Abstract
Systemic autoimmune diseases are a group of disorders characterized by a failure in self-tolerance to a wide variety of autoantigens. In genetically predisposed individuals, these diseases occur as a multistep process in which environmental factors have key roles in the development of abnormal innate and adaptive immune responses. Experimental evidence collected in the past decade suggests that neutrophils — the most abundant type of white blood cell — might have an important role in the pathogenesis of these diseases by contributing to the initiation and perpetuation of immune dysregulation through the formation of neutrophil extracellular traps (NETs), synthesis of proinflammatory cytokines and direct tissue damage. Many of the molecules externalized through NET formation are considered to be key autoantigens and might be involved in the generation of autoimmune responses in predisposed individuals. In several systemic autoimmune diseases, the imbalance between NET formation and degradation might increase the half-life of these lattices, which could enhance the exposure of the immune system to modified autoantigens and increase the capacity for NET-induced organ damage. This Review details the role of neutrophils and NETs in the pathophysiology of systemic autoimmune diseases, including their effect on renal damage, and discusses neutrophil targets as potential novel therapies for these diseases.
Neutrophils — the most abundant type of white blood cell in humans — have crucial roles in the innate immune response and act as a first line of defence against invading microorganisms1. Neutrophils target microorganisms through a number of processes including degranulation (the release of granular antimicrobial peptides such as myeloperoxidase (MPO), neutrophil elastase and matrix metalloproteinases (MMPs)), phagocytosis and degradation via synthesis of reactive oxygen species (ROS) inside phagolysosomes, and microbial trapping by extrusion of a meshwork of chromatin bound to granular peptides termed neutrophil extracellular traps (NETs)2.
NETs are structures composed of externalized, concentrated, antimicrobial molecules that can trap, immobilize, inactivate and kill microorganisms, and activate other immune cells3. They are generated and released during a distinct process of cell death called NETosis (FIG. 1), which differs from apoptosis and necrosis. NETosis is an important step of the innate immune response, and can be triggered by both infectious and ‘sterile’ stimuli (for example cytokines4, monosodium uric acid crystals5, cholesterol crystals, autoantibodies6 and immune complexes7). In addition to triggering innate immune responses, neutrophils regulate the adaptive immune response8. Indeed, neutrophils engage with B cells9,10, T cells11,12 and antigen presenting cells in lymphoid organs13,14. NETosis also contributes to the activation of the immune system during inflammation15,16. As discussed in this Review, these effects are mediated, at least in part, through protease-mediated effector functions and by modulating intercellular signal transduction.
Figure 1. NETosis pathways and potential therapeutic targets.
Stimulation of neutrophil receptors by microorganisms or sterile stimuli leads to release of calcium (Ca+) from the endoplasmic reticulum (ER), which results in activation of protein kinase C (PKC) and assembly of the nicotinamide adenine dinucleotide phosphate (NADPH) oxidase complex and/or mitochondrial activation, leading to generation of reactive oxygen species (ROS). Other cellular sources might also be important contributors of ROS. This ROS generation is followed by myeloperoxidase (MPO)-dependent migration of granular neutrophil elastase (NE) to the nucleus where it cleaves histones. In addition, peptidylarginine deiminase (PAD) 4 is activated and induces histone citrullination, which contributes to chromatin decondensation. Finally, the nuclear membrane is degraded and a mixture of chromatin and granular proteins is extruded from the cell. Extracellular DNase eventually degrades neutrophil extracellular traps (NETs). Modulation of critical steps in NET formation and degradation (shown by blocking arrows) might be beneficial for the treatment of autoimmune disorders. C5aR, C5a receptor; G-CSF: granulocyte colony stimulating factor; G-CSF-R, G-CSF receptor; FcγR, Fc γ receptor; TLR, Toll-like receptor.
Systemic autoimmune diseases are characterized by a breakdown in self-tolerance to a wide range of auto-antigens. Certain autoimmune conditions such as systemic lupus erythematosus (SLE)17, antineutrophil cytoplasmic antibody (ANCA)-associated vasculitis (AAV)18, rheumatoid arthritis19 and antiphospholipid antibody syndrome (APS)20 can affect the kidney.
Many of the molecules externalized through NETosis (for example MPO, double-stranded (ds)DNA, histones) are recognized autoantigens in systemic autoimmunity, suggesting that aberrant NET formation might be important for the initiation of autoimmune responses in susceptible individuals. In support of this hypothesis, neutrophils from patients with various autoimmune diseases are more prone than those from healthy controls or patients without autoimmune diseases to undergo NETosis and, in turn, various autoantibodies can promote the release of NETs. This relationship between NETosis and autoantibodies could therefore promote a vicious cycle whereby autoantigen release by NETs leads to autoantibody formation, which promotes further release of antigenic material.
In this Review, we address the contribution of neutrophils and NETosis to host defence and their important role in innate and adaptive immunity. We also discuss the evidence supporting the potential involvement of neutrophils and NETs in the development of various systemic autoimmune disorders that affect the kidneys and consider how various neutrophil functions and NET formation and/or clearance could be modulated to treat autoimmune disorders.
Neutrophils and NETosis: basic principles
Host-defence
Neutrophils are typically short-lived with a circulating half-life of approximately 1–8 h21; however, some reports suggest that their lifespan can extend to over 5 days22, especially once activated23,24. They are characterized by a multilobulated nucleus, as well as distinct types of cytoplasmic granules packed with microbicidal molecules and oxidative enzymes, which are an important armamentarium for host defence. Neutrophils develop in the bone marrow and are released in the circulation as terminally differentiated cells21. Once they are recruited to sites of injury or infection, a multistep inflammatory response is initiated in which β2-integrins and kindlin-3 on circulating neutrophils promote their adhesion to the activated vascular endothelium25, followed by their extravasation and migration towards inflammatory foci, ultimately leading to the destruction of foreign microorganisms26.
Neutrophil granules have been classified into three types: azurophilic (primary) granules, specific (secondary) granules and gelatinase (tertiary) granules27,28. Azurophilic granules can be distinguished from the other types by their uptake of basic dye azure A, owing to their acid mucopolysaccharide content, and contain MPO, azurocidin, bacterial permeability-increasing protein, cathepsin G, defensins, elastase and human neutrophil peptides. Secondary granules contain cathelicidin antibacterial peptide (also known as antibacterial peptide LL-37), lactoferrin and neutrophil gelatinase-associated lipocalin. Tertiary granules contain peptidoglycan recognition proteins. Lysozyme is present in all three types of granule. Together these antimicrobial granular proteins have an important role in the activation of innate and adaptive immunity29,30.
When they come in contact with a pathogen, neutrophils undergo a respiratory burst, a crucial phenomenon in host defence, during which activation of the nicotinamide adenine dinucleotide phosphate (NADPH)-oxidase complex leads to superoxide synthesis30. Superoxide and its downstream products, hydrogen peroxide and hypochlorous acid, have critical antimicrobial roles30. Other important sources of ROS in the neutrophil include mitochondria31 and peroxisomes32. Neutrophils can also deploy complex enzymatic and non-enzymatic antioxidant defence systems, which involve enzymes such as catalase, superoxide dismutase and glutathione peroxidase. A balance between oxidant and antioxidant activities in the neutrophil is therefore required to maintain homeostasis of the immune system.
Neutrophils also have an important role as regulators of the adaptive immune response. Uptake of apoptotic neutrophils by dendritic cells can enhance levels of dendritic antigen presentation13,14. Neutrophils can both stimulate and suppress T-cell responses. Indeed, neutrophils cross-prime CD8+ T-cells in a major histocompatibility complex class I dependent manner and activate γδ T cells through cross-presentation of bacterial antigens11. Conversely, proteases like elastase and cathepsin G, contained in neutrophil granules, inhibit the production of T-cell stimulating cytokines, including IL-2 and IL-6 (REF. 12). Neutrophils can also downregulate the ζ chain of the T-cell receptor by producing ROS and secreting arginase, which leads to T-cell arrest33. Lastly, neutrophils express PD-L1, which promotes interferon-dependent PD-1-mediated T-cell apoptosis34. With regard to B cells, neutrophils synthesize cytokines crucial for B-cell development, such as B-cell activating factor (BAFF)9 and A proliferation-inducing ligand (APRIL)10. In the spleen, neutrophils can act as B-cell helpers in a T-cell independent manner35.
Complexes of extracellular DNA and cathelicidin or HMGB1 can form stable structures that activate phagosomal Toll-like receptor (TLR) 9 in monocytes and dendritic cells15,36. Interestingly, histones in NETs also activate TLR2 and TLR416, suggesting that NETs stimulate the immune system through various ligands and receptors. NETs can also prime T cells; however, this interaction is not clearly defined37. Lastly, NETs participate in the defence against pathogens by promoting timely and enhanced thrombus formation, which prevents microorganism dissemination38–41.
Neutrophils in resolution of inflammation
In addition to immune system activation, NETs might help to control inflammation by modulating the function of monocyte-derived dendritic cells by promoting the expression of type 2 T helper (TH2) cytokines and down-regulating the synthesis of TH1 and TH17 cytokines42. The presence of NETs during dendritic cell maturation diminishes their capacity to induce T-lymphocyte proliferation and polarization. NETs also promote the resolution of neutrophilic inflammation by degrading cytokines and chemokines and by disrupting neutrophil recruitment and activation via serine proteases in acute neutrophilic reactions such as gout43.
NET formation and clearance
NETs target many pathogens, including Staphylococcus aureus44,45, Streptococcus pyogenes46, Shigella flexneri3, Mycobacterium tuberculosis47, Aspergillus fumigatus48, Toxoplasma gondii49 and HIV50. Although the molecular pathways leading to NET formation remain to be fully characterized, the process of NETosis is known to involve calcium release from the endoplasmic reticulum of neutrophils, activation of PKC, assembly of the NADPH-oxidase complex, and generation of ROS30,51. These events are followed by MPO-dependent migration of elastase from granules to the nucleus52,53, where it cleaves histones to elicit chromatin decondensation54. Concomitantly, protein-arginine deiminase type-4 (PAD4; also known as peptidylarginine deiminase IV) substitutes the positively charged histone arginine residues with neutrally charged citrulline residues in the nucleus, which decreases the electrostatic interaction between histones and DNA, and thus promotes nuclear decondensation54. Eventually, the nuclear and granular membranes are degraded, leading to the mixing of chromatin and granular proteins in the cytoplasm, followed by extrusion of this meshwork from the cell and neutrophil lysis52.
Reports also exist of a non-lytic process of NET formation, termed ‘vital’ NETosis. This process is characterized by the formation of an anuclear — but still viable — cell or by selective extrusion of mitochondrial DNA; these processes and their implications in health and disease remain to be fully characterized55,56.
NETs are eventually degraded by DNase 1, an endonuclease that cleaves chromatin within NETs57. NET degradation can also occur by engulfment of NETs by macrophages through an active endocytic process that leads to the lysosome-mediated degradation of these lattices58.
Systemic autoimmune diseases
Systemic autoimmune diseases develop as multistep processes through a complex interplay between genetic and environmental factors, including infections, ultraviolet light (in the case of SLE), smoking (in rheumatoid arthritis), and certain medications (in the case of SLE and AAV)59. These factors can induce cellular damage through various mechanisms and increase the exposure of the immune cells to autoantigens60. Subclinical immune dysfunction leads to a preclinical phase of autoantibody synthesis60. Eventually, clinically apparent disease develops, along with detectable evidence of immune dysregulation60.
In diseases characterized by autoantibody development, responses to antigens are fairly specific. Autoantibodies to dsDNA and to RNA-binding proteins are characteristic of SLE17, whereas autoantibodies that target MPO and proteinase-3 (PR3) are predominantly observed in patients with AAV18. Individuals with rheumatoid arthritis develop autoantibodies (anti-citrullinated protein antibodies; ACPAs) that target citrullinated intracellular and extracellular antigens, such as vimentin, enolase, and fibrinogen19. APS is characterized by antibodies to β2 glycoprotein-I (β2GP1)20.
Dysregulation of apoptosis and/or clearance of apoptotic material has been proposed to be an important source of modified and/or externalized autoantigens in autoimmune diseases61. This view is perhaps oversimplified, given that various other cell-death mechanisms (including necroptosis, pyroptosis, NETosis, autophagic- related cell death and caspase-independent cell death)62 could act as sources of modified autoantigens. In fact, nuclear material released from NETs could be more immunogenic than apoptotic material. The proximity of the genomic DNA to ROS during NETosis promotes nucleic acid oxidation. Oxidised DNA externalized in NETs, which is more resistant than non-oxidised nucleic acids to degradation by nucleases such as TREX-1, activates the cGAS–STING intracellular sensing pathway, leading to enhanced type I interferon synthesis and immune dysregulation57,63.
Patients with systemic autoimmune diseases have a markedly enhanced risk of developing cardiovascular atherosclerotic diseases64 and thromboembolic events65,66, which could be mediated by a proatherogenic67,68 and prothrombotic69 effect of neutrophils and NETs. Many autoimmune disorders also involve the kidneys and neutrophils might have a prominent role in inflammation, tissue vasculopathy and fibrosis in the renal manifestations of these diseases70.
NETosis and autoimmune disorders
Systemic lupus erythematosus
SLE is a systemic autoimmune disease that predominantly affects women of childbearing age and involves multiple organs17. Although the exact aetiology is unclear, the development of SLE is multifactorial and can be attributed to disruptions in innate and adaptive immunity and aberrant responses to nuclear self-antigens. Patients with SLE have an increased production of and/or response to type I interferons, a process considered critical in SLE pathogenesis71. Furthermore, SLE is characterized by accelerated atherosclerosis and an increased risk of premature cardiovascular diseases64.
Patients with SLE display a distinct granulocyte population, called low-density granulocytes (LDGs), which are found in the peripheral blood mononuclear cell fraction following density separation of whole blood72–74. LDGs are a subset of pathogenic granulocytes that synthesize more proinflammatory cytokines, including type I interferons, than their regular-density counterparts and are toxic to endothelial cells73,75. Importantly, LDGs have an enhanced capacity to form NETs at baseline in vitro that remains unchanged even in the presence of PMA, a strong NET inducer, unlike their normal density or healthy counterparts, suggesting that LDGs might already be maximally stimulated in vivo75 (FIG. 2). These NETs contain higher levels of autoantigens and immunostimulatory molecules, such as LL-37, MMP9 and dsDNA15,36,76,77, and are more immunostimulatory than are NETs generated by healthy control neutrophils77.
Figure 2. NETs in low-density granulocytes.
Low-density granulocytes from a patient with systemic lupus erythematosus show spontaneous formation of neutrophil extracellular traps (NETs). Immunofluorescence imaging reveals NETs as strands of DNA (blue (labelled with Hoechst stain)) with neutrophil granular proteins (red (myeloperoxidase)). Image obtained by Monica Purmalek, Systemic Autoimmunity Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, NIH, USA.
In addition to the presence of LDGs with enhanced capacity to undergo NETosis, NETs are not properly cleared from the circulation in a substantial proportion of patients with SLE. In some patients, this defect in NET clearance might result from complement activation within NETs or from modifications (oxidation) of nucleic acids externalized by the NETs63,76,78. In other patients the defect might be due to the presence of inhibitors of DNase 1, or of anti-NET antibodies, which hamper the access of the DNase 1 to the NETs57. This inability to degrade NETs has been associated with a higher incidence of lupus nephritis in patients with SLE57.
NETs can further trigger an autoimmune response in SLE by exposing cathelicidin–DNA complexes, which, through activation of endosomal TLRs, promote the synthesis of type I interferons by plasmacytoid dendritic cells15,36 (FIG. 3). NETs generated by LDGs are enriched in oxidized mitochondrial DNA. Oxidized nucleic acids in NETs can also activate type I interferons in myeloid cells through activation of the STING pathway78. Furthermore, cathelicidin, and perhaps other NET proteins, can activate the NLRP3 inflammasome in lipopolysaccharide-primed macrophages by interacting with their P2X7 receptor and inducing potassium efflux, which leads to release of IL-1 and IL-18. In turn, these cytokines further promote NETosis, amplifying inflammatory pathways in tissues79 (FIG. 3). Interestingly, cathelicidin-mediated activation of the inflammasome is increased in macrophages derived from patients with SLE, compared to healthy macrophages, which could promote further inflammation and organ damage. Skin and kidneys from patients with SLE can be infiltrated by neutrophils that undergo NETosis and externalize nucleic acids in these tissues, which, in turn, is associated with increased levels of serum anti-dsDNA antibodies, supporting a direct role of NETosis in SLE pathogenesis and associated organ dysfunction75.
Figure 3. Neutrophils and NETosis in the pathogenesis of autoimmune and renal diseases.
Upon exposure to various infectious and ‘sterile’ stimuli, neutrophils (and low density granulocytes (LDGs) in the setting of systemic lupus erythematosus (SLE)) release neutrophil extracellular traps (NETs). These NETs externalize granular peptides (such as LL-37) in complex with DNA, which, in turn, activate plasmacytoid dendritic cells (pDCs) to synthetize type I interferons (IFN), which have critical roles in SLE pathogenesis. Neutrophils also release inflammatory cytokines that activate T cells and B cells, which produce autoantibodies, potentially contributing to the development of systemic autoimmune and renal diseases. NETs promote the expression of tissue factor, which activates platelets and coagulation factors, thereby promoting thrombosis. This process might contribute to the development of renal diseases, particularly antiphospholipid antibody syndrome (APS) and ANCA-associated vasculitis (AAV). NET-bound proteins, such as metalloproteinases (MMPs) and histones, promote vasculopathy — a feature of renal disease, AAV, SLE and APS — by damaging endothelial cells. Autoantibodies, immune complexes, autoantigens, complement activation factors and cytokines can in turn cause NETosis. NET peptides stimulate NLRP3 inflammasome activation in macrophages, leading to release of IL-1 and IL-18, which promote neutrophil activation and NET formation.
Mouse models of SLE display enhanced bone-marrow NETosis and autoantibodies that recognize NET components. In these murine models, the administration of chemical inhibitors that target PADs decreased NET formation and significantly reduced endothelial dysfunction, the expression of type I interferon-associated genes and tissue damage80–82. NADPH oxidase (Nox) 2 deficiency increased disease severity in lupus-prone mice83, suggesting that inhibition of NADPH oxidase-dependent pathways might actually stimulate the immune response. These findings are similar to observations in patients with chronic granulomatous disease, a condition caused by defective phagocyte NADPH oxidase activity84, in which the incidence of autoimmune diseases is increased. In contrast, inhibition of mitochondrial ROS synthesis, which is enhanced in LDGs from patients with SLE, ameliorates murine SLE78. These observations suggest that the effects of NET inhibition strategies will have to be carefully characterized in the context of SLE before human studies are undertaken.
ANCA-associated vasculitis
ANCAs are associated with a distinct form of vasculitis, termed AAV, which affects small and medium vessels. AAV frequently affects the kidney, skin, upper and lower respiratory tract and peripheral nerves. Systemic AAV can be categorized into three groups, according to the localization of the vascular inflammation and the presence or absence of granulomatosis and asthma: granulomatosis with polyangiitis (GPA), microscopic polyangiitis (MPA) and eosinophilic granulomatosis with polyangiitis (EGPA)18.
In patients with vasculitis, antibodies to MPO85 and PR3 (REF. 86) are not only serological markers of AAV but might also activate neutrophils, leading to degranulation and production of oxygen radicals and to the development of destructive necrotizing vascular and extravascular inflammation87,88. Indeed, MPO-ANCA and PR3-ANCA can activate neutrophils that have been primed by a proinflammatory stimuli, such as TNF89, G-CSF90, lipopolysaccharide91 or the complement anaphylatoxin C5a92, which results in activation of the alternative complement pathway93. This pathway triggers an inflammatory amplification loop in the vessel wall that attracts and activates neutrophils. The activated neutrophils undergo a respiratory burst, thereby producing toxic oxygen radicals and releasing destructive enzymes94,95.
Neutrophils from patients with AAV display enhanced NET formation in vitro96,97. In addition, levels of NET remnants, such as MPO–DNA complexes96, and neutrophil granular proteins, such as calprotectin (a dimer of S100-A8 and S100-A9)98, were increased in sera from patients with AAV. High levels of NET remnants are found in patients with high AAV disease activity and high neutrophil count, and correlate inversely with levels of ANCA99. Furthermore, immunostaining of renal biopsy specimens from patients with AAV revealed the presence of NETs and NET-associated molecules in areas of inflammation, around areas of fibrinoid necrosis in kidneys with necrotizing glomerulonephritis, and along interlobular arterial walls96,100. These observations suggest that NET formation is involved in vascular damage and immune system activation in AAV96,97,99,100. Vascular inflammation could be initiated and perpetuated by ANCA-induced activation of primed neutrophils and monocytes.
NETosis might also induce endothelial cell damage and vasculitis in the glomerulus by concentrating active proteases, which cause direct injury to the cells. Additionally, the presentation of ANCA antigens and dsDNA enhances the ability of NETs to induce auto-antibody formation and furthers cell damage101. ANCA-induced NETs generated by C5a-primed neutrophils could lead to enhanced thrombosis and inflammation in AAV by promoting the expression of tissue factor, a protein required for coagulation94,95. NETs can also present PR3 and MPO to dendritic cells102. These dendritic cells internalize NET material and, when transferred to mice, induce ANCAs against MPO, PR3 and dsDNA, leading to autoimmune vasculitis and lupus-like syndrome. In a 2014 study, serum from patients with ANCA was shown to both induce NETosis and inhibit NET degradation103.
Rheumatoid arthritis
Rheumatoid arthritis is a chronic, systemic autoimmune disease characterized by synovial inflammation and hyperplasia, cartilage and bone destruction, auto-antibody production (including ACPAs), and systemic features, including cardiovascular and pulmonary disorders19. Although, rheumatoid arthritis rarely affects the kidney directly, it can present as focal mesangioproliferative glomerulonephritis, membranous nephropathy or rheumatoid vasculitis104. Patients with long-standing, uncontrolled, inflammatory disease may develop secondary renal amyloidosis.
ACPAs — highly specific autoantibodies that target citrullinated proteins — are found in a significant percentage of patients with rheumatoid arthritis105. Immune complexes containing ACPAs and citrullinated antigens display enhanced immunogenicity and arthritogenicity106. Enhanced NETosis is detected in circulating and synovial-fluid neutrophils, synovial tissue, rheumatoid nodules, and skin of affected patients. This increase in NETosis positively correlates with ACPA levels and with the presence of systemic inflammatory markers in patients with rheumatoid arthritis107.
Extracellular citrullinated autoantigens are found in the joints of patients with rheumatoid arthritis108. This presence could be explained by the increase in NETosis, as NETs are a source of extracellular citrullinated auto-antigens in rheumatoid arthritis107, and/or by the release of active PAD isoforms through NETosis, which could citrullinate extracellular histones and fibrinogen109. Indeed, PAD2 and PAD4 are overexpressed in neutrophils110 and these enzymes are detected in the synovium of patients with rheumatoid arthritis, in close association with neutrophilic infiltrates111.
NETs stimulate inflammatory responses in synovial fibroblasts from patients with rheumatoid arthritis, including the production of proinflammatory cytokines, chemokines, and adhesion molecules107,112. In a predisposed host, enhanced NET formation could, therefore, conceivably promote the generation of key autoantibodies and disturbances in adaptive immunity that generate a vicious cycle leading to further promotion of NETosis and amplification of inflammatory responses in the joint and other tissues. Whether NETs have similar roles in the extra-articular manifestations of rheumatoid arthritis, including kidney damage, remains to be determined.
Antiphospholipid antibody syndrome
Primary APS is an autoimmune disease of uncertain aetiology that can affect the venous and arterial circulation, and is associated with spontaneous thrombosis and/or pregnancy loss, and high titres of autoantibodies against phospholipids (aPLs)20,113. It is proposed that in APS, aPLs and anti-β2GP1 antibodies promote thrombosis by activating endothelial cells, platelets and monocytes. The kidneys are commonly involved in APS, with the possibility of clot formation within any part of the renal vasculature — renal arteries, glomerular capillaries and/or renal veins. Patients with APS and renal involvement may present with renal artery stenosis, renal infarction, renal vein thrombosis, thrombotic microangiopathy and/or hypertension. Renal pathology findings vary from ischaemic glomeruli with thrombosis to proliferative glomerulonephritis with immune complex deposition and inflammatory infiltrates114. Patients with other autoimmune diseases, primarily SLE, can also display an associated APS, known as secondary APS.
NETs are important activators of the coagulation cascade, and are integral components of arterial and venous thrombi38,39,115. As such, neutrophils and NETosis have been implicated in the pathogenesis of APS in the past couple of years. Similar to SLE, the serum of patients with APS displays a decreased ability to degrade NETs; this defect correlates with increased levels of antibodies against NETs and neutrophil remnants116. As in SLE, sera and plasma from patients with primary APS had elevated levels of both cell-free DNA and NET remnants, and neutrophils from patients formed spontaneous NETs ex vivo6. Mechanistically, aPLs promote the release of NETs in a ROS and TLR4-dependent manner. In addition, sera and IgG from patients with APS can stimulate NET release from control neutrophils6. Recently, an LDG population has also been described in primary APS but their role in the pathogenesis of the disease remains to be determined117.
Type 1 diabetes mellitus
Type 1 diabetes mellitus (T1DM) is characterized by the organ-specific autoimmune destruction of insulin-producing pancreatic β cells in genetically predisposed individuals, leading to hyperglycaemia and the need for lifelong exogenous insulin replacement therapy118. Renal involvement (diabetic nephropathy) is a major cause of morbidity and premature mortality in patients with T1DM119. Autoimmunity in T1DM has been primarily attributed to β-cell destruction by autoreactive T cells, followed by the production of autoantibodies to various β-cell antigens118. In the past few years, neutrophils and NETs have been implicated in the pathogenesis of T1DM120–123. In a mouse model of T1DM, neutrophils and NETs were abundant in pancreatic islets, and inhibition of neutrophil function mitigated T1DM development in these mice120.
A 2013 report indicates that patients with T1DM and individuals at risk of developing T1DM exhibit circulating neutropenia122, which is not caused by antineutrophil autoantibody production or by impaired neutrophil production or differentiation. Rather, this neutropenia appears to be related to neutrophil sequestration in pancreatic tissue121. In another study, neutropenia in patients with T1DM was associated with elevated levels and enzymatic activity of various neutrophil serine proteases and resulted from neutrophil death due to NET formation123. These observations implicate neutrophils and NETs in the pathogenesis of T1DM and future studies should address whether modulation of NET and neutrophil function has potential therapeutic implications123.
In addition to a potential role in the development of diabetes, NETosis might be involved in promoting the comorbidities of this disease. Patients with diabetes mellitus (type 1 or type 2) exhibit impaired wound healing and studies in humans and mice published in the past 2 years suggest that this defect could stem from enhanced NETosis124,125.
Renal inflammatory diseases
Glomerulonephritides encompass a varied collection of disorders, including proliferative and non-proliferative glomerulonephritis, which have similar mechanisms of immune-cell-mediated kidney damage. Proliferative glomerulonephritis is associated with increased glomerular cellularity caused by immune cell influx and local immune cell proliferation126.
Leukocytes are key players in the progression of proliferative glomerulonephritis as they influence humoral and adaptive immune responses and modulate local effector pathways, which are directly responsible for glomerular damage70. Neutrophil aggregation is observed in proliferative glomerulonephritis of different aetiologies, including glomerulonephritis caused by the deposition of circulating immune complexes (for example, in the context of infections or autoimmune diseases), the generation of immune complexes within the kidney (such as anti-glomerular basement membrane (anti-GBM) antibodies, ANCAs, and anti-donor antibodies in transplant rejection), and/or complement dysfunction. As described in previous sections, neutrophils, NETosis and NET products, visualized by immunostaining in renal biopsy samples, are prevalent in lupus-associated glomerulonephritis75 and in pauci-immune glomerulonephritis in patients with AAV96,99,100. Moreover, NETs might be important sources of modified autoantigens in the kidney127. The impaired ability of patients with autoimmune disorders to degrade NETs might further enhance the inflammatory effects of NETs and NET components57,103. Indeed, decreased NET degradation was associated with the development of glomerulonephritis in patients with SLE128.
In necrotizing crescentic glomerulonephritis, neutrophil serine proteases like cathepsin G, neutrophil elastase and PR3 promoted IL-1β generation and kidney injury129. Subnephritogenic levels of anti-GBM antibodies and ANCAs amplified neutrophil recruitment and NET formation in renal tissue, and promoted subsequent development of necrotizing crescentic glomerulonephritis130. IL-17 (REF. 131) and C-X-C motif chemokine 5 (CXCL5) have also been hypothesized to drive neutrophil recruitment to kidney tubules, also contributing to renal tissue injury in crescentic glomerulonephritis132. Similarly, endogenous MPO (possibly released through NETosis and/or neutrophil degranulation) promoted neutrophil-mediated renal injury, but attenuated T-cell immunity133, suggesting that endogenous MPO might contribute to local glomerular damage during neutrophil-mediated glomerulonephritis, but that MPO might also attenuate the initiation of the adaptive immune response. Endogenous MPO attenuated the development of pristane-induced lupus nephritis by inhibiting the early inflammatory response and regulating the response of CD4+ T cells and macrophages in secondary lymphoid organs134; however, these observations in animal models need to be interpreted with caution, given previous evidence that comparing neutrophil function between rodents and humans has several caveats and such comparisons can yield different and even opposite results68,135,136.
Immobilized immune complexes might be involved in the induction of NET release through activation of the low affinity immunoglobulin γ Fc region receptor FcγR IIIB, and Integrin α-M (encoded by Itgam)7. Mice depleted of neutrophils or lacking Itgam exhibit a significant reduction in endothelial injury, glomerular thrombosis, and acute renal failure, despite the robust production of renal cytokines137. Intriguingly, endogenous FcγR IIB negatively regulates autoimmunity in anti-MPO-associated glomerulonephritis by limiting ANCA production, T-cell responses, and neutrophil activation in mice with AAV138. Therefore, various neutrophil FcγR receptors might have important roles in regulating antibody-mediated glomerulonephritis139. The alternative complement pathway might also be involved in this disorder: C5a and its neutrophil receptor C5aR could constitute an amplification loop in which NETs activate the complement cascade with production of C5a that, in turn, mediates neutrophil activation and ANCA-induced glomerulonephritis92.
Overall, these findings suggest a potential common denominator in systemic autoimmunity whereby various immune stimuli, including autoantibodies, immune complexes and cytokines, promote the release of extracellular traps by neutrophils in a dysregulated manner. This process can amplify inflammatory responses, activate innate and adaptive immunity and promote externalization of important autoantigens, thereby promoting loss of immune tolerance.
Potential therapeutic modulation of NETosis
NETosis might be involved in the pathophysiology of a variety of autoimmune diseases, opening the possibility of modulating this process for therapeutic purposes (TABLE 1). In in vitro studies, inhibition of ROS production by targeting NADPH6,44,140 or mitochondria78, or use of ROS scavengers, such as N-acetyl cysteine (NAC), reduced NET release141. Interestingly, NAC reduced disease activity in patients with SLE in two separate studies142,143 and inhibition of mitochondrial ROS decreased lupus severity in mouse models78. MPO inhibitors, such as 4-aminobenzoic acid hydrazide141 or PF-1355 (REF. 144), reduce NETosis, neutrophil recruitment and levels of circulating cytokines. PF-1355 treatment reduced neutrophil accumulation in a mouse model of immune complex-mediated vasculitis and suppressed albuminuria and chronic renal dysfunction in a murine model of anti-GBM-induced glomerulonephritis. The use of such inhibitors might improve outcomes for patients with immune-complex-mediated diseases, such as SLE, vasculitis and various types of glomerulonephritis.
Table 1.
Potential therapeutics to modulate neutrophils and NETosis
| Disease | Molecule | Target/Function | Effects on neutrophils | Effect on disease/clinical use |
|---|---|---|---|---|
| Systemic lupus erythematosus | NAC | ROS scavenger | Decreased NET release | Reduced disease activity in patients142,143 |
| MitoTEMPO | Mitochondrial ROS scavenger |
|
Reduced disease severity in murine models78 | |
| Cl-amidine, BB-Cl-amidine | PAD inhibitors | Reduced NET formation leading to decreased IFN responses | Protection against renal, skin and vascular manifestations in mice models81,82 | |
| Ciclosporin A, Tacrolimus | Calcineurin inhibitors |
|
Improvement of renal disease in patients152,153 | |
| DNase 1 | DNA | Enzymatic degradation of NETs | ||
| Eculizumab (anti-C5a monoclonal antibody) | C5a | Reduced NET formation and neutrophil activation | ||
| Rheumatoid arthritis | Cl-amidine | PAD inhibitor | Reduced citrullinated synovial proteins | Reduced arthritis severity in mouse model145 |
| Neutralizing anti-TNF antibodies | TNF | Reduced NET formation | ||
| Neutralizing anti-IL-17 antibodies | IL-17 | Reduced NET formation | ||
| Rituximab | B cells | Decreased NET formation and neutrophil activation through decreased autoantibody production | FDA approved for treatment150 | |
| ANCA-associated vasculitis | Rituximab | B cells | Decreased NET formation and neutrophil activation through decreased autoantibody production | FDA approved for treatment151 |
|
C5aR inhibitor | Reduced NET formation and neutrophil activation | Reduced NET release by human neutrophils in vitro95 | |
| ANCA-induced glomerulonephritis | Anti-C5aR mAb | C5aR inhibitor | Reduced NET formation | Decreased albuminuria and glomerular neutrophil influx in murine model92 |
| Immune complex vasculitis Anti-GBM disease |
PF-1355 | MPO inhibitor | Reduction of plasma MPO activity, neutrophil recruitment, NET formation | Suppression of pulmonary vasculitis and chronic renal dysfunction in mouse models144 |
| Antiphospholipid antibody syndrome | TAK-242 | TLR4 inhibitor | Reduced NET formation | Inhibition of NET release by human neutrophils in vitro6 |
| Gout | Colchicine | Actin cytoskeleton |
|
Used in management of gout attacks158 |
ANCA, anti-neutrophil cytoplasmic antibody; GBM, glomerular basement membrane; mAb, monoclonal antibody; MPO, myeloperoxidase; NAC, N-acetyl cysteine; NET, neutrophil extracellular trap; PAD, protein-arginine deiminase; ROS, reactive oxygen species; TLR, Toll-like receptors; TNF, tumour necrosis factor.
TLR6 and PAD80–82,145 inhibitors might have similar effects to those of MPO inhibitors. In vitro studies on human neutrophils showed that TLR4 inhibition with TAK-242, a small molecule, or with an anti-TLR4 blocking antibody decreased NET formation and ROS production6. Similarly, Cl-amidine and BB-Cl-amidine, two PAD inhibitors, decreased NET formation and protected against renal, skin and vascular manifestations in murine models of lupus81,82. In another study, Cl-amidine reduced the severity of arthritis in a mouse model of inflammatory arthritis145.
Whether impairing NET formation through the inhibition of PAD activity or the modulation of other pathways will have deleterious effects on host defence in humans, remains to be determined. Studies in PAD-knockout mice, however, suggest that, even in the absence of NET formation owing to the loss of PAD enzymes, other antibacterial functions of neutrophils remain intact146,147. Importantly, observations in patients with a complete lack of molecules implicated in NETosis (including PAD)148, as well as in patients with Papillon–Lefevre syndrome (an autosomal recessive genetic disorder caused by a deficiency in cathepsin C)149, provide preliminary evidence that inhibition of NET formation is not overtly immunosuppressive. Whether disruption of NET degradation affects neutrophil antimicrobial functions is still unclear.
As modified histones appear to have immunomodulatory and vasculopathic roles, it is possible that strategies that block the response of tissues to histones might be efficacious for the treatment of inflammatory diseases and prevention of NET-mediated vascular damage54. Alternatively, targetting B cells and plasma cells has been effective in autoimmune diseases, potentially owing to a reduction of autoantibody-induced NET formation150,151.
As efficient NET induction requires the mobilization of intracellular and extracellular calcium pools, using inhibitors of calcineurin (such as cyclosporine A or tacrolimus) or of the G-protein-coupled receptor phospholipase C (such as staurosporine) might be an alternative strategy to suppress or modulate NETosis for therapeutic purposes51. In fact, ciclosporin A152 and tacrolimus153 have shown efficacy for the treatment of various manifestations of SLE.
In addition to suppressing NET formation, another potential approach to treat autoimmune diseases is to disrupt the architecture of already formed NETs with DNase 1 and enhance their clearance. In transgenic mouse models of breast cancer and insulinoma, this approach restored perfusion, reduced injury and improved oxygenation of affected kidney and heart154. Similarly, intrabronchial administration of DNase 1 reduced NETs and lung injury, and improved oxygenation in murine models of primary graft dysfunction after lung transplantation155. In lupus-prone mice, administration of exogenous recombinant DNase can hamper the development of autoantibodies, reduce proteinuria and delay mortality156. A phase Ib study showed that DNase was well tolerated; however, serum markers of disease were unchanged157 and no follow-up studies have been conducted. The use of drugs, such as colchicine, which destabilize the actin cytoskeleton, a structure implicated in NETosis, should also be explored158.
Inhibition of neutrophil recruitment and NET release by disrupting the cross-talk between kindlin-3 and β2-integrin, is also an interesting therapeutic avenue159. CXCL5 could also be targeted to restrict pathogenic neutrophil infiltration in TH17-mediated autoimmune diseases, especially crescentic glomerulonephritis, while retaining the protective function of neutrophils against invading pathogens132.
Inhibition of TNF and IL-17 decreases NET formation in neutrophils from patients with rheumatoid arthritis107. Anti-TNF monoclonal antibodies are now the mainstay of rheumatoid arthritis treatment160 and anti-IL-17 monoclonal antibodies have also shown some efficacy for the treatment of this disease161. Similarly, modulating IL-17 signalling, which is required for the development of lupus glomerulonephritis, with monoclonal antibodies is a potential therapeutic strategy for the treatment of SLE, especially in cases with kidney involvement131. Altering the alternative complement pathway might also yield novel therapeutic strategies such as the inhibition of NET formation by C5a-primed neutrophils. C5aR, which has an important role in ANCA-induced glomerulonephritis, is a potential therapeutic target and its inhibition could prevent NET formation from ANCA-primed neutrophils92,95. Anti-C5 mAb therapy delayed onset of proteinuria and improved survival in murine SLE; a phase I study showed that the treatment was safe in patients with SLE, but did not result in a clear improvement in disease activity162,163.
Conclusions
The involvement of NETs in autoimmune diseases is just beginning to be investigated. In addition to having an important role in the innate immune response against pathogens, ample evidence indicates that NETs can be generated through non-infectious stimuli in various clinical settings. In acute or chronic inflammatory disorders, enhanced NET formation and/or impaired NET degradation might be essential to trigger and propagate autoimmune responses and organ damage (FIG. 3). Understanding precisely how NET dysregulation contributes to different auto-immune diseases will likely require a better understanding of how NET composition varies following specific stimuli, how the interactions between genes and environment affect neutrophil responses and the resulting clinical phenotypes, and of the main pathways that modulate autoimmune NETosis versus microbial-induced NETosis107. Indeed, differences in post-translational modifications of these auto-antigens exposed in various diseases might also contribute to the development of disease-specific autoantibodies107.
Prospective studies are also needed to assess whether the presence of NETosis, NET components and/or LDGs can be used as biomarkers in patients with autoimmune diseases. Whether potential therapeutic interventions to inhibit aberrant NETosis or the production of neutrophilic proteins linked to the pathogenesis of autoimmune diseases will lead to improved treatment of these diseases without markedly decreasing host defence, also needs further examination.
Key points.
Neutrophils may have critical roles in the pathogenesis of autoimmune diseases by forming neutrophil extracellular traps (NETs), secreting proinflammatory cytokines and by directly causing tissue damage
NET formation externalizes autoantigens that might contribute to the pathophysiology and clinical manifestations of autoimmune diseases, either directly or by modulating other components of the immune system
An imbalance in NET formation and degradation might increase the half-life of NET products and augment the immune response
Novel therapies that target key pathways in neutrophil function and NET formation might improve the treatment of autoimmune diseases.
Footnotes
Author contributions
M.J.K. and S.G. researched the data for the article, discussed its content and wrote the Review. M.J.K. reviewed and edited the manuscript before submission.
Competing interests statement
The authors declare no competing interests.
References
- 1.Nauseef WM, Borregaard N. Neutrophils at work. Nat Immunol. 2014;15:602–611. doi: 10.1038/ni.2921. [DOI] [PubMed] [Google Scholar]
- 2.Bardoel BW, Kenny EF, Sollberger G, Zychlinsky A. The balancing act of neutrophils. Cell Host Microbe. 2014;15:526–536. doi: 10.1016/j.chom.2014.04.011. [DOI] [PubMed] [Google Scholar]
- 3.Brinkmann V, et al. Neutrophil extracellular traps kill bacteria. Science. 2004;303:1532–1535. doi: 10.1126/science.1092385. [DOI] [PubMed] [Google Scholar]
- 4.Keshari RS, et al. Cytokines induced neutrophil extracellular traps formation: implication for the inflammatory disease condition. PLoS ONE. 2012;7:e48111. doi: 10.1371/journal.pone.0048111. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Schorn C, et al. Monosodium urate crystals induce extracellular DNA traps in neutrophils, eosinophils, and basophils but not in mononuclear cells. Front Immunol. 2012;3:277. doi: 10.3389/fimmu.2012.00277. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Yalavarthi S, et al. Antiphospholipid antibodies promote the release of neutrophil extracellular traps: a new mechanism of thrombosis in the antiphospholipid syndrome. Arthritis Rheumatol. 2015;67:2990–3003. doi: 10.1002/art.39247. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Behnen M, et al. Immobilized immune complexes induce neutrophil extracellular trap release by human neutrophil granulocytes via FcγRIIIB and Mac-1. J Immunol. 2014;193:1954–1965. doi: 10.4049/jimmunol.1400478. [DOI] [PubMed] [Google Scholar]
- 8.Mantovani A, Cassatella MA, Costantini C, Jaillon S. Neutrophils in the activation and regulation of innate and adaptive immunity. Nat Rev Immunol. 2011;11:519–531. doi: 10.1038/nri3024. [DOI] [PubMed] [Google Scholar]
- 9.Scapini P, Bazzoni F, Cassatella MA. Regulation of B-cell-activating factor (BAFF)/B lymphocyte stimulator (BLyS) expression in human neutrophils. Immunol Lett. 2008;116:1–6. doi: 10.1016/j.imlet.2007.11.009. [DOI] [PubMed] [Google Scholar]
- 10.Huard B, et al. APRIL secreted by neutrophils binds to heparan sulfate proteoglycans to create plasma cell niches in human mucosa. J Clin Invest. 2008;118:2887–2895. doi: 10.1172/JCI33760. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Tvinnereim AR, Hamilton SE, Harty JT. Neutrophil involvement in cross-priming CD8+ T cell responses to bacterial antigens. J Immunol. 2004;173:1994–2002. doi: 10.4049/jimmunol.173.3.1994. [DOI] [PubMed] [Google Scholar]
- 12.Fazio J, Kalyan S, Wesch D, Kabelitz D. Inhibition of human γδ T cell proliferation and effector functions by neutrophil serine proteases. Scand J Immunol. 2014;80:381–389. doi: 10.1111/sji.12221. [DOI] [PubMed] [Google Scholar]
- 13.Megiovanni AM, et al. Polymorphonuclear neutrophils deliver activation signals and antigenic molecules to dendritic cells: a new link between leukocytes upstream of T lymphocytes. J Leukoc Biol. 2006;79:977–988. doi: 10.1189/jlb.0905526. [DOI] [PubMed] [Google Scholar]
- 14.Yang CW, Strong BS, Miller MJ, Unanue ER. Neutrophils influence the level of antigen presentation during the immune response to protein antigens in adjuvants. J Immunol. 2010;185:2927–2934. doi: 10.4049/jimmunol.1001289. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Garcia-Romo GS, et al. Netting neutrophils are major inducers of type I IFN production in pediatric systemic lupus erythematosus. Sci Transl Med. 2011;3:73ra20. doi: 10.1126/scitranslmed.3001201. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Semeraro F, et al. Extracellular histones promote thrombin generation through platelet-dependent mechanisms: involvement of platelet TLR2 and TLR4. Blood. 2011;118:1952–1961. doi: 10.1182/blood-2011-03-343061. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Rahman A, Isenberg DA. Systemic lupus erythematosus. N Engl J Med. 2008;358:929–939. doi: 10.1056/NEJMra071297. [DOI] [PubMed] [Google Scholar]
- 18.Jennette JC, et al. 2012 revised International Chapel Hill Consensus Conference Nomenclature of Vasculitides. Arthritis Rheum. 2013;65:1–11. doi: 10.1002/art.37715. [DOI] [PubMed] [Google Scholar]
- 19.McInnes IB, Schett G. The pathogenesis of rheumatoid arthritis. N Engl J Med. 2011;365:2205–2219. doi: 10.1056/NEJMra1004965. [DOI] [PubMed] [Google Scholar]
- 20.Giannakopoulos B, Krilis SA. The pathogenesis of the antiphospholipid syndrome. N Engl J Med. 2013;368:1033–1044. doi: 10.1056/NEJMra1112830. [DOI] [PubMed] [Google Scholar]
- 21.Kolaczkowska E, Kubes P. Neutrophil recruitment and function in health and inflammation. Nat Rev Immunol. 2013;13:159–175. doi: 10.1038/nri3399. [DOI] [PubMed] [Google Scholar]
- 22.Pillay J, et al. In vivo labeling with 2H2O reveals a human neutrophil lifespan of 5.4 days. Blood. 2010;116:625–627. doi: 10.1182/blood-2010-01-259028. [DOI] [PubMed] [Google Scholar]
- 23.Summers C, et al. Neutrophil kinetics in health and disease. Trends Immunol. 2010;31:318–324. doi: 10.1016/j.it.2010.05.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Colotta F, Re F, Polentarutti N, Sozzani S, Mantovani A. Modulation of granulocyte survival and programmed cell death by cytokines and bacterial products. Blood. 1992;80:2012–2020. [PubMed] [Google Scholar]
- 25.Herter J, Zarbock A. Integrin regulation during leukocyte recruitment. J Immunol. 2013;190:4451–4457. doi: 10.4049/jimmunol.1203179. [DOI] [PubMed] [Google Scholar]
- 26.Henson PM. Pathologic mechanisms in neutrophil-mediated injury. Am J Pathol. 1972;68:593–612. [PMC free article] [PubMed] [Google Scholar]
- 27.Borregaard N, Cowland JB. Granules of the human neutrophilic polymorphonuclear leukocyte. Blood. 1997;89:3503–3521. [PubMed] [Google Scholar]
- 28.Lominadze G, et al. Proteomic analysis of human neutrophil granules. Mol Cell Proteomics. 2005;4:1503–1521. doi: 10.1074/mcp.M500143-MCP200. [DOI] [PubMed] [Google Scholar]
- 29.Faurschou M, Borregaard N. Neutrophil granules and secretory vesicles in inflammation. Microbes Infect. 2003;5:1317–1327. doi: 10.1016/j.micinf.2003.09.008. [DOI] [PubMed] [Google Scholar]
- 30.Kobayashi SD, DeLeo FR. Role of neutrophils in innate immunity: a systems biology-level approach. Wiley Interdiscip Rev Syst Biol Med. 2009;1:309–333. doi: 10.1002/wsbm.32. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Fossati G, et al. The mitochondrial network of human neutrophils: role in chemotaxis, phagocytosis, respiratory burst activation, and commitment to apoptosis. J Immunol. 2003;170:1964–1972. doi: 10.4049/jimmunol.170.4.1964. [DOI] [PubMed] [Google Scholar]
- 32.Fransen M, Nordgren M, Wang B, Apanasets O. Role of peroxisomes in ROS/RNS-metabolism: implications for human disease. Biochim Biophys Acta. 2012;1822:1363–1373. doi: 10.1016/j.bbadis.2011.12.001. [DOI] [PubMed] [Google Scholar]
- 33.Rodriguez PC, et al. Arginase I production in the tumor microenvironment by mature myeloid cells inhibits T-cell receptor expression and antigen-specific T-cell responses. Cancer Res. 2004;64:5839–5849. doi: 10.1158/0008-5472.CAN-04-0465. [DOI] [PubMed] [Google Scholar]
- 34.Bowers NL, et al. Immune suppression by neutrophils in HIV-1 infection: role of PD-L1/PD-1 pathway. PLoS Pathog. 2014;10:e1003993. doi: 10.1371/journal.ppat.1003993. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Puga I, et al. B cell-helper neutrophils stimulate the diversification and production of immunoglobulin in the marginal zone of the spleen. Nat Immunol. 2012;13:170–180. doi: 10.1038/ni.2194. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Lande R, et al. Neutrophils activate plasmacytoid dendritic cells by releasing self-DNA-peptide complexes in systemic lupus erythematosus. Sci Transl Med. 2011;3:73ra19. doi: 10.1126/scitranslmed.3001180. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Tillack K, Breiden P, Martin R, Sospedra M. T lymphocyte priming by neutrophil extracellular traps links innate and adaptive immune responses. J Immunol. 2012;188:3150–3159. doi: 10.4049/jimmunol.1103414. [DOI] [PubMed] [Google Scholar]
- 38.Fuchs TA, et al. Extracellular DNA traps promote thrombosis. Proc Natl Acad Sci USA. 2010;107:15880–15885. doi: 10.1073/pnas.1005743107. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Massberg S, et al. Reciprocal coupling of coagulation and innate immunity via neutrophil serine proteases. Nat Med. 2010;16:887–896. doi: 10.1038/nm.2184. [DOI] [PubMed] [Google Scholar]
- 40.Clark SR, et al. Platelet TLR4 activates neutrophil extracellular traps to ensnare bacteria in septic blood. Nat Med. 2007;13:463–469. doi: 10.1038/nm1565. [DOI] [PubMed] [Google Scholar]
- 41.von Bruhl ML, et al. Monocytes, neutrophils, and platelets cooperate to initiate and propagate venous thrombosis in mice in vivo. J Exp Med. 2012;209:819–835. doi: 10.1084/jem.20112322. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Barrientos L, et al. Neutrophil extracellular traps downregulate lipopolysaccharide-induced activation of monocyte-derived dendritic cells. J Immunol. 2014;193:5689–5698. doi: 10.4049/jimmunol.1400586. [DOI] [PubMed] [Google Scholar]
- 43.Schauer C, et al. Aggregated neutrophil extracellular traps limit inflammation by degrading cytokines and chemokines. Nat Med. 2014;20:511–517. doi: 10.1038/nm.3547. [DOI] [PubMed] [Google Scholar]
- 44.Fuchs TA, et al. Novel cell death program leads to neutrophil extracellular traps. J Cell Biol. 2007;176:231–241. doi: 10.1083/jcb.200606027. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Pilsczek FH, et al. A novel mechanism of rapid nuclear neutrophil extracellular trap formation in response to Staphylococcus aureus. J Immunol. 2010;185:7413–7425. doi: 10.4049/jimmunol.1000675. [DOI] [PubMed] [Google Scholar]
- 46.Buchanan JT, et al. DNase expression allows the pathogen group A Streptococcus to escape killing in neutrophil extracellular traps. Curr Biol. 2006;16:396–400. doi: 10.1016/j.cub.2005.12.039. [DOI] [PubMed] [Google Scholar]
- 47.Ramos-Kichik V, et al. Neutrophil extracellular traps are induced by Mycobacterium tuberculosis. Tuberculosis (Edinb) 2009;89:29–37. doi: 10.1016/j.tube.2008.09.009. [DOI] [PubMed] [Google Scholar]
- 48.Bruns S, et al. Production of extracellular traps against Aspergillus fumigatus in vitro and in infected lung tissue is dependent on invading neutrophils and influenced by hydrophobin RodA. PLoS Pathog. 2010;6:e1000873. doi: 10.1371/journal.ppat.1000873. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Abi Abdallah DS, et al. Toxoplasma gondii triggers release of human and mouse neutrophil extracellular traps. Infect Immun. 2012;80:768–777. doi: 10.1128/IAI.05730-11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Saitoh T, et al. Neutrophil extracellular traps mediate a host defense response to human immunodeficiency virus-1. Cell Host Microbe. 2012;12:109–116. doi: 10.1016/j.chom.2012.05.015. [DOI] [PubMed] [Google Scholar]
- 51.Gupta AK, Giaglis S, Hasler P, Hahn S. Efficient neutrophil extracellular trap induction requires mobilization of both intracellular and extracellular calcium pools and is modulated by cyclosporine A. PLoS ONE. 2014;9:e97088. doi: 10.1371/journal.pone.0097088. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Metzler KD, Goosmann C, Lubojemska A, Zychlinsky A, Papayannopoulos V. A myeloperoxidase-containing complex regulates neutrophil elastase release and actin dynamics during NETosis. Cell Rep. 2014;8:883–896. doi: 10.1016/j.celrep.2014.06.044. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Palmer LJ, et al. Hypochlorous acid regulates neutrophil extracellular trap release in humans. Clin Exp Immunol. 2012;167:261–268. doi: 10.1111/j.1365-2249.2011.04518.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Tessarz P, Kouzarides T. Histone core modifications regulating nucleosome structure and dynamics. Nat Rev Mol Cell Biol. 2014;15:703–708. doi: 10.1038/nrm3890. [DOI] [PubMed] [Google Scholar]
- 55.Yipp BG, et al. Infection-induced NETosis is a dynamic process involving neutrophil multitasking in vivo. Nat Med. 2012;18:1386–1393. doi: 10.1038/nm.2847. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Yousefi S, Mihalache C, Kozlowski E, Schmid I, Simon HU. Viable neutrophils release mitochondrial DNA to form neutrophil extracellular traps. Cell Death Differ. 2009;16:1438–1444. doi: 10.1038/cdd.2009.96. [DOI] [PubMed] [Google Scholar]
- 57.Hakkim A, et al. Impairment of neutrophil extracellular trap degradation is associated with lupus nephritis. Proc Natl Acad Sci USA. 2010;107:9813–9818. doi: 10.1073/pnas.0909927107. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Farrera C, Fadeel B. Macrophage clearance of neutrophil extracellular traps is a silent process. J Immunol. 2013;191:2647–2656. doi: 10.4049/jimmunol.1300436. [DOI] [PubMed] [Google Scholar]
- 59.Canas CA, Canas F, Bonilla-Abadia F, Ospina FE, Tobon GJ. Epigenetics changes associated to environmental triggers in autoimmunity. Autoimmunity. 2016;49:1–11. doi: 10.3109/08916934.2015.1086996. [DOI] [PubMed] [Google Scholar]
- 60.Davidson A, Diamond B. Autoimmune diseases. N Engl J Med. 2001;345:340–350. doi: 10.1056/NEJM200108023450506. [DOI] [PubMed] [Google Scholar]
- 61.Cocca BA, Cline AM, Radic MZ. Blebs and apoptotic bodies are B cell autoantigens. J Immunol. 2002;169:159–166. doi: 10.4049/jimmunol.169.1.159. [DOI] [PubMed] [Google Scholar]
- 62.Tait SW, Ichim G, Green DR. Die another way — non-apoptotic mechanisms of cell death. J Cell Sci. 2014;127:2135–2144. doi: 10.1242/jcs.093575. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Gehrke N, et al. Oxidative damage of DNA confers resistance to cytosolic nuclease TREX1 degradation and potentiates STING-dependent immune sensing. Immunity. 2013;39:482–495. doi: 10.1016/j.immuni.2013.08.004. [DOI] [PubMed] [Google Scholar]
- 64.Abou-Raya A, Abou-Raya S. Inflammation: a pivotal link between autoimmune diseases and atherosclerosis. Autoimmun Rev. 2006;5:331–337. doi: 10.1016/j.autrev.2005.12.006. [DOI] [PubMed] [Google Scholar]
- 65.Tektonidou MG, Laskari K, Panagiotakos DB, Moutsopoulos HM. Risk factors for thrombosis and primary thrombosis prevention in patients with systemic lupus erythematosus with or without antiphospholipid antibodies. Arthritis Rheum. 2009;61:29–36. doi: 10.1002/art.24232. [DOI] [PubMed] [Google Scholar]
- 66.Merkel PA, et al. Brief communication: high incidence of venous thrombotic events among patients with Wegener granulomatosis: the Wegener’s Clinical Occurrence of Thrombosis (WeCLOT) Study. Ann Intern Med. 2005;142:620–626. doi: 10.7326/0003-4819-142-8-200505030-00011. [DOI] [PubMed] [Google Scholar]
- 67.Sokolove J, et al. Brief report: citrullination within the atherosclerotic plaque: a potential target for the anti-citrullinated protein antibody response in rheumatoid arthritis. Arthritis Rheum. 2013;65:1719–1724. doi: 10.1002/art.37961. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Smith CK, et al. Neutrophil extracellular trap-derived enzymes oxidize high-density lipoprotein: an additional proatherogenic mechanism in systemic lupus erythematosus. Arthritis Rheumatol. 2014;66:2532–2544. doi: 10.1002/art.38703. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Kambas K, Mitroulis I, Ritis K. The emerging role of neutrophils in thrombosis — the journey of TF through NETs. Front Immunol. 2012;3:385. doi: 10.3389/fimmu.2012.00385. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Mayadas TN, Rosetti F, Ernandez T, Sethi S. Neutrophils: game changers in glomerulonephritis? Trends Mol Med. 2010;16:368–378. doi: 10.1016/j.molmed.2010.06.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Crow MK. Type I interferon in the pathogenesis of lupus. J Immunol. 2014;192:5459–5468. doi: 10.4049/jimmunol.1002795. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Hacbarth E, Kajdacsy-Balla A. Low density neutrophils in patients with systemic lupus erythematosus, rheumatoid arthritis, and acute rheumatic fever. Arthritis Rheum. 1986;29:1334–1342. doi: 10.1002/art.1780291105. [DOI] [PubMed] [Google Scholar]
- 73.Denny MF, et al. A distinct subset of proinflammatory neutrophils isolated from patients with systemic lupus erythematosus induces vascular damage and synthesizes type I IFNs. J Immunol. 2010;184:3284–3297. doi: 10.4049/jimmunol.0902199. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Bennett L, et al. Interferon and granulopoiesis signatures in systemic lupus erythematosus blood. J Exp Med. 2003;197:711–723. doi: 10.1084/jem.20021553. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Villanueva E, et al. Netting neutrophils induce endothelial damage, infiltrate tissues, and expose immunostimulatory molecules in systemic lupus erythematosus. J Immunol. 2011;187:538–552. doi: 10.4049/jimmunol.1100450. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Leffler J, et al. Neutrophil extracellular traps that are not degraded in systemic lupus erythematosus activate complement exacerbating the disease. J Immunol. 2012;188:3522–3531. doi: 10.4049/jimmunol.1102404. [DOI] [PubMed] [Google Scholar]
- 77.Carmona-Rivera C, Zhao W, Yalavarthi S, Kaplan MJ. Neutrophil extracellular traps induce endothelial dysfunction in systemic lupus erythematosus through the activation of matrix metalloproteinase-2. Ann Rheum Dis. 2015;74:1417–1424. doi: 10.1136/annrheumdis-2013-204837. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Lood C, et al. Neutrophil extracellular traps enriched in oxidized mitochondrial DNA are interferogenic and contribute to lupus-like disease. Nat Med. 2016;22:146–153. doi: 10.1038/nm.4027. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Kahlenberg JM, Carmona-Rivera C, Smith CK, Kaplan MJ. Neutrophil extracellular trap-associated protein activation of the NLRP3 inflammasome is enhanced in lupus macrophages. J Immunol. 2013;190:1217–1226. doi: 10.4049/jimmunol.1202388. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Knight JS, et al. Peptidylarginine deiminase inhibition reduces vascular damage and modulates innate immune responses in murine models of atherosclerosis. Circ Res. 2014;114:947–956. doi: 10.1161/CIRCRESAHA.114.303312. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Knight JS, et al. Peptidylarginine deiminase inhibition disrupts NET formation and protects against kidney, skin and vascular disease in lupus-prone MRL/lpr mice. Ann Rheum Dis. 2015;74:2199–2206. doi: 10.1136/annrheumdis-2014-205365. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Knight JS, et al. Peptidylarginine deiminase inhibition is immunomodulatory and vasculoprotective in murine lupus. J Clin Invest. 2013;123:2981–2993. doi: 10.1172/JCI67390. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Campbell AM, Kashgarian M, Shlomchik MJ. NADPH oxidase inhibits the pathogenesis of systemic lupus erythematosus. Sci Transl Med. 2012;5:157ra141. doi: 10.1126/scitranslmed.3004801. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Winkelstein JA, et al. Chronic granulomatous disease. Report on a national registry of 368 patients. Medicine (Baltimore) 2000;79:155–169. doi: 10.1097/00005792-200005000-00003. [DOI] [PubMed] [Google Scholar]
- 85.Falk RJ, Jennette JC. Anti-neutrophil cytoplasmic autoantibodies with specificity for myeloperoxidase in patients with systemic vasculitis and idiopathic necrotizing and crescentic glomerulonephritis. N Engl J Med. 1988;318:1651–1657. doi: 10.1056/NEJM198806233182504. [DOI] [PubMed] [Google Scholar]
- 86.Niles JL, McCluskey RT, Ahmad MF, Arnaout MA. Wegener’s granulomatosis autoantigen is a novel neutrophil serine proteinase. Blood. 1989;74:1888–1893. [PubMed] [Google Scholar]
- 87.Jennette JC, Falk RJ. Pathogenesis of antineutrophil cytoplasmic autoantibody-mediated disease. Nat Rev Rheumatol. 2014;10:463–473. doi: 10.1038/nrrheum.2014.103. [DOI] [PubMed] [Google Scholar]
- 88.Charles LA, Caldas ML, Falk RJ, Terrell RS, Jennette JC. Antibodies against granule proteins activate neutrophils in vitro. J Leukoc Biol. 1991;50:539–546. doi: 10.1002/jlb.50.6.539. [DOI] [PubMed] [Google Scholar]
- 89.Falk RJ, Terrell RS, Charles LA, Jennette JC. Anti-neutrophil cytoplasmic autoantibodies induce neutrophils to degranulate and produce oxygen radicals in vitro. Proc Natl Acad Sci USA. 1990;87:4115–4119. doi: 10.1073/pnas.87.11.4115. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Freeley SJ, Coughlan AM, Popat RJ, Dunn-Walters DK, Robson MG. Granulocyte colony stimulating factor exacerbates antineutrophil cytoplasmic antibody vasculitis. Ann Rheum Dis. 2013;72:1053–1058. doi: 10.1136/annrheumdis-2012-202160. [DOI] [PubMed] [Google Scholar]
- 91.Huugen D, et al. Aggravation of anti-myeloperoxidase antibody-induced glomerulonephritis by bacterial lipopolysaccharide: role of tumor necrosis factor-α. Am J Pathol. 2005;167:47–58. doi: 10.1016/s0002-9440(10)62952-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Schreiber A, et al. C5a receptor mediates neutrophil activation and ANCA-induced glomerulonephritis. J Am Soc Nephrol. 2009;20:289–298. doi: 10.1681/ASN.2008050497. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Wang H, Wang C, Zhao MH, Chen M. Neutrophil extracellular traps can activate alternative complement pathways. Clin Exp Immunol. 2015;181:518–527. doi: 10.1111/cei.12654. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Kambas K, et al. Tissue factor expression in neutrophil extracellular traps and neutrophil derived microparticles in antineutrophil cytoplasmic antibody associated vasculitis may promote thromboinflammation and the thrombophilic state associated with the disease. Ann Rheum Dis. 2014;73:1854–1863. doi: 10.1136/annrheumdis-2013-203430. [DOI] [PubMed] [Google Scholar]
- 95.Huang YM, Wang H, Wang C, Chen M, Zhao MH. C5a inducing tissue factor expressing microparticles and neutrophil extracellular traps promote hypercoagulability in ANCA-associated vasculitis. Arthritis Rheumatol. 2015;67:2780–2790. doi: 10.1002/art.39239. [DOI] [PubMed] [Google Scholar]
- 96.Kessenbrock K, et al. Netting neutrophils in autoimmune small-vessel vasculitis. Nat Med. 2009;15:623–625. doi: 10.1038/nm.1959. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Tang S, et al. Neutrophil extracellular trap formation is associated with autophagy-related signalling in ANCA-associated vasculitis. Clin Exp Immunol. 2015;180:408–418. doi: 10.1111/cei.12589. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Pepper RJ, et al. Leukocyte and serum S100A8/S100A9 expression reflects disease activity in ANCA-associated vasculitis and glomerulonephritis. Kidney Int. 2013;83:1150–1158. doi: 10.1038/ki.2013.2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Soderberg D, et al. Increased levels of neutrophil extracellular trap remnants in the circulation of patients with small vessel vasculitis, but an inverse correlation to anti-neutrophil cytoplasmic antibodies during remission. Rheumatology. 2015;54:2085–2094. doi: 10.1093/rheumatology/kev217. [DOI] [PubMed] [Google Scholar]
- 100.Yoshida M, Sasaki M, Sugisaki K, Yamaguchi Y, Yamada M. Neutrophil extracellular trap components in fibrinoid necrosis of the kidney with myeloperoxidase-ANCA-associated vasculitis. Clin Kidney J. 2013;6:308–312. doi: 10.1093/ckj/sft048. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.Nakazawa D, et al. Abnormal conformation and impaired degradation of propylthiouracil-induced neutrophil extracellular traps: implications of disordered neutrophil extracellular traps in a rat model of myeloperoxidase antineutrophil cytoplasmic antibody-associated vasculitis. Arthritis Rheum. 2012;64:3779–3787. doi: 10.1002/art.34619. [DOI] [PubMed] [Google Scholar]
- 102.Sangaletti S, et al. Neutrophil extracellular traps mediate transfer of cytoplasmic neutrophil antigens to myeloid dendritic cells toward ANCA induction and associated autoimmunity. Blood. 2012;120:3007–3018. doi: 10.1182/blood-2012-03-416156. [DOI] [PubMed] [Google Scholar]
- 103.Nakazawa D, et al. Enhanced formation and disordered regulation of NETs in myeloperoxidase-ANCA-associated microscopic polyangiitis. J Am Soc Nephrol. 2014;25:990–997. doi: 10.1681/ASN.2013060606. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Vinicki JP, et al. Analysis of 65 renal biopsies from patients with rheumatoid arthritis: change in treatment strategies decreased frequency and modified histopathological findings. J Clin Rheumatol. 2015;21:335–340. doi: 10.1097/RHU.0000000000000302. [DOI] [PubMed] [Google Scholar]
- 105.Makrygiannakis D, et al. Citrullination is an inflammation-dependent process. Ann Rheum Dis. 2006;65:1219–1222. doi: 10.1136/ard.2005.049403. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106.Kidd BA, et al. Epitope spreading to citrullinated antigens in mouse models of autoimmune arthritis and demyelination. Arthritis Res Ther. 2008;10:R119. doi: 10.1186/ar2523. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Khandpur R, et al. NETs are a source of citrullinated autoantigens and stimulate inflammatory responses in rheumatoid arthritis. Sci Transl Med. 2013;5:178ra140. doi: 10.1126/scitranslmed.3005580. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108.van Beers JJ, et al. The rheumatoid arthritis synovial fluid citrullinome reveals novel citrullinated epitopes in apolipoprotein E, myeloid nuclear differentiation antigen, and β-actin. Arthritis Rheum. 2013;65:69–80. doi: 10.1002/art.37720. [DOI] [PubMed] [Google Scholar]
- 109.Spengler J, et al. Release of active peptidyl arginine deiminases by neutrophils can explain production of extracellular citrullinated autoantigens in rheumatoid arthritis synovial fluid. Arthritis Rheumatol. 2015;67:3135–3145. doi: 10.1002/art.39313. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110.Jones JE, Causey CP, Knuckley B, Slack-Noyes JL, Thompson PR. Protein arginine deiminase 4 (PAD4): current understanding and future therapeutic potential. Curr Opin Drug Discov Devel. 2009;12:616–627. [PMC free article] [PubMed] [Google Scholar]
- 111.Foulquier C, et al. Peptidyl arginine deiminase type 2 (PAD-2) and PAD-4 but not PAD-1, PAD-3, and PAD-6 are expressed in rheumatoid arthritis synovium in close association with tissue inflammation. Arthritis Rheum. 2007;56:3541–3553. doi: 10.1002/art.22983. [DOI] [PubMed] [Google Scholar]
- 112.Pratesi F, et al. Antibodies from patients with rheumatoid arthritis target citrullinated histone 4 contained in neutrophils extracellular traps. Ann Rheum Dis. 2014;73:1414–1422. doi: 10.1136/annrheumdis-2012-202765. [DOI] [PubMed] [Google Scholar]
- 113.Miyakis S, et al. International consensus statement on an update of the classification criteria for definite antiphospholipid syndrome (APS) J Thromb Haemost. 2006;4:295–306. doi: 10.1111/j.1538-7836.2006.01753.x. [DOI] [PubMed] [Google Scholar]
- 114.Sciascia S, Cuadrado MJ, Khamashta M, Roccatello D. Renal involvement in antiphospholipid syndrome. Nat Rev Nephrol. 2014;10:279–289. doi: 10.1038/nrneph.2014.38. [DOI] [PubMed] [Google Scholar]
- 115.Brill A, et al. Neutrophil extracellular traps promote deep vein thrombosis in mice. J Thromb Haemost. 2012;10:136–144. doi: 10.1111/j.1538-7836.2011.04544.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116.Leffler J, et al. Degradation of neutrophil extracellular traps is decreased in patients with antiphospholipid syndrome. Clin Exp Rheumatol. 2014;32:66–70. [PubMed] [Google Scholar]
- 117.van den Hoogen LL, et al. Low-density granulocytes are increased in antiphospholipid syndrome and are associated with anti-β2-glycoprotein I antibodies: comment on the article by Yalavarthi et al. Arthritis Rheumatol. 2016 doi: 10.1002/art.39576. http://dx.doi.org/10.1002/art.39576. [DOI] [PubMed]
- 118.Atkinson MA, Eisenbarth GS, Michels AW. Type 1 diabetes. Lancet. 2014;383:69–82. doi: 10.1016/S0140-6736(13)60591-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119.Bjornstad P, Cherney D, Maahs DM. Early diabetic nephropathy in type 1 diabetes: new insights. Curr Opin Endocrinol Diabetes Obes. 2014;21:279–286. doi: 10.1097/MED.0000000000000074. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120.Diana J, et al. Crosstalk between neutrophils, B-1a cells and plasmacytoid dendritic cells initiates autoimmune diabetes. Nat Med. 2013;19:65–73. doi: 10.1038/nm.3042. [DOI] [PubMed] [Google Scholar]
- 121.Valle A, et al. Reduction of circulating neutrophils precedes and accompanies type 1 diabetes. Diabetes. 2013;62:2072–2077. doi: 10.2337/db12-1345. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122.Harsunen MH, et al. Reduced blood leukocyte and neutrophil numbers in the pathogenesis of type 1 diabetes. Horm Metab Res. 2013;45:467–470. doi: 10.1055/s-0032-1331226. [DOI] [PubMed] [Google Scholar]
- 123.Wang Y, et al. Increased neutrophil elastase and proteinase 3 and augmented NETosis are closely associated with β-cell autoimmunity in patients with type 1 diabetes. Diabetes. 2014;63:4239–4248. doi: 10.2337/db14-0480. [DOI] [PubMed] [Google Scholar]
- 124.Wong SL, et al. Diabetes primes neutrophils to undergo NETosis, which impairs wound healing. Nat Med. 2015;21:815–819. doi: 10.1038/nm.3887. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125.Fadini GP, et al. NETosis delays diabetic wound healing in mice and humans. Diabetes. 2016 doi: 10.2337/db15-0863. http://dx.doi.org/10.2337/db15-0863. [DOI] [PubMed]
- 126.Hricik DE, Chung-Park M, Sedor JR. Glomerulonephritis. N Engl J Med. 1998;339:888–899. doi: 10.1056/NEJM199809243391306. [DOI] [PubMed] [Google Scholar]
- 127.O’Sullivan KM, et al. Renal participation of myeloperoxidase in antineutrophil cytoplasmic antibody (ANCA)-associated glomerulonephritis. Kidney Int. 2015;88:1030–1046. doi: 10.1038/ki.2015.202. [DOI] [PubMed] [Google Scholar]
- 128.Leffler J, et al. Degradation of neutrophil extracellular traps co-varies with disease activity in patients with systemic lupus erythematosus. Arthritis Res Ther. 2013;15:R84. doi: 10.1186/ar4264. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129.Schreiber A, et al. Neutrophil serine proteases promote IL-1β generation and injury in necrotizing crescentic glomerulonephritis. J Am Soc Nephrol. 2012;23:470–482. doi: 10.1681/ASN.2010080892. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130.Kanzaki G, et al. Impact of anti-glomerular basement membrane antibodies and glomerular neutrophil activation on glomerulonephritis in experimental myeloperoxidase-antineutrophil cytoplasmic antibody vasculitis. Nephrol Dial Transplant. 2016;31:574–585. doi: 10.1093/ndt/gfv384. [DOI] [PubMed] [Google Scholar]
- 131.Pisitkun P, et al. Interleukin-17 cytokines are critical in development of fatal lupus glomerulonephritis. Immunity. 2012;37:1104–1115. doi: 10.1016/j.immuni.2012.08.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132.Disteldorf EM, et al. CXCL5 drives neutrophil recruitment in TH17-mediated GN. J Am Soc Nephrol. 2015;26:55–66. doi: 10.1681/ASN.2013101061. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 133.Odobasic D, Kitching AR, Semple TJ, Holdsworth SR. Endogenous myeloperoxidase promotes neutrophil-mediated renal injury, but attenuates T cell immunity inducing crescentic glomerulonephritis. J Am Soc Nephrol. 2007;18:760–770. doi: 10.1681/ASN.2006040375. [DOI] [PubMed] [Google Scholar]
- 134.Odobasic D, et al. Suppression of autoimmunity and renal disease in pristane-induced lupus by myeloperoxidase. Arthritis Rheumatol. 2015;67:1868–1880. doi: 10.1002/art.39109. [DOI] [PubMed] [Google Scholar]
- 135.Brennan ML, et al. Increased atherosclerosis in myeloperoxidase-deficient mice. J Clin Invest. 2001;107:419–430. doi: 10.1172/JCI8797. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 136.Mangold A, et al. Coronary neutrophil extracellular trap burden and deoxyribonuclease activity in ST-elevation acute coronary syndrome are predictors of ST-segment resolution and infarct size. Circ Res. 2015;116:1182–1192. doi: 10.1161/CIRCRESAHA.116.304944. [DOI] [PubMed] [Google Scholar]
- 137.Hirahashi J, et al. Mac-1 (CD11b/CD18) links inflammation and thrombosis after glomerular injury. Circulation. 2009;120:1255–1265. doi: 10.1161/CIRCULATIONAHA.109.873695. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 138.Ooi JD, et al. FcγRIIB regulates T-cell autoreactivity, ANCA production, and neutrophil activation to suppress anti-myeloperoxidase glomerulonephritis. Kidney Int. 2014;86:1140–1149. doi: 10.1038/ki.2014.189. [DOI] [PubMed] [Google Scholar]
- 139.Tsuboi N, Asano K, Lauterbach M, Mayadas TN. Human neutrophil Fcγ receptors initiate and play specialized nonredundant roles in antibody-mediated inflammatory diseases. Immunity. 2008;28:833–846. doi: 10.1016/j.immuni.2008.04.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 140.Remijsen Q, et al. Neutrophil extracellular trap cell death requires both autophagy and superoxide generation. Cell Res. 2011;21:290–304. doi: 10.1038/cr.2010.150. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 141.Patel S, et al. Nitric oxide donors release extracellular traps from human neutrophils by augmenting free radical generation. Nitric Oxide. 2010;22:226–234. doi: 10.1016/j.niox.2010.01.001. [DOI] [PubMed] [Google Scholar]
- 142.Garcia RJ, et al. Attention deficit and hyperactivity disorder scores are elevated and respond to N-acetylcysteine treatment in patients with systemic lupus erythematosus. Arthritis Rheum. 2013;65:1313–1318. doi: 10.1002/art.37893. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 143.Lai ZW, et al. N-acetylcysteine reduces disease activity by blocking mammalian target of rapamycin in T cells from systemic lupus erythematosus patients: a randomized, double-blind, placebo-controlled trial. Arthritis Rheum. 2012;64:2937–2946. doi: 10.1002/art.34502. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 144.Zheng W, et al. PF-1355, a mechanism-based myeloperoxidase inhibitor, prevents immune complex vasculitis and anti-glomerular basement membrane glomerulonephritis. J Pharmacol Exp Ther. 2015;353:288–298. doi: 10.1124/jpet.114.221788. [DOI] [PubMed] [Google Scholar]
- 145.Willis VC, et al. N-α-benzoyl-N5-(2-chloro-1-iminoethyl)-L-ornithine amide, a protein arginine deiminase inhibitor, reduces the severity of murine collagen-induced arthritis. J Immunol. 2011;186:4396–4404. doi: 10.4049/jimmunol.1001620. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 146.Martinod K, et al. PAD4-deficiency does not affect bacteremia in polymicrobial sepsis and ameliorates endotoxemic shock. Blood. 2015;125:1948–1956. doi: 10.1182/blood-2014-07-587709. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 147.Li P, et al. PAD4 is essential for antibacterial innate immunity mediated by neutrophil extracellular traps. J Exp Med. 2010;207:1853–1862. doi: 10.1084/jem.20100239. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 148.Sulem P, et al. Identification of a large set of rare complete human knockouts. Nat Genet. 2015;47:448–452. doi: 10.1038/ng.3243. [DOI] [PubMed] [Google Scholar]
- 149.Sorensen OE, et al. Papillon–Lèfevre syndrome patient reveals species-dependent requirements for neutrophil defenses. J Clin Invest. 2014;124:4539–4548. doi: 10.1172/JCI76009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 150.Cohen SB, et al. Rituximab for rheumatoid arthritis refractory to anti-tumor necrosis factor therapy: results of a multicenter, randomized, double-blind, placebo-controlled, phase III trial evaluating primary efficacy and safety at twenty-four weeks. Arthritis Rheum. 2006;54:2793–2806. doi: 10.1002/art.22025. [DOI] [PubMed] [Google Scholar]
- 151.Stone JH, et al. Rituximab versus cyclophosphamide for ANCA-associated vasculitis. N Engl J Med. 2010;363:221–232. doi: 10.1056/NEJMoa0909905. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 152.Zavada J, et al. Cyclosporine A or intravenous cyclophosphamide for lupus nephritis: the Cyclofa-Lune study. Lupus. 2010;19:1281–1289. doi: 10.1177/0961203310371155. [DOI] [PubMed] [Google Scholar]
- 153.Lee YH, Lee HS, Choi SJ, Dai Ji J, Song GG. Efficacy and safety of tacrolimus therapy for lupus nephritis: a systematic review of clinical trials. Lupus. 2011;20:636–640. doi: 10.1177/0961203310389486. [DOI] [PubMed] [Google Scholar]
- 154.Cedervall J, et al. Neutrophil extracellular traps accumulate in peripheral blood vessels and compromise organ function in tumor-bearing animals. Cancer Res. 2015;75:2653–2662. doi: 10.1158/0008-5472.CAN-14-3299. [DOI] [PubMed] [Google Scholar]
- 155.Sayah DM, et al. Neutrophil extracellular traps are pathogenic in primary graft dysfunction after lung transplantation. Am J Respir Crit Care Med. 2015;191:455–463. doi: 10.1164/rccm.201406-1086OC. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 156.Macanovic M, et al. The treatment of systemic lupus erythematosus (SLE) in NZB/W F1 hybrid mice; studies with recombinant murine DNase and with dexamethasone. Clin Exp Immunol. 1996;106:243–252. doi: 10.1046/j.1365-2249.1996.d01-839.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 157.Davis JC, Jr, et al. Recombinant human Dnase I (rhDNase) in patients with lupus nephritis. Lupus. 1999;8:68–76. doi: 10.1191/096120399678847380. [DOI] [PubMed] [Google Scholar]
- 158.Neeli I, Dwivedi N, Khan S, Radic M. Regulation of extracellular chromatin release from neutrophils. J Innate Immun. 2009;1:194–201. doi: 10.1159/000206974. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 159.Xu Z, et al. Interaction of kindlin-3 and β2-integrins differentially regulates neutrophil recruitment and NET release in mice. Blood. 2015;126:373–377. doi: 10.1182/blood-2015-03-636720. [DOI] [PubMed] [Google Scholar]
- 160.Taylor PC, Feldmann M. Anti-TNF biologic agents: still the therapy of choice for rheumatoid arthritis. Nat Rev Rheumatol. 2009;5:578–582. doi: 10.1038/nrrheum.2009.181. [DOI] [PubMed] [Google Scholar]
- 161.Kunwar S, Dahal K, Sharma S. Anti-IL-17 therapy in treatment of rheumatoid arthritis: a systematic literature review and meta-analysis of randomized controlled trials. Rheumatol Int. 2016 doi: 10.1007/s00296-016-3480-9. http://dx.doi.org/10.1007/s00296-016-3480-9. [DOI] [PubMed]
- 162.Wang Y, et al. Amelioration of lupus-like autoimmune disease in NZB/WF1 mice after treatment with a blocking monoclonal antibody specific for complement component C5. Proc Natl Acad Sci USA. 1996;93:8563–8568. doi: 10.1073/pnas.93.16.8563. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 163.Furie RML, Rollins SA, Mojcik CF. 64th Annual Scientific Meeting of the American College of Rheumatology; San Francisco. 2001. [Google Scholar]



