Jci 131 144918
Jci 131 144918
The etiopathogenesis of systemic lupus erythematosus (SLE), a clinically heterogeneous multisystemic syndrome that
derives its name from the initial characterization of facial lesions that resemble the bite of a wolf, is considered a complex,
multifactorial interplay between underlying genetic susceptibility factors and the environment. Prominent pathogenic factors
include the induction of aberrant cell death pathways coupled with defective cell death clearance mechanisms that promote
excessive externalization of modified cellular and nuclear debris with subsequent loss of tolerance to a wide variety of
autoantigens and innate and adaptive immune dysregulation. While abnormalities in adaptive immunity are well recognized
and are key to the pathogenesis of SLE, recent findings have emphasized fundamental roles of the innate immune system in
the initiation and propagation of autoimmunity and the development of organ damage in this disease. This Review focuses
on recent discoveries regarding the role of components of the innate immune system, specifically neutrophils and interferons,
in promoting various aspects of lupus pathogenesis, with potential implications for novel therapeutic strategies.
Introduction predisposed hosts, this imbalance in dead cell handling can con-
Systemic lupus erythematosus (SLE) is an autoimmune syndrome tribute to modification and externalization of nucleic acids and
of unclear etiology that predominantly affects women and dispro- other autoantigens, to promotion of innate and adaptive immune
portionately afflicts minorities (1). Lupus derives its name from dysregulation, to the development of autoantibodies that predom-
the Latin word for wolf, and early descriptions of this disease used inantly recognize nucleic acids and/or proteins binding to nucle-
the term to describe the facial lesions that look like a wolf ’s bite. ic acids, and to the formation of immune complexes (ICs) that
SLE has pleiotropic clinical manifestations and profound clinical deposit in various organs and promote damage (3). Autoantibody
heterogeneity, making its diagnosis and treatment very challeng- formation can precede clinical disease by many years, suggesting
ing. Indeed, what we call SLE may be driven by heterogeneous that immune dysregulation is an early event in disease pathogene-
pathways of immune dysregulation that eventually converge into sis (4). Notably, aspects of immune dysregulation characteristic of
a loosely shared clinical phenotype. SLE affects many organ sys- SLE have been described in unaffected relatives (5), further sup-
tems, including skin, kidneys, brain, and the vasculature. Char- porting a role for both genetic and environmental factors in sus-
acteristically, SLE presents with periods of clinical and serologic ceptibility to autoimmunity.
flare interspersed among phases of clinical remission, which can The innate and adaptive arms of the immune system are
occur spontaneously or modulated by exposure to environmen- altered and almost every immune cell type becomes dysregulated
tal stimuli, such as ultraviolet light or infections. Recent advanc- in SLE and its various murine models (3, 6). The role of the adap-
es in immunomodulatory/immunosuppressive treatments have tive immune system in SLE has been thoroughly studied and can-
improved morbidity and mortality in SLE. However, as patients not be overemphasized, and various current therapeutic strategies
live longer, prolonged inflammation and exposure to enhanced target these immune responses (7). In contrast, the role of various
oxidative stress promote the development of chronic complica- components of the innate immune system in SLE pathogenesis is
tions, including increased incidence of premature cardiovascu- less well characterized, but recent work emphasizes their crucial
lar disease (CVD) (2). Furthermore, the ability to predict which role in initiating and perpetuating autoimmunity in this disease.
patients will respond to specific medications or develop end-organ Abnormalities in phenotype and function of monocytes, mac-
complications remains an area of need to optimize SLE outcomes. rophages (8, 9), myeloid and plasmacytoid DCs (10, 11), baso-
SLE develops in a multistep process in which genetic, epigen- phils (12), neutrophils (13), NK cells, and γδ T cells (6) have been
etic, and environmental factors can promote aberrant cell death described in SLE. Furthermore, dysregulation of fundamental
and ineffective clearance of dead cell debris (3). In genetically innate immune strategies, such as the complement pathway, syn-
thesis of and response to IFNs, and mechanisms of neural regula-
tion of immune responses, contributes to autoimmunity and tis-
Conflict of interest: The NIAMS has collaborative research agreements with MedIm- sue damage in SLE (3, 14–16). In addition, genetic variations that
mune/AstraZeneca, Pfizer, and Bristol Myers Squibb. MJK is on the scientific advisory
alter various components of innate immune responses have been
boards of Neutrolis, Inc. and Cytrill.
Copyright: © 2021, American Society for Clinical Investigation.
reported to contribute to the risk and/or severity of SLE (Table 1).
Reference information: J Clin Invest. 2021;131(3):e144918. This Review focuses on the putative roles that the crosstalk
https://doi.org/10.1172/JCI144918. between aberrant cell death (particularly the formation of neutro-
1
REVIEW The Journal of Clinical Investigation
Table 1. Genes that modulate the innate immune system and are associated with SLE risk
phil extracellular traps), dysregulation of neutrophils, and the IFN as discussed below, play prominent roles in SLE pathogenesis (30,
pathway plays in the pathogenesis of SLE and its associated compli- 31). In particular, dysregulation in neutrophil cell death and the
cations, in the context of current and potential therapeutic targets. role of NETs in SLE, as discussed later, display high immunogenic
and proinflammatory potential.
SLE and the aberrant handling of cell death
Dysregulation in cell death pathways, defective clearance of dying Neutrophils, NETs, and SLE pathogenesis
cells and their products, and aberrant response to this cell debris by Neutrophil functions and plasticity. Over the past decade, neutro-
various immune cell subsets are considered fundamental aspects phils have reemerged as presumed culprits in various aspects of
of SLE pathogenesis (Figure 1). This process has been recently SLE pathogenesis (Figure 1 and ref. 13). As the first responders
reviewed in detail (17). Cell death is associated with modifica- against invading microorganisms (32), neutrophils develop and ter-
tion and redistribution to the cell surface and extracellular space minally differentiate in the bone marrow and are then released into
of many ubiquitously expressed autoantigens, which may affect the circulation, where they patrol and sense danger signals (32, 33).
immunologic self-tolerance. Dysregulation in many forms of cell Under conditions of homeostasis, neutrophils are also found in the
death, including apoptosis, necrosis, necroptosis, pyroptosis, and lung, spleen, and liver, and they follow tightly regulated circadian
neutrophil death through formation of neutrophil extracellular rhythms that allow for disarming strategies prior to their migration
traps (NETs), may have diverse effects on immune responses and to various tissues (33, 34). Whether disruptions in neutrophils’ nor-
tissue damage in the context of autoimmunity (17–21). It is import- mal circadian rhythm modulate autoimmune responses remains to
ant to consider that whether an elicited immune response to the be determined. In addition to their antimicrobial roles, neutrophils
dead cell becomes tolerogenic or immunogenic is determined not are responsible for fundamental homeostatic functions, including
only by whether cells die or not, but also by which cells die, and roles in coagulation, angiogenesis, resolution of inflammation, and
how and where they die (17). A combination of genetic (Table 1), tissue repair (33, 35–39). Some of these neutrophil properties are
epigenetic, and environmental factors may be involved in driving induced and modulated by specific tissues, with profound neutro-
aberrant cell death, impaired clearance, and/or altered immuno- phil intertissue heterogeneity in the steady state (33). Under condi-
logic response to the cell death products, including presentation tions of injury or infection, neutrophils display their antimicrobial
of modified autoantigens (Figure 1 and refs. 17, 19, 22–25). Vari- and proinflammatory strategies in tissues (33, 40), including the
ous mouse models that fail to clear apoptotic cells develop lupus- release of neutrophil-derived serine and matrix metalloproteinas-
like autoimmunity (26–28), and non-phagocytosed dead cells are es (MMPs) that cleave extracellular matrix components, thereby
detected in SLE germinal centers, indicative of impaired clearance disrupting existing tissue architecture (41). Neutrophils use a com-
(29). These autoantigens may be presented by germinal center plex armamentarium of strategies to immobilize and kill microbes,
follicular DCs to B and T lymphocytes in secondary lymphoid including phagocytosis and bacterial degradation through synthe-
tissues, resulting in loss of tolerance. The formation of ICs con- sis of ROS; release of antimicrobial peptides from granules; and the
taining autoantibodies that recognize nuclear and/or cytoplasmic synthesis of NETs, which are lattices of chromatin bound to gran-
material released from dead cells can lead to potent downstream ule peptides that have the ability to immobilize and potentially kill
inflammatory effects, including the synthesis of type I IFNs that, various microorganisms (42–44).
Figure 1. Role of neutrophils, NETs, and IFNs in SLE pathogenesis. Various stimuli can trigger neutrophils to undergo neutrophil extracellular trap (NET)
formation. NETs, in turn, can externalize self-antigens, including oxidized DNA and/or DNA–antimicrobial peptide complexes that can be presented to
antigen-presenting cells (APCs) and activate plasmacytoid DCs (pDCs) to synthesize type I IFNs. NETs have the ability to activate the NLRP3 inflam-
masome in macrophages, resulting in increased release of IL-1 and IL-18, which further prime neutrophils to undergo NET formation and perpetuate tissue
damage. Different exogenous and endogenous stimuli can promote type I IFN generation. The synthesis of type I IFNs further modulates other APCs,
tissue-resident cells, and T and B cell functions. NET products and IFNs modulate T cell responses and can also activate B cells to undergo class switching
and secrete autoantibodies against a wide range of self-antigens. DNA–antimicrobial peptide complexes (like LL37-DNA) released from NETs have the abil-
ity to directly activate B cells via TLR9 and promote autoantibody generation. NETs directly stimulate T cells by decreasing their activation threshold via
Zap70-mediated phosphorylation of the T cell receptor (TCR). Activated T cells release IL-17 and other proinflammatory cytokines that can result in endo-
thelial cell damage as well prime neutrophils to undergo further NET formation and migrate to inflamed tissues. NETs and IFNs can promote direct tissue
damage and vascular inflammation through their effect on endothelial cells and platelets. APRIL, a proliferation-inducing ligand; BAFF, B cell activating
factor; BLyS, B lymphocyte stimulator; LDG, low-density granulocyte.
NETs are typically generated and released through a distinct rial (61). This process is followed by degradation of nuclear and
programmed inflammatory cell death process (Figure 2 and refs. granule membranes and extrusion of NETs into the extracellular
13, 43), although mechanisms of NET formation that do not result space (Figure 2). Gasdermin D, a pore-forming protein considered
in cell death have been described (45, 46). NETs contain many a key executioner of pyroptosis, has been implicated in certain
potent antimicrobial molecules that can stimulate other neu- forms of NET formation (62, 63) but not in NETs formed in lupus
trophils and immune cells (43, 47, 48). Besides microbial stimu- animal models (64).
li, NETs are induced by sterile inflammatory stimuli, including The role of aberrant NET formation and clearance in various
platelets (49, 50), cytokines (51), uric acid and cholesterol crystals homeostatic and pathogenic conditions is an area of increased
(52, 53), various autoantibodies (54–56), and ICs (55, 57, 58). While interest. Compared with males, neutrophils from healthy young
the exact mechanisms that lead to NET formation continue to be adult females display enhanced ability to form NETs, both sponta-
further refined, this process can occur through several pathways neously and under microbial or sterile inflammatory stimuli (65).
that may or may not involve the NADPH oxidase (NOX) pathway This suggests that, in women, neutrophils have a higher propensity
of ROS formation and/or mitochondrial ROS synthesis (55, 59). to generate and externalize modified autoantigens. In individuals
Once initiated, posttranslational modifications of histones — with genetic predisposition to autoimmunity, this enhanced abil-
including citrullination by the peptidylarginine deiminase (PAD) ity to form NETs may contribute to breaking tolerance and may
family of enzymes, particularly PAD4 (60) — promote changes in explain, in part, why women are more prone to develop autoim-
electrostatic interactions of DNA and histones that disrupt chro- munity but also mount more robust responses to infectious agents
matin’s structure and promote decondensation of nuclear mate- than men. Female neutrophils are also more mature, activated,
Figure 2. Pathways of NET formation and targets for therapies. Stimulation of neutrophils in an individual genetically predisposed to SLE by various
stimuli (microbial, autoantibodies, cell products, etc.) mobilizes calcium from the endoplasmic reticulum that results in activation of protein kinase C
(PKC) and NADPH and/or mitochondrial ROS production. This leads to migration of granule protein to the nucleus and activation of peptidylarginine deimi-
nase-4 (PAD4), which induce citrullination, resulting in changes in electrostatic interactions of DNA and histones, which disrupt chromatin’s structure and
promote decondensation of nuclear material. This process is followed by degradation of nuclear and granule membranes with mixture of granular protein
with chromatin and eventual extrusion of NETs into the extracellular space. Gasdermin D (GSDMD) is implicated in some forms of NET formation, in which
ROS-mediated release of neutrophil elastase (NE) processes GSDMD, which further facilitates release and activation of NE. GSDMD also localizes to the
plasma membrane to form pores that promote cell lysis to release NETs. Possible therapies targeting critical steps in this pathway (represented by blunt
arrows) may be beneficial for SLE. FcγR, Fcγ receptor; IFNAR, type I IFN receptor; JAK, Janus kinases; MPO, myeloperoxidase; TLR, Toll-like receptor.
and readily primed to respond to danger signals than neutrophils ligand (APRIL) (79, 80), molecules that are highly relevant to B
from male counterparts, properties that appear to be regulated by cell development and function.
sex hormones (66). Several neural pathways activated by microbes or endogenous
Neutrophils regulate other innate and adaptive immune cells stimuli modulate neutrophil biology (16) through various neuro-
in fundamental ways (Figure 1 and refs. 67, 68). These cells can peptides that inhibit neutrophil function, including calcitonin
stimulate and suppress T cell responses in context-dependent gene–related peptide and somatostatin (81). Adenosine, a purine
manners (69). Various neutrophil granule proteases inhibit T nucleoside released by neutrophils at sites of inflammation, has
cell cytokine synthesis, including IL-2 and IL-6 (70). Neutro- complex effects on neutrophil biology, including NET formation,
phils can downregulate the ζ chain of the T cell receptor through which depend on the concentration of and signaling through its
synthesis of arginase and ROS, promoting T cell arrest (71), and four receptors (82–84).
they can express PD-L1 to induce IFN-dependent PD-1–medi- Attempts at characterizing neutrophil diversity in recent years
ated T cell apoptosis (72, 73). Conversely, neutrophils activate have highlighted the presence of neutrophil plasticity and neutro-
γδ T cells through cross-presentation of microbial antigens and phil subsets based on transcriptional, epigenetic, proteomic, and
cross-prime CD8+ T cells in an MHC class I–dependent manner functional analyses that suggest diverse physiological and patho-
(74, 75). Through T cell–independent mechanisms, splenic neutro- genic roles (Table 2 and refs. 39, 85–90). In healthy individuals,
phils function as B cell helpers (76) and are important sources of B several distinct circulating neutrophil subsets were recently identi-
cell activating factor (BAFF) (77, 78) and a proliferation-inducing fied based on single-cell transcriptomic analysis (66). It remains to
be better defined whether this heterogeneity is genetically driven healthy control neutrophils at the transcriptomic, epigenetic,
and/or secondary to differences in microbiome composition, circa- and functional levels (19, 85, 86, 90). LDGs are key drivers of the
dian rhythms, hormonal challenges, transmigration, senescence, characteristic type I IFN gene signature of SLE and contribute to
and/or specific tissue microenvironments. Identifying distinct synthesis of type I IFNs and other proinflammatory cytokines.
neutrophil subsets that play prominent roles in disease states is Lupus LDGs differ in protein content and have distinct modula-
key in selectively targeting pathogenic granulocytes without ham- tion of their cytoskeleton compared with normal-density control
pering important homeostatic functions of the neutrophil com- and lupus neutrophils. This translates into differences in modula-
partment as a whole. In this regard, the identification of a distinct tion of biomechanical properties that enhance their retention in
subset of proinflammatory neutrophils, called low-density granulo- microvasculature mimetics, suggesting that these cells may be
cytes (LDGs) because of their tendency to fractionate with PBMCs more prone to become trapped in organs like the lung and induce
during density separation of whole blood, has contributed to high- tissue damage (90). LDGs are not a homogenous population and
lighting the putative role of neutrophil heterogeneity and dysregu- display transcriptional, epigenetic, and functional heterogeneity,
lated neutrophil death in autoimmune diseases (85, 86, 91). comprising two main subpopulations of intermediate-mature and
NETs display many putative proinflammatory properties that immature neutrophils, with various degrees of chromatin accessi-
will be discussed in detail in the next section. In addition to their bility and distinct differences in transcription factor motif analy-
role in activation of the immune system, neutrophils and NETs sis (Table 2 and ref. 86). These two LDG subsets differ in various
contribute to resolution of inflammation by degrading chemokines functional readouts, including phagocytosis, responses to type
and cytokines and activating serine proteases that decrease neu- I IFN stimulation, NET formation, and chemotaxis. Overall, the
trophil recruitment (92). NETs can enhance Th2 cytokine expres- bulk of SLE LDGs do not represent immature neutrophils, and
sion, decrease Th1/Th17 cytokines (93), and induce both pro- and most of them have enhanced proinflammatory roles. Further-
antiinflammatory effects on DCs (Figure 1 and refs. 30, 94). Neu- more, they do not display suppressive capabilities characteristic of
trophils and NETs play a role in clearance of damaged endothelial myeloid-derived suppressor cells (97).
cells and remodeling of senescent vessels in some tissues (95). LDGs isolated from subjects with SLE (85) and other auto-
Role of neutrophils, LDGs, and NETs in SLE pathogenesis. A immune and autoinflammatory conditions (54, 56, 83, 98–100)
plethora of phenotypic and functional abnormalities have been exhibit enhanced propensity to form NETs and damage endothe-
described in lupus neutrophils (Figure 1), including increased lial cells, striated muscle cells, and other targets (19, 56). NETs
cell death, impaired phagocytosis, and dysregulated oxidative also promote endothelial dysfunction by inducing vascular leak-
activity (19, 47, 55, 85, 86). In SLE bone marrow, neutrophils con- age and endothelial-mesenchymal transition through their ability
tribute to type I IFN synthesis and B cell dysregulation through to degrade vascular endothelial cadherin and activate β-catenin
BAFF-mediated effects (96). signaling, with implications for lupus nephritis (101). LDG NETs
Lupus LDGs are considered a distinct neutrophil subset with are more immunostimulatory and proinflammatory than NETs
pathogenic roles. They differ from normal-density lupus and generated by other stimuli, with a higher abundance of modified
self-antigens, including oxidized nucleic acids, and immunogenic/ drial dysfunction, and NET formation in SLE. This is supported by
proinflammatory molecules such as LL37 and MMP-9 (19, 55, 102). observations that inhibitors of mitochondrial dysfunction or mito-
NETs activate the NLRP3 inflammasome in macrophages, promot- chondrial ROS generation ameliorate murine lupus (55, 112–114).
ing enhanced release of IL-1 and IL-18, a process that is heightened Similarly, the effectiveness of targeting PAD4 as a mechanism to
in lupus macrophages and can perpetuate tissue damage (103). decrease NET generation and lupus in animal models appears to
Dysregulated NET formation plays a number of putative cru- be strain and model dependent, suggesting that models associat-
cial pathogenic roles in SLE. In genetically predisposed individu- ed with more aberrant innate immune dysfunction improve with
als, key autoantigens that are externalized by NET formation may NET inhibition through PAD4 modulation (115).
be presented to the immune system. Several genetic polymor- NETs can directly modulate adaptive immune responses
phisms associated with SLE risk can enhance NET generation. For in SLE. LL37-DNA ICs activate endosomal TLRs to drive poly-
example, genetic alterations in TNFAIP3, encoding A20, disrupt clonal B cell activation and expand self-reactive memory B cells
the protein’s deubiquitinase domain, promoting upregulation (116). B cells can internalize and process NET components, and
of protein citrullination and NET formation through increased present them on class II MHC molecules to autoreactive T cells,
PADI4 (104). Increased circulating NET complexes have been promoting generation of memory T cells. NETs directly affect T
reported in SLE patients with a STAT4 risk allele (105, 106). cells by decreasing their activation threshold via Zap70-mediated
Healthy individuals who are homozygous for the major SLE risk phosphorylation of the T cell receptor, promoting enhanced T cell
haplotype of IRF5 display increased spontaneous NET formation, responses to suboptimal dosage of antigens (117). Importantly, the
supporting dysregulated neutrophil function as an early mecha- internalization of NETs through various ligand-receptor interac-
nism promoting autoimmunity (Table 1 and ref. 107). tions can also drastically alter the function of other cells, including
Oxidation of nucleic acids and certain posttranslational mod- DCs and fibroblasts (30, 58, 94).
ifications of proteins distinctly occur during NET formation and In addition to enhanced capacity of LDGs to form NETs, a
may promote enhanced immunogenicity. Both genomic and subset of SLE patients have impaired clearance of circulating
mitochondrial DNA become oxidized during NET formation. NET components, which leads to increased half-life of modified
Enhanced nucleic acid oxidation enhances the interferogenic self-antigens and their immunostimulatory effects and association
potential of NETs by extending the nucleic acids’ half-life through with higher incidence of lupus nephritis (24). Various culprits for
resistance to degradation by nucleases, thereby effectively engag- disruptions in NET clearance have been proposed, including com-
ing intracellular DNA sensors that promote type I IFN responses, plement activation within NETs, nucleic acid oxidation (108, 118,
including the cGAS/STING pathway (55, 108). Ribonuclear protein 119), DNase I inhibitors, and anti-NET antibodies (24). Genetic
ICs characteristic of SLE enhance mitochondrial ROS generation, polymorphisms that impact the function of molecules involved in
which promotes oxidation of nucleic acids and NET formation (55). removal of nucleic acids can impair NET clearance. For example,
LDG NETs also display higher levels of oxidized nucleic acids than loss-of-function variants in DNASE1L3, encoding an extracellular
NETs purified from other sources, leading to higher induction of nuclease endowed with the ability to degrade NETs, have been
type I IFN responses in target cells (55). As such, the generation of reported in familial forms of severe SLE (120).
modified nucleic acids by oxidation during pathogenic NET forma- Dysregulated NET formation/clearance appears to be oper-
tion in SLE may be a key mechanism enhancing autoantigen half- ational in vivo, as neutrophils and NETs are increased in affect-
life and induction of type I IFN and other inflammatory respons- ed lupus skin, kidneys, and placenta, in association with tissue
es, promoting loss of tolerance. LDG NETs also display enhanced inflammation and organ damage (19, 121, 122). At this point, it has
expression of ubiquitinated proteins that increase activation of not been possible to differentiate which neutrophil subsets infil-
proinflammatory responses in SLE macrophages (109). Overall, trate lupus tissues, as no distinct cell surface markers differenti-
these observations indicate that not all NETs are generated equal, ate LDGs from other lupus neutrophils. Given recent advances in
as lupus NETs are substantially more immunogenic and proinflam- differentiating these cells at the transcriptional, epigenetic, and
matory than NETs generated in other conditions. protein levels (86, 90), it may be possible in the future to expand
The exact stimuli and pathways that lead to generation of these tools to identify the role of LDGs in vivo at the tissue level.
NETs (59) may determine their role in lupus pathogenesis. For
example, lupus-prone mice that lack NOX2, an enzyme involved LDGs and NETs in vascular damage in SLE
in certain types of NET formation, develop exacerbated SLE Accumulating evidence implicates NETs in the development of
(110), and patients with chronic granulomatous disease (CGD), vascular damage and atherothrombosis (2, 19, 85, 102, 123, 124).
characterized by impaired NOX activity, have a higher prevalence NETs induce endothelial cell apoptosis through an MMP-9/
of autoimmunity. CGD patients also display an LDG subset with MMP-2 axis that is enhanced in lupus LDG NETs (102). NETs
enhanced ability to form NETs with enhanced mitochondrial ROS harm vascular smooth muscle cells through a histone-dependent
synthesis and enhanced generation of IFN responses (55). In addi- process that induces lytic cell death and promotes instability of
tion, genetic polymorphisms that impair NOX activity (NCF2) are atherosclerotic plaques (125). Various mouse models of athero-
associated with enhanced risk of developing SLE (111) and with sclerosis and vasculopathy suggest that inhibiting NET formation
accelerated lupus and enhanced NET activity in mouse models. can alter susceptibility to plaque formation/inflammation and
As such, it is possible that a dysfunctional NOX pathway promotes thrombosis (124, 126–128). Overall, dysregulated NET formation
synthesis of alternative sources of ROS (such as the mitochondria) in the context of SLE may play prominent roles in the accelerated
that lead to aberrant pathways of nucleic acid oxidation, mitochon- vascular damage characteristic of this disease.
SLE subjects demonstrate vascular inflammation, arterial dys- sclerosis characteristic of SLE. Elevated levels of IFN-regulated
function, and increased noncalcified plaque burden, which may proteins and ISGs associate with SLE vascular disease (119) inde-
explain the higher reported risk for acute coronary syndromes (2, pendent of traditional Framingham risk factors (153). Type I IFNs
123). Further supporting the role of LDGs in accelerated vascular impair endothelial function (154) and hamper vascular repair in
damage in SLE, circulating numbers of these cells, as well as an SLE (155). In lupus and atherosclerosis mouse models, type I IFNs
LDG transcriptional signature, associate with increased coronary impair vasculogenesis and promote endothelial dysfunction, ath-
plaque and aortic wall inflammation, independent of other vascular erothrombosis, and plaque progression (137). IFNs inhibit endo-
risk factors (86, 123). Furthermore, NETs can oxidize HDL and dis- thelial NO synthase and impair NO production by endothelial
rupt its atheroprotective role, in part by modifying the ability of this cells (156). Additionally, type I IFNs promote diet-induced insulin
lipoprotein to remove cholesterol from macrophages (cholesterol resistance by triggering liver accumulation of CD8+ T cells with
efflux capacity [CEC]) (129). HDL oxidation by NETs promotes subsequent glucose dysregulation and hepatic inflammation (157).
proinflammatory responses in macrophages because the modified Type I IFNs stimulate macrophage recruitment to arteries (158),
lipoprotein signals through the oxidized LDL receptor-1 (LOX-1), promote foam cell formation (159), and predict worse outcome
enhances NF-κΒ signaling, and disrupts the ability of HDL to block during myocardial infarction (160). ISGs are upregulated in bone
TLR-induced pathways (129). Overall, these observations suggest marrow neutrophil/monocyte progenitors and circulating and
pleiotropic vasculopathic roles of lupus LDGs and NETs. cardiac neutrophils soon after myocardial infarction (161). Block-
ing type I IFN signaling using an anti-IFNAR1 mAb (anifrolumab)
Pathogenic crosstalk between IFNs and decreases circulating NET complexes and improves CEC, imply-
neutrophils in SLE ing that type I IFN inhibition may improve cardiometabolic risk in
IFN pathway dysregulation plays critical roles in lupus pathogenesis SLE patients (138).
(130). A significant proportion of individuals diagnosed with SLE Other IFNs — type II (IFN-γ) and type III (IFN-λ1, IFN-λ2,
display an elevated IFN signature in peripheral blood and/or affect- IFN-λ3) — may contribute to SLE pathogenesis. Variations in cir-
ed tissues. Altered regulation of this pathway is likely an early event, culating levels of type I, II, and III IFNs and tissue responses may
since genetic polymorphisms that impact type I IFN signaling asso- help explain heterogeneity in pattern of organ involvement and
ciate with SLE risk (Table 1 and refs. 105, 131, 132). Genetically pre- IFN signature in SLE (15, 162, 163). Exogenous administration
disposed individuals may have a lower threshold to activate the type of IFN-γ can worsen human SLE (164), and elevations in IFN-γ
I IFN pathway when exposed to exogenous stimuli (viral, ultraviolet associate with disease activity. An IFN-γ signaling pathway is
light) or endogenous stimuli (dead cells, ICs, NETs) (55, 133, 134). activated in SLE PBMCs (165), and elevated IFN-γ in SLE T cells
In vitro and in vivo human and murine studies highlight the type I promotes monocyte/macrophage induction of BAFF (166). IFN-γ
IFN pathway’s importance in immune dysregulation and vascular may affect lupus B cell function and germinal center responses
and tissue damage (106, 135–139). Plasmacytoid DC activation is a (167, 168). Increases in type III IFNs have been reported in SLE,
main driver for enhanced production of IFNs, but other cell types, in correlation with disease activity (169, 170). In a lupus mouse
particularly of myeloid origin, also contribute (55, 85, 140, 141). model, type III IFNs were shown to have a pathogenic role that
IFNs enhance expression of costimulatory molecules and antigen is nonredundant relative to type I IFNs by promoting systemic
presentation by antigen-presenting cells (142) and alter B cell toler- immune dysregulation and local inflammatory effects in skin and
ance, plasma cell differentiation, immunoglobulin class switching, kidneys (171). B cells are the main immune responders to type III
antibody production, and generation of long-lasting memory B cells IFNs in humans, with potentially important implications in SLE
(143–147). Type I IFNs prolong T cell survival, inhibit T cell apopto- that require further investigation (172).
sis, enhance activity of cytotoxic T cells, suppress Treg activity, and Overall, IFNs play key roles in SLE pathogenesis, and the rele-
modify T cell polarization (142, 148, 149). vance of the various types of IFNs in explaining the heterogeneity
In the context of neutrophil dysregulation, type I IFNs can of SLE continues to be elucidated and may be of great importance
prime neutrophils to form NETs (47, 86, 150) and may enhance in the design of therapies that can target their deleterious effects
their response to TLR agonists (151). Conversely, as NETs induce (Table 3). A pathogenic crosstalk between IFNs and neutrophils
IFN production, this may lead to a vicious cycle of persistent may play fundamental roles in initiating, amplifying, and main-
inflammation (Figure 1). Compared with other immune cell types, taining various aspects of lupus immune dysregulation, as well as
LDGs display increased expression of ISGs (86) and increased in long-term outcomes driven by the development of vasculopathy
type I IFN–induced proteins (90). Healthy young adult females and end-stage organ damage (Figure 1).
display a neutrophil-specific increased ISG expression compared
with males, with associated hyperresponsiveness to type I IFNs Targeting neutrophils, NETs, and IFNs in SLE
(66). This enhanced priming of female neutrophils may lead to Given the growing evidence for a fundamental role of the patho-
increased responses to various danger signals. The neutrophils genic interplay between type I IFNs, neutrophils, and NETs in
that express enhanced ISGs represent a distinct circulating neu- SLE, there is interest in targeting them for therapeutic purposes.
trophil subset detected in healthy individuals, suggesting a puta- Decreasing the formation, release, and/or half-life of NETs can
tive critical role in antimicrobial and proinflammatory responses be attempted by a variety of strategies (Table 3). Targeting of oxi-
(Table 2 and refs. 66, 152). dative pathways implicated in NET formation using NOX (54,
Synergizing with the effects of neutrophils, type I IFNs may 172) or mitochondrial ROS (55) inhibitors, or ROS scavengers like
play a key role in triggering vasculopathy and premature athero- N-acetylcysteine (NAC) (173), has been explored in vitro and in
Table 3. Potential therapeutics that target the IFN/neutrophil pathogenic crosstalk in SLE
vivo. NAC decreased disease activity in SLE patients (174, 175), ease markers, perhaps owing more to the half-life of the molecule
while inhibition of mitochondrial ROS or targeting of aberrant used than to a lack of biological effects (183). DNaseIL3 is involved
mitochondrial function hampered murine lupus severity (55, 112, in degradation of extracellular DNA, like in NETs, and genetic
113). In contrast, NOX inhibition may exacerbate SLE as a result of deficiency of this enzyme in mice and humans is associated with
complex immunoregulatory effects (110). Inhibition of the func- autoantibody production and lupus features (184). Future studies
tion of various granule proteins can decrease neutrophil recruit- could assess the role of this enzyme in improving NET clearance
ment, NET formation, and inflammatory cytokine synthesis (176) in SLE. Other drugs that can affect NET structures by destabiliz-
and may be explored in future human studies. In preclinical stud- ing the neutrophil’s actin cytoskeleton have also been proposed as
ies, genetic deletion of PAD2 or PAD4 reduced ISGs, autoantibod- potential therapies (185).
ies, vascular dysfunction, and clinical manifestations in TLR7- Targeting IL-17 signaling can modulate neutrophil trafficking
dependent murine lupus, but not in other lupus models (115, 177). and NET formation and may mitigate lupus glomerulonephritis
Inhibiting PAD enzymes using small molecules also decreases (186, 187). Inhibition of neutrophil recruitment and NET release
NET formation and protects from organ damage and vascular dis- in tissues may also be accomplished by interruption of interactions
ease in various mouse models of lupus and vascular damage (124, between endothelial ligands and neutrophil-expressed kindlin-3
126, 127). Given the heterogeneity of response in murine systems, and β2 integrin and other similar modalities (188). Targeting var-
future studies in humans will lead to further clarification of the ious chemokines may also limit pathogenic neutrophil infiltration
role of PAD inhibition in SLE. in tissues (189). While inhibition of NET formation induced by
The putative deleterious effects on antimicrobial responses C5a priming is an important pathway to consider, early clinical
following NET inhibition in humans remain to be determined. trials using anti-C5 antibodies, although well tolerated, did not
PAD4-knockout mice maintain other antibacterial neutrophil promote improvements in disease activity even if they were effec-
functions even if NETs are inhibited. Even in mouse strains in tive in murine lupus (190, 191). In contrast, decreasing autoanti-
which PAD inhibition does not totally abolish NET formation, bodies and ICs through a combination of mAbs targeting CD20
the immunogenic and vasculopathic effects of these NETs are (rituximab) and BLyS/BAFF (belimumab) showed some benefits
diminished, suggesting that partial enzymatic blockade may be in lupus biomarkers and reduced circulating NETs (192), although
sufficient (177, 178). Patients with genetic disorders that lead to the benefit of combined therapy versus single therapy remains to
deficiency of molecules implicated in NET formation (PADs, be better defined.
cathepsin C) are not overtly immunosuppressed (179, 180). How- Targeting the IFN pathway holds promise for SLE man-
ever, additional preclinical and clinical studies are warranted to agement (Table 3). Blocking mAbs that target IFN-α, IFN-γ, or
assess the safety profile, in different patient populations, of strat- IFNAR1 have been studied in SLE (193). Anifrolumab, which
egies that specifically inhibit NETs, without considerably altering blocks IFNAR1, thus inhibiting the action of all type I IFNs, has
other essential neutrophil functions. shown promising results in one of two phase III trials, with a good
Degradation of already formed NETs to enhance their remov- tolerability profile and improvement in disease activity (139, 194).
al is another potential approach (181). Administration of DNase I Anifrolumab decreased NET formation in SLE subjects when com-
to lupus-prone mice was beneficial (182), but early phase Ib stud- pared with placebo and improved the function of lupus HDL and
ies of DNase I, though well tolerated, showed no changes in dis- other cardiometabolic parameters (138). These findings suggest
that suppressing the IFN/neutrophil axis may help prevent long- various environmental stimuli on IFN responses, neutrophil het-
term vascular complications in SLE and other diseases in which erogeneity, and the composition of NETs may help to explain the
these pathways are dysregulated. Several medications currently variability of SLE manifestations and the diverse autoantibody
used to treat SLE can modulate — at different levels — NET forma- responses. Future studies should focus on better defining the
tion and/or the type I IFN signature. Antimalarials— drugs with mechanisms that regulate the cross-play between IFN and myeloid
proven benefit in SLE, including CVD benefits — can decrease cells, and downstream consequences in autoimmunity, in order to
NET formation in vitro (129) and are weak type I IFN inhibitors, identify promising therapies. This has implications for other chron-
potentially through modulation of endosomal TLRs (195) and the ic inflammatory diseases in which type I IFNs, neutrophils, and
cGAS/STING pathway (196). NETs are proposed to play important pathogenic roles.
JAKs play fundamental roles in downstream signaling of mul- Several important questions remain, and the exact role of
tiple proinflammatory cytokines, including all IFNs (197). Addi- these immune players in disease pathogenesis may only be under-
tionally, JAKs directly modulate neutrophil biology. Tofacitinib, stood if clinical trials that specifically target neutrophil dysregula-
a pan-JAK inhibitor, reduced NET formation in vitro and in vivo tion, aberrant NET formation, and the neutrophil/IFN crosstalk
and decreased disease manifestations, including vasculopathy, in SLE come to fruition. One important aspect is whether it will
in lupus-prone mice (135). The JAK inhibitor baricitinib improved be feasible to specifically target LDGs, while leaving other neu-
lupus clinical features in a phase II study (198), with phase III trophil subsets untouched. This will depend on further advances
studies ongoing (NCT03616964, NCT03616912, ClinicalTrials. in the identification of unique markers of this neutrophil subset
gov). A phase Ia/IIb clinical trial using tofacitinib in mild to mod- and a better understanding of the genetic, epigenetic, and envi-
erate SLE showed significant decreases in type I IFN signature ronmental factors that modulate granulocyte plasticity in health
and circulating LDGs and NETs, with associated improvement in and disease states. While significant advances have occurred in
vascular function and cardiometabolic parameters (106). These characterizing LDGs, the factors that promote their formation,
observations support that targeting the neutrophil/IFN crosstalk release, and pathogenicity (genetic or acquired) remain unclear.
with JAK/STAT inhibitors may modulate lupus vasculopathy and The extent to which NETs contribute to the various aspects of
should be further explored in CVD prevention in SLE (106). lupus pathogenesis and why only certain autoantibodies pro-
mote NET formation remain to be clarified. Understanding why
Concluding remarks and remaining questions different autoimmune diseases, characterized by enhanced NET
SLE is complex and heterogeneous, with clear dysfunction in most formation, are associated with distinct autoantibody responses
components of the immune system and therapeutic challenges giv- that target specific NET components will help better characterize
en pathogenic and clinical heterogeneity. Dysregulation in crucial heterogeneity of the NET protein and nucleic acid cargo and the
innate immune pathways implicated in host defense has profound role that genetics play in the adaptive immune response to these
implications in various aspects of lupus pathogenesis, including structures. This may lead to the identification of better molecular
break of tolerance, induction of IFNs and other proinflammatory candidates as therapies to hopefully improve clinical outcomes.
cytokines, aberrant adaptive immunity, and tissue damage. Accu-
mulating in vitro and in vivo evidence, using human and murine Acknowledgments
systems, supports a central role for neutrophil dysregulation in SLE This research was supported by the Intramural Research Program
pathogenesis, spanning loss of tolerance, induction and amplifica- of the National Institute of Arthritis and Musculoskeletal and Skin
tion of inflammatory pathways, tissue damage, vascular disease, Diseases of the NIH.
and cardiometabolic dysfunction. Future studies will hopefully
help to better define subsets of patients in whom neutrophil/IFN Address correspondence to: Mariana J. Kaplan, Systemic Autoim-
interactions play particularly fundamental roles, in order to design munity Branch, NIAMS/NIH, 10 Center Drive, 12N248C, Bethes-
better personalized therapeutic approaches. Further understand- da, Maryland 20892, USA. Phone: 301.496.0517; Email: mariana.
ing the effects of the interaction between genetic, epigenetic, and kaplan@nih.gov.
1. Tsokos GC. Systemic lupus erythematosus. bution to systemic lupus erythematosus. Front Immunity. 2016;44(3):683–697.
N Engl J Med. 2011;365(22):2110–2121. Immunol. 2019;10:772. 11. Ding D, et al. Aberrant phenotype and function of
2. Liu Y, Kaplan MJ. Cardiovascular disease in sys- 7. Durcan L, et al. Management strategies and future myeloid dendritic cells in systemic lupus erythe-
temic lupus erythematosus: an update. Curr Opin directions for systemic lupus erythematosus in matosus. J Immunol. 2006;177(9):5878–5889.
Rheumatol. 2018;30(5):441–448. adults. Lancet. 2019;393(10188):2332–2343. 12. Charles N, et al. Basophils and the T helper 2
3. Tsokos GC, et al. New insights into the immuno- 8. Katsiari CG, et al. The pathophysiologic role of environment can promote the development of
pathogenesis of systemic lupus erythematosus. monocytes and macrophages in systemic lupus lupus nephritis. Nat Med. 2010;16(6):701–707.
Nat Rev Rheumatol. 2016;12(12):716–730. erythematosus: a reappraisal. Semin Arthritis 13. Gupta S, Kaplan MJ. The role of neutrophils and
4. Arbuckle MR, et al. Development of auto- Rheum. 2010;39(6):491–503. NETosis in autoimmune and renal diseases. Nat
antibodies before the clinical onset of sys- 9. Iwata Y, et al. Aberrant macrophages mediate Rev Nephrol. 2016;12(7):402–413.
temic lupus erythematosus. N Engl J Med. defective kidney repair that triggers nephri- 14. Botto M, Walport MJ. C1q, autoimmunity and apop-
2003;349(16):1526–1533. tis in lupus-susceptible mice. J Immunol. tosis. Immunobiology. 2002;205(4–5):395–406.
5. Niewold TB, et al. High serum IFN-alpha activity 2012;188(9):4568–4580. 15. Oke V, et al. High levels of circulating interferons
is a heritable risk factor for systemic lupus ery- 10. Menon M, et al. A regulatory feedback between type I, type II and type III associate with distinct
thematosus. Genes Immun. 2007;8(6):492–502. plasmacytoid dendritic cells and regulatory B clinical features of active systemic lupus erythe-
6. Herrada AA, et al. Innate immune cells’ contri- cells is aberrant in systemic lupus erythematosus. matosus. Arthritis Res Ther. 2019;21(1):107.
16. Nicholls AJ, et al. Activation of the sympathetic 35. Noubouossie DF, et al. Neutrophils: 2015;67(11):2990–3003.
nervous system modulates neutrophil function. back in the thrombosis spotlight. Blood. 55. Lood C, et al. Neutrophil extracellular traps
J Leukoc Biol. 2018;103(2):295–309. 2019;133(20):2186–2197. enriched in oxidized mitochondrial DNA are
17. Elkon KB. Review: Cell death, nucleic acids, 36. Fuchs TA, et al. Extracellular DNA traps pro- interferogenic and contribute to lupus-like dis-
and immunity: inflammation beyond the grave. mote thrombosis. Proc Natl Acad Sci U S A. ease. Nat Med. 2016;22(2):146–153.
Arthritis Rheumatol. 2018;70(6):805–816. 2010;107(36):15880–15885. 56. Seto N, et al. Neutrophil dysregulation is patho-
18. Mistry P, Kaplan MJ. Cell death in the pathogen- 37. Massena S, et al. Identification and characteriza- genic in idiopathic inflammatory myopathies.
esis of systemic lupus erythematosus and lupus tion of VEGF-A-responsive neutrophils express- JCI Insight. 2020;5(3):e134189.
nephritis. Clin Immunol. 2017;185:59–73. ing CD49d, VEGFR1, and CXCR4 in mice and 57. Behnen M, et al. Immobilized immune complex-
19. Villanueva E, et al. Netting neutrophils induce humans. Blood. 2015;126(17):2016–2026. es induce neutrophil extracellular trap release by
endothelial damage, infiltrate tissues, and 38. Wang J, et al. Visualizing the function and fate of human neutrophil granulocytes via FcγRIIIB and
expose immunostimulatory molecules in neutrophils in sterile injury and repair. Science. Mac-1. J Immunol. 2014;193(4):1954–1965.
systemic lupus erythematosus. J Immunol. 2017;358(6359):111–116. 58. Khandpur R, et al. NETs are a source of citrulli-
2011;187(1):538–552. 39. Christoffersson G, Phillipson M. The neu- nated autoantigens and stimulate inflammatory
20. Kahlenberg JM, et al. An essential role of caspase trophil: one cell on many missions or many responses in rheumatoid arthritis. Sci Transl Med.
1 in the induction of murine lupus and its asso- cells with different agendas? Cell Tissue Res. 2013;5(178):178ra40.
ciated vascular damage. Arthritis Rheumatol. 2018;371(3):415–423. 59. Kenny EF, et al. Diverse stimuli engage different
2014;66(1):152–162. 40. Herter J, Zarbock A. Integrin regulation neutrophil extracellular trap pathways. Elife.
21. Kahlenberg JM, et al. Inflammasome activation during leukocyte recruitment. J Immunol. 2017;6:e24437.
of IL-18 results in endothelial progenitor cell dys- 2013;190(9):4451–4457. 60. Wang Y, et al. Histone hypercitrullination
function in systemic lupus erythematosus. 41. Henson PM. Pathologic mechanisms in mediates chromatin decondensation and neu-
J Immunol. 2011;187(11):6143–6156. neutrophil-mediated injury. Am J Pathol. trophil extracellular trap formation. J Cell Biol.
22. Denny MF, et al. Accelerated macrophage apop- 1972;68(3):593–612. 2009;184(2):205–213.
tosis induces autoantibody formation and organ 42. Ley K, et al. Neutrophils: new insights and open 61. Tessarz P, Kouzarides T. Histone core mod-
damage in systemic lupus erythematosus. questions. Sci Immunol. 2018;3(30):eaat4579. ifications regulating nucleosome struc-
J Immunol. 2006;176(4):2095–2104. 43. Brinkmann V, et al. Neutrophil extracellular traps ture and dynamics. Nat Rev Mol Cell Biol.
23. Munoz LE, et al. The role of defective clearance kill bacteria. Science. 2004;303(5663):1532–1535. 2014;15(11):703–708.
of apoptotic cells in systemic autoimmunity. Nat 44. Rorvig S, et al. Proteome profiling of human neu- 62. Sollberger G, et al. Gasdermin D plays a vital
Rev Rheumatol. 2010;6(5):280–289. trophil granule subsets, secretory vesicles, and role in the generation of neutrophil extracellular
24. Hakkim A, et al. Impairment of neutrophil cell membrane: correlation with transcriptome traps. Sci Immunol. 2018;3(26):eaar6689.
extracellular trap degradation is associated profiling of neutrophil precursors. J Leukoc Biol. 63. Chen KW, et al. Noncanonical inflammasome
with lupus nephritis. Proc Natl Acad Sci U S A. 2013;94(4):711–721. signaling elicits gasdermin D-dependent
2010;107(21):9813–9818. 45. Yipp BG, et al. Infection-induced NETosis is a neutrophil extracellular traps. Sci Immunol.
25. Dieker J, et al. Circulating apoptotic micropar- dynamic process involving neutrophil multitask- 2018;3(26):eaar6676.
ticles in systemic lupus erythematosus patients ing in vivo. Nat Med. 2012;18(9):1386–1393. 64. Wang X, et al. Effects of gasdermin D in modulat-
drive the activation of dendritic cell subsets and 46. Yousefi S, et al. Viable neutrophils release mito- ing murine lupus and its associated organ dam-
prime neutrophils for NETosis. Arthritis Rheuma- chondrial DNA to form neutrophil extracellular age. Arthritis Rheumatol. 2020;72(12):2118–2129.
tol. 2016;68(2):462–472. traps. Cell Death Differ. 2009;16(11):1438–1444. 65. Blazkova J, et al. Multicenter systems analysis
26. Watanabe-Fukunaga R, et al. Lymphoprolifer- 47. Garcia-Romo GS, et al. Netting neutrophils are of human blood reveals immature neutrophils
ation disorder in mice explained by defects in major inducers of type I IFN production in pedi- in males and during pregnancy. J Immunol.
Fas antigen that mediates apoptosis. Nature. atric systemic lupus erythematosus. Sci Transl 2017;198(6):2479–2488.
1992;356(6367):314–317. Med. 2011;3(73):73ra20. 66. Gupta S, et al. Sex differences in neutrophil biol-
27. Wilber A, et al. Dnase1l3 deficiency in lupus- 48. Semeraro F, et al. Extracellular histones promote ogy modulate response to type I interferons and
prone MRL and NZB/W F1 mice. Clin Exp Immu- thrombin generation through platelet-dependent immunometabolism. Proc Natl Acad Sci U S A.
nol. 2003;134(1):46–52. mechanisms: involvement of platelet TLR2 and 2020;117(28):16481–16491.
28. Strasser A, et al. Enforced BCL2 expression in TLR4. Blood. 2011;118(7):1952–1961. 67. Megiovanni AM, et al. Polymorphonuclear neu-
B-lymphoid cells prolongs antibody responses 49. Clark SR, et al. Platelet TLR4 activates neutrophil trophils deliver activation signals and antigenic
and elicits autoimmune disease. Proc Natl Acad extracellular traps to ensnare bacteria in septic molecules to dendritic cells: a new link between
Sci U S A. 1991;88(19):8661–8665. blood. Nat Med. 2007;13(4):463–469. leukocytes upstream of T lymphocytes. J Leukoc
29. Baumann I, et al. Impaired uptake of apoptotic 50. Carestia A, et al. Mediators and molecular path- Biol. 2006;79(5):977–988.
cells into tingible body macrophages in germinal ways involved in the regulation of neutrophil 68. Yang CW, et al. Neutrophils influence the level
centers of patients with systemic lupus erythe- extracellular trap formation mediated by activat- of antigen presentation during the immune
matosus. Arthritis Rheum. 2002;46(1):191–201. ed platelets. J Leukoc Biol. 2016;99(1):153–162. response to protein antigens in adjuvants.
30. Lande R, et al. Neutrophils activate plasmacytoid 51. Keshari RS, et al. Cytokines induced neutrophil J Immunol. 2010;185(5):2927–2934.
dendritic cells by releasing self-DNA-peptide extracellular traps formation: implication for 69. Schonrich G, et al. Devilishly radical NETwork
complexes in systemic lupus erythematosus. Sci the inflammatory disease condition. PLoS One. in COVID-19: oxidative stress, neutrophil extra-
Transl Med. 2011;3(73):73ra19. 2012;7(10):e48111. cellular traps (NETs), and T cell suppression. Adv
31. Leonard D, et al. Activated T cells enhance inter- 52. Schorn C, et al. Monosodium urate crystals Biol Regul. 2020;77:100741.
feron-α production by plasmacytoid dendritic cells induce extracellular DNA traps in neutrophils, 70. Fazio J, et al. Inhibition of human γδ T cell prolifer-
stimulated with RNA-containing immune com- eosinophils, and basophils but not in mononucle- ation and effector functions by neutrophil serine
plexes. Ann Rheum Dis. 2016;75(9):1728–1734. ar cells. Front Immunol. 2012;3:277. proteases. Scand J Immunol. 2014;80(6):381–389.
32. Nauseef WM, Borregaard N. Neutrophils at work. 53. Munoz LE, et al. Neutrophil extracellular 71. Rodriguez PC, et al. Arginase I production in the
Nat Immunol. 2014;15(7):602–611. traps initiate gallstone formation. Immunity. tumor microenvironment by mature myeloid
33. Ballesteros I, et al. Co-option of neutro- 2019;51(3):443–450.e4. cells inhibits T-cell receptor expression and
phil fates by tissue environments. Cell. 54. Yalavarthi S, et al. Antiphospholipid antibodies antigen-specific T-cell responses. Cancer Res.
2020;183(5):1282–1297.e18. promote the release of neutrophil extracellular 2004;64(16):5839–5849.
34. Aroca-Crevillen A, et al. Circadian features of traps: a new mechanism of thrombosis in the 72. Bowers NL, et al. Immune suppression by neu-
neutrophil biology. Front Immunol. 2020;11:576. antiphospholipid syndrome. Arthritis Rheumatol. trophils in HIV-1 infection: role of PD-L1/PD-1
pathway. PLoS Pathog. 2014;10(3):e1003993. 91. Hacbarth E, Kajdacsy-Balla A. Low density immune sensing. Immunity. 2013;39(3):482–495.
73. Curran CS, et al. PD-1 immunobiology in systemic neutrophils in patients with systemic lupus 109. Barrera-Vargas A, et al. Differential ubiquitina-
lupus erythematosus. J Autoimmun. 2019;97:1–9. erythematosus, rheumatoid arthritis, and tion in NETs regulates macrophage responses in
74. Tvinnereim AR, et al. Neutrophil involvement in acute rheumatic fever. Arthritis Rheum. systemic lupus erythematosus. Ann Rheum Dis.
cross-priming CD8+ T cell responses to bacterial 1986;29(11):1334–1342. 2018;77(6):944–950.
antigens. J Immunol. 2004;173(3):1994–2002. 92. Schauer C, et al. Aggregated neutrophil extracellu- 110. Campbell AM, et al. NADPH oxidase inhibits the
75. Lim K, et al. In situ neutrophil efferocytosis lar traps limit inflammation by degrading cytokines pathogenesis of systemic lupus erythematosus.
shapes T cell immunity to influenza infection. and chemokines. Nat Med. 2014;20(5):511–517. Sci Transl Med. 2012;4(157):157ra41.
Nat Immunol. 2020;21(9):1046–1057. 93. Barrientos L, et al. Neutrophil extracellular traps 111. Jacob CO, et al. Lupus-associated causal muta-
76. Puga I, et al. B cell-helper neutrophils stimulate downregulate lipopolysaccharide-induced acti- tion in neutrophil cytosolic factor 2 (NCF2)
the diversification and production of immuno- vation of monocyte-derived dendritic cells. brings unique insights to the structure and func-
globulin in the marginal zone of the spleen. Nat J Immunol. 2014;193(11):5689–5698. tion of NADPH oxidase. Proc Natl Acad Sci U S A.
Immunol. 2012;13(2):170–180. 94. Sangaletti S, et al. Neutrophil extracellular traps 2012;109(2):E59–E67.
77. Parsa R, et al. BAFF-secreting neutrophils drive mediate transfer of cytoplasmic neutrophil anti- 112. Blanco LP, et al. Improved mitochondrial metab-
plasma cell responses during emergency granu- gens to myeloid dendritic cells toward ANCA olism and reduced inflammation following
lopoiesis. J Exp Med. 2016;213(8):1537–1553. induction and associated autoimmunity. Blood. attenuation of murine lupus with coenzyme
78. Giordano D, et al. BAFF produced by neutrophils 2012;120(15):3007–3018. Q10 analog idebenone. Arthritis Rheumatol.
and dendritic cells is regulated differently and 95. Binet F, et al. Neutrophil extracellular traps target 2020;72(3):454–464.
has distinct roles in antibody responses and pro- senescent vasculature for tissue remodeling in 113. Fortner KA, et al. Targeting mitochondrial oxi-
tective immunity against west nile virus. J Immu- retinopathy. Science. 2020;369(6506):eaay5356. dative stress with MitoQ reduces NET formation
nol. 2020;204(6):1508–1520. 96. Palanichamy A, et al. Neutrophil-mediated IFN and kidney disease in lupus-prone MRL-lpr mice.
79. Huard B, et al. APRIL secreted by neutrophils activation in the bone marrow alters B cell devel- Lupus Sci Med. 2020;7(1):e000387.
binds to heparan sulfate proteoglycans to create opment in human and murine systemic lupus 114. Kim J, et al. VDAC oligomers form mitochon-
plasma cell niches in human mucosa. J Clin erythematosus. J Immunol. 2014;192(3):906–918. drial pores to release mtDNA fragments
Invest. 2008;118(8):2887–2895. 97. Rahman S, et al. Low-density granulocytes acti- and promote lupus-like disease. Science.
80. Manfroi B, et al. CXCL-8/IL8 produced by vate T cells and demonstrate a non-suppressive 2019;366(6472):1531–1536.
diffuse large B-cell lymphomas recruits neutro- role in systemic lupus erythematosus. Ann Rheum 115. Gordon RA, et al. Lupus and proliferative nephri-
phils expressing a proliferation-inducing ligand Dis. 2019;78(7):957–966. tis are PAD4 independent in murine models. JCI
APRIL. Cancer Res. 2017;77(5):1097–1107. 98. Kessenbrock K, et al. Netting neutrophils in Insight. 2017;2(10):e92926.
81. Chiu IM, et al. Bacteria activate sensory neurons autoimmune small-vessel vasculitis. Nat Med. 116. Gestermann N, et al. Netting neutrophils acti-
that modulate pain and inflammation. Nature. 2009;15(6):623–625. vate autoreactive B cells in lupus. J Immunol.
2013;501(7465):52–57. 99. Lin AM, et al. Mast cells and neutrophils release 2018;200(10):3364–3371.
82. Barletta KE, et al. Regulation of neutrophil func- IL-17 through extracellular trap formation in pso- 117. Tillack K, et al. T lymphocyte priming by
tion by adenosine. Arterioscler Thromb Vasc Biol. riasis. J Immunol. 2011;187(1):490–500. neutrophil extracellular traps links innate
2012;32(4):856–864. 100.Mistry P, et al. Dysregulated neutrophil respons- and adaptive immune responses. J Immunol.
83. Carmona-Rivera C, et al. Deficiency of adenosine es and neutrophil extracellular trap formation 2012;188(7):3150–3159.
deaminase 2 triggers adenosine-mediated NETo- and degradation in PAPA syndrome. Ann Rheum 118. Leffler J, et al. Neutrophil extracellular traps
sis and TNF production in patients with DADA2. Dis. 2018;77(12):1825–1833. that are not degraded in systemic lupus erythe-
Blood. 2019;134(4):395–406. 101. Pieterse E, et al. Neutrophil extracellular traps matosus activate complement exacerbating the
84. Ali RA, et al. Adenosine receptor agonism drive endothelial-to-mesenchymal transition. Arte- disease. J Immunol. 2012;188(7):3522–3531.
protects against NETosis and thrombosis in rioscler Thromb Vasc Biol. 2017;37(7):1371–1379. 119. Lood C, et al. Platelet transcriptional profile and
antiphospholipid syndrome. Nat Commun. 102. Carmona-Rivera C, et al. Neutrophil extracel- protein expression in patients with systemic
2019;10(1):1916. lular traps induce endothelial dysfunction in lupus erythematosus: up-regulation of the type
85. Denny MF, et al. A distinct subset of proinflam- systemic lupus erythematosus through the acti- I interferon system is strongly associated with
matory neutrophils isolated from patients with vation of matrix metalloproteinase-2. Ann Rheum vascular disease. Blood. 2010;116(11):1951–1957.
systemic lupus erythematosus induces vascular Dis. 2015;74(7):1417–1424. 120. Al-Mayouf SM, et al. Loss-of-function vari-
damage and synthesizes type I IFNs. J Immunol. 103. Kahlenberg JM, et al. Neutrophil extracellular ant in DNASE1L3 causes a familial form of
2010;184(6):3284–3297. trap-associated protein activation of the NLRP3 systemic lupus erythematosus. Nat Genet.
86. Mistry P, et al. Transcriptomic, epigenetic, inflammasome is enhanced in lupus macro- 2011;43(12):1186–1188.
and functional analyses implicate neutrophil phages. J Immunol. 2013;190(3):1217–1226. 121. Marder W, et al. Placental histology and
diversity in the pathogenesis of systemic 104.Odqvist L, et al. Genetic variations in A20 neutrophil extracellular traps in lupus and
lupus erythematosus. Proc Natl Acad Sci U S A. DUB domain provide a genetic link to citrul- pre-eclampsia pregnancies. Lupus Sci Med.
2019;116(50):25222–25228. lination and neutrophil extracellular traps in 2016;3(1):e000134.
87. Fridlender ZG, et al. Polarization of tumor-associ- systemic lupus erythematosus. Ann Rheum Dis. 122. Frangou E, et al. REDD1/autophagy pathway
ated neutrophil phenotype by TGF-beta: “N1” ver- 2019;78(10):1363–1370. promotes thromboinflammation and fibrosis
sus “N2” TAN. Cancer Cell. 2009;16(3):183–194. 105. Remmers EF, et al. STAT4 and the risk of rheu- in human systemic lupus erythematosus (SLE)
88. Ma Y, et al. Temporal neutrophil polarization matoid arthritis and systemic lupus erythemato- through NETs decorated with tissue factor (TF)
following myocardial infarction. Cardiovasc Res. sus. N Engl J Med. 2007;357(10):977–986. and interleukin-17A (IL-17A). Ann Rheum Dis.
2016;110(1):51–61. 106. Hasni S, et al. A phase 1b/2a trial of tofacitinib, 2019;78(2):238–248.
89. Sagiv JY, et al. Phenotypic diversity and plasticity an oral janus kinase inhibitor, in systemic 123. Carlucci PM, et al. Neutrophil subsets and their
in circulating neutrophil subpopulations in can- lupus erythematosus. Lupus Sci Med. gene signature associate with vascular inflam-
cer. Cell Rep. 2015;10(4):562–573. 2019;6(suppl 1):A139–A140. mation and coronary atherosclerosis in lupus. JCI
90. Bashant KR, et al. Proteomic, biomechanical and 107. Li D, et al. IRF5 genetic risk variants drive Insight. 2018;3(8):e99276.
functional analyses define neutrophil hetero myeloid-specific IRF5 hyperactivation and pre- 124. Knight JS, et al. Peptidylarginine deiminase
geneity in systemic lupus erythematosus symptomatic SLE. JCI Insight. 2020;5(2):124020. inhibition disrupts NET formation and protects
[published online September 28, 2020]. Ann 108. Gehrke N, et al. Oxidative damage of DNA against kidney, skin and vascular disease in
Rheum Dis. https://doi.org/10.1136/ confers resistance to cytosolic nuclease TREX1 lupus-prone MRL/lpr mice. Ann Rheum Dis.
annrheumdis-2020-218338. degradation and potentiates STING-dependent 2015;74(12):2199–2206.
125. Silvestre-Roig C, et al. Externalized histone H4 functions of type I interferons. Nat Rev Immunol. marrow and is regulated by Nrf2-activated mac-
orchestrates chronic inflammation by inducing 2012;12(2):125–135. rophages. Sci Immunol. 2020;5(51):eaaz1974.
lytic cell death. Nature. 2019;569(7755):236–240. 143. Care MA, et al. Network analysis identifies proin- 162. Balomenos D, et al. Interferon-gamma is required
126. Knight JS, et al. Peptidylarginine deiminase flammatory plasma cell polarization for secretion for lupus-like disease and lymphoaccumulation in
inhibition is immunomodulatory and vas- of ISG15 in human autoimmunity. J Immunol. MRL-lpr mice. J Clin Invest. 1998;101(2):364–371.
culoprotective in murine lupus. J Clin Invest. 2016;197(4):1447–1459. 163. Seery JP, et al. Antinuclear autoantibodies and
2013;123(7):2981–2993. 144. Vazquez MI, et al. B cells responses and cytokine lupus nephritis in transgenic mice expressing
127. Knight JS, et al. Peptidylarginine deiminase inhi- production are regulated by their immune micro- interferon gamma in the epidermis. J Exp Med.
bition reduces vascular damage and modulates environment. Cytokine. 2015;74(2):318–326. 1997;186(9):1451–1459.
innate immune responses in murine models of 145. Kiefer K, et al. Role of type I interferons in the 164. Machold KP, Smolen JS. Interferon-gamma
atherosclerosis. Circ Res. 2014;114(6):947–956. activation of autoreactive B cells. Immunol Cell induced exacerbation of systemic lupus erythe-
128. Liu Y, et al. Myeloid-specific deletion of peptidy- Biol. 2012;90(5):498–504. matosus. J Rheumatol. 1990;17(6):831–832.
larginine deiminase 4 mitigates atherosclerosis. 146.Giltiay NV, et al. The plasticity of newly formed B 165. Karonitsch T, et al. Activation of the interfer-
Front Immunol. 2018;9:1680. cells. J Immunol. 2019;203(12):3095–3104. on-gamma signaling pathway in systemic lupus
129. Smith CK, et al. Neutrophil extracellular trap- 147. Domeier PP, et al. B-cell-intrinsic type 1 interfer- erythematosus peripheral blood mononuclear
derived enzymes oxidize high-density lipopro- on signaling is crucial for loss of tolerance and cells. Arthritis Rheum. 2009;60(5):1463–1471.
tein: an additional proatherogenic mechanism in the development of autoreactive B cells. Cell Rep. 166. Harigai M, et al. Excessive production of
systemic lupus erythematosus. Arthritis Rheuma- 2018;24(2):406–418. IFN-gamma in patients with systemic lupus
tol. 2014;66(9):2532–2544. 148. Lopez P, et al. A pathogenic IFNα, BLyS and IL-17 erythematosus and its contribution to induction
130. Crow MK. Type I interferon in the pathogenesis axis in systemic lupus erythematosus patients. Sci of B lymphocyte stimulator/B cell-activating
of lupus. J Immunol. 2014;192(12):5459–5468. Rep. 2016;6:20651. factor/TNF ligand superfamily-13B. J Immunol.
131. Langefeld CD, et al. Transancestral mapping and 149. Alunno A, et al. Balance between regulatory 2008;181(3):2211–2219.
genetic load in systemic lupus erythematosus. T and Th17 cells in systemic lupus erythema- 167. Chodisetti SB, et al. Type II but not type I IFN
Nat Commun. 2017;8:16021. tosus: the old and the new. Clin Dev Immunol. signaling is indispensable for TLR7-promoted
132. International Consortium for Systemic Lupus 2012;2012:823085. development of autoreactive B cells and systemic
Erythematosus Genetics, et al. Genome-wide 150. Gul E, et al. Type I IFN-related NETosis in ataxia autoimmunity. J Immunol. 2020;204(4):796–809.
association scan in women with systemic lupus telangiectasia and Artemis deficiency. J Allergy 168. Munroe ME, et al. Altered type II interferon
erythematosus identifies susceptibility variants Clin Immunol. 2018;142(1):246–257. precedes autoantibody accrual and elevated
in ITGAM, PXK, KIAA1542 and other loci. Nat 151. Zimmermann M, et al. IFNα enhances the pro- type I interferon activity prior to systemic lupus
Genet. 2008;40(2):204–210. duction of IL-6 by human neutrophils activated erythematosus classification. Ann Rheum Dis.
133. Mavragani CP, et al. Expression of long inter- via TLR8. Sci Rep. 2016;6:19674. 2016;75(11):2014–2021.
spersed nuclear element 1 retroelements and 152. Xie X, et al. Single-cell transcriptome profiling 169. Wu Q, et al. Interferon-lambda1 induces periph-
induction of type I interferon in patients with sys- reveals neutrophil heterogeneity in homeostasis eral blood mononuclear cell-derived chemokines
temic autoimmune disease. Arthritis Rheumatol. and infection. Nat Immunol. 2020;21(9):1119–1133. secretion in patients with systemic lupus ery-
2016;68(11):2686–2696. 153. Somers EC, et al. Type I interferons are associat- thematosus: its correlation with disease activity.
134. Robinson T, et al. Autoimmune disease risk vari- ed with subclinical markers of cardiovascular dis- Arthritis Res Ther. 2011;13(3):R88.
ant of IFIH1 is associated with increased sensitiv- ease in a cohort of systemic lupus erythematosus 170. Zickert A, et al. Interferon (IFN)-λ is a potential
ity to IFN-α and serologic autoimmunity in lupus patients. PLoS One. 2012;7(5):e37000. mediator in lupus nephritis. Lupus Sci Med.
patients. J Immunol. 2011;187(3):1298–1303. 154. Tyden H, et al. Endothelial dysfunction is associ- 2016;3(1):e000170.
135. Furumoto Y, et al. Tofacitinib ameliorates murine ated with activation of the type I interferon sys- 171. Goel RR, et al. Interferon lambda promotes
lupus and its associated vascular dysfunction. tem and platelets in patients with systemic lupus immune dysregulation and tissue inflammation
Arthritis Rheumatol. 2017;69(1):148–160. erythematosus. RMD Open. 2017;3(2):e000508. in TLR7-induced lupus. Proc Natl Acad Sci U S A.
136. Moore E, Putterman C. Are lupus animal mod- 155. Denny MF, et al. Interferon-alpha promotes 2020;117(10):5409–5419.
els useful for understanding and developing abnormal vasculogenesis in lupus: a potential 172. Remijsen Q, et al. Neutrophil extracellular trap
new therapies for human SLE? J Autoimmun. pathway for premature atherosclerosis. Blood. cell death requires both autophagy and superox-
2020;112:102490. 2007;110(8):2907–2915. ide generation. Cell Res. 2011;21(2):290–304.
137. Thacker SG, et al. Type I interferons modulate 156. Buie JJ, et al. IFN-α negatively regulates the 173. Patel S, et al. Nitric oxide donors release extra-
vascular function, repair, thrombosis, and plaque expression of endothelial nitric oxide synthase cellular traps from human neutrophils by aug-
progression in murine models of lupus and athero- and nitric oxide production: implications for menting free radical generation. Nitric Oxide.
sclerosis. Arthritis Rheum. 2012;64(9):2975–2985. systemic lupus erythematosus. J Immunol. 2010;22(3):226–234.
138. Casey KA, et al. Modulation of cardiometabolic 2017;199(6):1979–1988. 174. Garcia RJ, et al. Attention deficit and hyperac-
disease markers by type I interferon inhibition 157. Ghazarian M, et al. Type I interferon responses tivity disorder scores are elevated and respond
in systemic lupus erythematosus [published drive intrahepatic T cells to promote metabolic to N-acetylcysteine treatment in patients with
online September 10, 2020]. Arthritis Rheumatol. syndrome. Sci Immunol. 2017;2(10):eaai7616. systemic lupus erythematosus. Arthritis Rheum.
https://doi.org/10.1002/art.41518. 158. Goossens P, et al. Myeloid type I interferon sig- 2013;65(5):1313–1318.
139. Morand EF, et al. Trial of anifrolumab in active naling promotes atherosclerosis by stimulating 175. Lai ZW, et al. N-acetylcysteine reduces disease
systemic lupus erythematosus. N Engl J Med. macrophage recruitment to lesions. Cell Metab. activity by blocking mammalian target of rapa-
2020;382(3):211–221. 2010;12(2):142–153. mycin in T cells from systemic lupus erythe-
140. Murayama G, et al. Inhibition of mTOR 159. Li J, et al. Interferon-α priming promotes lipid matosus patients: a randomized, double-blind,
suppresses IFNalpha productionthe STING uptake and macrophage-derived foam cell placebo-controlled trial. Arthritis Rheum.
pathway in monocytes from systemic lupus formation: a novel link between interferon-α 2012;64(9):2937–2946.
erythematosus patients. Rheumatology (Oxford). and atherosclerosis in lupus. Arthritis Rheum. 176. Zheng W, et al. PF-1355, a mechanism-based
2020;59(10):2992–3002. 2011;63(2):492–502. myeloperoxidase inhibitor, prevents immune
141. Buechler MB, et al. Cutting edge: Type I IFN 160. King KR, et al. IRF3 and type I interferons fuel a complex vasculitis and anti-glomerular base-
drives emergency myelopoiesis and peripheral fatal response to myocardial infarction. Nat Med. ment membrane glomerulonephritis. J Pharma-
myeloid expansion during chronic TLR7 signal- 2017;23(12):1481–1487. col Exp Ther. 2015;353(2):288–298.
ing. J Immunol. 2013;190(3):886–891. 161. Calcagno DM, et al. The myeloid type I interferon 177. Liu Y, et al. Peptidylarginine deiminases 2
142. Gonzalez-Navajas JM, et al. Immunomodulatory response to myocardial infarction begins in bone and 4 modulate innate and adaptive immune
responses in TLR-7-dependent lupus. JCI Insight. matin release from neutrophils. J Innate Immun. their pathways in systemic lupus erythematosus.
2018;3(23):e124729. 2009;1(3):194–201. Autoimmun Rev. 2018;17(1):44–52.
178. Martinod K, et al. PAD4-deficiency does not 186.Pisitkun P, et al. Interleukin-17 cytokines are 194.Furie RA, et al. Type I interferon inhibitor
affect bacteremia in polymicrobial sepsis critical in development of fatal lupus glomerulo- anifrolumab in active systemic lupus erythemato-
and ameliorates endotoxemic shock. Blood. nephritis. Immunity. 2012;37(6):1104–1115. sus (TULIP-1): a randomised, controlled, phase 3
2015;125(12):1948–1956. 187. Satoh Y, et al. A case of refractory lupus trial. Lancet Rheumatology. 2019;1(4):e208–e219.
179. Sulem P, et al. Identification of a large set of nephritis complicated by psoriasis vulgaris 195. Sacre K, et al. Hydroxychloroquine is associated
rare complete human knockouts. Nat Genet. that was controlled with secukinumab. Lupus. with impaired interferon-alpha and tumor necro-
2015;47(5):448–452. 2018;27(7):1202–1206. sis factor-alpha production by plasmacytoid
180. Sorensen OE, et al. Papillon-Lefèvre syndrome 188. Xu Z, et al. Interaction of kindlin-3 and β2-integrins dendritic cells in systemic lupus erythematosus.
patient reveals species-dependent require- differentially regulates neutrophil recruitment and Arthritis Res Ther. 2012;14(3):R155.
ments for neutrophil defenses. J Clin Invest. NET release in mice. Blood. 2015;126(3):373–377. 196. An J, et al. Cutting edge: antimalarial drugs inhib-
2014;124(10):4539–4548. 189. Disteldorf EM, et al. CXCL5 drives neutrophil it IFN-β production through blockade of cyclic
181. Jimenez-Alcazar M, et al. Host DNases prevent recruitment in TH17-mediated GN. J Am Soc GMP-AMP synthase-DNA interaction. J Immu-
vascular occlusion by neutrophil extracellular Nephrol. 2015;26(1):55–66. nol. 2015;194(9):4089–4093.
traps. Science. 2017;358(6367):1202–1206. 190. Wang Y, et al. Amelioration of lupus-like autoim- 197. Gadina M, et al. Translational and clinical advanc-
182. Macanovic M, et al. The treatment of systemic mune disease in NZB/WF1 mice after treatment es in JAK-STAT biology: the present and future of
lupus erythematosus (SLE) in NZB/W F1 hybrid with a blocking monoclonal antibody specific for jakinibs. J Leukoc Biol. 2018;104(3):499–514.
mice; studies with recombinant murine DNase complement component C5. Proc Natl Acad Sci 198.Wallace DJ, et al. Baricitinib for systemic lupus
and with dexamethasone. Clin Exp Immunol. U S A. 1996;93(16):8563–8568. erythematosus: a double-blind, randomised,
1996;106(2):243–252. 191. Rother RP, et al. Inhibition of terminal comple- placebo-controlled, phase 2 trial. Lancet.
183. Davis JC Jr, et al. Recombinant human Dnase I ment: a novel therapeutic approach for the treat- 2018;392(10143):222–231.
(rhDNase) in patients with lupus nephritis. Lupus. ment of systemic lupus erythematosus. Lupus. 199. Moulton VR, et al. Pathogenesis of human sys-
1999;8(1):68–76. 2004;13(5):328–334. temic lupus erythematosus: a cellular perspec-
184.Sisirak V, et al. Digestion of chromatin in apop- 192.Kraaij T, et al. The NET-effect of combining rit- tive. Trends Mol Med. 2017;23(7):615–635.
totic cell microparticles prevents autoimmunity. uximab with belimumab in severe systemic lupus 200.Zavada J, et al. Cyclosporine A or intravenous cyclo-
Cell. 2016;166(1):88–101. erythematosus. J Autoimmun. 2018;91:45–54. phosphamide for lupus nephritis: the Cyclofa-Lune
185. Neeli I, et al. Regulation of extracellular chro- 193. Chasset F, Arnaud L. Targeting interferons and study. Lupus. 2010;19(11):1281–1289.