Skip to main content
Access keys NCBI Homepage MyNCBI Homepage Main Content Main Navigation
J Leukoc Biol. 2016 Feb; 99(2): 253–264.
Published online 2015 Oct 2. doi: 10.1189/jlb.5BT0615-247R
PMCID: PMC4718195
PMID: 26432901

At the Bench: Neutrophil extracellular traps (NETs) highlight novel aspects of innate immune system involvement in autoimmune diseases

Basic Research Review for Clinicians: Immunogenic and vasculopathic roles of NETs in the pathogenesis of SLE and other related autoimmune diseases.

Keywords: systemic lupus erythematosus, ANCA-associated vasculitis, atherosclerosis, thrombosis

Abstract

The putative role of neutrophils in host defense against pathogens is a well-recognized aspect of neutrophil function. The discovery of neutrophil extracellular traps has expanded the known range of neutrophil defense mechanisms and catalyzed a discipline of research focused upon ways in which neutrophils can shape the immunologic landscape of certain autoimmune diseases, including systemic lupus erythematosus. Enhanced neutrophil extracellular trap formation and impaired neutrophil extracellular trap clearance may contribute to immunogenicity in systemic lupus erythematosus and other autoimmune diseases by promoting the externalization of modified autoantigens, inducing synthesis of type I IFNs, stimulating the inflammasome, and activating both the classic and alternative pathways of the complement system. Vasculopathy is a central feature of many autoimmune diseases, and neutrophil extracellular traps may contribute directly to endothelial cell dysfunction, atherosclerotic plaque burden, and thrombosis. The elucidation of the subcellular events of neutrophil extracellular trap formation may generate novel, therapeutic strategies that target the innate immune system in autoimmune and vascular diseases.

Keywords: systemic lupus erythematosus, ANCA-associated vasculitis, atherosclerosis, thrombosis

Introduction

The objective of this review is to highlight the role of neutrophils in the pathogenesis of autoimmune diseases, with a particular focus on the potential immunogenic and vasculopathic roles that NETs may play in the development and maintenance of autoimmunity. Although increasing evidence links NETs to a wide array of autoimmune disorders and other conditions, this review will focus primarily on the potential role of NETs in the pathogenesis of SLE as an example of a prototypical autoimmune disease. Definitions of key terminology used throughout the manuscript and the relevance to NET formation are provided in Table 1.

TABLE 1.

Selected index of key terminology

TermDefinitionRelevance to NETs
Cellular events
 ApoptosisForm of programmed cell death characterized, in part, by cell shrinkage, chromatin condensation, and DNA fragmentationDistinct form of cell death different from NETosis
 NecrosisForm of premature cell death caused by external factors with resultant detrimental effectsDistinct form of cell death different from NETosis
 NETosisForm of programmed cell death characterized by release of decondensed chromatin bound to cytosolic contentsCell death related to extrusion of NETs
 Vital NETosisForm of NETosis in which the neutrophil remains intact and functional after extrusion of nuclear contentsPotentially different form of NETosis that does not result in immediate cell death
Mechanisms of NETosis
 ROSChemically reactive molecules containing oxygenPlays important role in cell signaling during NETosis
 NADPH oxidaseMembrane-bound enzyme complex activated during neutrophil respiratory burstA critical enzyme complex in NETosis
 DPIInhibitor of flavoenzymes, particularly NADPH oxidaseAbrogates NETosis in in vitro experiments
 Citrullination/deiminationConversion of positively charged, protein-bound arginine/methylarginine residues to the uncharged, nonstandard amino acid citrullineCritical step in NETosis to promote histone decondensation
 PADsA family of enzymes that catalyze citrullinationPAD4 inhibitors abrogate NETosis in animal and in vitro models
Pathways of NETosis
 AutophagyPhysiologic process whereby cell degrades unnecessary or dysfunctional componentsPathway implicated in NET formation
 AKTProtein kinase plays role in multiple cellular processes, such as apoptosis and proliferationMay direct neutrophil toward NETosis rather than apoptosis
 PI3KFamily of enzymes involved in cellular functions, such as cell growth, differentiation, and survivalInvolved in subcellular signaling in NETosis
 RAF-MEK-ERK pathwayChain of proteins in the cell involved in cell surface receptor signalingActivation of pathway is required for NET formation
Relevant animal models
 MRL/lpr miceMurine model of spontaneous lupus as a result of FAS mutation with glomerulonephritis, skin disease, autoantibody production, and vascular diseaseEnhanced rate of spontaneous NETosis/inhibition of NETs improves phenotype
 NZM2328 miceMurine model of female-predominant, spontaneous lupus with glomerulonephritis, autoantibody production, vascular disease, and strong dependence on IFNsEnhanced rate of spontaneous NETosis/inhibition of NETs improves phenotype
  ApoE/Murine model of spontaneous atherosclerosisInhibition of NETosis can improve atherosclerosis
Other relevant biology
 LDGsA subset of proinflammatory neutrophils identified in SLEProne to spontaneous NETosis in absence of stimulation
 TIIFNsGroup of proteins involved in innate immunity; overproduced in some patients with SLENET products can trigger production of TIIFNs
 DNase1An endonuclease that cleaves DNA; can be dysfunctional in patients with SLEPrimary nuclease to degrade NETs
 InflammasomeA multiprotein structure within myeloid cells; a component of the innate immune systemNETs can activate the inflammasome to produce cytokines
 Complement systemPart of the innate immune system that aids clearance of pathogensNETs can activate the classic and alternative pathways
 TFMajor initiator of physiologic coagulation and trigger of thrombosisExpressed within NETs; contributes to hypercoagulability
Neutrophil proteins
 LL-37 (CRAMP)Antimicrobial peptide found in lysosomes of neutrophilsCommon protein found within NETs; triggers TIIFNs
 ElastaseA protease enzyme found within neutrophil granular proteinsTranslocates to nucleus during NETosis
 MPOA peroxidase enzyme abundantly expressed in neutrophils with antimicrobial functionTranslocates to nucleus during NETosis
 MMPsA group of enzymes capable of degrading extracellular matrix proteinsMediates an interaction between NETs and vascular disease
 Citrullinated peptidesA group of proteins, including vimentin, that includes major autoantigens in rheumatoid arthritisProteins are found citrullinated within NETs

Neutrophils are the primary custodians of the innate immune system and play a critical role in host defense against microorganisms. Neutrophils respond to infectious insults through myriad defense mechanisms, including phagocytosis of microbes and toxic degranulation of cytoplasmic granular proteins that are microbicidal [1]. Impairments in neutrophil quantity or function render the host susceptible to a wide array of potentially life-threatening pathogens, highlighting the critical relevance of neutrophils in immune system homeostasis. Besides phagocytosis and degranulation, neutrophils stimulated by specific sterile and nonsterile stimuli can undergo a distinct form of cell death characterized by the extrusion of granular proteins bound to a meshwork of chromatin and other nuclear material [2]. These complexes of intermixed nuclear and cytoplasmic neutrophil contents released into the extracellular space have been termed NETs, and the process of NET formation as a novel form of cell death has been dubbed NETosis [3].

Despite the importance of neutrophils as agents of host defense, the current understanding of neutrophil function is superficial relative to the functional and phenotypical characterization of other immune cell populations. Part of these gaps in knowledge of neutrophil development and function reflect challenges inherent to studying these cells. Upon release from bone marrow, mature neutrophils are terminally differentiated cells that do not divide and cannot be genetically modified. In circulation, neutrophils are short-lived cells that do not readily survive freeze-thaw methods and die promptly in culture. As such, ideally, neutrophil functional studies must be carried out expeditiously on freshly isolated samples rather than samples collected and stored in existing bio-repositories. Murine neutrophil biology appears to differ critically from their human counterparts, somewhat limiting the use of mouse models of disease [4], and there are few established neutrophil cell lines [5]. Despite these limitations, the last decade has seen a revival of interest in neutrophil biology in the context of autoimmune disease.

SLE is an archetypal, idiopathic autoimmune syndrome characterized by profound clinical heterogeneity, the formation of autoantibodies to nucleic acids and associated nuclear proteins, multisystem organ involvement, unpredictable clinical course with periods of disease flare and remission, and potentially life-threatening end-organ complications. Pleiotropic abnormalities in both innate and adaptive immune responses characterize human and murine models of SLE.

A potential relationship between neutrophils and SLE was suggested >50 yr ago with the discovery of the lupus erythematosus cell phenomenon, defined as a neutrophil or macrophage that has phagocytized apoptotic nuclear material from another cell [6, 7]. Over the last decade, abnormalities in the phenotype and function of neutrophils, monocytes, macrophages, DCs, and other aspects of the innate immune system have been characterized in patients with SLE [811]. Accordingly, disease paradigms are shifting toward an increased awareness that profound abnormalities in both the innate and adaptive immune systems, triggered by genetic and environmental factors and a complex interplay between innate and adaptive immunity, likely shape disease susceptibility, pathogenesis, and outcomes in SLE.

NETs

In 2004, Brinkmann et al. [2] reported the identification of neutrophil-generated extracellular fibers, which they termed NETs. These fibers are decorated with domains containing proteins from primary, secondary, and tertiary granules. In the original descriptions, NETs were induced in activated neutrophils upon stimulation with IL-8, PMA, or LPS and were visualized trapping gram-positive and -negative bacteria and fungi by electron microscopy [2]. Time-lapse video microscopy illustrates the sequenced, morphologic events of NETosis. Minutes after activation, neutrophils undergo a series of changes, including delobulation of the nucleus, chromatin decondensation, detachment of the nuclear membrane, disintegration of granular membranes, and intermixing of the nuclear and cytoplasmic contents [2, 12]. Loss of intracellular membranes before the integrity of the plasma membrane is compromised is a defining feature of NETosis. Ejection of cellular contents through the ruptured cell membrane results in extrusion of NETs that occupy a space 10- to 15-fold bigger than the volume of cells from which they originate [13] (Fig. 1).

An external file that holds a picture, illustration, etc.
Object name is jleub5BT0615-247Rf1.jpg
Immunofluorescence microscopy demonstrating NET formation.

LDGs isolated from a patient with SLE undergo spontaneous NET formation. NETs are visualized by confocal microscopy as wispy strands of DNA decorated with neutrophil granular proteins (blue, Hoechst stain for DNA; red, MPO). Image courtesy of Carmelo Carmona-Rivera, Systemic Autoimmunity Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, U.S. National Institutes of Health.

Some of the initial, functional studies of NETs were performed in animal models of infection, where NETs are regarded to play a role in host defense via the immobilization of microorganisms and facilitation of pathogen elimination [2, 14, 15]. Many different pathogens are known to induce NETs, including gram-positive and -negative bacteria, fungi, and parasites [16]. Containment of pathogens to sites of initial infections may be an important function of NETs, and a variety of pathogens may have adapted to evade NET-mediated elimination [15]. Neutrophils from neonates are less capable of forming NETs in response to various microbial stimuli compared with neutrophils from adult subjects, which may underscore some of the enhanced susceptibility to infections observed in this age group [17]. The putative role of extracellular traps in host defense is exemplified by the conservation of these structures in various animals [18, 19], insects [20], and even plants [21].

Beyond a role in combating infectious disease, NETs have been detected in association with malignancy [22, 23], atherosclerosis [24], and autoimmune diseases. The presence of NETs has been demonstrated in synovial fluid from patients with gout and rheumatoid arthritis [25, 26], in psoriatic skin lesions [27, 28], in diabetic wounds [29], and in affected glomeruli from patients with SLE and ANCA-associated vasculitis [30, 31], indirectly suggesting that NETs play a role in immune-mediated disease.

MECHANISMS OF NET FORMATION

NETosis is considered a unique form of cell death that is morphologically distinct from apoptosis, necrosis, and other forms of cell death [27]. The mechanisms regulating NET formation and the subcellular events of NETosis have not been fully elucidated and are likely dependent on the type of triggering stimuli and the context of stimulation. There are a few cellular events proposed to be critical in NET formation, namely production of ROS, migration of neutrophil elastase and later, MPO to the nucleus, and histone modification and decondensation.

As neutrophil granular proteins are potentially harmful to the host, activation of neutrophils and release of their cellular contents are strongly regulated by production of ROS. Generation of superoxide by the NADPH oxidase enzyme complex at the phagosomal membrane, a process known as respiratory burst, is considered a critical, early step in NET formation. Patients with chronic granulomatous disease display genetically determined defects in subunits of the NADPH oxidase enzyme complex, have decreased capacity to form ROS, and have impairments in NET formation following in vitro stimulation with specific stimuli [12, 32]. Pretreatment of neutrophils with DPI, a potent inhibitor of NADPH oxidase, can abrogate NET formation in human and murine neutrophils [12, 33, 34]. However, in 1 study, lupus prone MRL/lpr mice, genetically modified to be deficient in NADPH oxidase, surprisingly developed markedly exacerbated lupus with increased renal disease and elevated and altered autoantibody profiles [35]. Additionally, loss-of-function mutations in NADPH oxidase have been associated with SLE in humans [36, 37]. These conflicting observations may reflect the fact that NADPH oxidase, although involved in NET formation, is involved in other neutrophil-related functions, such as phagocytosis, and also has anti-inflammatory roles. Therefore, pharmacologic inhibition of superoxide production may have unintended consequences and seems unlikely to be a viable therapeutic strategy in SLE. More recently, pathways of NETosis that are NADPH oxidase independent and mediated by mitochondrial ROS have been described [38]. Use of medications that target mitochondrial ROS production may perhaps constitute a potentially more selective therapeutic approach for diseases associated with enhanced NET formation.

Citrullination, also termed deimination, of histones is another important step in NET formation that occurs downstream of superoxide production. PADs are a family of 5 calcium-dependent enzymes that catalyze the process of citrullination of various target proteins [39]. PAD2 and -4 have been reported to be present in myeloid cells and can citrullinate histones [40, 41]. A PAD4-null strain of mice failed to produce NETs [42, 43]. As evidence of the potential therapeutic implications of targeting PADs, inhibition of these enzymes in 2 different lupus-prone mouse models (MRL/lpr and NZM2328) has been shown to reduce NET formation in vivo and to protect against lupus-related organ damage, including amelioration of rash, reduction of proteinuria and immune complex deposition in the kidneys, improvement of endothelial function, and alteration of circulating autoantibody profiles and complement levels [44, 45].

The Raf-MEK-ERK pathway, which has been implicated in NET formation upstream of NADPH oxidase, modulates expression of antiapoptotic proteins, suggesting that neutrophils might block apoptosis to allow NETosis [34]. Activation of protein kinase B, also known as Akt, is essential for NADPH oxidase-mediated NETosis, and Akt-specific inhibitors can direct neutrophils away from NET formation and toward apoptosis [46]. Therefore, Akt may serve as a molecular switch that regulates between an axis of NETosis and apoptosis. The autophagy pathway has also been implicated in NET formation. When neutrophils are stimulated by PMA, they develop large vacuoles that resemble autophagosomes [12]. Wortmannin, which inhibits PI3Ks and related enzymes, can block vacuolization and intracellular chromatin decondensation, thereby preventing NET formation and promoting neutrophil apoptosis [47]. Signal inhibitory receptor on leukocyte 1, a surface molecule exclusive to phagocytes, may represent a therapeutic target to inhibit NET formation by abrogating MEK-ERK pathway signaling and suppressing the formation of ROS [48]. The Src/Syk signaling pathway has been implicated in NET formation induced by β-glucan, 1 of the surface components of yeasts [49]. Whether Src/Syk signaling plays a role in NET formation in the context of autoimmune diseases is unknown. Mammalian target of rapamycin can regulate NET formation by post-transcriptional control of expression of hypoxia-inducible factor 1a, suggesting additional, potential points of molecular intervention in strategies to modify NET formation in disease [50].

Interestingly, NET formation may not always be synonymous with cell death [51]. A few groups have raised the idea that NETosis may occur in the absence of cell death [5254]. Delivery of the NET without neutrophil membrane lysis may result in an enucleated neutrophil that retains some functions of host defense [55]. This process, termed “vital NETosis,” has been described in the context of neutrophil defense against gram-positive infections and warrants further study.

DEGRADATION OF NETs

Both enhanced NET formation and impairments in degradation of NETs may contribute to neutrophil-mediated immunogenicity. Defects in the clearance of apoptotic material have been demonstrated in human SLE [56]. In a parallel observation, clearance of NETs is also seemingly altered in patients with SLE. DNase1, a serum endonuclease, is essential to degrade chromatin within NETs [57]. Rare mutations in DNase1 have been reported in association with SLE [58], and mice that lack DNase1 develop a lupus-like phenotype [59]. In vitro studies identify impairments in NET degradation in serum from a subset of patients with SLE (30–40%), and these impairments can vary over time within individual patients in association with disease activity [57, 60]. Among patients with SLE, impaired NET degradation has been associated with lupus nephritis, hypocomplementemia, production of TIIFNs, and elevated autoantibody levels [60]. Potential mechanisms for impaired DNase1 activity in SLE include the presence of DNase1 inhibitors [61] and the ability of autoantibodies to NET components to stabilize NETs from degradation [57]. rDNase has been considered a potential therapy for SLE [62]; however, a phase I study in human patients demonstrated safety but did not demonstrate improvement in serum markers of disease activity [63]. More recently, contributions of macrophages to NET clearance have been described. Macrophages actively engulf NETs opsonized by C1q, an event that is facilitated by the preprocessing of NETs by DNase1 [64]. In addition to SLE, impairments in NET degradation have been reported in association with ANCA-associated vasculitis [65], antiphospholipid antibody syndrome [66], and drug-induced vasculitis [67]. In each of these conditions, defects in clearance of NETs may allow NETs to persist longer in vivo and thereby, facilitate NET-mediated immunogenic and pathogenic effects.

LDGs AND OTHER CELL SUBSETS THAT FORM EXTRACELLULAR TRAPS

Unlike other leukocytes, subsets of neutrophils with differing function are only recently starting to be identified. One of these recently characterized neutrophil subsets has been termed LDGs, based on their identification within the PBMC fraction in density gradient preparations [68]. LDGs are present in peripheral blood from patients with SLE in association with disease activity and are characterized in vitro as proinflammatory neutrophils that are toxic to endothelial cells and display enhanced ability to synthesize TIIFN and type II IFNs and other proinflammatory cytokines [69]. Compared with normal density neutrophils, LDGs have a strikingly enhanced capacity to form NETs in the absence of any triggering stimulus in ex vivo studies [31]. These cells have also been described in ANCA-associated vasculitis, a condition that shares many phenotypic features with SLE, where LDGs correlate with disease activity and may be a useful biomarker to predict response to therapy [70]. Although LDGs have a different gene-expression profile from autologous normal density neutrophils [31], distinct cell surface markers or epigenetic markings to readily identify and differentiate these cells remain elusive [71].

Clinical Questions:

What is the extent of human diseases in which LDGs are present? Is there a way to readily distinguish and selectively target LDGs? Does quantity of LDGs serve as a biomarker to predict clinical outcomes in SLE and ANCA-associated vasculitis?

Neutrophils, eosinophils, mast cells, and basophils all contain cytoplasmic granular proteins, and each of these cell types has the capacity to form extracellular traps [27, 72, 73]. Expulsion of mitochondrial, rather than nuclear DNA, has been reported as a novel mechanism of extracellular trap formation in eosinophils and neutrophils [53, 73]. Associations between extracellular traps formed from eosinophils, mast cells, or basophils and autoimmune diseases have not been firmly established.

Clinical Question:

Do extracellular traps originating from immune cell populations other than neutrophils play a role in human disease?

IMMUNOGENIC ROLE OF NETs

Source of autoantigens

Despite the fact that neutrophils have various types of granules containing hundreds of proteins, only a limited number of proteins (20–30) have been detected within NETs [26, 74]. Many of the proteins found within NETs represent the major autoantigenic targets in rheumatologic diseases: SLE (dsDNA, histones), ANCA-associated vasculitis (MPO, PR3), and rheumatoid arthritis (vimentin, enolase, histones). Several groups have proposed a “vicious loop” theory, whereby modified proteins found within NETs contribute as antigenic targets for autoantibody and immune complex formation that can, in turn, induce further NETosis. For example, MPO and PR3, which are the major antigenic targets of ANCA, have been demonstrated within NETs isolated from patients with ANCA-associated vasculitis, and ANCA antibodies can further induce NET formation in these patients [30]. Novel autoantibodies to NET-related proteins have been described in association with various autoimmune diseases. LL-37 and MMP-9 have been demonstrated within NETs, and autoantibodies to these proteins have been identified in patients with SLE and characterized as immunogenic and vasculopathic [75, 76]. Protein cargo within NETs may be influenced directly by the nature of the inciting stimuli. In rheumatoid arthritis, disease-associated autoantibodies induced a wider array of NET-protein content compared with NET induction with proinflammatory cytokines [26]. Ultimately, detailed analysis of NET contents across different diseases may reveal differences in protein abundance and post-translational modifications that could help to explain the differing autoantibody profiles associated with various autoimmune diseases.

Interaction between NET contents and APCs in the context of autoantibody production merits investigation. In coculture experiments, myeloid DCs more avidly uploaded components of netting neutrophils compared with contents from apoptotic or necrotic neutrophils [77]. Transfer of neutrophil granular proteins was observed by confocal microscopy and was inhibited by pretreatment with DNase. Immunization with NET-loaded DCs induced ANCA formation and vasculitis in murine models genetically predisposed to autoimmunity, whereas DCs loaded with apoptotic neutrophil debris did not induce pathology. A subset of neutrophils identified around the marginal zone of the spleen is prone to spontaneous NET formation and can activate B cells in a T cell-independent manner to induce Ig class-switching and antibody production [78]. Upon direct contact with T cells, NETs can reduce activation thresholds and increase T cell responses to specific antigens [79]. These findings support the concept that the complex of neutrophil cytoplasmic proteins and nucleic acids may play an important role in NET-mediated immunogenicity.

Clinical Questions:

Does protein content within NETs differ across autoimmune diseases in association with distinct autoantibody profiles? Are novel autoantibodies targeting NET contents differentially expressed in autoimmune conditions, and what is the potential functional significance of such antibodies?

NETs and TIIFNs

Overproduction of TIIFNs or increased responses to these cytokines are a feature of SLE, and this phenomenon is considered critical for SLE pathogenesis [80]. Three groups have independently shown in SLE that NETs can activate pDCs to synthesize IFN-α in a TLR-dependent manner [31, 76, 81]. Immune complexes found in the sera of patients with SLE can stabilize DNA from extracellular degradation. These complexes, comprised of self-DNA bound to NET-related peptides, such as LL-37 or HMGB1, facilitate access of DNA to the intracellular compartments of pDCs with subsequent enhancement of IFN-α production. Autoantibodies to NET-derived self-DNA-peptide complexes can further promote transport of these complexes into pDCs and subsequently amplify production of IFN-α [76]. A similar process of NET-mediated TIIFN production has also been reported in psoriasis [28, 82] and Type 1 diabetes [83]. As an additional mechanism for NET-mediated TIIFN production, oxidized DNA present in NETs can activate the intracellular stimulator of IFN genes pathway [84]. These observations highlight a connection between neutrophils and the innate activation of pDCs and link 2 fundamental components of the pathogenesis of SLE and other autoimmune diseases.

Clinical Question:

Are amplification of TIIFN responses by NETs unique to SLE or similarly found in other related autoimmune diseases?

Other immunogenic mechanisms

Activation of the inflammasome machinery by NETs may represent another important amplifier of inflammatory pathways. Both NETs and LL-37 (a protein contained in NETs) can activate caspase-1, the central enzyme of the inflammasome, in human and murine macrophages [85]. Activation of the NLRP3 inflammasome by NETs results in the synthesis and release of active IL-1β and IL-18, and these effects are enhanced in macrophages derived from patients with SLE. This process appears to be mediated, at least in part, by LL-37 binding to P2X7R [85]. A feed-forward loop has been proposed, whereby NETs stimulate the inflammasome, triggering increased IL-18 and IL-1β production, which in turn, induces further NET formation.

NETs can activate the classic and alternative pathways of the complement system. Activation and consumption of components within the classic complement pathway are a hallmark of SLE. C1q can bind to NETs and activate complement with resultant deposition of C3b on NETs [61]. Thus, nondegraded NETs, particularly in the subset of patients with SLE who demonstrated impaired clearance of NETs, may play a role in complement consumption in SLE. Activation of the complement system via the alternative pathway rather than the classic pathway has been implicated in the pathogenesis of ANCA-associated vasculitis [86]. Serum from patients with ANCA-positive vasculitis can induce production of NETs that contain components that can activate the alternative complement pathway, including factor B and properdin [87].

NET formation in inflammatory diseases may not always have a deleterious effect. Gout is a painful crystal arthropathy caused by an acute inflammatory reaction and accumulation of neutrophils in response to MSU crystals deposited within synovial tissue. MSU crystals can induce NET formation and activate the inflammasome with resultant production of IL-1β [88, 89]. Interestingly, aggregation of NETs in gouty joints may serve an anti-inflammatory function by degrading cytokines and chemokines and by disruption of neutrophil recruitment and activation [89].

Clinical Questions:

In addition to potential deleterious effects, do NETs play a beneficial or anti-inflammatory role in certain autoimmune diseases? Does enhanced NET formation confer a protective effect against pathogens?

VASCULOPATHIC ROLE OF NETs

Toxicity to endothelial cells

Endothelial dysfunction and damage are a fairly shared finding across different systemic autoimmune diseases. Endothelial dysfunction can precede accelerated atherosclerosis in SLE and rheumatoid arthritis, and overt endothelial damage is a hallmark of systemic vasculitis [90]. Compared with normal-density neutrophils from healthy controls and patients with SLE, LDGs isolated from patients with SLE have a heightened capacity to induce endothelial cell cytotoxicity, and this toxic effect on the endothelium is abrogated by disrupting NETs with a nuclease [31]. Direct incubation of endothelial cell lines with NETs isolated from SLE LDGs promotes endothelial cell death. The mechanism by which enhanced NET formation by LDGs contributes to endothelial damage is, at least in part, mediated by MMPs [75]. MMP-9, externalized in NETs, activates MMP-2 in endothelial cells and induces a cell death program. Neutralization of MMP-9 prevents MMP-2 activation, decreases NET-mediated cytotoxicity, and restores endothelium-dependent vasorelaxation. The vasculopathic effect of NETs is supported by in vivo models. In murine lupus, use of chemical PAD inhibitors that abrogate NET formation improves endothelium-dependent vasorelaxation and endothelial progenitor cell phenotype and function [44, 45]. Whether biomarkers of NETosis correlate with measures of endothelial dysfunction in humans remains to be shown.

Clinical Question:

Can a standardized biomarker for in vivo NET formation be developed and used to predict cardiovascular events in autoimmune and vasculopathic diseases and target preventative approaches to minimize future cardiovascular events?

Atherosclerosis

Accelerated atherosclerosis, unexplained by traditional risk factors, is a feature of many rheumatologic conditions, and cardiovascular disease is a leading cause of mortality among patients with SLE and rheumatoid arthritis [91]. Several lines of evidence suggest a proatherogenic role of neutrophils and NETs. Inflammatory cells, including neutrophils and NETs, infiltrate human and murine atherosclerotic plaques. Citrullinated peptides and PAD4 are present in human atherosclerotic plaques [92]. Murine knockouts of CRAMP (the murine ortholog of LL-37) in the ApoE−/− mouse model of atherosclerosis protect these mice against atherosclerosis [93]. Likewise, antibody neutralization of HMGB1 or depletion of S100A8/S100A9, 2 neutrophil-related molecules, reduces atherosclerotic burden in ApoE−/−mice [94, 95]. Increased levels of IFN-α and autoantibodies to CRAMP/DNA complexes have been reported in murine models of atherosclerosis, and CRAMP/DNA complexes can stimulate IFN-α production in pDCs within murine atherosclerotic plaques [24], promoting proinflammatory responses in the vasculature. In the ApoE−/−mice, administration of chemical PAD inhibitors can reduce arterial-wall NET formation and IFN-α expression and significantly reduce arterial lesions, lipid-rich plaque, and prothrombotic phenotype in these mice in a TIIFN- and neutrophil-dependent manner [96]. These results support a role for NET formation in the pathogenesis of atherosclerosis through modulation of innate immune responses.

An additional proatherogenic mechanism of neutrophils was described recently in SLE [97]. Oxidation of HDL is considered a risk factor for cardiovascular disease through impairment in the function of this lipoprotein and altering macrophage cholesterol efflux. Levels of oxidized HDL are substantially higher in patients with SLE and in lupus-prone mice compared with healthy controls. Neutrophils possess the machinery to oxidize HDL via NADPH oxidase, NOS, and MPO. These enzymes are externalized within NETs and strongly promote HDL oxidation. In vivo blockade of NET formation in lupus-prone mice by PAD inhibition significantly decreased HDL oxidation. As such, NET-mediated lipoprotein modification may be an additional mechanism to promote accelerated atherosclerosis in SLE.

Clinical Question:

To what extent do markers of NETosis explain cardiovascular risk factors in autoimmune diseases independent of traditional risk factors?

Thrombosis

Many autoimmune diseases, characterized by dysregulated NET formation, including SLE and ANCA-associated vasculitis, have an associated, increased risk for thromboembolic events [98, 99]. The role of neutrophils as mediators of thrombosis is historically a controversial issue; however, accumulating evidence linking NETs with clot formation and propagation supports the involvement of neutrophils in thrombosis and suggests potential mechanisms for neutrophil-driven thrombogenicity [100]. NETs have been detected within thrombus isolated from patients with ANCA-associated vasculitis [101]. TF, a protein present in endothelial tissue and leukocytes, is the major initiator of coagulation and thrombin formation [102]. Whereas it has been controversial whether neutrophils synthesize functional TF or acquire TF produced by neighboring macrophages [103105], the presence of TF has been demonstrated consistently within NETs in many different clinical settings. NETs, isolated from serum of patients with active small vessel vasculitis or patients with sepsis, contain TF [106, 107], as do NETs found within thrombi from coronary arteries during acute myocardial infarction [108]. TF within activated neutrophils is engulfed in autophagosomes and translocated to NETs, implicating autophagy-dependent, extracellular delivery of TF [107]. NETs containing TF may provide a scaffold for fibrin deposition and platelet entrapment and activation, and in a feed-forward loop, activated platelets may trigger further NET formation [109]. Additionally, serine proteases derived from NETs can activate the coagulation cascade by degrading TF pathway inhibitor [110] and promoting the activation of factor XII [111].

Animal models suggest the potential for neutrophil-targeted therapeutics to ameliorate thrombotic disease. Neutrophil depletion attenuates thrombus formation in an animal model of DVT [111]. In 2 murine models of lupus and a murine model of atherosclerosis, inhibition of PAD enzymes blocked NET formation and delayed time to carotid artery thrombosis induced by photochemical injury [44, 45, 96]. In addition, DNase can delay thrombus development in murine models of lupus and prevent extension of thrombus in a murine model of DVT [45, 111]. Therefore, the targeting of NET formation could have therapeutic efficacy to prevent the initiation and propagation of arterial or venous thrombosis [112].

Clinical Questions:

Would inhibition of NET formation be a useful therapeutic strategy to prevent initiation and propagation of arterial or venous thrombosis? If so, would inhibition of NETs have broad efficacy in all types or specific types of thromboembolic disease?

POTENTIAL THERAPEUTICS

Medications that target key subcellular events in NET formation or that enhance NET clearance may provide much-needed novel therapeutic strategies in SLE and related autoimmune diseases. Potential therapeutic targets include inhibitors of NADPH oxidase, mitochondrial ROS production, actin cytoskeleton, and PAD enzymes. Whereas the safety of targeting NETosis as a therapeutic strategy will need to be evaluated rigorously, initial evidence from animal models and humans suggests that the targeting of PAD enzymes and other neutrophil-related targets may be effective to prevent NET formation without incurring substantial risks for treatment-related infectious sequelae [44, 45, 96, 113116].

Additionally, medications with proven therapeutic efficacy in SLE should be investigated in the context of their potential effects on NET formation. Physiologically relevant concentrations of antimalarials, a mainstay of therapy for patients with SLE, can inhibit NET formation in vitro [97]. Cyclosporine A, a medication that is efficacious, albeit infrequently used to treat SLE, has also been shown to suppress NETosis via antagonism of the calcineurin pathway [117].

Clinical Questions:

Would inhibition of specific critical subcellular events of NETosis be efficacious to treat SLE or other related autoimmune diseases, and what would be the potential infectious and other risks of such approaches? What is the role of current effective therapies in autoimmunity with regard to modulation of NETosis?

CONCLUDING REMARKS

Research focused on neutrophils, NETs, and how the innate immune system contributes to autoimmune and vascular diseases has generated novel insights into disease pathogenesis in SLE and other autoimmune conditions (Fig. 2). Given the crucial role that neutrophils play in host defense and the potentially devastating clinical consequences of neutrophil depletion, development of therapeutics that target neutrophils in autoimmune diseases may seem ill advised. However, an improved understanding in the molecular checkpoints that guide different types of neutrophil programmed cell death and elucidation of the critical mechanisms that govern pathways of NETosis may usher in a wave of novel therapeutics that target the innate immune system in autoimmune diseases. Identification of pathogenic subsets of neutrophils may facilitate the development of therapeutics that safely target specific components of the innate immune system, while persevering critical aspects of host defense (Table 2). Ongoing research conducted at the bench to fill in gaps in understanding about neutrophil development and function and well-designed clinical trials will be essential toward translating these findings effectively and safely into clinical practice.

An external file that holds a picture, illustration, etc.
Object name is jleub5BT0615-247Rf2.jpg
Overview of NETs involvement in autoimmunity and vasculopathy.

Neutrophils (PMNs) release NETs with potential immunogenic roles. Protein-DNA complexes within NETs (e.g., LL-37/DNA) activate pDCs to synthesize TIIFNs. NETs interact with B and T lymphocytes, linking the innate and adaptive immune systems and promoting their activation. NETs can activate platelets and coagulation factors and promote thrombosis. NET contents are potentially toxic to endothelial cells (EC), promoting vasculopathy and apoptosis through MMPs and histones. Several amplification loops of NETosis exist, whereby products found within NETs (autoantigens, complement activating factors, proinflammatory cytokines) or antibodies (Abs) and immune complexes (ICs) to/containing NET-related proteins stimulate neutrophils to undergo NET formation. In a feed-forward loop, NET proteins stimulate the inflammasome (NLRP3) in macrophages (Mϕ) with resultant release of proinflammatory cytokines (IL-1 and IL-18) that promote additional NET formation in granulocytes.

TABLE 2.

Summary of questions for future clinical-translational research

• What is the extent of human diseases in which LDGs are present? Is there a way to readily distinguish and selectively target LDGs? Does quantity of LDGs serve as a biomarker to predict clinical outcomes in SLE and ANCA-associated vasculitis?
• Do extracellular traps originating from immune cell populations other than neutrophils play a role in human disease?
• Does protein content within NETs differ across autoimmune diseases in association with distinct autoantibody profiles? Are novel autoantibodies targeting NET contents differentially expressed in autoimmune conditions, and what is the potential functional significance of such antibodies?
• Are amplification of TIIFN responses by NETs unique to SLE or similarly found in other related autoimmune diseases?
• In addition to potential deleterious effects, do NETs play a beneficial or anti-inflammatory role in certain autoimmune diseases? Does enhanced NET formation confer a protective effect against pathogens?
• Can a standardized biomarker for in vivo NET formation be developed and used to predict cardiovascular events in autoimmune and vasculopathic diseases and target preventative approaches to minimize future cardiovascular events?
• To what extent do markers of NETosis explain cardiovascular risk factors in autoimmune diseases independent of traditional risk factors?
• Would inhibition of NET formation be a useful therapeutic strategy to prevent initiation and propagation of arterial or venous thrombosis? If so, would inhibition of NETs have broad efficacy in all types or specific types of thromboembolic disease?
• Would inhibition of specific critical subcellular events of NETosis be efficacious to treat SLE or other related autoimmune diseases, and what would be the potential infectious and other risks of such approaches? What is the role of current effective therapies in autoimmunity with regard to modulation of NETosis?

AUTHORSHIP

P.C.G. and M.J.K. provided the study conception and design; acquisition, analysis, and interpretation of data; and drafting and review of the manuscript.

ACKNOWLEDGMENTS

This research was supported by the Intramural Research Program at the National Institute of Arthritis and Musculoskeletal and Skin Diseases, U.S. National Institutes of Health.

Glossary

AKTprotein kinase B
ANCAantineutrophil cytoplasmic antibody
ApoE−/−apolipoprotein E-deficient
CRAMPcathelicidin-related antimicrobial peptide
DCdendritic cell
DPIdiphenyleneiodonium
DVTdeep vein thrombosis
HMGB1high-mobility group box 1
LDGlow-density granulocyte
LL-37cathelicidin
MMPmatrix metalloproteinase
MPOmyeloperoxidase
MSUmonosodium urate
NETneutrophil extracellular trap
NETosisprocess of neutrophil extracellular trap formation
NLRP3nucleotide-binding oligomerization domain-like receptor family, pyrin domain containing 3
PADpeptidylarginine deiminase
pDCplasmacytoid dendritic cell
PR3proteinase 3
ROSreactive oxygen species
SLEsystemic lupus erythematosus
TFtissue factor
TIIFNtype I IFN

Footnotes

SEE CORRESPONDING ARTICLE ON PAGE 265

DISCLOSURES

The authors declare no conflicts of interest.

REFERENCES

1. Bardoel B. W., Kenny E. F., Sollberger G., Zychlinsky A. (2014) The balancing act of neutrophils. Cell Host Microbe 15, 526–536. [PubMed] [Google Scholar]
2. Brinkmann V., Reichard U., Goosmann C., Fauler B., Uhlemann Y., Weiss D. S., Weinrauch Y., Zychlinsky A. (2004) Neutrophil extracellular traps kill bacteria. Science 303, 1532–1535. [PubMed] [Google Scholar]
3. Steinberg B. E., Grinstein S. (2007) Unconventional roles of the NADPH oxidase: signaling, ion homeostasis, and cell death. Sci. STKE 2007, pe11. [PubMed] [Google Scholar]
4. Mestas J., Hughes C. C. (2004) Of mice and not men: differences between mouse and human immunology. J. Immunol. 172, 2731–2738. [PubMed] [Google Scholar]
5. Collins S. J. (1987) The HL-60 promyelocytic leukemia cell line: proliferation, differentiation, and cellular oncogene expression. Blood 70, 1233–1244. [PubMed] [Google Scholar]
6. Hargraves M. M., Richmond H., Morton R. (1948) Presentation of two bone marrow elements; the tart cell and the L.E. cell. Proc. Staff Meet. Mayo Clin. 23, 25–28. [PubMed] [Google Scholar]
7. Tsokos G. C. (2011) Systemic lupus erythematosus. N. Engl. J. Med. 365, 2110–2121. [PubMed] [Google Scholar]
8. Denny M. F., Chandaroy P., Killen P. D., Caricchio R., Lewis E. E., Richardson B. C., Lee K. D., Gavalchin J., Kaplan M. J. (2006) Accelerated macrophage apoptosis induces autoantibody formation and organ damage in systemic lupus erythematosus. J. Immunol. 176, 2095–2104. [PubMed] [Google Scholar]
9. Blanco P., Palucka A. K., Gill M., Pascual V., Banchereau J. (2001) Induction of dendritic cell differentiation by IFN-alpha in systemic lupus erythematosus. Science 294, 1540–1543. [PubMed] [Google Scholar]
10. Ding D., Mehta H., McCune W. J., Kaplan M. J. (2006) Aberrant phenotype and function of myeloid dendritic cells in systemic lupus erythematosus. J. Immunol. 177, 5878–5889. [PubMed] [Google Scholar]
11. Kaplan M. J. (2011) Neutrophils in the pathogenesis and manifestations of SLE. Nat. Rev. Rheumatol. 7, 691–699. [PMC free article] [PubMed] [Google Scholar]
12. Fuchs T. A., Abed U., Goosmann C., Hurwitz R., Schulze I., Wahn V., Weinrauch Y., Brinkmann V., Zychlinsky A. (2007) Novel cell death program leads to neutrophil extracellular traps. J. Cell Biol. 176, 231–241. [PMC free article] [PubMed] [Google Scholar]
13. Brinkmann V., Zychlinsky A. (2012) Neutrophil extracellular traps: is immunity the second function of chromatin? J. Cell Biol. 198, 773–783. [PMC free article] [PubMed] [Google Scholar]
14. Buchanan J. T., Simpson A. J., Aziz R. K., Liu G. Y., Kristian S. A., Kotb M., Feramisco J., Nizet V. (2006) DNase expression allows the pathogen group A Streptococcus to escape killing in neutrophil extracellular traps. Curr. Biol. 16, 396–400. [PubMed] [Google Scholar]
15. Beiter K., Wartha F., Albiger B., Normark S., Zychlinsky A., Henriques-Normark B. (2006) An endonuclease allows Streptococcus pneumoniae to escape from neutrophil extracellular traps. Curr. Biol. 16, 401–407. [PubMed] [Google Scholar]
16. Kaplan M. J., Radic M. (2012) Neutrophil extracellular traps: double-edged swords of innate immunity. J. Immunol. 189, 2689–2695. [PMC free article] [PubMed] [Google Scholar]
17. Yost C. C., Cody M. J., Harris E. S., Thornton N. L., McInturff A. M., Martinez M. L., Chandler N. B., Rodesch C. K., Albertine K. H., Petti C. A., Weyrich A. S., Zimmerman G. A. (2009) Impaired neutrophil extracellular trap (NET) formation: a novel innate immune deficiency of human neonates. Blood 113, 6419–6427. [PMC free article] [PubMed] [Google Scholar]
18. Ermert D., Urban C. F., Laube B., Goosmann C., Zychlinsky A., Brinkmann V. (2009) Mouse neutrophil extracellular traps in microbial infections. J. Innate Immun. 1, 181–193. [PMC free article] [PubMed] [Google Scholar]
19. Palić D., Ostojić J., Andreasen C. B., Roth J. A. (2007) Fish cast NETs: neutrophil extracellular traps are released from fish neutrophils. Dev. Comp. Immunol. 31, 805–816. [PubMed] [Google Scholar]
20. Altincicek B., Stötzel S., Wygrecka M., Preissner K. T., Vilcinskas A. (2008) Host-derived extracellular nucleic acids enhance innate immune responses, induce coagulation, and prolong survival upon infection in insects. J. Immunol. 181, 2705–2712. [PubMed] [Google Scholar]
21. Wen F., White G. J., VanEtten H. D., Xiong Z., Hawes M. C. (2009) Extracellular DNA is required for root tip resistance to fungal infection. Plant Physiol. 151, 820–829. [PMC free article] [PubMed] [Google Scholar]
22. Boone B. A., Orlichenko L., Schapiro N. E., Loughran P., Gianfrate G. C., Ellis J. T., Singhi A. D., Kang R., Tang D., Lotze M. T., Zeh H. J. (2015) The receptor for advanced glycation end products (RAGE) enhances autophagy and neutrophil extracellular traps in pancreatic cancer. Cancer Gene Ther. 22, 326–334. [PMC free article] [PubMed] [Google Scholar]
23. Demers M., Krause D. S., Schatzberg D., Martinod K., Voorhees J. R., Fuchs T. A., Scadden D. T., Wagner D. D. (2012) Cancers predispose neutrophils to release extracellular DNA traps that contribute to cancer-associated thrombosis. Proc. Natl. Acad. Sci. USA 109, 13076–13081. [PMC free article] [PubMed] [Google Scholar]
24. Döring Y., Manthey H. D., Drechsler M., Lievens D., Megens R. T., Soehnlein O., Busch M., Manca M., Koenen R. R., Pelisek J., Daemen M. J., Lutgens E., Zenke M., Binder C. J., Weber C., Zernecke A. (2012) Auto-antigenic protein-DNA complexes stimulate plasmacytoid dendritic cells to promote atherosclerosis. Circulation 125, 1673–1683. [PubMed] [Google Scholar]
25. Mitroulis I., Kambas K., Chrysanthopoulou A., Skendros P., Apostolidou E., Kourtzelis I., Drosos G. I., Boumpas D. T., Ritis K. (2011) Neutrophil extracellular trap formation is associated with IL-1β and autophagy-related signaling in gout. PLoS One 6, e29318. [PMC free article] [PubMed] [Google Scholar]
26. Khandpur R., Carmona-Rivera C., Vivekanandan-Giri A., Gizinski A., Yalavarthi S., Knight J. S., Friday S., Li S., Patel R. M., Subramanian V., Thompson P., Chen P., Fox D. A., Pennathur S., Kaplan M. J. (2013) NETs are a source of citrullinated autoantigens and stimulate inflammatory responses in rheumatoid arthritis. Sci. Transl. Med. 5, 178ra40. [PMC free article] [PubMed] [Google Scholar]
27. Lin A. M., Rubin C. J., Khandpur R., Wang J. Y., Riblett M., Yalavarthi S., Villanueva E. C., Shah P., Kaplan M. J., Bruce A. T. (2011) Mast cells and neutrophils release IL-17 through extracellular trap formation in psoriasis. J. Immunol. 187, 490–500. [PMC free article] [PubMed] [Google Scholar]
28. Skrzeczynska-Moncznik J., Wlodarczyk A., Zabieglo K., Kapinska-Mrowiecka M., Marewicz E., Dubin A., Potempa J., Cichy J. (2012) Secretory leukocyte proteinase inhibitor-competent DNA deposits are potent stimulators of plasmacytoid dendritic cells: implication for psoriasis. J. Immunol. 189, 1611–1617. [PubMed] [Google Scholar]
29. Wong S. L., Demers M., Martinod K., Gallant M., Wang Y., Goldfine A. B., Kahn C. R., Wagner D. D. (2015) Diabetes primes neutrophils to undergo NETosis, which impairs wound healing. Nat. Med. 21, 815–819. [PMC free article] [PubMed] [Google Scholar]
30. Kessenbrock K., Krumbholz M., Schönermarck U., Back W., Gross W. L., Werb Z., Gröne H. J., Brinkmann V., Jenne D. E. (2009) Netting neutrophils in autoimmune small-vessel vasculitis. Nat. Med. 15, 623–625. [PMC free article] [PubMed] [Google Scholar]
31. Villanueva E., Yalavarthi S., Berthier C. C., Hodgin J. B., Khandpur R., Lin A. M., Rubin C. J., Zhao W., Olsen S. H., Klinker M., Shealy D., Denny M. F., Plumas J., Chaperot L., Kretzler M., Bruce A. T., Kaplan M. J. (2011) Netting neutrophils induce endothelial damage, infiltrate tissues, and expose immunostimulatory molecules in systemic lupus erythematosus. J. Immunol. 187, 538–552. [PMC free article] [PubMed] [Google Scholar]
32. Bianchi M., Hakkim A., Brinkmann V., Siler U., Seger R. A., Zychlinsky A., Reichenbach J. (2009) Restoration of NET formation by gene therapy in CGD controls aspergillosis. Blood 114, 2619–2622. [PMC free article] [PubMed] [Google Scholar]
33. Palmer L. J., Cooper P. R., Ling M. R., Wright H. J., Huissoon A., Chapple I. L. (2012) Hypochlorous acid regulates neutrophil extracellular trap release in humans. Clin. Exp. Immunol. 167, 261–268. [PMC free article] [PubMed] [Google Scholar]
34. Hakkim A., Fuchs T. A., Martinez N. E., Hess S., Prinz H., Zychlinsky A., Waldmann H. (2011) Activation of the Raf-MEK-ERK pathway is required for neutrophil extracellular trap formation. Nat. Chem. Biol. 7, 75–77. [PubMed] [Google Scholar]
35. Campbell A. M., Kashgarian M., Shlomchik M. J. (2012) NADPH oxidase inhibits the pathogenesis of systemic lupus erythematosus. Sci. Transl. Med. 4, 157ra141. [PMC free article] [PubMed] [Google Scholar]
36. Manzi S., Urbach A. H., McCune A. B., Altman H. A., Kaplan S. S., Medsger T. A. Jr., Ramsey-Goldman R. (1991) Systemic lupus erythematosus in a boy with chronic granulomatous disease: case report and review of the literature. Arthritis Rheum. 34, 101–105. [PubMed] [Google Scholar]
37. Cale C. M., Morton L., Goldblatt D. (2007) Cutaneous and other lupus-like symptoms in carriers of X-linked chronic granulomatous disease: incidence and autoimmune serology. Clin. Exp. Immunol. 148, 79–84. [PMC free article] [PubMed] [Google Scholar]
38. Douda D. N., Khan M. A., Grasemann H., Palaniyar N. (2015) SK3 channel and mitochondrial ROS mediate NADPH oxidase-independent NETosis induced by calcium influx. Proc. Natl. Acad. Sci. USA 112, 2817–2822. [PMC free article] [PubMed] [Google Scholar]
39. Wang S., Wang Y. (2013) Peptidylarginine deiminases in citrullination, gene regulation, health and pathogenesis. Biochim. Biophys. Acta 1829, 1126–1135. [PMC free article] [PubMed] [Google Scholar]
40. Zhang X., Bolt M., Guertin M. J., Chen W., Zhang S., Cherrington B. D., Slade D. J., Dreyton C. J., Subramanian V., Bicker K. L., Thompson P. R., Mancini M. A., Lis J. T., Coonrod S. A. (2012) Peptidylarginine deiminase 2-catalyzed histone H3 arginine 26 citrullination facilitates estrogen receptor α target gene activation. Proc. Natl. Acad. Sci. USA 109, 13331–13336. [PMC free article] [PubMed] [Google Scholar]
41. Martinod K., Demers M., Fuchs T. A., Wong S. L., Brill A., Gallant M., Hu J., Wang Y., Wagner D. D. (2013) Neutrophil histone modification by peptidylarginine deiminase 4 is critical for deep vein thrombosis in mice. Proc. Natl. Acad. Sci. USA 110, 8674–8679. [PMC free article] [PubMed] [Google Scholar]
42. Li P., Wang D., Yao H., Doret P., Hao G., Shen Q., Qiu H., Zhang X., Wang Y., Chen G., Wang Y. (2010) Coordination of PAD4 and HDAC2 in the regulation of p53-target gene expression. Oncogene 29, 3153–3162. [PMC free article] [PubMed] [Google Scholar]
43. Hemmers S., Teijaro J. R., Arandjelovic S., Mowen K. A. (2011) PAD4-mediated neutrophil extracellular trap formation is not required for immunity against influenza infection. PLoS One 6, e22043. [PMC free article] [PubMed] [Google Scholar]
44. Knight J. S., Subramanian V., O’Dell A. A., Yalavarthi S., Zhao W., Smith C. K., Hodgin J. B., Thompson P. R., Kaplan M. J. (2014) Peptidylarginine deiminase inhibition disrupts NET formation and protects against kidney, skin and vascular disease in lupus-prone MRL/lpr mice. Ann. Rheum. Dis. [Epub ahead of print]. [PMC free article] [PubMed] [Google Scholar]
45. Knight J. S., Zhao W., Luo W., Subramanian V., O’Dell A. A., Yalavarthi S., Hodgin J. B., Eitzman D. T., Thompson P. R., Kaplan M. J. (2013) Peptidylarginine deiminase inhibition is immunomodulatory and vasculoprotective in murine lupus. J. Clin. Invest. 123, 2981–2993. [PMC free article] [PubMed] [Google Scholar]
46. Douda D. N., Yip L., Khan M. A., Grasemann H., Palaniyar N. (2014) Akt is essential to induce NADPH-dependent NETosis and to switch the neutrophil death to apoptosis. Blood 123, 597–600. [PubMed] [Google Scholar]
47. Remijsen Q., Vanden Berghe T., Wirawan E., Asselbergh B., Parthoens E., De Rycke R., Noppen S., Delforge M., Willems J., Vandenabeele P. (2011) Neutrophil extracellular trap cell death requires both autophagy and superoxide generation. Cell Res. 21, 290–304. [PMC free article] [PubMed] [Google Scholar]
48. Van Avondt K., Fritsch-Stork R., Derksen R. H., Meyaard L. (2013) Ligation of signal inhibitory receptor on leukocytes-1 suppresses the release of neutrophil extracellular traps in systemic lupus erythematosus. PLoS One 8, e78459. [PMC free article] [PubMed] [Google Scholar]
49. Nanì S., Fumagalli L., Sinha U., Kamen L., Scapini P., Berton G. (2015) Src family kinases and Syk are required for neutrophil extracellular trap formation in response to β-glucan particles. J. Innate Immun. 7, 59–73. [PMC free article] [PubMed] [Google Scholar]
50. McInturff A. M., Cody M. J., Elliott E. A., Glenn J. W., Rowley J. W., Rondina M. T., Yost C. C. (2012) Mammalian target of rapamycin regulates neutrophil extracellular trap formation via induction of hypoxia-inducible factor 1 α. Blood 120, 3118–3125. [PMC free article] [PubMed] [Google Scholar]
51. Yipp B. G., Kubes P. (2013) NETosis: how vital is it? Blood 122, 2784–2794. [PubMed] [Google Scholar]
52. Clark S. R., Ma A. C., Tavener S. A., McDonald B., Goodarzi Z., Kelly M. M., Patel K. D., Chakrabarti S., McAvoy E., Sinclair G. D., Keys E. M., Allen-Vercoe E., Devinney R., Doig C. J., Green F. H., Kubes P. (2007) Platelet TLR4 activates neutrophil extracellular traps to ensnare bacteria in septic blood. Nat. Med. 13, 463–469. [PubMed] [Google Scholar]
53. Yousefi S., Mihalache C., Kozlowski E., Schmid I., Simon H. U. (2009) Viable neutrophils release mitochondrial DNA to form neutrophil extracellular traps. Cell Death Differ. 16, 1438–1444. [PubMed] [Google Scholar]
54. Pilsczek F. H., Salina D., Poon K. K., Fahey C., Yipp B. G., Sibley C. D., Robbins S. M., Green F. H., Surette M. G., Sugai M., Bowden M. G., Hussain M., Zhang K., Kubes P. (2010) A novel mechanism of rapid nuclear neutrophil extracellular trap formation in response to Staphylococcus aureus. J. Immunol. 185, 7413–7425. [PubMed] [Google Scholar]
55. Yipp B. G., Petri B., Salina D., Jenne C. N., Scott B. N., Zbytnuik L. D., Pittman K., Asaduzzaman M., Wu K., Meijndert H. C., Malawista S. E., de Boisfleury Chevance A., Zhang K., Conly J., Kubes P. (2012) Infection-induced NETosis is a dynamic process involving neutrophil multitasking in vivo. Nat. Med. 18, 1386–1393. [PMC free article] [PubMed] [Google Scholar]
56. Shao W. H., Cohen P. L. (2011) Disturbances of apoptotic cell clearance in systemic lupus erythematosus. Arthritis Res. Ther. 13, 202. [PMC free article] [PubMed] [Google Scholar]
57. Hakkim A., Fürnrohr B. G., Amann K., Laube B., Abed U. A., Brinkmann V., Herrmann M., Voll R. E., Zychlinsky A. (2010) Impairment of neutrophil extracellular trap degradation is associated with lupus nephritis. Proc. Natl. Acad. Sci. USA 107, 9813–9818. [PMC free article] [PubMed] [Google Scholar]
58. Bodaño A., González A., Ferreiros-Vidal I., Balada E., Ordi J., Carreira P., Gómez-Reino J. J., Conde C. (2006) Association of a non-synonymous single-nucleotide polymorphism of DNASEI with SLE susceptibility. Rheumatology (Oxford) 45, 819–823. [PubMed] [Google Scholar]
59. Jacob M., Napirei M., Ricken A., Dixkens C., Mannherz H. G. (2002) Histopathology of lupus-like nephritis in Dnase1-deficient mice in comparison to NZB/W F1 mice. Lupus 11, 514–527. [PubMed] [Google Scholar]
60. Leffler J., Gullstrand B., Jönsen A., Nilsson J. A., Martin M., Blom A. M., Bengtsson A. A. (2013) Degradation of neutrophil extracellular traps co-varies with disease activity in patients with systemic lupus erythematosus. Arthritis Res. Ther. 15, R84. [PMC free article] [PubMed] [Google Scholar]
61. Leffler J., Martin M., Gullstrand B., Tydén H., Lood C., Truedsson L., Bengtsson A. A., Blom A. M. (2012) Neutrophil extracellular traps that are not degraded in systemic lupus erythematosus activate complement exacerbating the disease. J. Immunol. 188, 3522–3531. [PubMed] [Google Scholar]
62. Martínez Valle F., Balada E., Ordi-Ros J., Vilardell-Tarres M. (2008) DNase 1 and systemic lupus erythematosus. Autoimmun. Rev. 7, 359–363. [PubMed] [Google Scholar]
63. Verthelyi D., Dybdal N., Elias K. A., Klinman D. M. (1998) DNAse treatment does not improve the survival of lupus prone (NZB x NZW)F1 mice. Lupus 7, 223–230. [PubMed] [Google Scholar]
64. Farrera C., Fadeel B. (2013) Macrophage clearance of neutrophil extracellular traps is a silent process. J. Immunol. 191, 2647–2656. [PubMed] [Google Scholar]
65. Nakazawa D., Shida H., Tomaru U., Yoshida M., Nishio S., Atsumi T., Ishizu A. (2014) Enhanced formation and disordered regulation of NETs in myeloperoxidase-ANCA-associated microscopic polyangiitis. J. Am. Soc. Nephrol. 25, 990–997. [PMC free article] [PubMed] [Google Scholar]
66. Leffler J., Stojanovich L., Shoenfeld Y., Bogdanovic G., Hesselstrand R., Blom A. M. (2014) Degradation of neutrophil extracellular traps is decreased in patients with antiphospholipid syndrome. Clin. Exp. Rheumatol. 32, 66–70. [PubMed] [Google Scholar]
67. Nakazawa D., Tomaru U., Suzuki A., Masuda S., Hasegawa R., Kobayashi T., Nishio S., Kasahara M., Ishizu A. (2012) 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. 64, 3779–3787. [PubMed] [Google Scholar]
68. Carmona-Rivera C., Kaplan M. J. (2013) Low-density granulocytes: a distinct class of neutrophils in systemic autoimmunity. Semin. Immunopathol. 35, 455–463. [PMC free article] [PubMed] [Google Scholar]
69. Denny M. F., Yalavarthi S., Zhao W., Thacker S. G., Anderson M., Sandy A. R., McCune W. J., Kaplan M. J. (2010) A distinct subset of proinflammatory neutrophils isolated from patients with systemic lupus erythematosus induces vascular damage and synthesizes type I IFNs. J. Immunol. 184, 3284–3297. [PMC free article] [PubMed] [Google Scholar]
70. Grayson P. C., Carmona-Rivera C., Xu L., Lim N., Gao Z., Asare A. L., Specks U., Stone J. H., Seo P., Spiera R. F., Langford C. A., Hoffman G. S., Kallenberg C. G., St Clair E. W., Tchao N. K., Ytterberg S. R., Phippard D. J., Merkel P. A., Kaplan M. J., Monach P. A., Rituximab in ANCA-Associated Vasculitis-Immune Tolerance Network Research Group (2015) Neutrophil-related gene expression and low-density granulocytes associated with disease activity and response to treatment in ANCA-associated vasculitis. Arthritis Rheumatol. 67, 1922–1932. [PMC free article] [PubMed] [Google Scholar]
71. Coit P., Yalavarthi S., Ognenovski M., Zhao W., Hasni S., Wren J. D., Kaplan M. J., Sawalha A. H. (2015) Epigenome profiling reveals significant DNA demethylation of interferon signature genes in lupus neutrophils. J. Autoimmun. 58, 59–66. [PMC free article] [PubMed] [Google Scholar]
72. Morshed M., Hlushchuk R., Simon D., Walls A. F., Obata-Ninomiya K., Karasuyama H., Djonov V., Eggel A., Kaufmann T., Simon H. U., Yousefi S. (2014) NADPH oxidase-independent formation of extracellular DNA traps by basophils. J. Immunol. 192, 5314–5323. [PubMed] [Google Scholar]
73. Yousefi S., Gold J. A., Andina N., Lee J. J., Kelly A. M., Kozlowski E., Schmid I., Straumann A., Reichenbach J., Gleich G. J., Simon H. U. (2008) Catapult-like release of mitochondrial DNA by eosinophils contributes to antibacterial defense. Nat. Med. 14, 949–953. [PubMed] [Google Scholar]
74. Urban C. F., Ermert D., Schmid M., Abu-Abed U., Goosmann C., Nacken W., Brinkmann V., Jungblut P. R., Zychlinsky A. (2009) Neutrophil extracellular traps contain calprotectin, a cytosolic protein complex involved in host defense against Candida albicans. PLoS Pathog. 5, e1000639. [PMC free article] [PubMed] [Google Scholar]
75. Carmona-Rivera C., Zhao W., Yalavarthi S., Kaplan M. J. (2014) Neutrophil extracellular traps induce endothelial dysfunction in systemic lupus erythematosus through the activation of matrix metalloproteinase-2. Ann. Rheum. Dis. 74, 1417–1424. [PMC free article] [PubMed] [Google Scholar]
76. Lande R., Ganguly D., Facchinetti V., Frasca L., Conrad C., Gregorio J., Meller S., Chamilos G., Sebasigari R., Riccieri V., Bassett R., Amuro H., Fukuhara S., Ito T., Liu Y. J., Gilliet M. (2011) Neutrophils activate plasmacytoid dendritic cells by releasing self-DNA-peptide complexes in systemic lupus erythematosus. Sci. Transl. Med. 3, 73ra19. [PMC free article] [PubMed] [Google Scholar]
77. Sangaletti S., Tripodo C., Chiodoni C., Guarnotta C., Cappetti B., Casalini P., Piconese S., Parenza M., Guiducci C., Vitali C., Colombo M. P. (2012) Neutrophil extracellular traps mediate transfer of cytoplasmic neutrophil antigens to myeloid dendritic cells toward ANCA induction and associated autoimmunity. Blood 120, 3007–3018. [PubMed] [Google Scholar]
78. Puga I., Cols M., Barra C. M., He B., Cassis L., Gentile M., Comerma L., Chorny A., Shan M., Xu W., Magri G., Knowles D. M., Tam W., Chiu A., Bussel J. B., Serrano S., Lorente J. A., Bellosillo B., Lloreta J., Juanpere N., Alameda F., Baró T., de Heredia C. D., Torán N., Català A., Torrebadell M., Fortuny C., Cusí V., Carreras C., Diaz G. A., Blander J. M., Farber C. M., Silvestri G., Cunningham-Rundles C., Calvillo M., Dufour C., Notarangelo L. D., Lougaris V., Plebani A., Casanova J. L., Ganal S. C., Diefenbach A., Aróstegui J. I., Juan M., Yagüe J., Mahlaoui N., Donadieu J., Chen K., Cerutti A. (2012) B cell-helper neutrophils stimulate the diversification and production of immunoglobulin in the marginal zone of the spleen. Nat. Immunol. 13, 170–180. [PMC free article] [PubMed] [Google Scholar]
79. Tillack K., Breiden P., Martin R., Sospedra M. (2012) T lymphocyte priming by neutrophil extracellular traps links innate and adaptive immune responses. J. Immunol. 188, 3150–3159. [PubMed] [Google Scholar]
80. Banchereau J., Pascual V. (2006) Type I interferon in systemic lupus erythematosus and other autoimmune diseases. Immunity 25, 383–392. [PubMed] [Google Scholar]
81. Garcia-Romo G. S., Caielli S., Vega B., Connolly J., Allantaz F., Xu Z., Punaro M., Baisch J., Guiducci C., Coffman R. L., Barrat F. J., Banchereau J., Pascual V. (2011) Netting neutrophils are major inducers of type I IFN production in pediatric systemic lupus erythematosus. Sci. Transl. Med. 3, 73ra20. [PMC free article] [PubMed] [Google Scholar]
82. Skrzeczynska-Moncznik J., Wlodarczyk A., Banas M., Kwitniewski M., Zabieglo K., Kapinska-Mrowiecka M., Dubin A., Cichy J. (2013) DNA structures decorated with cathepsin G/secretory leukocyte proteinase inhibitor stimulate IFNI production by plasmacytoid dendritic cells. Am. J. Clin. Exp. Immunol. 2, 186–194. [PMC free article] [PubMed] [Google Scholar]
83. Diana J., Simoni Y., Furio L., Beaudoin L., Agerberth B., Barrat F., Lehuen A. (2013) Crosstalk between neutrophils, B-1a cells and plasmacytoid dendritic cells initiates autoimmune diabetes. Nat. Med. 19, 65–73. [PubMed] [Google Scholar]
84. Gehrke N., Mertens C., Zillinger T., Wenzel J., Bald T., Zahn S., Tüting T., Hartmann G., Barchet W. (2013) Oxidative damage of DNA confers resistance to cytosolic nuclease TREX1 degradation and potentiates STING-dependent immune sensing. Immunity 39, 482–495. [PubMed] [Google Scholar]
85. Kahlenberg J. M., Carmona-Rivera C., Smith C. K., Kaplan M. J. (2013) Neutrophil extracellular trap-associated protein activation of the NLRP3 inflammasome is enhanced in lupus macrophages. J. Immunol. 190, 1217–1226. [PMC free article] [PubMed] [Google Scholar]
86. Xiao H., Schreiber A., Heeringa P., Falk R. J., Jennette J. C. (2007) Alternative complement pathway in the pathogenesis of disease mediated by anti-neutrophil cytoplasmic autoantibodies. Am. J. Pathol. 170, 52–64. [PMC free article] [PubMed] [Google Scholar]
87. Wang H., Wang C., Zhao M. H., Chen M. (2015) Neutrophil extracellular traps can activate alternative complement pathways. Clin. Exp. Immunol. 181, 518–527. [PMC free article] [PubMed] [Google Scholar]
88. Martinon F., Pétrilli V., Mayor A., Tardivel A., Tschopp J. (2006) Gout-associated uric acid crystals activate the NALP3 inflammasome. Nature 440, 237–241. [PubMed] [Google Scholar]
89. Schauer C., Janko C., Munoz L. E., Zhao Y., Kienhöfer D., Frey B., Lell M., Manger B., Rech J., Naschberger E., Holmdahl R., Krenn V., Harrer T., Jeremic I., Bilyy R., Schett G., Hoffmann M., Herrmann M. (2014) Aggregated neutrophil extracellular traps limit inflammation by degrading cytokines and chemokines. Nat. Med. 20, 511–517. [PubMed] [Google Scholar]
90. Knight J. S., Kaplan M. J. (2013) Cardiovascular disease in lupus: insights and updates. Curr. Opin. Rheumatol. 25, 597–605. [PMC free article] [PubMed] [Google Scholar]
91. Abou-Raya A., Abou-Raya S. (2006) Inflammation: a pivotal link between autoimmune diseases and atherosclerosis. Autoimmun. Rev. 5, 331–337. [PubMed] [Google Scholar]
92. Sokolove J., Brennan M. J., Sharpe O., Lahey L. J., Kao A. H., Krishnan E., Edmundowicz D., Lepus C. M., Wasko M. C., Robinson W. H. (2013) Brief report: citrullination within the atherosclerotic plaque: a potential target for the anti-citrullinated protein antibody response in rheumatoid arthritis. Arthritis Rheum. 65, 1719–1724. [PMC free article] [PubMed] [Google Scholar]
93. Döring Y., Drechsler M., Wantha S., Kemmerich K., Lievens D., Vijayan S., Gallo R. L., Weber C., Soehnlein O. (2012) Lack of neutrophil-derived CRAMP reduces atherosclerosis in mice. Circ. Res. 110, 1052–1056. [PubMed] [Google Scholar]
94. Kanellakis P., Agrotis A., Kyaw T. S., Koulis C., Ahrens I., Mori S., Takahashi H. K., Liu K., Peter K., Nishibori M., Bobik A. (2011) High-mobility group box protein 1 neutralization reduces development of diet-induced atherosclerosis in apolipoprotein e-deficient mice. Arterioscler. Thromb. Vasc. Biol. 31, 313–319. [PubMed] [Google Scholar]
95. Croce K., Gao H., Wang Y., Mooroka T., Sakuma M., Shi C., Sukhova G. K., Packard R. R., Hogg N., Libby P., Simon D. I. (2009) Myeloid-related protein-8/14 is critical for the biological response to vascular injury. Circulation 120, 427–436. [PMC free article] [PubMed] [Google Scholar]
96. Knight J. S., Luo W., O’Dell A. A., Yalavarthi S., Zhao W., Subramanian V., Guo C., Grenn R. C., Thompson P. R., Eitzman D. T., Kaplan M. J. (2014) Peptidylarginine deiminase inhibition reduces vascular damage and modulates innate immune responses in murine models of atherosclerosis. Circ. Res. 114, 947–956. [PMC free article] [PubMed] [Google Scholar]
97. Smith C. K., Vivekanandan-Giri A., Tang C., Knight J. S., Mathew A., Padilla R. L., Gillespie B. W., Carmona-Rivera C., Liu X., Subramanian V., Hasni S., Thompson P. R., Heinecke J. W., Saran R., Pennathur S., Kaplan M. J. (2014) Neutrophil extracellular trap-derived enzymes oxidize high-density lipoprotein: an additional proatherogenic mechanism in systemic lupus erythematosus. Arthritis Rheumatol. 66, 2532–2544. [PMC free article] [PubMed] [Google Scholar]
98. Tektonidou M. G., Laskari K., Panagiotakos D. B., Moutsopoulos H. M. (2009) Risk factors for thrombosis and primary thrombosis prevention in patients with systemic lupus erythematosus with or without antiphospholipid antibodies. Arthritis Rheum. 61, 29–36. [PubMed] [Google Scholar]
99. Merkel P. A., Lo G. H., Holbrook J. T., Tibbs A. K., Allen N. B., Davis J. C. Jr., Hoffman G. S., McCune W. J., St Clair E. W., Specks U., Spiera R., Petri M., Stone J. H.; Wegener’s Granulomatosis Etanercept Trial Research Group (2005) 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. 142, 620–626. [PubMed] [Google Scholar]
100. Kambas K., Mitroulis I., Ritis K. (2012) The emerging role of neutrophils in thrombosis-the journey of TF through NETs. Front. Immunol. 3, 385. [PMC free article] [PubMed] [Google Scholar]
101. Nakazawa D., Tomaru U., Yamamoto C., Jodo S., Ishizu A. (2012) Abundant neutrophil extracellular traps in thrombus of patient with microscopic polyangiitis. Front. Immunol. 3, 333. [PMC free article] [PubMed] [Google Scholar]
102. Manly D. A., Boles J., Mackman N. (2011) Role of tissue factor in venous thrombosis. Annu. Rev. Physiol. 73, 515–525. [PMC free article] [PubMed] [Google Scholar]
103. Østerud B. (2004) Tissue factor in neutrophils: no. J. Thromb. Haemost. 2, 218–220. [PubMed] [Google Scholar]
104. Egorina E. M., Sovershaev M. A., Olsen J. O., Østerud B. (2008) Granulocytes do not express but acquire monocyte-derived tissue factor in whole blood: evidence for a direct transfer. Blood 111, 1208–1216. [PubMed] [Google Scholar]
105. Maugeri N., Brambilla M., Camera M., Carbone A., Tremoli E., Donati M. B., de Gaetano G., Cerletti C. (2006) Human polymorphonuclear leukocytes produce and express functional tissue factor upon stimulation. J. Thromb. Haemost. 4, 1323–1330. [PubMed] [Google Scholar]
106. Kambas K., Chrysanthopoulou A., Vassilopoulos D., Apostolidou E., Skendros P., Girod A., Arelaki S., Froudarakis M., Nakopoulou L., Giatromanolaki A., Sidiropoulos P., Koffa M., Boumpas D. T., Ritis K., Mitroulis I. (2014) 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. 73, 1854–1863. [PubMed] [Google Scholar]
107. Kambas K., Mitroulis I., Apostolidou E., Girod A., Chrysanthopoulou A., Pneumatikos I., Skendros P., Kourtzelis I., Koffa M., Kotsianidis I., Ritis K. (2012) Autophagy mediates the delivery of thrombogenic tissue factor to neutrophil extracellular traps in human sepsis. PLoS One 7, e45427. [PMC free article] [PubMed] [Google Scholar]
108. Stakos D. A., Kambas K., Konstantinidis T., Mitroulis I., Apostolidou E., Arelaki S., Tsironidou V., Giatromanolaki A., Skendros P., Konstantinides S., Ritis K. (2015) Expression of functional tissue factor by neutrophil extracellular traps in culprit artery of acute myocardial infarction. Eur. Heart J. 36, 1405–1414. [PMC free article] [PubMed] [Google Scholar]
109. Fuchs T. A., Brill A., Duerschmied D., Schatzberg D., Monestier M., Myers D. D. Jr., Wrobleski S. K., Wakefield T. W., Hartwig J. H., Wagner D. D. (2010) Extracellular DNA traps promote thrombosis. Proc. Natl. Acad. Sci. USA 107, 15880–15885. [PMC free article] [PubMed] [Google Scholar]
110. Massberg S., Grahl L., von Bruehl M. L., Manukyan D., Pfeiler S., Goosmann C., Brinkmann V., Lorenz M., Bidzhekov K., Khandagale A. B., Konrad I., Kennerknecht E., Reges K., Holdenrieder S., Braun S., Reinhardt C., Spannagl M., Preissner K. T., Engelmann B. (2010) Reciprocal coupling of coagulation and innate immunity via neutrophil serine proteases. Nat. Med. 16, 887–896. [PubMed] [Google Scholar]
111. Von Brühl M. L., Stark K., Steinhart A., Chandraratne S., Konrad I., Lorenz M., Khandoga A., Tirniceriu A., Coletti R., Köllnberger M., Byrne R. A., Laitinen I., Walch A., Brill A., Pfeiler S., Manukyan D., Braun S., Lange P., Riegger J., Ware J., Eckart A., Haidari S., Rudelius M., Schulz C., Echtler K., Brinkmann V., Schwaiger M., Preissner K. T., Wagner D. D., Mackman N., Engelmann B., Massberg S. (2012) Monocytes, neutrophils, and platelets cooperate to initiate and propagate venous thrombosis in mice in vivo. J. Exp. Med. 209, 819–835. [PMC free article] [PubMed] [Google Scholar]
112. Martinod K., Wagner D. D. (2014) Thrombosis: tangled up in NETs. Blood 123, 2768–2776. [PMC free article] [PubMed] [Google Scholar]
113. Martinod K., Fuchs T. A., Zitomersky N. L., Wong S. L., Demers M., Gallant M., Wang Y., Wagner D. D. (2015) PAD4-deficiency does not affect bacteremia in polymicrobial sepsis and ameliorates endotoxemic shock. Blood 125, 1948–1956. [PMC free article] [PubMed] [Google Scholar]
114. Li P., Li M., Lindberg M. R., Kennett M. J., Xiong N., Wang Y. (2010) PAD4 is essential for antibacterial innate immunity mediated by neutrophil extracellular traps. J. Exp. Med. 207, 1853–1862. [PMC free article] [PubMed] [Google Scholar]
115. Sulem P., Helgason H., Oddson A., Stefansson H., Gudjonsson S. A., Zink F., Hjartarson E., Sigurdsson G. T., Jonasdottir A., Jonasdottir A., Sigurdsson A., Magnusson O. T., Kong A., Helgason A., Holm H., Thorsteinsdottir U., Masson G., Gudbjartsson D. F., Stefansson K. (2015) Identification of a large set of rare complete human knockouts. Nat. Genet. 47, 448–452. [PubMed] [Google Scholar]
116. Sørensen O. E., Clemmensen S. N., Dahl S. L., Østergaard O., Heegaard N. H., Glenthøj A., Nielsen F. C., Borregaard N. (2014) Papillon-Lefèvre syndrome patient reveals species-dependent requirements for neutrophil defenses. J. Clin. Invest. 124, 4539–4548. [PMC free article] [PubMed] [Google Scholar]
117. Gupta A. K., Giaglis S., Hasler P., Hahn S. (2014) Efficient neutrophil extracellular trap induction requires mobilization of both intracellular and extracellular calcium pools and is modulated by cyclosporine A. PLoS One 9, e97088. [PMC free article] [PubMed] [Google Scholar]

Articles from Journal of Leukocyte Biology are provided here courtesy of The Society for Leukocyte Biology

-