is variable in the CNS; some sion32–34. For example, in response to stress signals (that
neuronal populations cannot store these molecules and is, damage-associated molecular patterns) of dying or
damaged neurons via pattern recognition receptors,
activated microglia release cytokines and chemokines to
Table 2 | Increased incidence of LSDs in specific populations before carrier screening recruit immune cells at affected sites or to activate neigh-
Disorder Region or population Incidence bouring neural cells in a paracrine and/or autocrine
(per 100,000 manner, hence amplifying the inflammatory response35.
individuals) In line with these findings, anti-inflammatory therapies
Gaucher disease type I Ashkenazi Jews 100 (ref.9) have demonstrated benefits in some animal models of
LSDs34,36 (such as Niemann–Pick disease type C (NPC)
Tay–Sachs disease (infantile) Ashkenazi Jews 25 (ref.114)
and Sandhoff disease), and trials of biological agents that
Niemann–Pick disease type A Ashkenazi Jews 2.5 (ref.9) target tumour necrosis factor (TNF) are in clinical tri-
GM1 gangliosidosis Rudari isolate 10 (ref.198) als in some forms of mucopolysaccharidosis (MPS; see
Metachromatic leukodystrophy Western Navajo Nation 40 (ref.199) Management). In addition, adaptive immune responses
(for example, autoantibody production) have been
Pompe disease The Netherlands 2.5 (ref.200)
described in some LSDs37, and the precise pathophys-
Cystinosis Brittany 3.8 (ref.201) iological role of a dysregulated immune system mer-
Salla disease Finland or Sweden 2.5 (ref.202) its greater investigation as it is a potentially tractable
Aspartylglucosaminuria Finland 54 (ref.12) therapeutic target.
Notably, the common secondary pathways of LSDs
Niemann–Pick disease type C Nova Scotia 100 (ref.203)
(French Acadian) have also been implicated in other, more common adult
conditions that are mostly associated with ageing, includ-
Fabry disease Nova Scotia 9.3a ing cancer and neurodegenerative diseases. Moreover,
Hermansky–Pudlak (types 1 and 3) Puerto Rico 56 (refs13,14) unexpected roles of lysosomal enzymes and their sub-
a
M. West, personal communication. GM1, GM1-ganglioside; LSD, lysosomal storage disease. strates have been demonstrated in cellular pathways and
NATUre RevIewS | DISeASe PRImeRS | Article citation ID: (2018) 4:27 9
Primer
Exocytosis
LYNUS machinery
Mature LRO
Lysosomal membrane SLC38A9
proteins
V-type Transporters
H+ ATPase
Lysosome LAMP2
Cytosol NPC1
Late endosome
Pre-LRO MVB
Early Lysosome membrane
LAMP1 ?
endosome Glycocalyx Lumen
Golgi
LIMP1
Recycling Microtubules
endosome
Traffic and
MCOLN1
fusion proteins
Hydrolases
Proteases: cathepsins Ion channels
Glycosidases
Biogenesis
Lipases Nucleases
Cargo transport
Fig. 2 | Complexity of lysosomal proteins and their functions. Lysosomes are involved in the breakdown and recycling of
several cellular macromolecules, including nucleic acids, lipids, proteins and carbohydrates, through the action of lysosomal
hydrolases. Substances are delivered to the lysosome for degradation via fusion with late endosomes (extracellular
material) or autophagosomes (intracellular material); a process that is mediated by SNARE (soluble N-ethylmaleimide-
sensitive factor activating protein receptor) and Rab proteins (traffic and fusion proteins). Lysosomes can be transported to
required cellular compartments via microtubules such as dynein and kinesins and actin-based motor myosins193.
Transporters such as NPC1, sodium-coupled neutral amino acid transporter 9 (SLC38A9) and lysosome-associated
membrane protein 2 (L AMP2) are responsible for the transport of substrates between the lysosome and the cytoplasm.
NPC1 facilitates the transport of lipid substrates and amines, SLC38A9 transports arginine and L AMP2 transports cytosolic
substrates. Ion channels, including mucolipin 1 (MCOLN1), act to regulate the ionic environment of lysosomes and
contribute to cell signalling. The lysosomal nutrient-sensing (LYNUS) machinery is composed of protein complexes that
sense lysosomal nutrient composition and transfer signals to the nucleus. Lysosomal membrane proteins are involved in
lysosome–plasma membrane docking (L AMP1) and the sorting of lysosomal enzymes (such as lysosome membrane protein 1
(LIMP1)). L AMP1 binds to motor myosin, which is, in turn, speculated to bind to microtubules (denoted by ?). Illustrative
examples of lysosomal proteins are provided. Solid arrows reflect organelle maturation and/or biogenesis and dashed
arrows represent the movement of macromolecular cargo. LRO, lysosome-related organelle; MVB, multivesicular body.
Adapted with permission from ref.103, Annual Reviews.
regulatory networks that go beyond canonical lysosomal and glucosylsphingosine (GlcSph). GlcCer and GlcSph
degradation or recycling38,39. Importantly, this research accumulate in mononuclear phagocytes (due to the phago-
led to the discovery of parallel mechanisms between rare, cytic activity of these cells), primarily macrophages, which
paediatric LSDs and common, adult diseases, includ- have a foamy appearance and are referred to as Gaucher
ing age-related neurodegenerative diseases (such as, cells51,52. Investigations into the chronic inflammation and
Parkinson disease (PD) and Alzheimer disease (AD)), in haematologic abnormalities in patient-derived primary
addition to fibrosis and cancer40–45. These findings have macrophages53 have identified defects in macroautophagy
fostered the development of improved model systems and degradation, but not the initial uptake, of apoptotic
used to evaluate pathogenesis and new treatments for or necrotic cells (efferocytosis)49, likely as a result of gly-
LSDs, including models using patient-derived induced cosphingolipid storage54,55. In acute and chronic Gaucher
pluripotent stem cells46–48 differentiated into specific cel- disease types II and III, which are neuropathic, microglial
lular lineages49,50. Overall, research into LSDs has sparked activation and neuroinflammation have been associated
the development of therapies that might benefit both with neuronal dysfunction and neuronal loss in the brain
patients with LSDs and those with other disorders. Below, and spinal cord56,57. The mechanism of neuronal loss and
we give some illustrative examples of LSDs in which fairly inflammation in murine models of Gaucher disease is
detailed pathogenetic pathways have been elucidated. deregulated processing and release of pro-inflammatory
cytokines (that is, IL-1α, IL-1β, TNF, IL-6, CXC-chemokine
Gaucher disease ligand 10 (CXCL10) and type I interferon58,59), altered
Gaucher disease, a prototypical LSD, is caused by the defi- chemotaxis of inflammatory and immune cells and pro-
ciency of the lysosomal hydrolase β-glucocerebrosidase duction of reactive oxygen species60–65. This pathogenetic
(GCase), encoded by GBA51 (Fig. 3), leading to the accumu- cascade has been studied in a conditional knockout model
lation of the glycosphingolipids glucosylceramide (GlcCer) of Gaucher disease (Gbaflox/flox Nestin–Cre mouse), which
Fig. 3 | Selected examples of cellular pathogenesis in LSDs. The molecular pathways involved in lysosomal storage
diseases (LSDs) might become applicable to other adult diseases of ageing. Thus, deficiency of lysosomal ß-glucocerebro-
sidase (GCase) and accumulation of glucosylceramide (GlcCer), leading to Gaucher disease, are also associated with
accumulation and/or deposition of α-synuclein, a characteristic of Parkinson disease and dementia with Lewy bodies. The
deficiency of α-N-acetylglucosaminidase (NaGlu) and resulting accumulation of heparan sulfate (HS-GAG) in mucopoly-
saccharidosis (MPS) IIIB leads to deposition of hyperphosphorylated tau (P-tau), which is a cause of neurodegenerative
diseases, including Alzheimer disease. In addition, in Niemann–Pick disease type C (NPC) deficiency of NPC1 leads to
cholesterol and sphingolipid accumulation, and the cell biology of NPC parallels aspects of Alzheimer disease. In sialidosis,
excessive lysosomal exocytosis downstream of neuraminidase-1 (NEU1) deficiency and accumulation of the lysosome-
associated membrane glycoprotein 1 (L AMP1) have a key pathogenetic role and might contribute to Alzheimer disease,
fibrosis and cancer. Interestingly , aberrant chaperone-mediated autophagy (CMA), associated with protective protein
cathepsin A (PPCA , also known as CTSA) deficiency in patients with galactosialidosis and impaired L AMP2 degradation,
might have a role in frontotemporal dementia. In addition, the accumulation of GM1-ganglioside (GM1) in intracellular
membranes and the disturbance of calcium homeostasis are pathogenetic in GM1 gangliosidosis but might also occur in
Alzheimer disease. The occurrence of a lysosomal form of subunit c of mitochondrial ATP synthase (SCMAS) in MPS IIIA ,
IIIB and IIIC as well as in the neuronal ceroid lipofuscinosis diseases suggests a common pathology between these
conditions, but whether it reflects a pathogenetic sign is still unclear. Lastly , the fact that Lassa and Ebola viruses use
lysosomal integral membrane proteins L AMP1 and NPC1, respectively , as their intracellular receptors to potentiate their
infectivity has added to the multiplicity of functions and complexity of the eclectic lysosomal organelle (not shown)194,195.
Furthermore, Mycobacterium tuberculosis actively inhibits NPC1 in order to persist in host cells196, adding another twist to
the complex relationship between pathogens and lysosomal proteins (not shown). APP, amyloid precursor protein; ER ,
endoplasmic reticulum; FNGN, focal necrotizing glomerulonephritis; GLB1, β-galactosidase.
suggests that neuroinflammation is elicited by damage- Natural history and population genetic studies have
associated molecular patterns released from dying neu- revealed that patients with Gaucher disease type I or
rons, astrocytes and oligodendrocytes58,66,67. These effectors carriers of GBA mutations have an increased risk of PD
promote pro-inflammatory activity in microglia, which or Lewy body disorders45,68,69. These neurodegenerative
amplifies their neurotoxic process, ultimately leading to conditions are characterized by the presence of insolu-
neurodegeneration35. In the same Gaucher mouse model, ble, oligomeric or fibrillar α-synuclein inclusions in neu-
the upregulation of receptor-interacting serine/threonine- rons of the substantia nigra, the hippocampus and the
protein kinases 1 and 3 (RIPK1 and RIPK3, respectively), cerebral cortex55,70. Interestingly, an inverse relationship
coupled with necrotic cell death and inflammasome acti- between the levels of GCase and α-synuclein aggregates
vation, have also been implicated in neurodegeneration31. has been demonstrated experimentally in cellular mod-
In addition, the chronic inflammation in experimental and els and mice, but the mode of interaction between these
clinical Gaucher disease could be due to the production molecules and their contribution to the development of
of complement-activating GlcCer-specific immunoglob- synucleinopathies have not been fully elucidated55,71,72. In
ulin G (IgG) autoantibodies, which have been proposed addition, GlcCer directly controls a reversible change
to drive a cycle of GlcCer and GlcSph accumulation and in α-synuclein that promotes its aggregation and toxicity,
complement activation62. opening the possibility of therapeutic intervention by
NATUre RevIewS | DISeASe PRImeRS | Article citation ID: (2018) 4:27 11
Primer
NATUre RevIewS | DISeASe PRImeRS | Article citation ID: (2018) 4:27 13
Primer
or abnormal movements are common presenting fea- affect the CNS, is characterized by proximal tubular dys-
tures and are easily confused with symptoms of more function owing to cystine accumulation, which leads to
common adult-onset neurodegenerative disorders. renal Fanconi syndrome and renal failure in >90% of
Psychosis or depression can also be a presenting feature patients106. The prompt diagnosis and treatment with
in adult-onset disorders, such as late-onset Tay–Sachs cysteamine prevents renal failure. However, cystinosis
disease, and can delay diagnosis by years to decades105. is a systemic disorder, with other symptoms including
Substrate storage in specific organ systems is characteris- photophobia and visual impairment owing to corneal
tic of some enzyme deficiency disorders, such as storage deposition of cystine crystals (which can be treated
in the kidney in Fabry disease, the heart in Pompe dis- effectively with cysteamine eye drops), endocrine abnor-
ease or the mononuclear phagocyte system in Gaucher malities (such as hypothyroidism and growth hormone
disease. Similarly, common constellations of findings deficiency) and skeletal deformities (including scoliosis,
such as hepatosplenomegaly, coarsening facial features stress fractures and joint pain). Even with cysteamine
and joint contractures (with or without corneal cloud- therapy, patients with cystinosis can also develop a sec-
ing) should suggest a MPS disorder. However, more- ondary phenotype of progressive myopathy and muscle
subtle presentations such as mild muscle weakness in weakness, dysarthria and swallowing difficulties. NPC
adults (which can occur in Pompe disease) or a clinical type 1 or 2 should be suspected in a child with isolated
sign with more common aetiologies such as proteinu- splenomegaly or neonatal liver disease, as patients can
ria, diminished renal function or cardiac hypertrophy have no other clinical findings for years before develop-
(which can occur in Fabry disease) can make diagnosis ing the characteristic supranuclear gaze palsy followed
more challenging. by intellectual decline. Neuropsychiatric symptoms and
cognitive decline can predominate in NPC with onset in
Disorders of post-translational modification. Disorders adolescence and adulthood, resulting in long delays
of post-translational modification include multiple sul- in diagnosis107,108.
fatase deficiency and mucolipidoses II and III and result
from mutations in genes that have a role in biochemi NCLs. The NCLs involve the CNS and were initially
cally modifying lysosomal hydrolases. Therefore, the classified on the basis of age at onset109 (Table 1). More
clinical manifestations of these disorders are more recently, the classification was updated to include the
generalized than other forms of LSDs and have over- genetic mutation as well as the age of onset (for exam-
lapping features with disorders caused by defects in ple, CLN3 disease, juvenile onset). Symptom progres-
single lysosomal hydrolases (Table 1). For example, sion is variable10; some disorders present initially with
multiple sulfatase deficiency is caused by mutations progressive visual loss (for example, CLN3 disease,
in SUMF1, which encodes formylglycine-generating juvenile onset)110, followed by mental decline and sei-
enzyme (FGE). FGE modifies sulfatases (a class of zures10. Indeed, most patients with CLN3 disease are
lysosomal enzymes), which include the enzymes that diagnosed by ophthalmologists owing to the early visual
degrade mucopolysaccharides that are affected in loss with characteristic retinal pathologies110. Other
MPS II, IIIA, IIID, IVA and VI. As a result, the clinical NCLs present with developmental delay (for example,
manifestations of multiple sulfatase deficiency over- CLN2 disease, late-infantile onset), with visual loss
lap with the MPS disorders, making diagnosis chal- occurring at later stages111. In general, the NCLs involve
lenging. Similarly, mucolipidoses II and III are caused progressive movement disorders, epilepsy, dementia
by mutations in GNPTAB and GNPTG, respectively, and early death.
encoding the subunits of N- a cetylglucosamine-1-
phosphotransferase, which tags lysosomal hydrolases Disorders of LROs. The most notable LRO disor-
with mannose 6-phosphate for targeting to the lysosome. ders include the Hermansky–Pudlak, Griscelli and
As a result, hydrolases that require mannose 6-phos- Chédiak–Higashi syndromes, which are all character-
phate tagging (which include degradative enzymes for ized by hypopigmentation (owing to a melanosome
most sphingolipids, mucopolysaccharides and glycopro- defect) and prolonged bleeding (owing to a platelet δ
teins) are not trafficked to the lysosome and are secreted granule defect)103. The classic form of Chédiak–Higashi
into the extracellular space. This process leads to the syndrome also includes recurrent life-threatening infec-
accumulation of multiple complex substrates in lyso- tions due to immunodeficiency, and 85% of patients
somes, which leads to disorders with severe phenotypes develop a life-threatening hyperinflammatory condi-
that share clinical features with the sphingolipidoses, tion (haemophagocytic lymphohistiocytosis). Untreated
glycoproteinoses and mucopolysaccharidoses, but, most patients die, usually of overwhelming bacterial infec-
notably the MPS disorders. tion, in infancy or early childhood103. Most LRO dis-
orders manifest during infancy, with the exception of
Disorders of integral membrane proteins. Similar to the Chédiak–Higashi disease, which has juvenile and adult-
enzyme deficiency disorders, most disorders of integral onset forms. Haematopoietic stem cell transplantation
membrane proteins can present during infancy, child- (HSCT) facilitates improved survival of patients with
hood and adulthood and can affect the CNS. Common classic Chédiak–Higashi syndrome into adulthood,
symptoms include intellectual disability, ataxia, seizures although a secondary phenotype similar to the adult-
and spasticity. With the exception of NPC and cystinosis, onset form subsequently emerges, including tremor,
most integral membrane protein disorders are rare in the ataxia, peripheral neuropathy and cognitive decline that
general population. Cystinosis, which does not primarily is life-limiting112.
NATUre RevIewS | DISeASe PRImeRS | Article citation ID: (2018) 4:27 15
Primer
Box 1 | Newborn screening for LSDs enzyme activity is currently being used rather than
DNA sequencing owing to poor phenotype–genotype
Universal screening for lysosomal storage diseases (LSDs) correlations in many disorders119. However, some con-
is mandated in the following regions and is under troversy surrounds screening for LSDs, as screening
development or in pilot phases in several other regions:
assays identify newborn babies with enzyme defi-
• Pompe disease: Taiwan and, in the United States, ciencies who might have adult-onset disease and who
Illinois, Kentucky, Minnesota, Missouri, New York, might not require treatment for decades, or at all, such
Pennsylvania and Tennessee
as Fabry disease and Gaucher disease. Ultimately,
• Mucopolysaccharidosis I: Illinois, Kentucky, Minnesota, newborn screening using next-generation sequencing
Missouri, New York, Pennsylvania and Tennessee
(whole-exome or whole-genome sequencing) will mag-
• Krabbe disease: Illinois, Kentucky, New Mexico, New nify this dilemma, as practitioners try to interpret vari-
York and Pennsylvania
ants of uncertain significance in disease-causing genes.
• Fabry disease: Illinois and Missouri The unmet needs for newborn screening in LSDs are
• Gaucher disease: Illinois and Missouri robust, cost-effective assays, more pilot programmes
• Niemann–Pick disease types A and B: Illinois demonstrating utility and increased understanding of
the benefits of early treatment.
common mutations that cause Tay–Sachs disease in
Ashkenazi Jewish individuals has been proved superior Prenatal screening and prevention. In families who
to traditional enzyme testing, although the detection already have a child with a diagnosed LSD and the spe-
of variants of unknown significance has restricted the cific disease-causing mutations are ascertained, using
sole use of next-generation sequencing for carrier detec- IVF with preimplantation genetic diagnosis (PGD) to
tion117. Despite the success of carrier screening, owing to identify unaffected embryos for implantation offers
the dispersion of high-risk individuals across the world at-risk couples the chance to have additional children
due to increased migration, some individuals might not who do not have the disorder. For families with chil-
realize they are at increased risk of Tay–Sachs disease, dren with a juvenile-onset disease or when a child with
which emphasizes the need for cost-effective universal early-onset disease has a protracted diagnostic odyssey,
screening. using IVF with PGD is not an option and often results
in multiple children in a sibship with the LSD. PGD is
Newborn screening. The rationale for mandatory new- expensive, but is approved in the United Kingdom by
born screening has historically included disorders with the Human Fertilization and Embryology Authority
an effective therapy, a cost-effective and relatively high- for several LSDs. PGD is available in the United States,
throughput detection method and disorders for which Canada and Western Europe but might not be covered
early detection (therefore allowing timely treatment) is by health insurance, and PGD is typically not available
essential; phenylketonuria is the prototype. Newborn in low-resource countries. As an alternative to PGD, at-
screening can identify patients rapidly using testing risk couples can utilize prenatal diagnosis during the first
for increased levels of metabolites, such as phenyl trimester of pregnancy, with termination of fetuses with
alanine in the case of phenylketonuria, or identifying LSDs. Using donor sperm or eggs to produce an embryo
common disease-causing mutations, such as ΔF508 in without LSD-causing mutations and the use of adoption
cystic fibrosis. Newborn screening is correctly viewed are additional choices for at-risk couples.
from the perspective of the affected child but can also
affect subsequent reproductive choices for the parents. Management
The goal of newborn screening is better public health Some LSDs are treatable; for example, the missing or
outcomes and requires not only a reliable detection defective enzyme can be provided exogenously through
assay but also parental education, clinical follow-up ERT, the amount of stored material within lysosomes
and available treatments for all positive newborn babies. can be reduced by substrate reduction therapy (SRT) or
Although universal newborn screening is mandated for compounds can improve the function of the defective
some disorders in most developed countries, the num- enzyme using chaperone therapy and proteostasis mod-
ber of specific disorders varies by country and, within ulators. Alternatively, genetic therapies might be a pos-
the United States, varies by state. In addition, manda- sibility, such as gene therapy or stop codon readthrough
tory screening for LSDs is limited, although the tech- drugs (see Outlook). Although treatments are currently
nology and availability for such screening is expanding available for a number of LSDs, most disorders cannot
rapidly (Box 1). be treated and are managed symptomatically (Table 3).
Until 2006 when alglucosidase alfa was approved by Additional therapeutic strategies might be developed
the US FDA for the treatment of Pompe disease, none for specific LSDs, although this will require a thorough
of the LSDs met the criteria for newborn screening as no understanding of their pathophysiology120. Treatment
effective therapies were available; however, this has now outcomes vary between LSDs and range from very effec-
changed with the development of treatments for some tive to minimally effective. As new therapies are devel-
disorders. Owing to the development of these treat- oped, there might be a choice between different therapies;
ments, parents and advocacy groups continue to lobby unless a medical or scientific reason exists for proceeding
to include LSDs on newborn screening panels. Newborn with one drug, the choice might be based on cost and/or
screening for LSDs using fluorometric enzymatic assays the expertise of the attending physicians. Combination
and more recently tandem mass spectrometry to measure therapy is also likely given that a combination of
drugs is more likely to mitigate the many symptoms development, whereas transplantation of older patients
of these complex diseases. (26 months of age) allowed peripheral improvement
but did not prevent cognitive decline124. HSCT has also
Available therapies been used for other LSDs for which no other treatment
HSCT. Bone marrow transplantation was initially per- was available, the most prominent being metachromatic
formed on a 1-year-old patient with Hurler syndrome leukodystrophy and Krabbe disease; however, the results
(the severe form of MPS I) in 1981 (ref.121). The ration- have been less promising than with Hurler syndrome.
ale for using bone marrow transplantation was that
this therapy would provide a lasting source of bone- ERT. ERT is based on the ability of cells to take up macro
marrow-derived cells with normal enzyme levels that molecules (such as enzymes) by endocytosis, which
would donate enzyme to the patient’s deficient cells. This can be packaged into lysosomes through endosome
therapy was the first treatment of an LSD to show any and lysosome fusion. Receptor-mediated endocytosis
success and was continued to be used despite its high underlies the cellular uptake and lysosomal targeting of
morbidity and mortality. Over time, the use of umbili- several lysosomal enzymes; for example, GCase (which
cal cord blood instead of bone marrow, as well as better is deficient in Gaucher disease) contains mannose resi-
conditioning regimens and donor selection, reduced dues, which bind to mannose receptors on the surface of
the adverse effects of transplantation122 and improved macrophages, whereas other soluble lysosomal enzymes
efficacy. HSCT is currently the standard of care for contain mannose 6-phosphate groups, which bind to
infants with Hurler syndrome, owing to the benefit of mannose 6-phosphate receptors on the surface of most
this therapy for preventing the development of neuro- cells125. Thus, a lysosomal enzyme, made in recombi-
logical symptoms, which do not respond to intravenous nant form and containing an appropriate carbohydrate
ERT123. Younger patients have better results from HSCT modification, will be taken up by cells, incorporated
than older patients; the transplantation of umbilical cord into lysosomes and, accordingly, can be used to treat a
blood in patients with Hurler syndrome <9 months of lysosomal enzyme deficiency. Non-carbohydrate mod-
age prevented the onset of both neurological and ifications are also being tested, such as insulin growth
peripheral symptoms and allowed normal cognitive factor 2, which also binds to the mannose 6-phosphate
NATUre RevIewS | DISeASe PRImeRS | Article citation ID: (2018) 4:27 17
Primer
receptor and can be used when there is inadequate man- type I136–139, and miglustat is approved for NPC140,141.
nose 6-phosphate on the enzyme126. ERT is feasible only Both drugs block GlcCer synthase, which inhibits the
with enzymes that are soluble or easily solubilized and first step in glycosphingolipid biosynthesis142,143.
does not work for membrane-bound enzymes. SRT drugs are orally administered and are stable at
ERT has been developed and approved by the FDA ambient temperatures. Thus, in contrast to ERT, SRT
or the European Medicines Agency (EMA) for the treat- does not involve a labile product and invasive delivery,
ment of several enzyme deficiency disorders (Table 3), and owing to their low molecular mass, SRT drugs are
including Gaucher disease, Fabry disease, Pompe dis- non-immunogenic. Miglustat can cross the blood–brain
ease, lysosomal acid lipase deficiency and MPS I, II, IV, barrier and, accordingly, can slow the progression of
VI and VII, and therapies are under development for neurological symptoms of NPC, including improving
other LSDs (Table 4). Most clinical manifestations of swallowing, which reduces the number of deaths result-
type I Gaucher disease can be treated or prevented with ing from aspiration pneumonia144,145. However, some
ERT (which can be started in childhood or adulthood), SRT drugs such as eliglustat do not cross the blood–
allowing the patients to live essentially normal lives127,128. brain barrier and are restricted to non-neuronopathic
However, the use of ERT for other LSDs has more limited glycosphingolipidoses, in which, for example, they
results. A major limitation of ERT is that not all organs reduce visceral disease and improve haematological
are accessible to the exogenously administered enzyme; parameters in Gaucher disease type I146,147.
orthopaedic, cardiovascular, neurological and ocular SRT drugs have a slower onset of efficacy than ERT,
symptoms are especially difficult to treat. In addition, as substrate turnover (a particularly slow process in
the blood–brain barrier prevents entry of the exogenous the brain but which is more rapid in organs with cell
enzyme into brain parenchyma; as a result, the enzyme turnover, such as the liver) must occur before clinical
requires modification to take advantage of existing trans- benefit. As with all small-molecule drugs, SRT drugs
port systems (such as the insulin or transferrin recep- have adverse effects or complications relating to drug
tors) or requires direct administration into the brain or metabolism. For example, miglustat causes osmotic diar-
spinal canal. Accordingly, intracerebroventricular and rhoea148, whereas eliglustat requires cytochrome P450
intrathecal methods of administration through which 2D6 (CYP2D6) status to be determined to avoid the
recombinant enzymes are infused into the CSF through use of this drug in CYP2D6 ultra-rapid metabolizers149.
catheters inserted into the lateral ventricle or the intrath- This requirement is because eliglustat is a CYP2D6 and
ecal space, respectively, are used126,129. ERT directed to CYP3A substrate. If eliglustat is used to treat patients
the cerebral ventricles has been recently approved for with ultra-rapid CYP2D6 metabolism status, they will
CLN2 (ref.130) (Table 3). For peripheral symptoms, ERT not achieve an effective dose. Also, if eliglustat is com-
is administered intravenously. Depending on the com- bined with drugs that inhibit these cytochrome P450
plexity of the required approach, ERT infusions can be system enzymes (CYP2D6 and CYP3A), it increases
carried out at the patient’s home. eliglustat exposure and can potentially cause cardiac
Another limitation of ERT is that the therapeu- arrhythmias. SRT drugs are expensive, and, as with
tic enzyme often elicits the production of antibodies, ERT, access globally is restricted to relatively affluent
which can reduce the efficacy of the treatment and lead health-care systems.
to adverse events, including anaphylaxis. The likelihood In principle, SRT could be developed for all LSDs but
of antibody production and adverse immune responses currently is restricted to glycosphingolipidoses because,
can be a major issue associated with ERT in patients with to date, only biosynthetic inhibitors specific to this path-
Pompe disease and varies depending on the nature of the way have been identified. Trials in other sphingolipi-
mutation; it occurs in nearly all patients who have a null doses are in progress (Table 4). Several other SRT drugs
mutation and no residual protein131. In addition, the high are currently being evaluated in clinical trials, including
cost of ERT might discourage the use of this therapy, genistein (an isoflavone that indirectly reduces proteo-
particularly in developing countries with limited health glycan biosynthesis) for MPS III, ibiglustat for Gaucher
budgets. As the dose of the therapeutic enzyme is deter- disease and Fabry disease and lucerastat for Fabry dis-
mined by the patient’s body mass, the cost of treatment ease (Table 4). In addition to small molecules, another
increases as the patient grows and can reach costs of technique for achieving SRT is ASO-mediated suppres-
several hundred thousand US dollars annually132. sion of biosynthetic enzymes, which is in preclinical
The therapeutic benefit of ERT is transient and, accord- evaluation150. Also, in principle, SRT drugs can be com-
ingly, this therapy must be repeated throughout life. bined with other therapies that target different steps in
However, lysosomal enzymes and recombinant enzymes the pathogenetic cascade.
for ERT generally have a long half-life in vivo, and adminis-
tration of therapeutic enzymes can therefore be performed Chaperone therapy. Many mutations in the genes encod-
weekly, biweekly or monthly. As LSDs are progressive, ERT ing lysosomal enzymes result in an unstable protein
appears to be most effective if started early124,133. that is prone to rapid turnover and/or reduced cata-
lytic activity151. Subinhibitory concentrations of active
SRT. SRT uses small molecules to inhibit the build-up site inhibitors (known as chaperone therapy) can sta-
of macromolecules in the lysosome to reduce stor- bilize the mutant enzyme, which extends half-life and
age134,135 and includes drugs that inhibit the biosynthesis improves catalysis, thereby reducing storage. The only
of storage metabolites. SRT using miglustat and eliglus- approved chaperone drug is migalastat for the treat-
tat is approved for the treatment of Gaucher disease ment of Fabry disease152,153, which improves ventricular
hypertrophy and reduces the severity of diarrhoea, reflux progressive neurodegenerative disease. Together with
and indigestion154,155 (Table 3). physical, occupational and speech therapists, physiatrists
The inhibitory nature of chaperone therapies requires can provide patients with assistive devices for ambula-
dosing every other day to allow maximal enzyme tion, such as walkers or canes and a well-fitted wheel-
enhancement. Allosteric enhancers are in development chair for children and adults with progressive disease.
to avoid the use of active site inhibitors and will allow Providing patients with appropriate seating with trunk
conventional daily dosing156. Some of these chaperone support will prevent scoliosis and restrictive lung disease
drugs (including migalastat) can enter the CNS and, and requires periodic adjustments as the child grows. An
therefore, could potentially treat neurological symptoms occupational therapist can provide or recommend adap-
in neuronopathic LSDs. Similar to drugs for other LSDs, tive equipment that facilitates independence in feeding,
these chaperone therapies are expensive. Chaperone dressing and other activities of daily living. Physical
therapies for Tay–Sachs disease and Gaucher disease therapists can design a regimen of active and passive
are currently in clinical trials (Table 4). stretching for patients that maintains range of motion
and prevents joint contractures. Speech and language
Supportive care therapists can help patients with dysarthria maintain
Many LSDs that do not have an approved therapy can intelligible speech for as long as possible and can evalu-
be managed symptomatically, particularly in patients ate functional swallowing with various food textures to
who were diagnosed at an early stage. Regular follow-up facilitate safe feeding. In addition, speech and language
with a physiatrist or rehabilitative medicine specialist, in therapists can provide adaptive communication devices
addition to a neurologist, is important for patients with when speech is no longer intelligible. For patients with
NATUre RevIewS | DISeASe PRImeRS | Article citation ID: (2018) 4:27 19
Primer
Direct gene transfer Indirect gene transfer similar to the QOL of patients with more common dis-
orders, such as AD, PD and Huntington diseases. Some
Therapeutic Brain Stem cells
gene
late-onset LSDs, such as the GM2 gangliosidoses, can
include or even present with psychosis, which affects the
Liver
QOL of caregivers. For adult-onset LSDs without cog-
nitive dysfunction, depression might be a comorbid fea-
Delivery
vehicle ture, particularly for Fabry disease, in which the rate of
Delivery to
target tissues depression varies from 15% to 62% with the most com-
Cell mon associated factor being neuropathic pain162. Patients
expansion
Delivery to with Fabry disease also have a lower overall QOL than
target tissues healthy individuals163.
Seeking out health-care providers with specific exper-
Fig. 5 | Gene therapy methods in use for the treatment of LSDs. In general, there are tise in LSDs, usually within the context of an academic
two main methods for introducing a normal copy of the faulty transcript into patients. medical centre, is important for optimal management of
In direct gene transfer, a normal cDNA sequence, usually expressed from a ubiquitous therapies including supportive care. Clinical experience
promoter and containing a non-native poly(A) signal, is inserted into a vector (common
matters, particularly for patients who require anaesthe-
vectors include adeno-associated virus and lentivirus), tested for potency in animal
sia for surgery or other procedures164. Patient advocacy
models and/or cells and then manufactured for human application. In direct gene
transfer, the vector is infused into the bloodstream, the cerebrospinal fluid or target groups can aid in providing up-to-date information on
organs (for example, the brain). In indirect gene transfer, the vectors manufactured for clinical trials and research, as well as durable medical
human use are applied ex vivo to stem cells in culture. Cells expressing the gene product equipment and supportive therapies, and can instruct
are harvested and re-implanted back into the donor (autologous stem cell therapy). families how to navigate the health-care system to obtain
In indirect gene therapy , methods of bone marrow ablation (for example, partial or them. Knowing what to expect in the next phase of disease
complete) can affect the extent of grafting. LSD, lysosomal storage disease. progression and how to mitigate its effects on the QOL
can be empowering and transformational for patients and
visual impairment, vision specialists can recommend caregivers. Advocacy groups along with knowledgeable
large-print books or audio books as well as low-vision health-care providers can prepare families for the difficult
strategies for performing activities of daily living. discussions of gastrostomy or tracheostomy tube place-
ment and end-of-life care. As the parent of a child with
Quality of life infantile Tay–Sachs disease once shared, “Every life has
Diagnosing patients and providing them with a patient a beginning, a middle, and an end. Some are just shorter
advocacy community that has experience in caring for than others. We must plan for a full life for our children”.165
patients and family members with their LSD is impor-
tant and substantially improves the quality of life (QOL) Outlook
of patients and caregivers. Studies addressing QOL for Enormous progress has been made in LSD research
patients with LSDs or their caregivers are uncommon and therapy development over the past two decades,
and, accordingly, are still required. For QOL studies, and many successful clinical trials have resulted in
LSDs can be divided into disorders of childhood or FDA-approved and/or EMA-approved therapies that
adult onset and those with progressive neurodegener- have improved patient QOL and survival. However,
ation or those with preserved cognitive functioning. the majority of LSDs remain without an effective ther-
Of 266 parents with children with life-limiting conditions apy, particularly those with CNS involvement. As such,
(including LSDs), 95% identified receiving information major research efforts are in progress to develop new
from their physician on their children’s QOL as the most generations of therapies that effectively target the brain.
important factor in establishing trust in their child’s Progress has also been made in developing new cellu-
physician157. Parents with children with MPS disorders lar and animal models that better mirror the aberrant
feel the isolation and stigma of living with a rare disor- physiology and pathological processes of patients with
der, the disruption of typical family life, anxiety about LSDs than older models and that can be used to yield
their children’s well-being (both children with LSDs and new insights into disease mechanisms. These models are
healthy children) and an effect on their own physical also useful for understanding how the pathophysiology
and psychological health158. Among parents and children of LSDs converges with other human diseases and will
diagnosed with rare disorders, anxiety, social isolation, improve our ability to develop new therapies for rare
fear and uncertainty have been reported in addition to LSDs and more common neurodegenerative diseases.
symptoms of depression159,160. In addition, parents of
chronically ill children who have not been diagnosed New mechanisms of disease
identified increased anxiety and depression161. This The study of LSDs has led to a much greater under-
finding is relevant to the parents of children with LSDs standing of fundamental lysosome biology and has
because the time from onset of symptoms to diagnosis revealed the lysosome to be a highly complex signalling
is lengthy for many families. hub in addition to its role as the catabolic and recycling
Many adult-onset LSDs involve neurodegeneration, centre of the cell. The integration of lysosomal func-
including loss of the ability to ambulate or perform activ- tion with other aspects of cell metabolism explains why
ities of daily living, with some characterized by progres- lysosomal dysfunction causes damage to many aspects
sive dementia or disordered movement. Accordingly, of cellular homeostasis and damage to other organelles.
the QOL for these patients and their families might be Studying other functions of lysosomes is currently
a very active area of research and will likely hold the key to heat shock proteins such as heat shock protein 70, which
identification of novel clinical interventions in the future. is a protein that is synthesized in response to stress and is
part of the cellular network of molecular chaperones that
Therapeutics assist in protein folding166. To this end, one trial of ari-
ERTs remain the major class of approved therapies for moclomol, which stabilizes the transcription factor that
LSDs. The original ERT drugs were developed for dis- induces heat shock protein 70 expression, is ongoing in
orders that did not include CNS pathology and, there- patients with NPC, and could, in the future, be a general
fore, did not need to access the CNS to show efficacy. LSD therapy as this mechanism could be applicable to
However, the majority of LSDs involve the CNS and the multiple LSDs167 (Table 4).
peripheral nervous system, and drug development must
now include drugs that cross the blood–brain barrier. Stop codon readthrough therapy. In-frame premature
To accomplish this task, several drugs are being deliv- termination codons can be 'over-ridden' using small
ered directly to the CNS, and some small molecules that molecules such as gentamycin168, with many more com-
cross the blood–brain barrier are either approved or in pounds now under evaluation or development169. None
clinical trials for LSDs, including SRT, chaperones or are yet approved for an LSD and are not easily tested in
proteostasis modifiers (Table 4). Other major advances most knockout animal models as this would require the
are nucleic-acid-based or targeted approaches such as model to recapitulate the human stop codon mutation.
gene therapy that have now advanced to clinical trials One example in which this has been achieved in animal
for several LSDs. studies is in the Iduatm1Kmke mouse (a model of Hurler
syndrome), which carries a mutation homologous to the
Proteostasis modifiers. One potential therapeutic human W402X nonsense mutation170.
approach for LSDs is based on rescuing the function
of misfolded mutant proteins by harnessing the cell’s Disease-specific and anti-inflammatory therapies. For
endogenous proteostasis machinery via enhancement of some LSDs, such as NPC, disease-specific modifiers
are being evaluated in clinical trials (Table 4). For exam-
ple, intrathecal hydroxypropyl-β-cyclodextrin is being
a Genomic DNA evaluated in phase II/III clinical trials on the basis of
3' 5'
promising phase I/II clinical data from an observational
5' 3'
Target gene study171. The precise mechanism of action of this drug
has not been completely defined172,173, but the drug mobi-
Double-stranded break lizes storage of cholesterol and sphingolipids from the
brain in animal models of NPC173,174. Anti-inflammatory
therapies, such as adalimumab, an anti-TNF monoclonal
antibody, are also being evaluated in trials for several
Homology-directed repair Non-homologous end joining LSDs, including MPS I and MPS II (Table 4).
NATUre RevIewS | DISeASe PRImeRS | Article citation ID: (2018) 4:27 21
Primer
in which bone marrow stem cells isolated from patients Challenges of designing clinical trials
are transduced in vitro using lentiviral vectors encoding LSDs are individually rare and for many the natural
ARSA to overexpress the missing arylsulfatase A enzyme history is not well characterized. A major hurdle in
and are subsequently re-infused into the patient (Table 4). the approval of disease-modifying therapies is patient
After transplantation, the cells migrate to the brain and recruitment and a clinical trial design that can defin-
other organs, where they secrete arylsulfatase A. Patients itively determine whether a treatment is efficacious.
with metachromatic leukodystrophy who receive this In addition, the time from onset of symptoms to diag-
treatment have improved function and lifespan relative nosis is excessive, disease progression is variable even
to their untreated siblings184. Direct infusions of AAV within a given subtype and good natural history data
expressing the corrected gene into blood, CSF or tissues are lacking for many LSDs. Recruiting fairly homo
have also been tested in early clinical studies (Table 4). In geneous patient populations is therefore difficult, and
these studies, although only some cells would be geneti- robust outcome measures are often lacking. Trials for
cally corrected, the supposition is that a sufficient num- rare diseases are by definition small and tend to be
ber of cells will overexpress the gene product for cross underpowered, and for LSDs this is compounded by
correction of non-gene-corrected cells. clinical heterogeneity. What clinical parameters might
ASOs are often considered a tool for reducing the be responsive to a given therapy and in what time
expression of a mutated gene and act by binding to target frame clinical change will manifest is also unclear and
mRNAs to block the translation of the abnormal pro- can affect trial design. Efforts to improve the situation
tein or induce its degradation. ASOs could be useful for include creating patient registries, increasing awareness
inhibiting the proteins necessary for the production of to reduce the diagnostic delay and improving clinical
stored substrates (nucleic-acid-based SRT)185. However, trial design where possible.
ASOs can also promote splicing around mutations186,187.
Although ASO therapies for LSDs are only in early pre- Era of treatment leading to new symptoms
clinical testing using cellular and animal models, this LSDs are multisystem disorders. Therapies understand-
therapy has been approved for clinical use for other indi- ably target the most clinically meaningful (often life-
cations188,189, which might warrant further investigation altering) finding (for example, cardiovascular symptoms
for broader utility in LSDs. in infantile Pompe disease). With the primary symp-
Genome editing for treating an LSD can refer to cor- tom improved or ameliorated with effective therapies,
recting the mutated gene directly or editing the genome ‘secondary phenotypes’ are now emerging and require
to insert a new functional copy of the deficient gene190,191 treatment. Combination therapies to address the diverse
(Fig. 6a). For the correction of LSD-causing genes, the symptoms of LSDs will undoubtedly be required and are
target locus needs to be edited and a functional copy of now being employed.
the gene or a corrected copy of a portion of the mutant In summary, the LSD field is a vibrant basic research
locus inserted. These types of editing require homology- and translational arena with major advances in under-
directed repair and, regardless of the delivery method standing disease mechanisms and the development of
used (viral and non-viral approaches), can be done novel therapeutics to complement approved ERTs and
with zinc-finger nucleases, transcription activator-like small molecules. The convergence in pathogenetic
effector nucleases (TALENs) or CRISPR–Cas9-based pathways between LSDs and other diseases will shed
systems192 (Fig. 6b). The first gene therapy trials using much-needed light on fundamental disease mechanisms
editing for correcting an LSD are underway for MPS II and lead to some innovative cross-disease therapeutic
and involve the AAV-mediated correction of a mutant platforms in the future.
IDS sequence using zinc-finger nucleases (Table 4) based
on earlier preclinical work191. Published online xx xx xxxx
1. Di Fruscio, G. et al. Lysoplex: an efficient toolkit to 8. Meikle, P. J., Hopwood, J. J., Clague, A. E. & Carey, W. F. 16. van der Meijden, J. C. et al. Long-term follow-up of
detect DNA sequence variations in the autophagy- Prevalence of lysosomal storage disorders. JAMA. 17 patients with childhood Pompe disease treated
lysosomal pathway. Autophagy 11, 928–938 281, 249–254 (1999). with enzyme replacement therapy. J. Inherit Metab.
(2015). 9. US National Library of Medicine, Genetics Home Dis. https://doi.org/10.1007/s10545-018-0166-3
2. Palmieri, M. et al. Characterization of the CLEAR Reference. NIH https://ghr.nlm.nih.gov/ (2018). (2018).
network reveals an integrated control of cellular 10. Nita, D. A., Mole, S. E. & Minassian, B. A. Neuronal 17. Bley, A. E. et al. Natural history of infantile G(M2)
clearance pathways. Hum. Mol. Genet. 20, ceroid lipofuscinoses. Epilept. Disord. 18, 73–88 gangliosidosis. Pediatrics 128, e1233–e1241 (2011).
3852–3866 (2011). (2016). 18. Schulz, A. et al. Study of intraventricular cerliponase
3. Chapel, A. et al. An extended proteome map of the 11. Aronson, N. N. Jr Aspartylglycosaminuria: biochemistry alfa for CLN2 disease. N. Engl. J. Med. 378,
lysosomal membrane reveals novel potential and molecular biology. Biochim. Biophys. Acta 1455, 1898–1907 (2018).
transporters. Mol. Cell Proteom. 12, 1572–1588 139–154 (1999). 19. Jones, H. N. et al. Oropharyngeal dysphagia in infants
(2013). 12. Witkop, C. J. et al. Albinism and Hermansky-Pudlak and children with infantile Pompe disease. Dysphagia
4. Schroder, B. et al. Integral and associated syndrome in Puerto Rico. Bol. Asoc. Med. P R. 82, 25, 277–283 (2010).
lysosomal membrane proteins. Traffic 8, 333–339 (1990). 20. Nicolino, M. et al. Clinical outcomes after long-term
1676–1686 (2007). 13. Witkop, C. J., Almadovar, C., Pineiro, B. & treatment with alglucosidase alfa in infants and
5. Sleat, D. E. et al. Mass spectrometry-based protein Nunez Babcock, M. Hermansky-Pudlak syndrome children with advanced Pompe disease. Genet. Med.
profiling to determine the cause of lysosomal storage (HPS). An epidemiologic study. Ophthalm. Paediatr. 11, 210–219 (2009).
diseases of unknown etiology. Mol. Cell Proteom. 8, Genet. 11, 245–250 (1990). 21. Chakrapani, A., Vellodi, A., Robinson, P., Jones, S. &
1708–1718 (2009). 14. Kishnani, P. S. et al. A retrospective, multinational, Wraith, J. E. Treatment of infantile Pompe disease with
6. Szklarczyk, D. et al. STRING v10: protein-protein multicenter study on the natural history of alglucosidase alpha: the UK experience. J. Inherit
interaction networks, integrated over the tree of life. infantile-onset Pompe disease. J. Pediatr. 148, Metab. Dis. 33, 747–750 (2010).
Nucleic Acids Res. 43, D447–D452 (2015). 671–676 (2006). 22. Settembre, C., Fraldi, A., Medina, D. L. & Ballabio, A.
7. Parenti, G., Andria, G. & Ballabio, A. Lysosomal 15. Kishnani, P. S. et al. Recombinant human acid Signals from the lysosome: a control centre for cellular
storage diseases: from pathophysiology to therapy. α-glucosidase: major clinical benefits in infantile-onset clearance and energy metabolism. Nat. Rev. Mol.
Annu. Rev. Med. 66, 471–486 (2015). Pompe disease. Neurology 68, 99–109 (2007). Cell Biol. 14, 283–296 (2013).
23. Medina, D. L. & Ballabio, A. Lysosomal calcium https://doi.org/10.2174/187152731666617061512 73. Zunke, F. et al. Reversible conformational conversion
regulates autophagy. Autophagy 11, 970–971 (2015). 1753 (2017). of alpha-synuclein into toxic assemblies by
24. Todkar, K., Ilamathi, H. S. & Germain, M. 48. Lojewski, X. et al. Human iPSC models of neuronal glucosylceramide. Neuron 97, 92–107 (2018).
Mitochondria and lysosomes: discovering bonds. ceroid lipofuscinosis capture distinct effects of TPP1 74. Robak, L. A. et al. Excessive burden of lysosomal
Front. Cell Dev. Biol. 5, 106 (2017). and CLN3 mutations on the endocytic pathway. storage disorder gene variants in Parkinson’s disease.
25. Kilpatrick, B. S. et al. An endosomal NAADP-sensitive Hum. Mol. Genet. 23, 2005–2022 (2014). Brain 140, 3191–3203 (2017).
two-pore Ca(2+) channel regulates ER-endosome 49. Panicker, L. M. et al. Induced pluripotent stem cell 75. Suzuki, Y., Oshima, A. & Nanba, E. in The Metabolic
membrane contact sites to control growth factor model recapitulates pathologic hallmarks of and Molecular Bases of Inherited Disease Vol. 3
signaling. Cell Rep. 18, 1636–1645 (2017). Gaucher disease. Proc. Natl Acad. Sci. USA 109, (eds Scriver, C. R., Beadet, A. L., Valle, D. & Sly, W. S.)
26. Annunziata, I., Sano, R. & d’Azzo, A. 18054–18059 (2012). 3775–3809 (McGraw Hill, 2001).
Mitochondria-associated ER membranes (MAMs) 50. Aflaki, E. et al. A new glucocerebrosidase chaperone 76. d’Azzo, A., Tessitore, A. & Sano, R. Gangliosides as
and lysosomal storage diseases. Cell Death Dis. 9, reduces alpha-synuclein and glycolipid levels in apoptotic signals in ER stress response. Cell Death
328 (2018). iPSC-derived dopaminergic neurons from patients with Differ. 13, 404–414 (2006).
27. Wong, Y. C., Ysselstein, D. & Krainc, D. Gaucher disease and parkinsonism. J. Neurosci. 36, 77. Ledeen, R. W. & Wu, G. GM1 ganglioside: another
Mitochondria-lysosome contacts regulate 7441–7452 (2016). nuclear lipid that modulates nuclear calcium. GM1
mitochondrial fission via RAB7 GTP hydrolysis. 51. Mistry, P. K. et al. Gaucher disease: progress and potentiates the nuclear sodium-calcium exchanger.
Nature 554, 382–386 (2018). ongoing challenges. Mol. Genet. Metab. 120, 8–21 Can. J. Physiol. Pharmacol. 84, 393–402 (2006).
28. Grabowski, G. A. Overview of inflammation in (2017). 78. Ledeen, R. W. & Wu, G. The multi-tasked life of GM1
neurometabolic diseases. Semin. Pediatr. Neurol. 24, 52. Beutler, E. Gaucher’s disease. N. Engl. J. Med. 325, ganglioside, a true factotum of nature. Trends
207–213 (2017). 1354–1360 (1991). Biochem. Sci. 40, 407–418 (2015).
29. Rigante, D., Cipolla, C., Basile, U., Gulli, F. & 53. Borger, D. K., Sidransky, E. & Aflaki, E. New macrophage 79. Hahn, C. N. et al. Generalized CNS disease and
Savastano, M. C. Overview of immune abnormalities models of Gaucher disease offer new tools for drug massive GM1-ganglioside accumulation in mice
in lysosomal storage disorders. Immunol. Lett. 188, development. Macrophage (Houst) 2, e712 (2015). defective in lysosomal acid beta-galactosidase.
79–85 (2017). 54. Aflaki, E. et al. Lysosomal storage and impaired Hum. Mol. Genet. 6, 205–211 (1997).
30. Vitner, E. B., Platt, F. M. & Futerman, A. H. Common autophagy lead to inflammasome activation 80. Tessitore, A. et al. A GM1-ganglioside-mediated
and uncommon pathogenic cascades in lysosomal in Gaucher macrophages. Aging Cell 15, 77–88 activation of the unfolded protein response causes
storage diseases. J. Biol. Chem. 285, 20423–20427 (2016). neuronal death in a neurodegenerative gangliosidosis.
(2010). 55. Aflaki, E., Westbroek, W. & Sidransky, E. Mol. Cell. 15, 753–766 (2004).
31. Vitner, E. B. et al. RIPK3 as a potential therapeutic The complicated relationship between Gaucher disease 81. Sano, R. et al. GM1-ganglioside accumulation at the
target for Gaucher’s disease. Nat. Med. 20, 204–208 and parkinsonism: insights from a rare disease. Neuron mitochondria-associated ER membranes links ER
(2014). 93, 737–746 (2017). stress to Ca(2+)-dependent mitochondrial apoptosis.
32. Wada, R., Tifft, C. J. & Proia, R. L. Microglial activation 56. Wong, K. et al. Neuropathology provides clues to the Mol. Cell 36, 500–511 (2009).
precedes acute neurodegeneration in Sandhoff pathophysiology of Gaucher disease. Mol. Genet. Metab. 82. Yanagisawa, K., Odaka, A., Suzuki, N. & Ihara, Y.
disease and is supressed by bone marrow 82, 192–207 (2004). GM1 ganglioside-bound amyloid beta-protein
transplantation. Proc. Natl Acad. Sci. USA 97, 10954– 57. Vitner, E. B. & Futerman, A. H. in Sphingolipids in (A beta): a possible form of preamyloid in Alzheimer’s
10959 (2000). Disease. Handbook of Experimental Pharmacology, disease. Nat. Med. 1, 1062–1066 (1995).
33. Jeyakumar, M. et al. Central nervous system vol 216 (eds Gulbins E. & Petrache I.) 405–419 83. Yanagisawa, K. GM1 ganglioside and Alzheimer’s
inflammation is a hallmark of pathogenesis in mouse (Springer, Vienna 2013). disease. Glycoconj. J. 32, 87–91 (2015).
models of GM1 and GM2 gangliosidosis. Brain 126, 58. Vitner, E. B., Farfel-Becker, T., Eilam, R., Biton, I. & 84. Hirano-Sakamaki, W. et al. Alzheimer’s disease is
974–987 (2003). Futerman, A. H. Contribution of brain inflammation associated with disordered localization of ganglioside
34. Jeyakumar, M. et al. NSAIDs increase survival to neuronal cell death in neuronopathic forms of GM1 molecular species in the human dentate gyrus.
in the Sandhoff disease mouse: synergy with Gaucher’s disease. Brain 135, 1724–1735 (2012). FEBS Lett. 589, 3611–3616 (2015).
N-butyldeoxynojirimycin. Ann. Neurol. 56, 642–649 59. Vitner, E. B. et al. Induction of the type I interferon 85. Calamai, M. et al. Single molecule experiments
(2004). response in neurological forms of Gaucher disease. emphasize GM1 as a key player of the different
35. Bosch, M. E. & Kielian, T. Neuroinflammatory J. Neuroinflamm. 13, 104 (2016). cytotoxicity of structurally distinct Aβ1-42
paradigms in lysosomal storage diseases. 60. Allen, M. J., Myer, B. J., Khokher, A. M., Rushton, N. oligomers. Biochim. Biophys. Acta 1858, 386–392
Front. Neurosci. 9, 417 (2015). & Cox, T. M. Pro-inflammatory cytokines and the (2016).
36. Smith, D., Wallom, K. L., Williams, I. M., Jeyakumar, M. pathogenesis of Gaucher’s disease: increased release 86. Calamai, M. & Pavone, F. S. Partitioning and
& Platt, F. M. Beneficial effects of anti-inflammatory of interleukin-6 and interleukin-10. Q. J. Med. 90, confinement of GM1 ganglioside induced by amyloid
therapy in a mouse model of Niemann-Pick 19–25 (1997). aggregates. FEBS Lett. 587, 1385–1391 (2013).
disease type C1. Neurobiol. Dis. 36, 242–251 61. Cox, T. M. Gaucher disease: understanding the 87. d’Azzo, A., Machado, E. & Annunziata, I. Pathogenesis,
(2009). molecular pathogenesis of sphingolipidoses. J. Inherit. emerging therapeutic targets and treatment in sialidosis.
37. Yamaguchi, A. et al. Possible role of autoantibodies Metab. Dis. 24, 106–121 (2001). Expert Opin. Orphan Drugs 3, 491–504 (2015).
in the pathophysiology of GM2 gangliosidoses. 62. Pandey, M. K. et al. Complement drives 88. Bonten, E. J., Annunziata, I. & d’Azzo, A. Lysosomal
J. Clin. Invest. 113, 200–208 (2004). glucosylceramide accumulation and tissue multienzyme complex: pros and cons of working
38. Ballabio, A. & Gieselmann, V. Lysosomal disorders: inflammation in Gaucher disease. Nature 543, together. Cell. Mol. Life Sci. 71, 2017–2032 (2014).
from storage to cellular damage. Biochim. Biophys. 108–112 (2017). 89. de Geest, N. et al. Systemic and neurologic
Acta 1793, 684–696 (2009). 63. Aker, M., Zimran, A., Abrahamov, A., Horowitz, M. & abnormalities distinguish the lysosomal disorders
39. Saftig, P. in Fabry Disease: Perspectives from 5 Years of Matzner, Y. Abnormal neutrophil chemotaxis in sialidosis and galactosialidosis in mice. Hum. Mol.
FOS (eds Mehta, A., Beck, M. & Sunder-Plassmann, G.) Gaucher disease. Br. J. Haematol. 83, 187–191 Genet. 11, 1455–1464 (2002).
21–31 (2006). (1993). 90. Yogalingam, G. et al. Neuraminidase 1 is a negative
40. Sidransky, E. & Lopez, G. The link between the GBA 64. Deganuto, M. et al. Altered intracellular redox status regulator of lysosomal exocytosis. Dev. Cell 15, 74–86
gene and parkinsonism. Lancet Neurol. 11, 986–998 in Gaucher disease fibroblasts and impairment (2008).
(2012). of adaptive response against oxidative stress. This study is the first demonstration that excessive
This review summarizes the link between J. Cell. Physiol. 212, 223–235 (2007). lysosomal exocytosis downstream of NEU1
mutations in a rare disease-causing gene, GBA, 65. Thomas, A. S., Mehta, A. & Hughes, D. A. deficiency is responsible for the pathogenesis of
and a common neurological disorder, PD. Gaucher disease: haematological presentations and sialidosis.
41. Annunziata, I. et al. Lysosomal NEU1 deficiency affects complications. Br. J. Haematol. 165, 427–440 91. Wu, X. et al. Vacuolization and alterations of
amyloid precursor protein levels and amyloid-beta (2014). lysosomal membrane proteins in cochlear marginal
secretion via deregulated lysosomal exocytosis. 66. Enquist, I. B. et al. Murine models of acute cells contribute to hearing loss in neuraminidase
Nat. Commun. 4, 2734 (2013). neuronopathic Gaucher disease. Proc. Natl Acad. 1-deficient mice. Biochim. Biophys. Acta 1802,
42. Zanoteli, E. et al. Muscle degeneration in neuraminidase Sci. USA 104, 17483–17488 (2007). 259–268 (2010).
1-deficient mice results from infiltration of the 67. Farfel-Becker, T., Vitner, E. B. & Futerman, A. H. 92. Reddy, A., Caler, E. V. & Andrews, N. W. Plasma
muscle fibers by expanded connective tissue. Animal models for Gaucher disease research. membrane repair is mediated by Ca(2+)-regulated
Biochim. Biophys. Acta 1802, 659–672 (2010). Dis. Model. Mech. 4, 746–752 (2011). exocytosis of lysosomes. Cell 106, 157–169 (2001).
43. Machado, E. et al. Regulated lysosomal exocytosis 68. Tayebi, N. et al. Gaucher disease and parkinsonism: 93. Vanier, M. T. Niemann-Pick disease type C. Orphanet
mediates cancer progression. Sci. Adv. 1, e1500603 a phenotypic and genotypic characterization. J. Rare Dis. 5, 16 (2010).
(2015). Mol. Genet. Metab. 73, 313–321 (2001). 94. Lloyd-Evans, E. et al. Niemann-Pick disease type C1 is
44. Bellettato, C. M. & Scarpa, M. Pathophysiology of 69. Aflaki, E. et al. Efferocytosis is impaired in Gaucher a sphingosine storage disease that causes deregulation
neuropathic lysosomal storage disorders. J. Inherit. macrophages. Haematologica 102, 656–665 of lysosomal calcium. Nat. Med. 14, 1247–1255
Metab. Dis. 33, 347–362 (2010). (2017). (2008).
45. Sidransky, E. et al. Multicenter analysis of 70. Pringsheim, T., Jette, N., Frolkis, A. & Steeves, T. D. 95. Lee, H. et al. Bone marrow-derived mesenchymal stem
glucocerebrosidase mutations in Parkinson’s disease. The prevalence of Parkinson’s disease: a systematic cells prevent the loss of Niemann-Pick type C mouse
N. Engl. J. Med. 361, 1651–1661 (2009). review and meta-analysis. Mov Disord. 29, Purkinje neurons by correcting sphingolipid metabolism
This study proves a strong genetic relationship 1583–1590 (2014). and increasing sphingosine-1-phosphate. Stem Cells
between GBA mutations and risk of PD. 71. Mazzulli, J. R. et al. Gaucher disease glucocerebrosidase 28, 821–831 (2010).
46. Borger, D. K., Aflaki, E. & Sidransky, E. and alpha-synuclein form a bidirectional pathogenic 96. Visentin, S. et al. The stimulation of adenosine A2A
Applications of iPSC-derived models of Gaucher loop in synucleinopathies. Cell 146, 37–52 (2011). receptors ameliorates the pathological phenotype of
disease. Ann. Transl Med. 3, 295 (2015). 72. Cullen, V. et al. Acid beta-glucosidase mutants linked fibroblasts from Niemann-Pick type C patients.
47. De Filippis, L., Zalfa, C. & Ferrari, D. Neural stem cells to Gaucher disease, Parkinson disease, and Lewy J. Neurosci. 33, 15388–15393 (2013).
and human induced pluripotent stem cells to model body dementia alter alpha-synuclein processing. 97. Xu, M. et al. δ-Tocopherol reduces lipid accumulation
rare CNS diseases. CNS Neurol Disord Drug Targets. Ann. Neurol. 69, 940–953 (2011). in Niemann-Pick type C1 and Wolman cholesterol
NATUre RevIewS | DISeASe PRImeRS | Article citation ID: (2018) 4:27 23
Primer
storage disorders. J. Biol. Chem. 287, 39349–39360 120. Cox, T. M. Innovative treatments for lysosomal 142. Shayman, J. A. ELIGLUSTAT TARTRATE:
(2012). diseases. Best Pract. Res. Clin. Endocrinol. Metab. 29, glucosylceramide synthase inhibitor treatment of type 1
98. Liscum, L. Niemann-Pick type C mutations cause lipid 275–311 (2015). Gaucher disease. Drugs Future 35, 613–620 (2010).
traffic jam. Traffic 1, 218–225 (2000). 121. Hobbs, J. R. et al. Reversal of clinical features of 143. Platt, F. M., Neises, G. R., Dwek, R. A. & Butters, T. D.
99. Ioannou, Y. A. Multidrug permeases and subcellular Hurler’s disease and biochemical improvement after N-Butyldeoxynojirimycin is a novel inhibitor of
cholesterol transport. Nat. Rev. Mol. Cell Biol. 2, treatment by bone-marrow transplantation. Lancet 2, glycolipid biosynthesis. J. Biol. Chem. 269,
657–668 (2001). 709–712 (1981). 8362–8365 (1994).
100. Davies, J. P., Chen, F. W. & Ioannou, Y. A. This study is the first demonstration that Hurler 144. Walterfang, M. et al. Dysphagia as a risk factor for
Transmembrane molecular pump activity of syndrome is a treatable disease. mortality in Niemann-Pick disease type C: systematic
Niemann-Pick C1 protein. Science 290, 2295–2298 122. Aldenhoven, M. & Kurtzberg, J. Cord blood is the literature review and evidence from studies with
(2000). optimal graft source for the treatment of pediatric miglustat. Orphanet J. Rare Dis. 7, 76 (2012).
101. Goldman, S. D. & Krise, J. P. Niemann-Pick C1 patients with lysosomal storage diseases: clinical 145. Lyseng-Williamson, K. A. Miglustat: a review of its use
functions independently of Niemann-Pick C2 in the outcomes and future directions. Cytotherapy 17, in niemann-pick disease type C. Drugs 74, 61–74
initial stage of retrograde transport of membrane- 765–774 (2015). (2014).
impermeable lysosomal cargo. J. Biol. Chem. 285, 123. Boelens, J. J. et al. Outcomes of transplantation using 146. Cox, T. M. Eliglustat tartrate, an orally active
4983–4994 (2010). various hematopoietic cell sources in children with glucocerebroside synthase inhibitor for the potential
102. Auer, I. A. et al. Paired helical filament tau (PHFtau) in Hurler syndrome after myeloablative conditioning. treatment of Gaucher disease and other lysosomal
Niemann-Pick type C disease is similar to PHFtau in Blood 121, 3981–3987 (2013). storage diseases. Curr. Opin. Investig. Drugs 11,
Alzheimer’s disease. Acta Neuropathol. 90, 547–551 124. Poe, M. D., Chagnon, S. L. & Escolar, M. L. Early 1169–1181 (2010).
(1995). treatment is associated with improved cognition in 147. Cox, T. M. & Mistry, P. K. Therapeutic position
103. Huizing, M., Helip-Wooley, A., Westbroek, W., Hurler syndrome. Ann. Neurol. 76, 747–753 of eliglustat. Blood Cells Mol. Dis. 69, 117–118
Gunay-Aygun, M. & Gahl, W. A. Disorders of lysosome- (2014). (2018).
related organelle biogenesis: clinical and molecular 125. Sly, W. S., Kaplan, A., Achord, D. T., Brot, F. E. & 148. Lachmann, R. H. Miglustat. Oxford GlycoSciences/
genetics. Annu. Rev. Genom. Hum. Genet. 9, 359–386 Bell, C. E. Receptor-mediated uptake of lysosomal Actelion. Curr. Opin. Investig. Drugs 4, 472–479
(2008). enzymes. Prog. Clin. Biol. Res. 23, 547–551 (2003).
104. Raposo, G., Fevrier, B., Stoorvogel, W. & Marks, M. S. (1978). 149. Belmatoug, N. et al. Management and monitoring
Lysosome-related organelles: a view from immunity This paper reviews the evidence for recognition of recommendations for the use of eliglustat in adults
and pigmentation. Cell Struct. Funct. 27, 443–456 lysosomal enzymes by receptors for mannose and with type 1 Gaucher disease in Europe. Eur. J.
(2002). for mannose 6-phosphate. Intern. Med. 37, 25–32 (2017).
105. Neudorfer, O. et al. Late-onset Tay-Sachs disease: 126. Kan, S. H. et al. Delivery of an enzyme-IGFII fusion 150. Clayton, N. P. et al. Antisense oligonucleotide-
phenotypic characterization and genotypic protein to the mouse brain is therapeutic for mediated suppression of muscle glycogen synthase 1
correlations in 21 affected patients. Genet. Med. 7, mucopolysaccharidosis type IIIB. Proc. Natl Acad. synthesis as an approach for substrate reduction
119–123 (2005). Sci. USA 111, 14870–14875 (2014). therapy of Pompe disease. Mol. Ther. Nucleic Acids 3,
106. Veys, K. R., Elmonem, M. A., Arcolino, F. O., 127. Grabowski, G. A. Gaucher disease and other storage e206 (2014).
van den Heuvel, L. & Levtchenko, E. Nephropathic disorders. Hematol. Am. Soc. Hematol. Educ. Program 151. Parenti, G., Andria, G. & Valenzano, K. J.
cystinosis: an update. Curr. Opin. Pediatr. 29, 2012, 13–18 (2012). Pharmacological chaperone therapy: preclinical
168–178 (2017). 128. Zimran, A. How I treat Gaucher disease. Blood 118, development, clinical translation, and prospects
107. Patterson, M. C. et al. Recommendations for the 1463–1471 (2011). for the treatment of lysosomal storage disorders.
diagnosis and management of Niemann-Pick disease 129. Boado, R. J., Lu, J. Z., Hui, E. K., Lin, H. & Mol. Ther. 23, 1138–1148 (2015).
type C: an update. Mol. Genet. Metab. 106, 330–344 Pardridge, W. M. Insulin receptor Antibody-alpha- 152. Markham, A. Migalastat: first global approval. Drugs
(2012). N-Acetylglucosaminidase fusion protein penetrates 76, 1147–1152 (2016).
108. Evans, W. R. & Hendriksz, C. J. Niemann-Pick type C the primate blood-brain barrier and reduces 153. Hughes, D. A. et al. Oral pharmacological chaperone
disease - the tip of the iceberg? A review of glycosoaminoglycans in Sanfilippo Type B fibroblasts. migalastat compared with enzyme replacement
neuropsychiatric presentation, diagnosis and Mol. Pharm. 13, 1385–1392 (2016). therapy in Fabry disease: 18-month results from the
treatment. BJPsych Bull. 41, 109–114 (2017). 130. Markham, A. Cerliponase Alfa: first global approval. randomised phase III ATTRACT study. J. Med. Genet.
109. Cooper, J. D., Tarczyluk, M. A. & Nelvagal, H. R. Drugs 77, 1247–1249 (2017). 54, 288–296 (2017).
Towards a new understanding of NCL pathogenesis. 131. Broomfield, A., Jones, S. A., Hughes, S. M. & This clinical study compares the first-in-class
Biochim. Biophys. Acta 1852, 2256–2261 (2015). Bigger, B. W. The impact of the immune system on the approved small-molecule chaperone drug
110. Preising, M. N., Abura, M., Jager, M., Wassill, K. H. & safety and efficiency of enzyme replacement therapy in migalastat with standard-of-care ERT,
Lorenz, B. Ocular morphology and function in juvenile lysosomal storage disorders. J. Inherit Metab. Dis. 39, showing its potential as an alternative to ERT in
neuronal ceroid lipofuscinosis (CLN3) in the first 499–512 (2016). patients with Fabry disease with chaperonable
decade of life. Ophthalm. Genet. 38, 252–259 132. Afroze, B. & Brown, N. Ethical issues in managing mutations.
(2017). Lysosomal storage disorders in children in low and 154. Germain, D. P. et al. Treatment of Fabry’s disease with
111. Williams, R. E. et al. Management strategies for CLN2 middle income countries. Pak. J. Med. Sci. 33, the pharmacologic chaperone migalastat. N. Engl.
disease. Pediatr. Neurol. 69, 102–112 (2017). 1036–1041 (2017). J. Med. 375, 545–555 (2016).
112. Introne, W. J. et al. Neurologic involvement in patients 133. McGill, J. J. et al. Enzyme replacement therapy for 155. Schiffmann, R. et al. Migalastat improves diarrhea in
with atypical Chediak-Higashi disease. Neurology 88, mucopolysaccharidosis VI from 8 weeks of age — patients with Fabry disease: clinical-biomarker
e57–e65 (2017). a sibling control study. Clin. Genet. 77, 492–498 correlations from the phase 3 FACETS trial. Orphanet
113. Suzuki, K. Enzymic diagnosis of sphingolipidoses. (2010). J. Rare Dis. 13, 68 (2018).
Methods Enzymol. 50, 456–488 (1978). 134. Aerts, J. M., Hollak, C. E., Boot, R. G., Groener, J. E. & 156. Citro, V. et al. Identification of an allosteric binding
114. Kaback, M. et al. Tay-Sachs disease—carrier Maas, M. Substrate reduction therapy of site on human lysosomal alpha-galactosidase
screening, prenatal diagnosis, and the molecular era. glycosphingolipid storage disorders. J. Inherit Metab. opens the way to new pharmacological chaperones
An international perspective, 1970 to 1993. Dis. 29, 449–456 (2006). for Fabry disease. PLOS One 11, e0165463
The International TSD Data Collection Network. 135. Lachmann, R. H. & Platt, F. M. Substrate (2016).
JAMA. 270, 2307–2315 (1993). reduction therapy for glycosphingolipid storage 157. Huang, I. C. et al. Quality of life information and trust
This seminal paper discusses the high-risk disorders. Expert Opin. Investig. Drugs 10, 455–466 in physicians among families of children with life-
population screening that reduced the incidence of (2001). limiting conditions. Patient Relat. Outcome Meas.
a uniformly fatal disease by 90% within 10 years. 136. Cox, T. et al. Novel oral treatment of Gaucher’s disease 2010, 141–148 (2010).
115. Regier, D. S., Proia, R. L., D’Azzo, A. & Tifft, C. J. with N-butyldeoxynojirimycin (OGT 918) to decrease 158. Somanadhan, S. & Larkin, P. J. Parents’ experiences of
The GM1 and GM2 Gangliosidoses: natural history substrate biosynthesis. Lancet 355, 1481–1485 living with, and caring for children, adolescents and
and progress toward therapy. Pediatr. Endocrinol. Rev. (2000). young adults with Mucopolysaccharidosis (MPS).
13, 663–673 (2016). This study provides the first clinical evidence of the Orphanet J. Rare Dis. 11, 138 (2016).
116. van der Tol, L. et al. A systematic review on screening efficacy of SRT using the first approved SRT drug, 159. Pelentsov, L. J., Laws, T. A. & Esterman, A. J.
for Fabry disease: prevalence of individuals with miglustat, in Gaucher disease type I. The supportive care needs of parents caring for a child
genetic variants of unknown significance. J. Med. 137. Hollak, C. E., Hughes, D., van Schaik, I. N., Schwierin, B. with a rare disease: a scoping review. Disabil Health J.
Genet. 51, 1–9 (2014). & Bembi, B. Miglustat (Zavesca) in type 1 Gaucher 8, 475–491 (2015).
117. Hoffman, J. D. et al. Next-generation DNA sequencing disease: 5-year results of a post-authorisation safety 160. Besier, T. et al. Anxiety, depression, and life
of HEXA: a step in the right direction for carrier surveillance programme. Pharmacoepidemiol. Drug satisfaction in parents caring for children with
screening. Mol. Genet. Genom. Med. 1, 260–268 Safety 18, 770–777 (2009). cystic fibrosis. Pediatr. Pulmonol 46, 672–682
(2013). 138. Poole, R. M. Eliglustat: first global approval. Drugs 74, (2011).
118. [No authors listed]. Committee Opinion Number 691. 1829–1836 (2014). 161. McConkie-Rosell, A. et al. Psychosocial profiles of
American College of Obstetricians and Gynecologists 139. Belmatoug, N. et al. Management and monitoring parents of children with undiagnosed diseases:
https://www.acog.org/Clinical-Guidance-and- recommendations for the use of eliglustat in adults managing well or just managing? J. Genet. Couns. 27,
Publications/Committee-Opinions/Committee-on- with type 1 Gaucher disease in Europe. Eur. J. 935–946 (2018).
Genetics/Carrier-Screening-for-Genetic-Conditions Intern. Med. 37, 25–32 (2016). 162. Bolsover, F. E., Murphy, E., Cipolotti, L., Werring, D. J.
(2017). 140. Patterson, M. C. et al. Oral miglustat in Niemann-Pick & Lachmann, R. H. Cognitive dysfunction and
119. Schielen, P., Kemper, E. A. & Gelb, M. H. Newborn type C (NPC) disease. Rev. Neurol. (Separata) 43, 8 depression in Fabry disease: a systematic review.
screening for lysosomal storage diseases: a concise (2006). J. Inherit Metab. Dis. 37, 177–187 (2014).
review of the literature on screening methods, 141. Patterson, M. C., Vecchio, D., Prady, H., Abel, L. & 163. Arends, M., Hollak, C. E. & Biegstraaten, M. Quality of
therapeutic possibilities and regional programs. Wraith, J. E. Miglustat for treatment of Niemann-Pick C life in patients with Fabry disease: a systematic
Int. J. Neonatal Screen. https://doi.org/10.3390/ disease: a randomised controlled study. Lancet Neurol. review of the literature. Orphanet J. Rare Dis. 10, 77
ijns3020006 (2017). 6, 765–772 (2007). (2015).
164. Kloesel, B. & Holzman, R. S. Anesthetic management 183. Mendell, J. R. et al. Single-dose gene-replacement 202. Aula, N. et al. The spectrum of SLC17A5-gene
of patients with inborn errors of metabolism. therapy for spinal muscular atrophy. N. Engl. J. Med. mutations resulting in free sialic acid storage diseases
Anesth Analg. 125, 822–836 (2017). 377, 1713–1722 (2017). indicates some genotype-phenotype correlation.
165. Goldstein, R. (ed.) Cameron’s Arc: Creating a Full Life 184. Biffi, A. et al. Lentiviral hematopoietic stem cell gene Am. J. Hum. Genet. 67, 832–840 (2000).
(American Academy of Paediatrics, 2007). therapy benefits metachromatic leukodystrophy. 203. Greer, W. L. et al. The Nova Scotia (Type D) form of
166. Petersen, N. H. & Kirkegaard, T. HSP70 and Science 341, 1233158 (2013). Niemann-Pick disease is caused by a G3097 to T
lysosomal storage disorders: novel therapeutic 185. Goina, E., Peruzzo, P., Bembi, B., Dardis, A. & transversion in NPC1. Am. J. Hum. Genet. 63, 52–54
opportunities. Biochem. Soc. Trans. 38, 1479–1483 Buratti, E. Glycogen reduction in myotubes of (1998).
(2010). late-onset pompe disease patients using antisense
167. Kirkegaard, T. et al. Heat shock protein-based therapy technology. Mol. Ther. 25, 2117–2128 (2017). Acknowledgements
as a potential candidate for treating the 186. van der Wal, E., Bergsma, A. J., Pijnenburg, J. M., F.M.P. is a Royal Society Wolfson Merit award holder and a
sphingolipidoses. Sci. Transl. Med. 8, 355ra118 van der Ploeg, A. T. & Pijnappel, W. Antisense Wellcome Trust Investigator in Science. A.d.A. is supported by
(2016). oligonucleotides promote exon inclusion and correct NIH grants DK095169 and GM104981, the Assisi
168. Keeling, K. M., Xue, X., Gunn, G. & Bedwell, D. M. the common c.-32-13T>G GAA splicing variant in Foundation of Memphis, Ultragenyx Pharmaceutical and the
Therapeutics based on stop codon readthrough. pompe disease. Mol. Ther. Nucleic Acids 7, 90–100 American Lebanese Syrian Associated Charities (ALSAC) and
Annu. Rev. Genom. Hum. Genet. 15, 371–394 (2017). holds the Jewellers for Children Endowed Chair in Genetics
(2014). 187. Siva, K., Covello, G. & Denti, M. A. Exon-skipping and Gene Therapy. A.d.A. thanks B. Stelter (St. Jude
169. Keeling, K. M. Nonsense suppression as an approach antisense oligonucleotides to correct missplicing in Biomedical Communication) for help with the graphic design
to treat lysosomal storage diseases. Diseases 4, 32 neurogenetic diseases. Nucleic Acid. Ther. 24, 69–86 of Fig. 3 and I. Annunziata for her assistance with literature
(2016). (2014). screening and building the End Note library for the
170. Hein, L. K. et al. alpha-L-iduronidase premature 188. Corey, D. R. Nusinersen, an antisense oligonucleotide Mechanisms/pathophysiology section. B.L.D. is supported by
stop codons and potential read-through in drug for spinal muscular atrophy. Nat. Neurosci. 20, the Children’s Hospital of Philadelphia Research Institute
mucopolysaccharidosis type I patients. J. Mol. Biol. 497–499 (2017). and the NIH (NS76631, NS090390 and NS94355) and holds
338, 453–462 (2004). 189. Khvorova, A. & Watts, J. K. The chemical evolution the Arthur V. Meigs Chair in Pediatrics. C.J.T. is supported
171. Ory, D. S. et al. Intrathecal 2-hydroxypropyl-beta- of oligonucleotide therapies of clinical utility. by the Division of Intramural Research of the National Human
cyclodextrin decreases neurological disease Nat. Biotechnol. 35, 238–248 (2017). Genome Research Institute of the NIH, US Department of
progression in Niemann-Pick disease, type C1: 190. Schneller, J. L., Lee, C. M., Bao, G. & Venditti, C. P. Health and Human Services. C.J.T. thanks M. Huizing for
a non-randomised, open-label, phase 1–2 trial. Genome editing for inborn errors of metabolism: design of Fig. 4 and organization of the section on disorders
Lancet 390, 1758–1768 (2017). advancing towards the clinic. BMC Med. 15, 43 of LROs.
172. Chen, F. W., Li, C. & Ioannou, Y. A. Cyclodextrin (2017).
induces calcium-dependent lysosomal exocytosis. 191. Sharma, R. et al. In vivo genome editing of
PLOS One 5, e15054 (2010). the albumin locus as a platform for protein Author contributions
173. Dai, S. et al. Methyl-β-cyclodextrin restores impaired replacement therapy. Blood 126, 1777–1784 Introduction (all); Epidemiology (C.J.T.); Mechanisms/
autophagy flux in Niemann-Pick C1-deficient cells (2015). pathophysiology (A.d.A. and F.M.P.); Diagnosis, screening
through activation of AMPK. Autophagy 13, 192. Yin, H., Kauffman, K. J. & Anderson, D. G. and prevention (C.J.T.); Management (E.F.N., B.L.D. and
1435–1451 (2017). Delivery technologies for genome editing. Nat. Rev. F.M.P.); Quality of life (C.J.T.); Outlook (all); Overview of
174. Davidson, C. D. et al. Chronic cyclodextrin treatment Drug Discov. 16, 387–399 (2017). Primer (F.M.P.).
of murine Niemann-Pick C disease ameliorates 193. Pu, J., Guardia, C. M., Keren-Kaplan, T. &
neuronal cholesterol and glycosphingolipid storage Bonifacino, J. S. Mechanisms and functions of Competing interests
and disease progression. PLOS One 4, e6951 (2009). lysosome positioning. J. Cell Sci. 129, 4329–4339 F.M.P. is a trustee of Gordon Research Conferences, Chair of
175. Sands, M. S. & Davidson, B. L. Gene therapy for (2016). the Scientific Advisory Board of the National Tay-Sachs &
lysosomal storage diseases. Mol. Ther. 13, 839–849 194. Carette, J. E. et al. Ebola virus entry requires the Allied Diseases Association (NTSAD), a cofounder of and con-
(2006). cholesterol transporter Niemann-Pick C1. Nature sultant to IntraBio and a consultant to Actelion and
176. Aronovich, E. L. et al. Prolonged expression of secreted 477, 340–343 (2011). Orphazyme. A.d.A. has received research support from
enzymes in dogs after liver-directed delivery of sleeping 195. Hulseberg, C. E., Feneant, L., Szymanska, K. M. & Ultragenyx Pharmaceutical. B.L.D. founded Talee Bio, Inc.
beauty transposons: implications for non-viral gene White, J. M. Lamp1 increases the efficiency of lassa and Spark Therapeutics and is on the Scientific Advisory
therapy of systemic disease. Hum. Gene Ther. 28, virus infection by promoting fusion in less acidic Board of Homology Medicines, Intellia Therapeutics, Prevail
551–564 (2017). endosomal compartments. MBio. https://doi.org/ Therapeutics, Inc and Sarepta Therapeutics. E.F.N. has previ-
177. Sandrin, V., Russell, S. J. & Cosset, F. L. Targeting 10.1128/mBio.01818-17 (2018). ously received funding from BioMarin and is a member of the
retroviral and lentiviral vectors. Curr. Top. Microbiol. 196. Fineran, P. et al. Pathogenic mycobacteria achieve Scientific Advisory Board of the National MPS Society. C.J.T.
Immunol. 281, 137–178 (2003). cellular persistence by inhibiting the Niemann-Pick is supported by the Division of Intramural Research of the
178. Davidson, B. L. et al. Recombinant adeno-associated Type C disease cellular pathway. Wellcome Open Res. National Human Genome Research Institute of the NIH, US
virus type 2, 4, and 5 vectors: transduction of variant 1, 18 (2016). Department of Health and Human Services and is a member
cell types and regions in the mammalian central 197. Adam, M. P. et al. (eds) GeneReviews®. NCBI of the Scientific Advisory Board of NTSAD.
nervous system. Proc. Natl Acad. Sci. USA 97, https://www.ncbi.nlm.nih.gov/books/NBK1116/
3428–3432 (2000). (2018). Publisher’s note
179. Gilkes, J. A., Bloom, M. D. & Heldermon, C. D. 198. Sinigerska, I. et al. Founder mutation causing infantile Springer Nature remains neutral with regard to jurisdictional
Preferred transduction with AAV8 and AAV9 via GM1-gangliosidosis in the Gypsy population. Mol. claims in published maps and institutional affiliations.
thalamic administration in the MPS IIIB model: Genet. Metab. 88, 93–95 (2006).
a comparison of four rAAV serotypes. Mol. Genet. 199. Holve, S., Hu, D. & McCandless, S. E. Metachromatic
Reviewer information
Metab. Rep. 6, 48–54 (2016). leukodystrophy in the Navajo: fallout of the American-
Nature Reviews Disease Primers thanks M. Beck, J. Cooper, R.
180. Foust, K. D. et al. Intravascular AAV9 preferentially Indian wars of the nineteenth century. Am. J. Med.
Giugliani, C. Hollak, G. Pastores and other anonymous
targets neonatal neurons and adult astrocytes. Genet. 101, 203–208 (2001).
referee(s) for their contribution to the peer review of this work.
Nat. Biotechnol. 27, 59–65 (2009). 200. Ausems, M. G. et al. Frequency of glycogen storage
181. Bevan, A. K. et al. Systemic gene delivery in large disease type II in The Netherlands: implications for
species for targeting spinal cord, brain, and peripheral diagnosis and genetic counselling. Eur. J. Hum. Genet. Related links
tissues for pediatric disorders. Mol. Ther. 19, 7, 713–716 (1999). ClinicalTrials.gov: https://clinicaltrials.gov/
1971–1980 (2011). 201. Kalatzis V. et al. Characterization of a putative founder Genetics Home reference: https://ghr.nlm.nih.gov/
182. Deverman, B. E. et al. Cre-dependent selection yields mutation that accounts for the high incidence of Human Fertilization and Embryology Authority:
AAV variants for widespread gene transfer to the cystinosis in Brittany. J. Am. Soc. Nephrol. 12, https://www.hfea.gov.uk/pgd-conditions/
adult brain. Nat. Biotechnol. 34, 204–209 (2016). 2170–2174 (2001).
NATUre RevIewS | DISeASe PRImeRS | Article citation ID: (2018) 4:27 25