Next Article in Journal
Nitrogen Deficiency-Induced Decrease in Cytokinins Content Promotes Rice Seminal Root Growth by Promoting Root Meristem Cell Proliferation and Cell Elongation
Next Article in Special Issue
Neutrophils and Neutrophil Extracellular Traps Drive Necroinflammation in COVID-19
Previous Article in Journal
Inflammation Alters the Secretome and Immunomodulatory Properties of Human Skin-Derived Precursor Cells
Previous Article in Special Issue
Neutrophil Chemotaxis and NETosis in Murine Chronic Liver Injury via Cannabinoid Receptor 1/Gαi/o/ROS/p38 MAPK Signaling Pathway
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Neutrophil Extracellular Traps (NETs) Take the Central Stage in Driving Autoimmune Responses

Department of Rheumatology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
*
Author to whom correspondence should be addressed.
Cells 2020, 9(4), 915; https://doi.org/10.3390/cells9040915
Submission received: 16 March 2020 / Revised: 3 April 2020 / Accepted: 5 April 2020 / Published: 8 April 2020

Abstract

:
Following fifteen years of research, neutrophil extracellular traps (NETs) are widely reported in a large range of inflammatory infectious and non-infectious diseases. Cumulating evidences from in vitro, in vivo and clinical diagnostics suggest that NETs may play a crucial role in inflammation and autoimmunity in a variety of autoimmune diseases, such as rheumatoid arthritis (RA), systemic lupus erythematosus (SLE) and anti-neutrophil cytoplasmic antibodies (ANCA)-associated vasculitis (AAV). Most likely, NETs contribute to breaking self-tolerance in autoimmune diseases in several ways. During this review, we discuss the current knowledge on how NETs could drive autoimmune responses. NETs can break self-tolerance by being a source of autoantigens for autoantibodies found in autoimmune diseases, such as anti-citrullinated protein antibodies (ACPAs) in RA, anti-dsDNA in SLE and anti-myeloperoxidase and anti-protein 3 in AAV. Moreover, NET components could accelerate the inflammatory response by mediating complement activation, acting as danger-associated molecular patterns (DAMPs) and inflammasome activators, for example. NETs also can activate other immune cells, such as B cells, antigen-presenting cells and T cells. Additionally, impaired clearance of NETs in autoimmune diseases prolongs the presence of active NETs and their components and, in this way, accelerate immune responses. NETs have not only been implicated as drivers of inflammation, but also are linked to resolution of inflammation. Therefore, NETs may be central regulators of inflammation and autoimmunity, serve as biomarkers, as well as promising targets for future therapeutics of inflammatory autoimmune diseases.

1. Introduction

Known as one of the first responder cells of the innate immune system, neutrophils are described as phagocytes in textbooks that are involved in initial early host-defence responses during infection/injury. However, the discovery of neutrophil extracellular traps (NETs) has shifted the paradigm of our current understanding of neutrophil functions, and their significance during immune responses, quite drastically. Upon interaction with an invading microbe/cytokine, neutrophils release their chromatin material together with a wide range of granular enzymes to form net-like structures known as NETs [1]. NETs cannot only trap the invading pathogen but also degrade them with NET-associated proteolytic enzymes [1]. NETs are involved in numerous infectious/non-infectious diseases and are believed to be crucially involved during inflammation. While NETs are beneficial during infections, they may play a detrimental role in the case of inflammation, autoimmunity and other pathophysiological conditions. NETs accelerate the inflammatory processes by releasing a wide range of active molecules like danger associated molecular patterns (DAMPs), histones, as well as active lytic-enzymes in extracellular space, leading to further immune responses. NETs, therefore, also may serve as a potential source of auto-antigens against which the autoantibodies associated with a wide range of inflammatory autoimmune diseases are directed.
The functions and morphology of neutrophils undergo radical transformation during inflammation, injury and infection. Neutrophils migrate along vesicles by expressing a wide range of migratory protein cascades as well as start to express various pattern recognition receptors and secrete a wide range of cytokines in a process called ‘neutrophil activation’. Over the years, it has become clearer that only a fraction of neutrophils can make NETs, indicating the heterogeneity of the neutrophil population, especially during sterile inflammation [2,3] Therefore, it is important to speculate if only a specific subpopulation of neutrophils can undergo NET formation [2,4]. A distinct population of low-density neutrophils, for example, are known to be more vulnerable towards NET formation in systemic lupus erythematosus (SLE) patients [3,5], possibly explaining a link between this disease and NET formation. Interestingly, the composition of NETs may differ based on the stimuli and, therefore, the disease with which it is associated [6]. Furthermore, in certain situations, NETs also might have anti-inflammatory characteristics [7]. It is, therefore, important to characterize NETs in a disease-specific manner to understand their specific involvement during the development of autoimmunity and disease.

2. Composition of Neutrophil Extracellular Traps (NETs)

Neutrophil extracellular traps (NETs) formation can be triggered by a wide range of stimuli in vitro and in vivo during various pathophysiological conditions [6,8]. The protein cargo of NETs induced by different stimuli is heterogenous, making comparing research and drawing conclusions challenging. Due to this, there is an ongoing discussion about the precise mechanisms involved in NET formation, their composition and, thereby, their functional profile specifically their inflammatory/antimicrobial properties [6,9,10]. Recently, there have been new insights about how molecular mechanisms of NET formation may differ in a species specific manner [11,12] but, also based on the location of neutrophils in the blood stream or tissue, as well as local environmental alkaline or oxygen conditions [13]. Therefore, in the context of autoimmune diseases, detailed proteomic analysis of disease-specific NET protein composition (NETome) has the potential to elucidate novel mechanisms of disease onset and progression. The presence of DNase1 inhibitors in SLE)-associated NETs could potentially be demonstrated to lead to impairment of NET degradation [14]. Although disease-specific NETs may have different pathological roles, Chapman et al., recently compared the protein composition of SLE and rheumatoid arthritis (RA) NETs induced by the same stimulant, and showed that only a small number of NET proteins were significantly different between the two diseases [10]. Upon phorbol myristate acetate (PMA) stimulation, RNASE2 was higher in RA NETs, whereas myeloperoxidase (MPO), leukocyte elastase inhibitor and thymidine phosphorylase (TYMP) were higher in SLE NETs. Conversely, NETome comparison between NOX2-dependent (PMA) and NOX2-independent (A23187) showed more distinct protein profiles, irrespective of the disease background of the neutrophils [10]. PMA-induced NETs, for example, were decorated with the annexin proteins azurocidin and histone H3, whereas A23287-induced NETs contained more granule proteins, such as cathelicidin antimicrobial peptides CAMP/LL37 and matrix metalloproteinase-8 (MMP8) [10]. This finding indicates that the in vitro stimulant might be of more influence on the NETome compared to the phenotype of neutrophils. Another study, however, reported the presence of several proteins that varied between PMA–NETs of SLE, lupus nephritis (LN) and healthy neutrophils [15]. LN patient-derived PMA–NETs presented a high expression of α-enolase and annexin A1, compared to SLE PMA–NETs and healthy neutrophils. Accompanying these contradicting findings, however, it is important to take into account that most of these in vitro NETome studies are performed using non-physiological stimuli such as PMA or calcium ionophore A23187 [10,15]. Physiological stimulants might affect protein composition of NETs differently. More research and more critically designed experiments are needed in this direction to understand the role of NETs and the stimuli required to form them during autoimmune responses.

3. Contribution of Neutrophil Extracellular Traps (NETs) to Autoimmunity

Since neutrophils and NETs are present in abundance at the inflammatory sites of various autoimmune diseases, their active involvement in driving autoimmune responses is plausible. Recent evidences from various research groups indeed suggest that (the impaired clearance of) NETs themselves, as well as interactions of NETs with other immune cells could be very crucial for the development of ‘autoimmunity’ and breaking of self-tolerance (Figure 1). These various aspects associating NETs to autoimmunity are discussed in detail below.

3.1. Neutrophil Extracellular Traps (NETs) as a Source of Autoantigens

Various neutrophil granular enzymes, together with decondensed chromatin, are present on NETs besides the anti-microbial proteins captured by them. Nonetheless, many of these NET-associated proteins are of “autoantigenic nature” in various rheumatologic diseases (Table 1). Examples include NET-associated MPO and proteinase 3 (PR3) enzymes as major autoantigenic targets of anti-neutrophil cytoplasmic antibody (ANCA)-associated vasculitis (AAV); NET-derived extracellular nucleic acids and dsDNA as the targets of SLE autoantibodies; citrullinated proteins namely citrullinated histones as neoepitopes for anti-citrullinated protein antibodies (ACPA) in RA [16,17,18]. These NET-associated autoantigens and their contribution are discussed in a disease-specific manner below. It is noteworthy, despite numerous evidences, that the exact mechanisms of how exactly NET autoantigens drive autoimmunity are still in a pre-mature state. There are several missing links and a lack of knowledge so further research is required to define common mechanisms of NET-derived autoimmune responses. Evidences from different in vitro and in vivo studies currently support a theory of what we propose to be a ‘vicious loop of inflammation and autoimmunity’. It implies that inflammation-derived NET-components could be a source of autoantigens for autoantibody production. These autoantibodies and associated immune complexes in return may further induce NET formation, leading to a self-amplifying loop of autoimmune-inflammation [19].

3.2. NETs as Complement Activators

Complement activation and consumption is a hallmark of SLE [54]. Lower levels of complement factors C3 and C4, indicating classic complement consumption, was found in sera of a subset of SLE patients with impaired NETs clearance [26]. Seen in AAV, on the other hand, activation of the alternative complement pathway by NETs was demonstrated [25,55]. Tumour necrosis factor alpha (TNF-α)-primed neutrophils stimulated with ANCA lead to C3a, C5a and C5b-9 generation and could be due to granular protein properdin present on NETs [25]. Also, neutrophils stimulated with an anti-MPO antibody generated complement factors C5a and C3d, but not C1q [55]. This indicates that NETs are involved in different complement pathways in specific autoimmune diseases.

3.3. NETs as Damage-Associated Molecular Patterns (DAMPs)

NET-derived products could serve as damage-associated molecular patterns (DAMPs) to initiate further inflammatory response. Particularly, the presence of extracellular histones is perceived as a DAMP [56]. Other DAMPs secreted with NETs are the high-mobility group box 1 (HMGB1) and LL37. NET-derived HMGB1 is associated with SLE [36,37], but not with AAV [36]. Contrarily, HMGB1 contributed to ANCA-induced NET formation [57]. How NET-associated DAMPs may further contribute to tissue injury and inflammation needs to be investigated in detail for autoimmune-inflammatory diseases.

3.4. NETs as Inflammasome Activators

Inflammasomes are key players in the activation of inflammatory responses and NETs can mediate their activation. PMA–NETs activate caspase1 leading to nucleotide-binding oligomerization domain-like receptor protein 3 (NLRP3) inflammasome activation in macrophages of SLE patients [58]. Activation of inflammasome led to secretion of interleukin (IL)-18 and IL-1β. Besides being pro-inflammatory, these cytokines also can induce further NET formation. Adult-onset Still’s disease neutrophils, stimulated with PMA, also were found to activate NLRP3 inflammasome through caspase1 [59]. NET formation itself was found to be dependent on noncanonical inflammasome activation, as extrusion of NETs by neutrophils exposed to lipopolysaccharide (LPS) or Gram-negative bacteria relied on activated caspase11 and activated gasdermin D (GSDMD) [60,61]. Together, this implies that NET formation may be both dependent on inflammasome activation and also be able to activate inflammasome.

4. Impaired Clearance of Neutrophil Extracellular Traps (NETs)

NETs contain various active enzymes and DAMPs such as extracellular histones [10]. It is, therefore, important to degrade the NETs by phagocytic cells like macrophages [62]. DNase1 plays an important role to degrade NETs in physiology [14]. After NETs are pre-processed by DNase1, macrophages can ingest NETs for degradation [62]. PMA–NET degradation by macrophages takes place in lysosomes [63]. However, PMA–NET degradation in macrophages also occurs in the cytosol by three prime repair exonuclease (TREX1) enzymes (or DNaseIII), as well as extracellularly by DNase1L3 secreted by dendritic cells (DCs) [62]. PMA–NET clearance by healthy donor macrophages has been described as immunologically silent and without secretion of proinflammatory cytokines [63].
Interestingly, degradation of NETs is found to be impaired in several autoimmune diseases, such as SLE [14,26,64] and AAV [65]. Hakkim et al., reported that reduced DNase1 activity in a subset of SLE patients dysregulated clearance of NETs [14]. Furthermore, SLE-associated NETs activate complement, leading to deposition of C1q on NETs which further inhibits DNase1 activity [26]. A mutation in the DNase1L3 gene also is associated with a familiar form of SLE [66]. Additionally, PMA–NET uptake by monocyte-derived macrophages [67] and LPS–NETs cleared by SLE macrophages [68] led to a pro-inflammatory response, suggesting that clearance of activated NETs in autoimmune diseases also may amplify the inflammatory response. Compromised NET clearance generates the accumulation of active-NETs at inflammatory sites; leading to more inflammation and prolonged presence of NET autoantigens [69]. This could possibly break self-tolerance and, thereby, intensify the existing autoimmune response.

5. Interplay of Neutrophil Extracellular Traps (NETs) and Other Adaptive Immune Cells

5.1. NET and B Cells

NET formation by splenic neutrophils, but not circulating neutrophils, has been found to induce immunoglobulin class switching, hypermutation and secretion by activating c [70]. Also, type I interferons (IFN) α and B cell factors produced by bone marrow neutrophils have been shown to inhibit early B cell development and expansion in SLE, leading to possibly enhanced recruitment of autoreactive clones into the mature B cell repertoire [71]. Conversely, circulating neutrophils activated with ANCA caused the release of a B lymphocyte stimulator and promoted B cell survival [72]. Recently, it also was shown that LL37-DNA complexes in NETs can directly trigger self-reactive memory B cells to produce anti-LL37 antibodies in SLE [41]. Self-reactive B cells generally do not react to DNA, however, if the DNA originates from NETs in the form of LL37-DNA complex, it can trigger B cell activation in a toll-like receptor 9 (TLR9)-dependent manner [41]. Notably, SLE-immune complexes were found to induce NET formation recently [36]. Thus, it is conceivable that B cell-derived immune complexes may activate neutrophils by binding to the Fc gamma receptor IIIb (FcγRIIIb) and induce further NET formation [73], causing a feedback loop of autoimmune responses and disease progression.

5.2. NETs and Antigen Presenting Cells

IFNs play a key role in the pathogenesis of autoimmune diseases and are described as a hallmark for SLE [74]. Plasmacytoid dendritic cells (pDC), the main producers of IFN, can be activated by NETs [40]. Specifically, proteins LL37 and HMGB1 form complexes with NETs DNA, which further facilitates uptake and recognition of DNA by pDCs and increases production of IFN-α [42]. In g a RA mouse model, it was shown that T cell response in the presence of NETs is mediated through DC activation [75]. Spontaneous mouse and human NETs could directly activate DCs to induce costimulatory molecules CD80 and CD86, and proinflammatory cytokine IL-6. Furthermore, mouse NETs-treated DCs promoted CD4+ Th cell immune response to secrete INF-γ and IL-17. PMA–NETs activated DCs, on the other hand, were unable to activate CD4+ T cells [76].
NETs also can activate another type of antigen presenting cell (APC): macrophages [77]. However, prolonged exposure to NETs led to damage of the mitochondrial membrane of the macrophage, indicating macrophage death [77] Remarkably, the presence of LL37 and C1q on NETs increased activation of macrophages by RA NETs, leading to secretion of pro-inflammatory IL-8, IL-6 and TNF-α [7]. Activation of inflammatory macrophages also was seen in adult-onset Still’s disease [59]. Contrary, strongly activated macrophages exposed to NETs mediated an anti-inflammatory response with increased IL-10 secretion and inhibited IL-6 secretion [7].

5.3. NETs and T Cells

Recently, an indirect interaction between NETs and T cells was found in the joint of an RA patient [78]. Specifically, NETs were shown to be taken up by fibroblasts, which then presented NET-associated citrullinated peptides to T cells. Also, a direct interaction between PMA–NETs and T cells was proposed [76]. NETs directly prime CD4+ T cells and lower the activation threshold. Therefore, T cell response to specific antigens and suboptimal stimuli is increased, which would otherwise not lead to activation of resting T cells. Interestingly, this T cell priming by NETs seems to involve T cell receptor (TCR) mediation, but is not TLR9-dependent [76]. Direct interaction also was found between SLE low-density granulocytes (LDGs) and T cells [79]. Activation of T cells by SLE LDGs induced the release of proinflammatory cytokines INF-γ and TNF-α. This induction of proinflammatory cytokines was not detected by normal density granulocytes [79]. The study investigated in vitro activation of T cells by LDGs, not NETs. Considering that LDGs in SLE are known to form NETs [5], possibly NETs could play a role in the T cell activation.

6. Neutrophil Extracellular Traps (NETs) in Various Autoimmune Diseases

6.1. Rheumatoid Arthritis (RA)

RA is a chronic systemic disease characterized by joint inflammation and bone destruction. Being an autoimmune disease, a distinct feature of RA is the presence of specific autoantibodies against post-translationally modified proteins known as anti-modified protein antibodies (AMPA) in serum and synovial fluid samples of RA patients [80] Examples of AMPA include antibodies against post-translationally modified proteins like citrullinated proteins (ACPA), antibodies against protein carbamylation (anti-CarP), and antibodies against acetylated proteins (AAPA) [81,82,83] The presence of autoantibodies has been associated with disease progression and pathogenesis. ACPA can bind to citrullinate proteins to form immune complexes, inducing a pro-inflammatory response which further includes complement activation [84,85], for example. Interestingly, AMPA (for example ACPA) can be present in circulation for several years before RA onset [86]. These clinical observations indicate that systemic autoimmunity and development of autoimmune disease are not coupled, and factors driving the transition from pre-existing autoimmunity to RA pathogenesis need to be determined.
NET formation is strongly associated with inflammatory RA [18]. NETs extrude novel autoantigens, such as citrullinated histones, which can promote the autoimmune response in RA [18,32]. Certainly ACPAs are reported to recognize autoantigens on NETs [7,30]; in particular citrullinated histones [31]. The NET-protein MPO was found elevated in RA synovial fluid (SF), skin and rheumatoid nodules, indicating that NETs are present in these inflamed areas (joint/synovium). Neutrophils of RA patients show increased spontaneous NET propensity, compared to healthy control neutrophils in vitro [18,87]. NET propensity increases when neutrophils are stimulated with RA SF and ACPA RA serum [18,87]. Several research groups also have reported the potential of ACPA to drive NET formation in vitro, demonstrating the inflammatory potential of ACPA [18,87]. Elevated MPO–DNA complexes [88] and cell-free nucleosome levels [89] are detected in RA serum. These serum levels of cell-free nucleosome in RA patients also correlate with clinical parameters, such as C-reactive protein (CRP) and positivity for rheumatoid factor (RF) and ACPA [89]. The MPO–DNA complex level also correlated with the ACPA level in RA patient sera samples [88]. Thus, NETs and NET-derived products could serve as a biomarker for RA disease activity. Besides potential biomarkers, NET and NET-derived products also may be promising therapeutic targets for inflammatory RA (Table 2).

6.2. Anti-Neutrophil Cytoplasmic Antibodies (ANCA) Vasculitis (AAV)

AAV is a systemic autoimmune disorder, characterized by inflammation and destruction of small vessels in various organs. Presence of autoantibodies against MPO and PR3 are the key hallmarks that are believed to trigger the disease response.
PR3 is expressed on the membrane of resting neutrophils while MPO is stored within the granules of the neutrophils [122]. Their expression increases when neutrophils are activated by cytokines [123]. NETs also are decorated with MPO and PR3, as shown by several immunofluorescence studies on in vitro NETs and in vivo NETs in necrotizing lesions of AAV [17]. Colocalization of DNA, MPO and PR3 in kidney tissue of small-vessel vasculitis (SVV) glomerulonephritis patients, for example, indicates the presence of NETs and ANCA antigens in inflamed tissue [17]. MPO release in the presence of NETs also was found in kidney biopsies of MPO–ANCA-associated glomerulonephritis patients [49,50]. NETs not only were found in the kidneys of AAV patients, but in other types of vasculitis as well. Nerve samples of ANCA-associated vasculitic neuropathy, for instance, had citrullinated histones and MPO identified as NETs [47]. One recent study suggests that NET-protein neutrophil elastase can digest ANCA in a time-dependent manner, leading to pauci-immune lesions [124].
The presence of NETs in AAV patients is evident, however their potential as biomarkers for AAV disease activity is inconclusive. Regarding the serum of active AAV patients, higher MPO–DNA levels were detected compared to AAV patients in remission [125]. Another study, however, did not find a difference between the serum MPO–DNA levels in active and remissive AAV [126].
While it is becoming clear that NETs may be a crucial source of ANCA-autoantigens, some studies suggest that binding of ANCA to their target antigens, PR3 and MPO, further activate the neutrophils. NETs are released in response to ANCA stimulation [17]. A recent study, however, contrasts these findings. NET formation in vitro, through stimulation of healthy neutrophils with AAV serum, did not correlate with serum levels of ANCA [127]. Also, after depletion of immunoglobulin (Ig)-G and IgA in serum, NET formation remained unchanged, raising the question whether ANCA actually influences NET formation [127]. Added to being antigenic in nature, NETs also influence AAV disease progression by directly damaging vessels through cytotoxicity of NET-associated histone release [56]. NET-induced endothelial cell damage can be prevented when NETs are degraded with DNase1 [55]. Intravenous immunoglobulin (IVIG) is a potential treatment for AAV patients [121]. Intriguingly, the amount of PMA-induced NETs was significantly lower when neutrophils were pre-treated with IVIG before exposure to PMA [121]. Moreover, the inhibitory effect of IVIG on NETs, as well as decreased ANCA titers and pulmonary haemorrhage, also was seen in peritoneal tissue of MPO–AAV rats treated with IVIG [121]. To conclude, the presence of NETs and their autoantigenic properties in AAV patients is well-demonstrated. Therefore, NETs could represent informative biomarkers for disease diagnosis and targets for future therapeutics (Table 2). Conversely, the role of ANCA-mediated NET induction is not yet fully convincing. Further research is needed in understanding the interplay of ANCA and NETs in the pathophysiology of AAV.

6.3. Systemic Lupus Erythematosus (SLE)

Systemic lupus erythematosus (SLE) is a chronic autoimmune disease attacking healthy tissue across the body. SLE can manifest in milder forms, in skin and joints, while more serious manifestations impair functions of the kidneys and central nervous system. Regarding the kidneys, autoantigens and autoantibodies can deposit as immune complexes causing severe lupus nephritis. SLE progression is a result of autoantibodies against nucleic acids and dsDNA. These autoantigens can be sourced from apoptotic and necrotic material [128]. Clearance of this material is thought to be defective in SLE [129].
NETs are known to be a central source of SLE autoantigens [5,130,131,132,133]. The persistence of NETs also could extend the exposure time of autoantigens, due to impaired NET clearance and thereby contribute to SLE pathogenesis [14,133,134]. Removal of NETs by DNase1 was shown to be malfunctioning in in vitro NETs stimulated with SLE serum [14]. This finding indicates that DNase-1 inhibitors may either be present in NETs or (auto) antibodies bound to NETs could inhibit NET-degradation by protecting against proteases. Studies in SLE patients on the relation between NET degradation and disease manifestation support this hypothesis. SLE patients with impaired NET degradation were found more likely to develop lupus nephritis, compared to SLE patients with functioning NET degradation [14,135]. Impaired NET degradation also correlated with high levels of SLE-associated autoantibodies [135]. Other effects of nondegraded NETs are activation of the complement system, thereby driving inflammation forward [26], as well as priming other neutrophils into NET formation [130]. Notably, SLE patients possess a distinct set of neutrophils, called low-density granulocytes (LDGs) [3,136,137]. These specific neutrophils have an increased spontaneous NET propensity, compared to other neutrophils of SLE patients [5,29,134]. Endothelial damage is a common symptom of SLE and may be due to the actions of NETs [5], particularly those formed by LDGs [45]. A recent study characterized two subsets of SLE LDGs, CD10+ LDGs and CD10 LDGs [138]. The phenotype of CD10 LDGs display a more immature stage of neutrophil differentiation and is less prone to spontaneous NET formation, as compared to CD10+ LDGs [139]. Together, this points to the importance of NETs in SLE pathogenesis and how impaired clearance of NETs may prolong the (auto)inflammatory effects of NETs.

6.4. Antiphospholipid Syndrome (APS)

Antiphospholipid syndrome (APS) is an autoimmune disease that often occurs together with SLE, but also presents itself as a primary disease. Autoantibodies targeting phospholipids and phospholipid binding proteins, such as β2-glycoprotein 1 (β2-GP1) are the hallmarks of APS [140]. Particularly, APS patients showcase an increased risk of thrombosis. Thrombi consist of platelets and neutrophils, but also NETs, suggesting the possible role of NETs in APS development [141]. Furthermore, neutrophils from APS patients were shown to display enhanced spontaneous NET release [134,142]. NET response might be mediated through activation of TLR4 [100,142], together with reactive oxygen species (ROS) [100,142,143]. Remarkably, IgG purified from APS patients induced NET release from healthy neutrophils [142]. Furthermore, NET release did not correlate with APS disease activity, but rather with the presence of autoantibodies [134]. An increase in NET release in response to autoantibodies could be explained by the presence of β2-GP1 on the surface of neutrophils [142]. Several studies observed anti-β2-GP1 to induce NET formation in healthy neutrophils [100,142,143]. Evidences suggest that NETs may directly mediate APS pathology by featuring as a scaffold for platelets to aggregate [144], possibly by presenting a tissue factor [53,100]. Moreover, activated platelets can stimulate neutrophils to NET formation by releasing HMGB1 [141]. Accordingly, this highlights a central role for NETs in thrombosis and, thereby, APS development.

6.5. Multiple Sclerosis (MS)

Multiple sclerosis (MS) is a disease of the central nervous system with a strong autoimmune character. Few studies have been performed on the role of NETs in MS pathogenesis [145]. However, in a MS rodent model it became apparent that NETs could be of importance in MS, as depletion of MPO, attenuated rodent MS phenotypes and restored the blood–brain barrier integrity [146]. Elevated MPO–DNA complexes also were detected in the MS serum of patients, but these levels did not correlate with disease activity [147]. Within MS patients, differences in MPO–DNA complex levels correlated with the patient’s gender, suggesting that NETs may underlie gender-specific differences in MS pathogenesis. Moreover, LDGs, CD14-CD15high, were found in peripheral blood of MS patients, at levels comparable to SLE patients [148]. Whether the propensity of MS LDGs to form NETs differentiates from normal density neutrophils has not been investigated yet.

6.6. Anti-Inflammatory NETs

While NETs clearly enhance the inflammatory response in various ways, recent evidence suggests that they also may have anti-inflammatory properties or induce an anti-inflammatory response. Found in the presence of PMA–NETs, LPS-activated macrophages act in an anti-inflammatory manner [7]. A downregulation of IL-6 and an increase in IL-10 secretion by these macrophages shows the anti-inflammatory potential of NETs. This effect was enhanced in presence of C1q and LL37 [7]. Viable and apoptotic neutrophils also mediate an anti-inflammatory response in macrophages through NF-kB signalling suppression [149], suggesting that an anti-inflammatory response may be mediated by the type of neutrophil cell death. Certain proteases in NETs also could contribute to the anti-inflammatory potential of NETs. Regarding gout, where NETs are aggregated in the synovial joint, monosodium urate crystals (MSU)-induced aggregated NETs were found to degrade inflammatory mediators, such as IL-1β, IL-6 and TNF [150]. NET-proteases also could act on NET-proteins that are autoantigens [151]. Shown in the presence of the neutrophil protease inhibitor phenylmethylsulfonyl fluoride (PMSF), autoantigens in PMA–NETs remain present. While without PMSF, PMA–NET proteases degrade NET autoantigens. More research is needed toward understanding the anti-inflammatory role of NETs and their association with autoimmunity.

7. Conclusions and Future Directions

Neutrophils and NETs are present in abundance at the inflammatory sites of various autoimmune diseases and play an active role during the development and persistence of autoimmune responses. As a potential source of autoantigens and activators of immune-cells, NETs could be crucial attributors to the development of ‘autoimmunity’ and the breaking of self-tolerance. Further research is required to understand the physiology and responses of NETs in a disease-specific manner. Therefore, the use of physiological disease-specific stimulants to induce NETs in vitro, an improved understanding of the disease-specific NETome, as well as their impact on other immune cells and inflammation will be crucial for future research. These efforts would provide a substantial basis to target NETs as promising biomarkers or therapeutics to treat inflammatory autoimmune diseases.

Author Contributions

R.T. and J.D. designed the concept of the review. E.F. and J.D. wrote and significantly revised the manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by European Union’s Horizon 2020 research and innovation programme under the Marie Sklodowska Curie grant agreement No 707404.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Brinkmann, V.; Reichard, U.; Goosmann, C.; Fauler, B.; Uhlemann, Y.; Weiss, D.S.; Weinrauch, Y.; Zychlinsky, A. Neutrophil extracellular traps kill bacteria. Science 2004, 303, 1532–1535. [Google Scholar] [CrossRef]
  2. Ng, L.G.; Ostuni, R.; Hidalgo, A. Heterogeneity of neutrophils. Nat. Rev. Immunol. 2019, 19, 255–265. [Google Scholar] [CrossRef]
  3. Denny, M.F.; Yalavarthi, S.; Zhao, W.; Thacker, S.G.; Anderson, M.; Sandy, A.R.; McCune, W.J.; Kaplan, M.J. 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. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Christoffersson, G.; Phillipson, M. The neutrophil: One cell on many missions or many cells with different agendas? Cell Tissue Res. 2018, 371, 415–423. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. 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.; et al. Netting Neutrophils Induce Endothelial Damage, Infiltrate Tissues, and Expose Immunostimulatory Molecules in Systemic Lupus Erythematosus. J. Immunol. 2011, 187, 538–552. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  6. Petretto, A.; Bruschi, M.; Pratesi, F.; Croia, C.; Candiano, G.; Ghiggeri, G.; Migliorini, P. Neutrophil extracellular traps (NET) induced by different stimuli: A comparative proteomic analysis. PLoS ONE 2019, 14, e0218946. [Google Scholar] [CrossRef] [PubMed]
  7. Ribon, M.; Seninet, S.; Mussard, J.; Sebbag, M.; Clavel, C.; Serre, G.; Boissier, M.-C.; Semerano, L.; Decker, P. Neutrophil extracellular traps exert both pro- and anti-inflammatory actions in rheumatoid arthritis that are modulated by C1q and LL-37. J. Autoimmun. 2019, 98, 122–131. [Google Scholar] [CrossRef]
  8. Papayannopoulos, V. Neutrophil extracellular traps in immunity and disease. Nat. Rev. Immunol. 2018, 18, 134–147. [Google Scholar] [CrossRef]
  9. Boeltz, S.; Amini, P.; Anders, H.-J.; Andrade, F.; Bilyy, R.; Chatfield, S.; Cichon, I.; Clancy, D.M.; Desai, J.; Dumych, T.; et al. To NET or not to NET:current opinions and state of the science regarding the formation of neutrophil extracellular traps. Cell Death Differ. 2019, 26, 395–408. [Google Scholar] [CrossRef] [Green Version]
  10. Chapman, E.A.; Lyon, M.; Simpson, D.; Mason, D.; Beynon, R.J.; Moots, R.J.; Wright, H.L. Caught in a Trap? Proteomic Analysis of Neutrophil Extracellular Traps in Rheumatoid Arthritis and Systemic Lupus Erythematosus. Front. Immunol. 2019, 10, 423. [Google Scholar] [CrossRef]
  11. Amulic, B.; Cazalet, C.; Hayes, G.L.; Metzler, K.D.; Zychlinsky, A. Neutrophil function: From mechanisms to disease. Annu. Rev. Immunol. 2012, 30, 459–489. [Google Scholar] [CrossRef] [PubMed]
  12. Martinod, K.; Witsch, T.; Farley, K.; Gallant, M.; Remold-O’Donnell, E.; Wagner, D.D. Neutrophil elastase-deficient mice form neutrophil extracellular traps in an experimental model of deep vein thrombosis. J. Thromb. Haemost. JTH 2016, 14, 551–558. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Naffah de Souza, C.; Breda, L.C.D.; Khan, M.A.; de Almeida, S.R.; Câmara, N.O.S.; Sweezey, N.; Palaniyar, N. Alkaline pH Promotes NADPH Oxidase-Independent Neutrophil Extracellular Trap Formation: A Matter of Mitochondrial Reactive Oxygen Species Generation and Citrullination and Cleavage of Histone. Front. Immunol. 2018, 8, 1849. [Google Scholar] [CrossRef] [PubMed]
  14. Hakkim, A.; Fürnrohr, B.G.; Amann, K.; Laube, B.; Abed, U.A.; Brinkmann, V.; Herrmann, M.; Voll, R.E.; Zychlinsky, A. Impairment of neutrophil extracellular trap degradation is associated with lupus nephritis. Proc. Natl. Acad. Sci. USA 2010, 107, 9813–9818. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  15. Bruschi, M.; Petretto, A.; Santucci, L.; Vaglio, A.; Pratesi, F.; Migliorini, P.; Bertelli, R.; Lavarello, C.; Bartolucci, M.; Candiano, G.; et al. Neutrophil Extracellular Traps protein composition is specific for patients with Lupus nephritis and includes methyl-oxidized αenolase (methionine sulfoxide 93). Sci. Rep. 2019, 9, 1–13. [Google Scholar] [CrossRef]
  16. Jorch, S.K.; Kubes, P. An emerging role for neutrophil extracellular traps in noninfectious disease. Nat. Med. 2017, 23, 279–287. [Google Scholar] [CrossRef]
  17. Kessenbrock, K.; Krumbholz, M.; Schönermarck, U.; Back, W.; Gross, W.L.; Werb, Z.; Gröne, H.-J.; Brinkmann, V.; Jenne, D.E. Netting neutrophils in autoimmune small-vessel vasculitis. Nat. Med. 2009, 15, 623–625. [Google Scholar] [CrossRef]
  18. Khandpur, R.; Carmona-Rivera, C.; Vivekanandan-Giri, A.; Gizinski, A.; Yalavarthi, S.; Knight, J.S.; Friday, S.; Li, S.; Patel, R.M.; Subramanian, V.; et al. NETs are a source of citrullinated autoantigens and stimulate inflammatory responses in rheumatoid arthritis. Sci. Transl. Med. 2013, 5, 178ra40. [Google Scholar] [CrossRef] [Green Version]
  19. Grayson, P.C.; Kaplan, M.J. At the Bench: Neutrophil extracellular traps (NETs) highlight novel aspects of innate immune system involvement in autoimmune diseases. J. Leukoc. Biol. 2016, 99, 253–264. [Google Scholar] [CrossRef] [Green Version]
  20. Bonanni, A.; Vaglio, A.; Bruschi, M.; Sinico, R.A.; Cavagna, L.; Moroni, G.; Franceschini, F.; Allegri, L.; Pratesi, F.; Migliorini, P.; et al. Multi-antibody composition in lupus nephritis: Isotype and antigen specificity make the difference. Autoimmun. Rev. 2015, 14, 692–702. [Google Scholar] [CrossRef]
  21. Bruschi, M.; Sinico, R.A.; Moroni, G.; Pratesi, F.; Migliorini, P.; Galetti, M.; Murtas, C.; Tincani, A.; Madaio, M.; Radice, A.; et al. Glomerular autoimmune multicomponents of human lupus nephritis in vivo: α-enolase and annexin AI. J. Am. Soc. Nephrol. JASN 2014, 25, 2483–2498. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  22. Iaccarino, L.; Ghirardello, A.; Canova, M.; Zen, M.; Bettio, S.; Nalotto, L.; Punzi, L.; Doria, A. Anti-annexins autoantibodies: Their role as biomarkers of autoimmune diseases. Autoimmun. Rev. 2011, 10, 553–558. [Google Scholar] [CrossRef] [PubMed]
  23. Kretz, C.C.; Norpo, M.; Abeler-Dörner, L.; Linke, B.; Haust, M.; Edler, L.; Krammer, P.H.; Kuhn, A. Anti-annexin 1 antibodies: A new diagnostic marker in the serum of patients with discoid lupus erythematosus. Exp. Dermatol. 2010, 19, 919–921. [Google Scholar] [CrossRef]
  24. van Beers, J.J.B.C.; Schwarte, C.M.; Stammen-Vogelzangs, J.; Oosterink, E.; Božič, B.; Pruijn, G.J.M. 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. [Google Scholar] [CrossRef] [PubMed]
  25. Wang, H.; Wang, C.; Zhao, M.-H.; Chen, M. Neutrophil extracellular traps can activate alternative complement pathways. Clin. Exp. Immunol. 2015, 181, 518–527. [Google Scholar] [CrossRef] [Green Version]
  26. Leffler, J.; Martin, M.; Gullstrand, B.; Tydén, H.; Lood, C.; Truedsson, L.; Bengtsson, A.A.; Blom, A.M. Neutrophil extracellular traps that are not degraded in systemic lupus erythematosus activate complement exacerbating the disease. J. Immunol. Baltim. Md 1950 2012, 188, 3522–3531. [Google Scholar] [CrossRef] [Green Version]
  27. Schaller, M.; Bigler, C.; Danner, D.; Ditzel, H.J.; Trendelenburg, M. Autoantibodies against C1q in systemic lupus erythematosus are antigen-driven. J. Immunol. 2009, 183, 8225–8231. [Google Scholar] [CrossRef] [Green Version]
  28. Mansour, R.B.; Lassoued, S.; Gargouri, B.; El Gaïd, A.; Attia, H.; Fakhfakh, F. Increased levels of autoantibodies against catalase and superoxide dismutase associated with oxidative stress in patients with rheumatoid arthritis and systemic lupus erythematosus. Scand. J. Rheumatol. 2008, 37, 103–108. [Google Scholar] [CrossRef]
  29. Carmona-Rivera, C.; Kaplan, M.J. Low density granulocytes: A distinct class of neutrophils in systemic autoimmunity. Semin. Immunopathol. 2013, 35, 455–463. [Google Scholar] [CrossRef]
  30. Corsiero, E.; Bombardieri, M.; Carlotti, E.; Pratesi, F.; Robinson, W.; Migliorini, P.; Pitzalis, C. Single cell cloning and recombinant monoclonal antibodies generation from RA synovial B cells reveal frequent targeting of citrullinated histones of NETs. Ann. Rheum. Dis. 2016, 75, 1866–1875. [Google Scholar] [CrossRef] [Green Version]
  31. Lloyd, K.A.; Wigerblad, G.; Sahlström, P.; Garimella, M.G.; Chemin, K.; Steen, J.; Titcombe, P.J.; Marklein, B.; Zhou, D.; Stålesen, R.; et al. Differential ACPA Binding to Nuclear Antigens Reveals a PAD-Independent Pathway and a Distinct Subset of Acetylation Cross-Reactive Autoantibodies in Rheumatoid Arthritis. Front. Immunol. 2018, 9, 3033. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  32. Pratesi, F.; Dioni, I.; Tommasi, C.; Alcaro, M.C.; Paolini, I.; Barbetti, F.; Boscaro, F.; Panza, F.; Puxeddu, I.; Rovero, P.; et al. Antibodies from patients with rheumatoid arthritis target citrullinated histone 4 contained in neutrophils extracellular traps. Ann. Rheum. Dis. 2014, 73, 1414–1422. [Google Scholar] [CrossRef] [PubMed]
  33. Yaniv, G.; Twig, G.; Shor, D.B.-A.; Furer, A.; Sherer, Y.; Mozes, O.; Komisar, O.; Slonimsky, E.; Klang, E.; Lotan, E.; et al. A volcanic explosion of autoantibodies in systemic lupus erythematosus: A diversity of 180 different antibodies found in SLE patients. Autoimmun. Rev. 2015, 14, 75–79. [Google Scholar] [CrossRef] [PubMed]
  34. Isenberg, D.A.; Manson, J.J.; Ehrenstein, M.R.; Rahman, A. Fifty years of anti-ds DNA antibodies: Are we approaching journey’s end? Rheumatol. Oxf. Engl. 2007, 46, 1052–1056. [Google Scholar] [CrossRef] [Green Version]
  35. Gripenberg, M.; Helve, T.; Kurki, P. Profiles of antibodies to histones, DNA and IgG in patients with systemic rheumatic diseases determined by ELISA. J. Rheumatol. 1985, 12, 934–939. [Google Scholar]
  36. van Dam, L.S.; Kraaij, T.; Kamerling, S.W.A.; Bakker, J.A.; Scherer, U.H.; Rabelink, T.J.; van Kooten, C.; Teng, Y.K.O. Intrinsically Distinct Role of Neutrophil Extracellular Trap Formation in Antineutrophil Cytoplasmic Antibody-Associated Vasculitis Compared to Systemic Lupus Erythematosus. Arthritis Rheumatol. Hoboken NJ 2019. [Google Scholar]
  37. Whittall-García, L.P.; Torres-Ruiz, J.; Zentella-Dehesa, A.; Tapia-Rodríguez, M.; Alcocer-Varela, J.; Mendez-Huerta, N.; Gómez-Martín, D. Neutrophil extracellular traps are a source of extracellular HMGB1 in lupus nephritis: Associations with clinical and histopathological features. Lupus 2019, 28, 1549–1557. [Google Scholar] [CrossRef]
  38. Tang, S.; Zhang, Y.; Yin, S.-W.; Gao, X.-J.; Shi, W.-W.; Wang, Y.; Huang, X.; Wang, L.; Zou, L.-Y.; Zhao, J.-H.; et al. Neutrophil extracellular trap formation is associated with autophagy-related signalling in ANCA-associated vasculitis. Clin. Exp. Immunol. 2015, 180, 408–418. [Google Scholar] [CrossRef] [Green Version]
  39. Kain, R.; Tadema, H.; McKinney, E.F.; Benharkou, A.; Brandes, R.; Peschel, A.; Hubert, V.; Feenstra, T.; Sengölge, G.; Stegeman, C.; et al. High prevalence of autoantibodies to hLAMP-2 in anti-neutrophil cytoplasmic antibody-associated vasculitis. J. Am. Soc. Nephrol. JASN 2012, 23, 556–566. [Google Scholar] [CrossRef] [Green Version]
  40. Garcia-Romo, G.S.; Caielli, S.; Vega, B.; Connolly, J.; Allantaz, F.; Xu, Z.; Punaro, M.; Baisch, J.; Guiducci, C.; Coffman, R.L.; et al. Netting neutrophils are major inducers of type I IFN production in pediatric systemic lupus erythematosus. Sci. Transl. Med. 2011, 3, 73ra20. [Google Scholar] [CrossRef] [Green Version]
  41. Gestermann, N.; Di Domizio, J.; Lande, R.; Demaria, O.; Frasca, L.; Feldmeyer, L.; Di Lucca, J.; Gilliet, M. Netting Neutrophils Activate Autoreactive B Cells in Lupus. J. Immunol. Baltim. Md 1950 2018, 200, 3364–3371. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  42. Lande, R.; Ganguly, D.; Facchinetti, V.; Frasca, L.; Conrad, C.; Gregorio, J.; Meller, S.; Chamilos, G.; Sebasigari, R.; Riccieri, V.; et al. Neutrophils activate plasmacytoid dendritic cells by releasing self-DNA-peptide complexes in systemic lupus erythematosus. Sci. Transl. Med. 2011, 3, 73ra19. [Google Scholar] [CrossRef] [Green Version]
  43. Frasca, L.; Palazzo, R.; Chimenti, M.S.; Alivernini, S.; Tolusso, B.; Bui, L.; Botti, E.; Giunta, A.; Bianchi, L.; Petricca, L.; et al. Anti-LL37 Antibodies Are Present in Psoriatic Arthritis (PsA) Patients: New Biomarkers in PsA. Front. Immunol. 2018, 9. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Mattey, D.L.; Nixon, N.B.; Dawes, P.T. Association of circulating levels of MMP-8 with mortality from respiratory disease in patients with rheumatoid arthritis. Arthritis Res. Ther. 2012, 14, R204. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  45. Carmona-Rivera, C.; Zhao, W.; Yalavarthi, S.; Kaplan, M.J. Neutrophil extracellular traps induce endothelial dysfunction in systemic lupus erythematosus through the activation of matrix metalloproteinase-2. Ann. Rheum. Dis. 2015, 74, 1417–1424. [Google Scholar] [CrossRef] [Green Version]
  46. Nakazawa, D.; Shida, H.; Tomaru, U.; Yoshida, M.; Nishio, S.; Atsumi, T.; Ishizu, A. Enhanced formation and disordered regulation of NETs in myeloperoxidase-ANCA-associated microscopic polyangiitis. J. Am. Soc. Nephrol. JASN 2014, 25, 990–997. [Google Scholar] [CrossRef] [PubMed]
  47. Takeuchi, H.; Kawasaki, T.; Shigematsu, K.; Kawamura, K.; Oka, N. Neutrophil extracellular traps in neuropathy with anti-neutrophil cytoplasmic autoantibody-associated microscopic polyangiitis. Clin. Rheumatol. 2017, 36, 913–917. [Google Scholar] [CrossRef]
  48. Urban, C.F.; Ermert, D.; Schmid, M.; Abu-Abed, U.; Goosmann, C.; Nacken, W.; Brinkmann, V.; Jungblut, P.R.; Zychlinsky, A. Neutrophil extracellular traps contain calprotectin, a cytosolic protein complex involved in host defense against Candida albicans. PLoS Pathog. 2009, 5, e1000639. [Google Scholar] [CrossRef] [Green Version]
  49. 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. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  50. O’Sullivan, K.M.; Lo, C.Y.; Summers, S.A.; Elgass, K.D.; McMillan, P.J.; Longano, A.; Ford, S.L.; Gan, P.-Y.; Kerr, P.G.; Kitching, A.R.; et al. Renal participation of myeloperoxidase in antineutrophil cytoplasmic antibody (ANCA)-associated glomerulonephritis. Kidney Int. 2015, 88, 1030–1046. [Google Scholar] [CrossRef] [Green Version]
  51. Schultz, D.R.; Tozman, E.C. Antineutrophil cytoplasmic antibodies: Major autoantigens, pathophysiology, and disease associations. Semin. Arthritis Rheum. 1995, 25, 143–159. [Google Scholar] [CrossRef]
  52. Kambas, K.; Mitroulis, I.; Apostolidou, E.; Girod, A.; Chrysanthopoulou, A.; Pneumatikos, I.; Skendros, P.; Kourtzelis, I.; Koffa, M.; Kotsianidis, I.; et al. Autophagy Mediates the Delivery of Thrombogenic Tissue Factor to Neutrophil Extracellular Traps in Human Sepsis. PLoS ONE 2012, 7, e45427. [Google Scholar] [CrossRef] [PubMed]
  53. Frangou, E.; Chrysanthopoulou, A.; Mitsios, A.; Kambas, K.; Arelaki, S.; Angelidou, I.; Arampatzioglou, A.; Gakiopoulou, H.; Bertsias, G.K.; Verginis, P.; et al. REDD1/autophagy pathway promotes thromboinflammation and fibrosis in human systemic lupus erythematosus (SLE) through NETs decorated with tissue factor (TF) and interleukin-17A (IL-17A). Ann. Rheum. Dis. 2019, 78, 238–248. [Google Scholar] [CrossRef] [PubMed]
  54. Ramsey-Goldman, R.; Alexander, R.V.; Massarotti, E.M.; Wallace, D.J.; Narain, S.; Arriens, C.; Collins, C.E.; Saxena, A.; Putterman, C.; Kalunian, K.C.; et al. Complement activation occurs in patients with probable systemic lupus erythematosus and may predict progression to ACR classified SLE. Arthritis Rheumatol. 2020, 72, 78–88. [Google Scholar] [CrossRef] [Green Version]
  55. Schreiber, A.; Rousselle, A.; Becker, J.U.; von Mässenhausen, A.; Linkermann, A.; Kettritz, R. Necroptosis controls NET generation and mediates complement activation, endothelial damage, and autoimmune vasculitis. Proc. Natl. Acad. Sci. USA 2017, 114, E9618–E9625. [Google Scholar] [CrossRef] [Green Version]
  56. Kumar, S.V.R.; Kulkarni, O.P.; Mulay, S.R.; Darisipudi, M.N.; Romoli, S.; Thomasova, D.; Scherbaum, C.R.; Hohenstein, B.; Hugo, C.; Müller, S.; et al. Neutrophil Extracellular Trap-Related Extracellular Histones Cause Vascular Necrosis in Severe GN. J. Am. Soc. Nephrol. JASN 2015, 26, 2399–2413. [Google Scholar] [CrossRef] [Green Version]
  57. Ma, Y.-H.; Ma, T.; Wang, C.; Wang, H.; Chang, D.-Y.; Chen, M.; Zhao, M.-H. High-mobility group box 1 potentiates antineutrophil cytoplasmic antibody-inducing neutrophil extracellular traps formation. Arthritis Res. Ther. 2016, 18, 2. [Google Scholar] [CrossRef] [Green Version]
  58. Kahlenberg, J.M.; Carmona-Rivera, C.; Smith, C.K.; Kaplan, M.J. Neutrophil extracellular trap-associated protein activation of the NLRP3 inflammasome is enhanced in lupus macrophages. J. Immunol. Baltim. Md 1950 2013, 190, 1217–1226. [Google Scholar] [CrossRef] [Green Version]
  59. Hu, Q.; Shi, H.; Zeng, T.; Liu, H.; Su, Y.; Cheng, X.; Ye, J.; Yin, Y.; Liu, M.; Zheng, H.; et al. Increased neutrophil extracellular traps activate NLRP3 and inflammatory macrophages in adult-onset Still’s disease. Arthritis Res. Ther. 2019, 21, 9. [Google Scholar] [CrossRef] [Green Version]
  60. Chen, K.W.; Monteleone, M.; Boucher, D.; Sollberger, G.; Ramnath, D.; Condon, N.D.; von Pein, J.B.; Broz, P.; Sweet, M.J.; Schroder, K. Noncanonical inflammasome signaling elicits gasdermin D-dependent neutrophil extracellular traps. Sci. Immunol. 2018, 3, eaar6676. [Google Scholar] [CrossRef] [Green Version]
  61. Sollberger, G.; Choidas, A.; Burn, G.L.; Habenberger, P.; Di Lucrezia, R.; Kordes, S.; Menninger, S.; Eickhoff, J.; Nussbaumer, P.; Klebl, B.; et al. Gasdermin D plays a vital role in the generation of neutrophil extracellular traps. Sci. Immunol. 2018, 3, eaar6689. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  62. Lazzaretto, B.; Fadeel, B. Intra- and Extracellular Degradation of Neutrophil Extracellular Traps by Macrophages and Dendritic Cells. J. Immunol. 2019, 203, 2276–2290. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Farrera, C.; Fadeel, B. Macrophage clearance of neutrophil extracellular traps is a silent process. J. Immunol. 2013, 191, 2647–2656. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. Mahajan, A.; Herrmann, M.; Muñoz, L.E. Clearance Deficiency and Cell Death Pathways: A Model for the Pathogenesis of SLE. Front. Immunol. 2016, 7, 35. [Google Scholar] [CrossRef] [Green Version]
  65. Nakazawa, D.; Tomaru, U.; Suzuki, A.; Masuda, S.; Hasegawa, R.; Kobayashi, T.; Nishio, S.; Kasahara, M.; Ishizu, A. 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. [Google Scholar] [CrossRef]
  66. Al-Mayouf, S.M.; Sunker, A.; Abdwani, R.; Abrawi, S.A.; Almurshedi, F.; Alhashmi, N.; Al Sonbul, A.; Sewairi, W.; Qari, A.; Abdallah, E.; et al. Loss-of-function variant in DNASE1L3 causes a familial form of systemic lupus erythematosus. Nat. Genet. 2011, 43, 1186–1188. [Google Scholar] [CrossRef]
  67. Nakazawa, D.; Shida, H.; Kusunoki, Y.; Miyoshi, A.; Nishio, S.; Tomaru, U.; Atsumi, T.; Ishizu, A. The responses of macrophages in interaction with neutrophils that undergo NETosis. J. Autoimmun. 2016, 67, 19–28. [Google Scholar] [CrossRef]
  68. Barrera-Vargas, A.; Gómez-Martín, D.; Carmona-Rivera, C.; Merayo-Chalico, J.; Torres-Ruiz, J.; Manna, Z.; Hasni, S.; Alcocer-Varela, J.; Kaplan, M.J. Differential ubiquitination in NETs regulates macrophage responses in systemic lupus erythematosus. Ann. Rheum. Dis. 2018, 77, 944–950. [Google Scholar] [CrossRef]
  69. Lee, K.H.; Kronbichler, A.; Park, D.D.-Y.; Park, Y.; Moon, H.; Kim, H.; Choi, J.H.; Choi, Y.; Shim, S.; Lyu, I.S.; et al. Neutrophil extracellular traps (NETs) in autoimmune diseases: A comprehensive review. Autoimmun. Rev. 2017, 16, 1160–1173. [Google Scholar] [CrossRef]
  70. Puga, I.; Cols, M.; Barra, C.M.; He, B.; Cassis, L.; Gentile, M.; Comerma, L.; Chorny, A.; Shan, M.; Xu, W.; et al. B cell-helper neutrophils stimulate the diversification and production of immunoglobulin in the marginal zone of the spleen. Nat. Immunol. 2011, 13, 170–180. [Google Scholar] [CrossRef] [Green Version]
  71. Palanichamy, A.; Bauer, J.W.; Yalavarthi, S.; Meednu, N.; Barnard, J.; Owen, T.; Cistrone, C.; Bird, A.; Rabinovich, A.; Nevarez, S.; et al. Neutrophil-mediated IFN activation in the bone marrow alters B cell development in human and murine systemic lupus erythematosus. J. Immunol. 2014, 192, 906–918. [Google Scholar] [CrossRef] [PubMed]
  72. Holden, N.J.; Williams, J.M.; Morgan, M.D.; Challa, A.; Gordon, J.; Pepper, R.J.; Salama, A.D.; Harper, L.; Savage, C.O.S. ANCA-stimulated neutrophils release BLyS and promote B cell survival: A clinically relevant cellular process. Ann. Rheum. Dis. 2011, 70, 2229–2233. [Google Scholar] [CrossRef] [PubMed]
  73. Behnen, M.; Leschczyk, C.; Möller, S.; Batel, T.; Klinger, M.; Solbach, W.; Laskay, T. Immobilized immune complexes induce neutrophil extracellular trap release by human neutrophil granulocytes via FcγRIIIB and Mac-1. J. Immunol. 2014, 193, 1954–1965. [Google Scholar] [CrossRef] [PubMed]
  74. Crow, M.K. Type I Interferon in the Pathogenesis of Lupus. J. Immunol. 2014, 192, 5459–5468. [Google Scholar] [CrossRef]
  75. Papadaki, G.; Kambas, K.; Choulaki, C.; Vlachou, K.; Drakos, E.; Bertsias, G.; Ritis, K.; Boumpas, D.T.; Thompson, P.R.; Verginis, P.; et al. Neutrophil extracellular traps exacerbate Th1-mediated autoimmune responses in rheumatoid arthritis by promoting DC maturation. Eur. J. Immunol. 2016, 46, 2542–2554. [Google Scholar] [CrossRef] [Green Version]
  76. 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. [Google Scholar] [CrossRef]
  77. Donis-Maturano, L.; Sánchez-Torres, L.E.; Cerbulo-Vázquez, A.; Chacón-Salinas, R.; García-Romo, G.S.; Orozco-Uribe, M.C.; Yam-Puc, J.C.; González-Jiménez, M.A.; Paredes-Vivas, Y.L.; Calderón-Amador, J.; et al. Prolonged exposure to neutrophil extracellular traps can induce mitochondrial damage in macrophages and dendritic cells. SpringerPlus 2015, 4, 161. [Google Scholar] [CrossRef] [Green Version]
  78. Carmona-Rivera, C.; Carlucci, P.M.; Moore, E.; Lingampalli, N.; Uchtenhagen, H.; James, E.; Liu, Y.; Bicker, K.L.; Wahamaa, H.; Hoffmann, V.; et al. Synovial fibroblast-neutrophil interactions promote pathogenic adaptive immunity in rheumatoid arthritis. Sci. Immunol. 2017, 2, eaag3358. [Google Scholar] [CrossRef] [Green Version]
  79. Rahman, S.; Sagar, D.; Hanna, R.N.; Lightfoot, Y.L.; Mistry, P.; Smith, C.K.; Manna, Z.; Hasni, S.; Siegel, R.M.; Sanjuan, M.A.; et al. Low-density granulocytes activate T cells and demonstrate a non-suppressive role in systemic lupus erythematosus. Ann. Rheum. Dis. 2019, 78, 957–966. [Google Scholar] [CrossRef] [Green Version]
  80. Scott, D.L.; Wolfe, F.; Huizinga, T.W. Rheumatoid arthritis. Lancet 2010, 376, 1094–1108. [Google Scholar] [CrossRef]
  81. Juarez, M.; Bang, H.; Hammar, F.; Reimer, U.; Dyke, B.; Sahbudin, I.; Buckley, C.D.; Fisher, B.; Filer, A.; Raza, K. Identification of novel antiacetylated vimentin antibodies in patients with early inflammatory arthritis. Ann. Rheum. Dis. 2016, 75, 1099–1107. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  82. Schellekens, G.A.; De Jong, B.A.; Van den Hoogen, F.H.; Van de Putte, L.B.; van Venrooij, W.J. Citrulline is an essential constituent of antigenic determinants recognized by rheumatoid arthritis-specific autoantibodies. J. Clin. Investig. 1998, 101, 273–281. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  83. Shi, J.; Knevel, R.; Suwannalai, P.; van der Linden, M.P.; Janssen, G.M.C.; van Veelen, P.A.; Levarht, N.E.W.; van Mil, A.H.M.v.d.H.; Cerami, A.; Huizinga, T.W.J.; et al. Autoantibodies recognizing carbamylated proteins are present in sera of patients with rheumatoid arthritis and predict joint damage. Proc. Natl. Acad. Sci. USA 2011, 108, 17372–17377. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  84. Trouw, L.A.; Haisma, E.M.; Levarht, E.W.N.; van der Woude, D.; Ioan-Facsinay, A.; Daha, M.R.; Huizinga, T.W.J.; Toes, R.E. Anti-cyclic citrullinated peptide antibodies from rheumatoid arthritis patients activate complement via both the classical and alternative pathways. Arthritis Rheum. 2009, 60, 1923–1931. [Google Scholar] [CrossRef] [PubMed]
  85. Toes, R.; Pisetsky, D.S. Pathogenic effector functions of ACPA: Where do we stand? Ann. Rheum. Dis. 2019, 78, 716–721. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  86. Nielen, M.M.J.; van Schaardenburg, D.; Reesink, H.W.; van de Stadt, R.J.; van der Horst-Bruinsma, I.E.; de Koning, M.H.M.T.; Habibuw, M.R.; Vandenbroucke, J.P.; Dijkmans, B.A.C. Specific autoantibodies precede the symptoms of rheumatoid arthritis: A study of serial measurements in blood donors. Arthritis Rheum. 2004, 50, 380–386. [Google Scholar] [CrossRef] [PubMed]
  87. Sur Chowdhury, C.; Giaglis, S.; Walker, U.A.; Buser, A.; Hahn, S.; Hasler, P. Enhanced neutrophil extracellular trap generation in rheumatoid arthritis: Analysis of underlying signal transduction pathways and potential diagnostic utility. Arthritis Res. Ther. 2014, 16, R122. [Google Scholar] [CrossRef] [Green Version]
  88. Wang, W.; Peng, W.; Ning, X. Increased levels of neutrophil extracellular trap remnants in the serum of patients with rheumatoid arthritis. Int. J. Rheum. Dis. 2018, 21, 415–421. [Google Scholar] [CrossRef]
  89. Pérez-Sánchez, C.; Ruiz-Limón, P.; Aguirre, M.A.; Jiménez-Gómez, Y.; Arias-de la Rosa, I.; Ábalos-Aguilera, M.C.; Rodriguez-Ariza, A.; Castro-Villegas, M.C.; Ortega-Castro, R.; Segui, P.; et al. Diagnostic potential of NETosis-derived products for disease activity, atherosclerosis and therapeutic effectiveness in Rheumatoid Arthritis patients. J. Autoimmun. 2017, 82, 31–40. [Google Scholar] [CrossRef]
  90. Kusunoki, Y.; Nakazawa, D.; Shida, H.; Hattanda, F.; Miyoshi, A.; Masuda, S.; Nishio, S.; Tomaru, U.; Atsumi, T.; Ishizu, A. Peptidylarginine Deiminase Inhibitor Suppresses Neutrophil Extracellular Trap Formation and MPO-ANCA Production. Front. Immunol. 2016, 7, 227. [Google Scholar] [CrossRef] [Green Version]
  91. 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. 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. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  92. 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. Peptidylarginine deiminase inhibition is immunomodulatory and vasculoprotective in murine lupus. J. Clin. Investig. 2013, 123, 2981–2993. [Google Scholar] [CrossRef] [PubMed]
  93. Kawaguchi, H.; Matsumoto, I.; Osada, A.; Kurata, I.; Ebe, H.; Tanaka, Y.; Inoue, A.; Umeda, N.; Kondo, Y.; Tsuboi, H.; et al. Peptidyl arginine deiminase inhibition suppresses arthritis via decreased protein citrullination in joints and serum with the downregulation of interleukin-6. Mod. Rheumatol. 2019, 29, 964–969. [Google Scholar] [CrossRef] [PubMed]
  94. Lewis, H.D.; Liddle, J.; Coote, J.E.; Atkinson, S.J.; Barker, M.D.; Bax, B.D.; Bicker, K.L.; Bingham, R.P.; Campbell, M.; Chen, Y.H.; et al. Inhibition of PAD4 activity is sufficient to disrupt mouse and human NET formation. Nat. Chem. Biol. 2015, 11, 189–191. [Google Scholar] [CrossRef] [PubMed]
  95. Willis, V.C.; Banda, N.K.; Cordova, K.N.; Chandra, P.E.; Robinson, W.H.; Cooper, D.C.; Lugo, D.; Mehta, G.; Taylor, S.; Tak, P.P.; et al. Protein arginine deiminase 4 inhibition is sufficient for the amelioration of collagen-induced arthritis. Clin. Exp. Immunol. 2017, 188, 263–274. [Google Scholar] [CrossRef] [Green Version]
  96. Willis, V.C.; Gizinski, A.M.; Banda, N.K.; Causey, C.P.; Knuckley, B.; Cordova, K.N.; Luo, Y.; Levitt, B.; Glogowska, M.; Chandra, P.; 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. [Google Scholar] [CrossRef]
  97. Macanovic, M.; Sinicropi, D.; Shak, S.; Baughman, S.; Thiru, S.; Lachmann, P.J. 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. [Google Scholar] [CrossRef]
  98. Davis, J.C.; Manzi, S.; Yarboro, C.; Rairie, J.; Mcinnes, I.; Averthelyi, D.; Sinicropi, D.; Hale, V.G.; Balow, J.; Austin, H.; et al. Recombinant human Dnase I (rhDNase) in patients with lupus nephritis. Lupus 1999, 8, 68–76. [Google Scholar] [CrossRef]
  99. Meng, H.; Yalavarthi, S.; Kanthi, Y.; Mazza, L.F.; Elfline, M.A.; Luke, C.E.; Pinsky, D.J.; Henke, P.K.; Knight, J.S. In Vivo Role of Neutrophil Extracellular Traps in Antiphospholipid Antibody–Mediated Venous Thrombosis. Arthritis Rheumatol. 2017, 69, 655–667. [Google Scholar] [CrossRef] [Green Version]
  100. Zha, C.; Zhang, W.; Gao, F.; Xu, J.; Jia, R.; Cai, J.; Liu, Y. Anti-β2GPI/β2GPI induces neutrophil extracellular traps formation to promote thrombogenesis via the TLR4/MyD88/MAPKs axis activation. Neuropharmacology 2018, 138, 140–150. [Google Scholar] [CrossRef]
  101. Wang, Y.; Wang, Y.; Wu, J.; Liu, C.; Zhou, Y.; Mi, L.; Zhang, Y.; Wang, W. PRAK Is Required for the Formation of Neutrophil Extracellular Traps. Front. Immunol. 2019, 10, 1252. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  102. Vorobjeva, N.V.; Pinegin, B.V. Effects of the antioxidants Trolox, Tiron and Tempol on neutrophil extracellular trap formation. Immunobiology 2016, 221, 208–219. [Google Scholar] [CrossRef] [PubMed]
  103. Mohammed, B.M.; Fisher, B.J.; Kraskauskas, D.; Farkas, D.; Brophy, D.F.; Fowler, A.A.; Natarajan, R. Vitamin C: A novel regulator of neutrophil extracellular trap formation. Nutrients 2013, 5, 3131–3151. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  104. Merrill, J.T.; Wallace, D.J.; Petri, M.; Kirou, K.A.; Yao, Y.; White, W.I.; Robbie, G.; Levin, R.; Berney, S.M.; Chindalore, V.; et al. Safety profile and clinical activity of sifalimumab, a fully human anti-interferon α monoclonal antibody, in systemic lupus erythematosus: A phase I, multicentre, double-blind randomised study. Ann. Rheum. Dis. 2011, 70, 1905–1913. [Google Scholar] [CrossRef]
  105. Petri, M.; Wallace, D.J.; Spindler, A.; Chindalore, V.; Kalunian, K.; Mysler, E.; Neuwelt, C.M.; Robbie, G.; White, W.I.; Higgs, B.W.; et al. Sifalimumab, a human anti-interferon-α monoclonal antibody, in systemic lupus erythematosus: A phase I randomized, controlled, dose-escalation study. Arthritis Rheum. 2013, 65, 1011–1021. [Google Scholar] [CrossRef]
  106. Khamashta, M.; Merrill, J.T.; Werth, V.P.; Furie, R.; Kalunian, K.; Illei, G.G.; Drappa, J.; Wang, L.; Greth, W.; CD1067 study investigators. Sifalimumab, an anti-interferon-α monoclonal antibody, in moderate to severe systemic lupus erythematosus: A randomised, double-blind, placebo-controlled study. Ann. Rheum. Dis. 2016, 75, 1909–1916. [Google Scholar] [CrossRef] [Green Version]
  107. McBride, J.M.; Jiang, J.; Abbas, A.R.; Morimoto, A.; Li, J.; Maciuca, R.; Townsend, M.; Wallace, D.J.; Kennedy, W.P.; Drappa, J. Safety and pharmacodynamics of rontalizumab in patients with systemic lupus erythematosus: Results of a phase I, placebo-controlled, double-blind, dose-escalation study. Arthritis Rheum. 2012, 64, 3666–3676. [Google Scholar] [CrossRef]
  108. Furie, R.; Khamashta, M.; Merrill, J.T.; Werth, V.P.; Kalunian, K.; Brohawn, P.; Illei, G.G.; Drappa, J.; Wang, L.; Yoo, S.; et al. Anifrolumab, an Anti-Interferon-α Receptor Monoclonal Antibody, in Moderate-to-Severe Systemic Lupus Erythematosus. Arthritis Rheumatol. Hoboken NJ 2017, 69, 376–386. [Google Scholar] [CrossRef] [Green Version]
  109. Hair, P.S.; Enos, A.I.; Krishna, N.K.; Cunnion, K.M. Inhibition of Immune Complex Complement Activation and Neutrophil Extracellular Trap Formation by Peptide Inhibitor of Complement C1. Front. Immunol. 2018, 9, 558. [Google Scholar] [CrossRef]
  110. van Bijnen, S.T.A.; Wouters, D.; van Mierlo, G.J.; Muus, P.; Zeerleder, S. Neutrophil activation and nucleosomes as markers of systemic inflammation in paroxysmal nocturnal hemoglobinuria: Effects of eculizumab. J. Thromb. Haemost. JTH 2015, 13, 2004–2011. [Google Scholar] [CrossRef] [Green Version]
  111. Roberts, H.; White, P.; Dias, I.; McKaig, S.; Veeramachaneni, R.; Thakker, N.; Grant, M.; Chapple, I. Characterization of neutrophil function in Papillon-Lefèvre syndrome. J. Leukoc. Biol. 2016, 100, 433–444. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  112. Seren, S.; Rashed Abouzaid, M.; Eulenberg-Gustavus, C.; Hirschfeld, J.; Nasr Soliman, H.; Jerke, U.; N’Guessan, K.; Dallet-Choisy, S.; Lesner, A.; Lauritzen, C.; et al. Consequences of cathepsin C inactivation for membrane exposure of proteinase 3, the target antigen in autoimmune vasculitis. J. Biol. Chem. 2018, 293, 12415–12428. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  113. Murthy, P.; Singhi, A.D.; Ross, M.A.; Loughran, P.; Paragomi, P.; Papachristou, G.I.; Whitcomb, D.C.; Zureikat, A.H.; Lotze, M.T.; Zeh III, H.J.; et al. Enhanced Neutrophil Extracellular Trap Formation in Acute Pancreatitis Contributes to Disease Severity and Is Reduced by Chloroquine. Front. Immunol. 2019, 10, 28. [Google Scholar] [CrossRef]
  114. Handono, K.; Sidarta, Y.O.; Pradana, B.A.; Nugroho, R.A.; Hartono, I.A.; Kalim, H.; Endharti, A.T. Vitamin D prevents endothelial damage induced by increased neutrophil extracellular traps formation in patients with systemic lupus erythematosus. Acta Medica Indones. 2014, 46, 189–198. [Google Scholar]
  115. Reynolds, J.; Ray, D.; Alexander, M.Y.; Bruce, I. Role of vitamin D in endothelial function and endothelial repair in clinically stable systemic lupus erythematosus. Lancet Lond. Engl. 2015, 385 (Suppl. 1), S83. [Google Scholar] [CrossRef] [Green Version]
  116. Bornhöfft, K.F.; Viergutz, T.; Kühnle, A.; Galuska, S.P. Nanoparticles Equipped with α2,8-Linked Sialic Acid Chains Inhibit the Release of Neutrophil Extracellular Traps. Nanomaterials 2019, 9, E610. [Google Scholar]
  117. Galuska, C.E.; Dambon, J.A.; Kühnle, A.; Bornhöfft, K.F.; Prem, G.; Zlatina, K.; Lütteke, T.; Galuska, S.P. Artificial Polysialic Acid Chains as Sialidase-Resistant Molecular-Anchors to Accumulate Particles on Neutrophil Extracellular Traps. Front. Immunol. 2017, 8, 1229. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  118. Kühnle, A.; Lütteke, T.; Bornhöfft, K.F.; Galuska, S.P. Polysialic Acid Modulates the Binding of External Lactoferrin in Neutrophil Extracellular Traps. Biology 2019, 8, 20. [Google Scholar] [CrossRef] [Green Version]
  119. Ruiz-Limón, P.; Ortega, R.; de la Rosa, I.A.; del Carmen Abalos-Aguilera, M.; Perez-Sanchez, C.; Jimenez-Gomez, Y.; Peralbo-Santaella, E.; Font, P.; Ruiz-Vilches, D.; Ferrin, G.; et al. Tocilizumab improves the proatherothrombotic profile of rheumatoid arthritis patients modulating endothelial dysfunction, NETosis, and inflammation. Transl. Res. 2017, 183, 87–103. [Google Scholar] [CrossRef]
  120. Kraaij, T.; Kamerling, S.W.A.; de Rooij, E.N.M.; van Daele, P.L.A.; Bredewold, O.W.; Bakker, J.A.; Bajema, I.M.; Scherer, H.U.; Toes, R.E.M.; Huizinga, T.J.W.; et al. The NET-effect of combining rituximab with belimumab in severe systemic lupus erythematosus. J. Autoimmun. 2018, 91, 45–54. [Google Scholar] [CrossRef]
  121. Uozumi, R.; Iguchi, R.; Masuda, S.; Nishibata, Y.; Nakazawa, D.; Tomaru, U.; Ishizu, A. Pharmaceutical immunoglobulins reduce neutrophil extracellular trap formation and ameliorate the development of MPO-ANCA-associated vasculitis. Mod. Rheumatol. 2019, 1–7. [Google Scholar] [CrossRef]
  122. Cornec, D.; Cornec-Le Gall, E.; Fervenza, F.C.; Specks, U. ANCA-associated vasculitis—Clinical utility of using ANCA specificity to classify patients. Nat. Rev. Rheumatol. 2016, 12, 570–579. [Google Scholar] [CrossRef] [PubMed]
  123. Csernok, E.; Ernst, M.; Schmitt, W.; Bainton, D.F.; Gross, W.L. Activated neutrophils express proteinase 3 on their plasma membrane in vitro and in vivo. Clin. Exp. Immunol. 1994, 95, 244–250. [Google Scholar] [CrossRef] [PubMed]
  124. Futamata, E.; Masuda, S.; Nishibata, Y.; Tanaka, S.; Tomaru, U.; Ishizu, A. Vanishing Immunoglobulins: The Formation of Pauci-Immune Lesions in Myeloperoxidase-Antineutrophil Cytoplasmic Antibody-Associated Vasculitis. Nephron 2018, 138, 328–330. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  125. Söderberg, D.; Kurz, T.; Motamedi, A.; Hellmark, T.; Eriksson, P.; Segelmark, M. 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. Rheumatol. Oxf. Engl. 2015, 54, 2085–2094. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  126. Wang, H.; Sha, L.-L.; Ma, T.-T.; Zhang, L.-X.; Chen, M.; Zhao, M.-H. Circulating Level of Neutrophil Extracellular Traps Is Not a Useful Biomarker for Assessing Disease Activity in Antineutrophil Cytoplasmic Antibody-Associated Vasculitis. PLoS ONE 2016, 11, e0148197. [Google Scholar] [CrossRef] [Green Version]
  127. Kraaij, T.; Kamerling, S.W.A.; van Dam, L.S.; Bakker, J.A.; Bajema, I.M.; Page, T.; Brunini, F.; Pusey, C.D.; Toes, R.E.M.; Scherer, H.U.; et al. Excessive neutrophil extracellular trap formation in ANCA-associated vasculitis is independent of ANCA. Kidney Int. 2018, 94, 139–149. [Google Scholar] [CrossRef]
  128. Midgley, A.; Beresford, M. Cellular localization of nuclear antigen during neutrophil apoptosis: Mechanism for autoantigen exposure? Lupus 2011, 20, 641–646. [Google Scholar] [CrossRef]
  129. Ren, Y.; Tang, J.; Mok, M.Y.; Chan, A.W.K.; Wu, A.; Lau, C.S. Increased apoptotic neutrophils and macrophages and impaired macrophage phagocytic clearance of apoptotic neutrophils in systemic lupus erythematosus. Arthritis Rheum. 2003, 48, 2888–2897. [Google Scholar] [CrossRef]
  130. Dieker, J.; Tel, J.; Pieterse, E.; Thielen, A.; Rother, N.; Bakker, M.; Fransen, J.; Dijkman, H.B.P.M.; Berden, J.H.; de Vries, J.M.; et al. Circulating Apoptotic Microparticles in Systemic Lupus Erythematosus Patients Drive the Activation of Dendritic Cell Subsets and Prime Neutrophils for NETosis. Arthritis Rheumatol. Hoboken NJ 2016, 68, 462–472. [Google Scholar] [CrossRef] [Green Version]
  131. Liu, C.L.; Tangsombatvisit, S.; Rosenberg, J.M.; Mandelbaum, G.; Gillespie, E.C.; Gozani, O.P.; Alizadeh, A.A.; Utz, P.J. Specific post-translational histone modifications of neutrophil extracellular traps as immunogens and potential targets of lupus autoantibodies. Arthritis Res. Ther. 2012, 14, R25. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  132. Lood, C.; Blanco, L.P.; Purmalek, M.M.; Carmona-Rivera, C.; De Ravin, S.S.; Smith, C.K.; Malech, H.L.; Ledbetter, J.A.; Elkon, K.B.; Kaplan, M.J. Neutrophil extracellular traps enriched in oxidized mitochondrial DNA are interferogenic and contribute to lupus-like disease. Nat. Med. 2016, 22, 146–153. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  133. Wang, H.; Li, T.; Chen, S.; Gu, Y.; Ye, S. Neutrophil Extracellular Trap Mitochondrial DNA and Its Autoantibody in Systemic Lupus Erythematosus and a Proof-of-Concept Trial of Metformin. Arthritis Rheumatol. Hoboken NJ 2015, 67, 3190–3200. [Google Scholar] [CrossRef] [Green Version]
  134. van der Linden, M.; van den Hoogen, L.L.; Westerlaken, G.H.A.; Fritsch-Stork, R.D.E.; van Roon, J.A.G.; Radstake, T.R.D.J.; Meyaard, L. Neutrophil extracellular trap release is associated with antinuclear antibodies in systemic lupus erythematosus and anti-phospholipid syndrome. Rheumatol. Oxf. Engl. 2018, 57, 1228–1234. [Google Scholar] [CrossRef] [Green Version]
  135. Leffler, J.; Gullstrand, B.; Jönsen, A.; Nilsson, J.-Å.; Martin, M.; Blom, A.M.; Bengtsson, A.A. Degradation of neutrophil extracellular traps co-varies with disease activity in patients with systemic lupus erythematosus. Arthritis Res. Ther. 2013, 15, R84. [Google Scholar] [CrossRef] [Green Version]
  136. 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. [Google Scholar] [CrossRef]
  137. Yang, P.; Li, Y.; Xie, Y.; Liu, Y. Different Faces for Different Places: Heterogeneity of Neutrophil Phenotype and Function. J. Immunol. Res. 2019, 2019, 8016254. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  138. Mistry, P.; Nakabo, S.; O’Neil, L.; Goel, R.R.; Jiang, K.; Carmona-Rivera, C.; Gupta, S.; Chan, D.W.; Carlucci, P.M.; Wang, X.; et al. Transcriptomic, epigenetic, and functional analyses implicate neutrophil diversity in the pathogenesis of systemic lupus erythematosus. Proc. Natl. Acad. Sci. USA 2019, 116, 25222–25228. [Google Scholar] [CrossRef]
  139. Odqvist, L.; Jevnikar, Z.; Riise, R.; Öberg, L.; Rhedin, M.; Leonard, D.; Yrlid, L.; Jackson, S.; Mattsson, J.; Nanda, S.; et al. Genetic variations in A20 DUB domain provide a genetic link to citrullination and neutrophil extracellular traps in systemic lupus erythematosus. Ann. Rheum. Dis. 2019, 78, 1363–1370. [Google Scholar] [CrossRef] [Green Version]
  140. Radic, M.; Pattanaik, D. Cellular and Molecular Mechanisms of Anti-Phospholipid Syndrome. Front. Immunol. 2018, 9, 969. [Google Scholar] [CrossRef] [Green Version]
  141. Maugeri, N.; Campana, L.; Gavina, M.; Covino, C.; De Metrio, M.; Panciroli, C.; Maiuri, L.; Maseri, A.; D’Angelo, A.; Bianchi, M.E.; et al. Activated platelets present high mobility group box 1 to neutrophils, inducing autophagy and promoting the extrusion of neutrophil extracellular traps. J. Thromb. Haemost. 2014, 12, 2074–2088. [Google Scholar] [CrossRef] [PubMed]
  142. Yalavarthi, S.; Gould, T.J.; Rao, A.N.; Mazza, L.F.; Morris, A.E.; Núñez-Álvarez, C.; Hernández-Ramírez, D.; Bockenstedt, P.L.; Liaw, P.C.; Cabral, A.R.; et al. Release of Neutrophil Extracellular Traps by Neutrophils Stimulated With Antiphospholipid Antibodies: A Newly Identified Mechanism of Thrombosis in the Antiphospholipid Syndrome. Arthritis Rheumatol. 2015, 67, 2990–3003. [Google Scholar] [CrossRef] [PubMed]
  143. You, Y.; Liu, Y.; Li, F.; Mu, F.; Zha, C. Anti-β2GPI/β2GPI induces human neutrophils to generate NETs by relying on ROS. Cell Biochem. Funct. 2019, 37, 56–61. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  144. Fuchs, T.A.; Brill, A.; Duerschmied, D.; Schatzberg, D.; Monestier, M.; Myers, D.D.; Wrobleski, S.K.; Wakefield, T.W.; Hartwig, J.H.; Wagner, D.D. Extracellular DNA traps promote thrombosis. Proc. Natl. Acad. Sci. USA 2010, 107, 15880. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  145. Woodberry, T.; Bouffler, S.E.; Wilson, A.S.; Buckland, R.L.; Brüstle, A. The Emerging Role of Neutrophil Granulocytes in Multiple Sclerosis. J. Clin. Med. 2018, 7, 511. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  146. Yu, G.; Zheng, S.; Zhang, H. Inhibition of myeloperoxidase by N-acetyl lysyltyrosylcysteine amide reduces experimental autoimmune encephalomyelitis-induced injury and promotes oligodendrocyte regeneration and neurogenesis in a murine model of progressive multiple sclerosis. Neuroreport 2018, 29, 208–213. [Google Scholar] [CrossRef] [Green Version]
  147. Tillack, K.; Naegele, M.; Haueis, C.; Schippling, S.; Wandinger, K.-P.; Martin, R.; Sospedra, M. Gender differences in circulating levels of neutrophil extracellular traps in serum of multiple sclerosis patients. J. Neuroimmunol. 2013, 261, 108–119. [Google Scholar] [CrossRef] [Green Version]
  148. Ostendorf, L.; Mothes, R.; van Koppen, S.; Lindquist, R.L.; Bellmann-Strobl, J.; Asseyer, S.; Ruprecht, K.; Alexander, T.; Niesner, R.A.; Hauser, A.E.; et al. Low-Density Granulocytes Are a Novel Immunopathological Feature in Both Multiple Sclerosis and Neuromyelitis Optica Spectrum Disorder. Front. Immunol. 2019, 10. [Google Scholar] [CrossRef] [Green Version]
  149. Marwick, J.A.; Mills, R.; Kay, O.; Michail, K.; Stephen, J.; Rossi, A.G.; Dransfield, I.; Hirani, N. Neutrophils induce macrophage anti-inflammatory reprogramming by suppressing NF-κB activation. Cell Death Dis. 2018, 9, 665. [Google Scholar] [CrossRef]
  150. Schauer, C.; Janko, C.; Munoz, L.E.; Zhao, Y.; Kienhöfer, D.; Frey, B.; Lell, M.; Manger, B.; Rech, J.; Naschberger, E.; et al. Aggregated neutrophil extracellular traps limit inflammation by degrading cytokines and chemokines. Nat. Med. 2014, 20, 511–517. [Google Scholar] [CrossRef]
  151. de Bont, C.M.; Eerden, N.; Boelens, W.C.; Pruijn, G.J.M. Neutrophil proteases degrade autoepitopes of NET-associated proteins. Clin. Exp. Immunol. 2020, 199, 1–8. [Google Scholar] [CrossRef] [PubMed] [Green Version]
Figure 1. Neutrophil extracellular traps (NETs.) take the central stage in driving autoimmune responses. Abbreviations: AAV: Anti-neutrophil cytoplasmic antibodies associated vasculitis, ANCA: Anti-neutrophil cytoplasmic antibodies, C1q: complement factor 1q, C4: complement factor 3, C4: complement factor 4, DAMPs: damage associated molecular patterns, HMGB1: high mobility group box protein 1, IFN-α: interferon alpha, IFN-γ: interferon gamma, IL: interleukin, LL37: cathelicidin antimicrobial peptides, MSU: monosodium urate crystals, NETs: neutrophil extracellular traps, NLRP3: nucleotide-binding oligomerization domain-like receptor protein 3, RA: rheumatoid arthritis, SLE: systemic lupus erythematosus, TLR9: toll-like receptor 9, TNF-α: tumour necrosis factor alpha.
Figure 1. Neutrophil extracellular traps (NETs.) take the central stage in driving autoimmune responses. Abbreviations: AAV: Anti-neutrophil cytoplasmic antibodies associated vasculitis, ANCA: Anti-neutrophil cytoplasmic antibodies, C1q: complement factor 1q, C4: complement factor 3, C4: complement factor 4, DAMPs: damage associated molecular patterns, HMGB1: high mobility group box protein 1, IFN-α: interferon alpha, IFN-γ: interferon gamma, IL: interleukin, LL37: cathelicidin antimicrobial peptides, MSU: monosodium urate crystals, NETs: neutrophil extracellular traps, NLRP3: nucleotide-binding oligomerization domain-like receptor protein 3, RA: rheumatoid arthritis, SLE: systemic lupus erythematosus, TLR9: toll-like receptor 9, TNF-α: tumour necrosis factor alpha.
Cells 09 00915 g001
Table 1. Neutrophil extracellular traps (NETs)-associated molecules that are known autoantigens in various autoimmune diseases.
Table 1. Neutrophil extracellular traps (NETs)-associated molecules that are known autoantigens in various autoimmune diseases.
Which Autoantigens Are Found on Neutrophil Extracellular Traps (NETs)?To Which Autoimmune Diseases Are These Autoantigens Associated?
α-enolase[10,15]SLE[20]
Annexin A1[10,15]SLE[21,22,23]
RA[22]
Apolipoprotein A1[10]SLE[24]
Bb[25]AAV[25]
C1q[7,26]SLE[27]
Catalase[10]SLE[28]
RA[28]
Cathelicidin[10]SLE[21]
Citrullinated histones[7,18,29,30,31,32]RA[18],
SLE[33]
dsDNA SLE[33,34]
Histones[10]SLE[35]
HMGB1[36,37]SLE[33,36,37]
LAMP-2[38]AAV[39]
LL37[5,7,10,40,41,42]SLE[41]
Psoriasis[43]
MMP8[10]RA[44]
MMP9[10,45]SLE[45]
MPO[17,19,46,47,48,49]AAV[50,51]
PR3[17,19,48]AAV[38]
Properdin[25]AAV[25]
TF[52,53]SLE[53]
Abbreviations: AAV: anti-neutrophil cytoplasmic antibodies (ANCA) vasculitis, Bb: complement factor b, C1q: complement component 1q, HMGB1: high mobility group protein B, LAMP-2: Lysosomal membrane 2 protein, MMP8: matrix metalloproteinases, MMP9: matrix metalloproteinase 9, MPO: myeloperoxidase, PR3: proteinase 3, RA: rheumatoid arthritis, SLE: systemic lupus erythematosus; TF: tissue factor.
Table 2. Potential drug interventions targeting neutrophil extracellular traps (NETs) in autoimmune diseases (in vitro and in vivo evidences).
Table 2. Potential drug interventions targeting neutrophil extracellular traps (NETs) in autoimmune diseases (in vitro and in vivo evidences).
Potential Drug InterventionsMechanismStudy DesignEffect on Neutrophil Extracellular Traps (NET)Autoimmune DiseaseEffect on Disease Clinical EndpointReferences
PAD4
CI-amidinePAD4 enzymes inhibitorIn vitro PMA-induced NETsBlocked NET formationAAVNot tested[90]
Mouse model with MPO–ANCA productionReduced citrullination and reduced serum MPO–ANCA levelAAVNot tested[90]
SLE mouse modelDecreased histone citrullination and reduced release of NETsSLENot tested[91,92]
pGIA mouse modelReduced citrullinated proteinsRAReduced arthritis severity, but not significantly[93]
GSK199Low-calcium PAD4 enzymes inhibitorIn vitro ionomycin-induced mouse neutrophilsInhibition mouse NETs--[94]
In vitro S. aureus-induced human neutrophilsPartial NET formation remained--[94]
Collagen-induced arthritis mouse modelUnknownRAPrevented clinical and histological disease severity[95,96]
DNase
DNaseDNA degradationSLE mouse modelUnknownSLEProlongation of survival[97]
Phase Ib studyUnknownSLEUnaffected disease activity[14,98]
APS IgG treated miceDecreased NET formationAPSDecrease in thrombus formation[99]
Anti-β2-GP1/β2-GP1 treated ratsDecreased NET formationAPSDecrease in thrombus formation[100]
ROS
PRAK inhibitorInhibition of ROS-regulating PRAKIn vitro PMA-induced NETsIncreased neutrophil apoptosis over NET formation--[101]
TroloxAntioxidantIn vitro PMA-induced NETsInhibited ROS-dependent NET formation--[102]
TironAntioxidantIn vitro PMA-induced NETsRemained NET formation--[102]
TempolAntioxidantIn vitro PMA-induced NETsInhibited ROS-dependent NET formation--[102]
Vitamin CUnknownIn vitro PMA-induced NETsDecreased NET formation--[103]
IFN-alpha
SifalimumabBlocks IFN-α NETs stimulationPhase I studyUnknownSLEInhibited type I IFN signature and trend in improved SLEDAI[104,105]
Phase IIb studyUnknownSLEImproved SLE responder rate[106]
RontalizumabBlocks IFN-α NETs stimulationPhase I studyUnknownSLENo effect on IFN and anti-dsDNA levels[107]
AnifrolumabBlocks IFN-α NETs stimulationPhase IIb studyReduced plasma NETs complexesSLEImproved cholesterol efflux capacityWhite et al., 2018 (unpublished, conference abstract)
Phase IIb studyUnknownSLEReduced SLE disease activity[108]
Complement
PA-dPEG24C1 inhibitorIn vitro PMA, MPO or immune complex activated human seraInhibited complement activation and inhibited NET formation--[109]
EculizumabAntibody against c5aPNH patientsDecreased neutrophil activation-Unknown[110]
Proteases
IcatCCatC inhibition blocks PR3 activity and NET formationNeutrophil differentiated CD34+ HSCIcatC leads to absence of PR3 and suppression of PR3-ANCA antigen--[111,112]
ChloroquineInhibits autophagyAP patients NETsDecreased NET formation--[113]
AP mouse modelDecreased NET formation and improved survival--[113]
Vitamin D
Vitamin DUnknownIn vitro PMA-induced NETs and endothelial cellsReduced NET formationSLEReduced endothelial apoptosis[114]
Vitamin DUnknownSLE patients with low vitamin DUnknownSLEImproved endothelial function[115]
Nanoparticles
A2,8-sialylated nanoparticlesReduce PMA-initiated ROS productionIn vitro PMA-induced NETsInhibited NET release--[116]
Polysialylated vesiclesCounteract cytotoxic characteristic of extracellular histones, possibly through lactoferrin5B8 cells in presence of histonesReduced cytotoxicity reduced in 5B8 cells. Vesicles bind to PMA–NETs--[117,118]
Existing autoimmune disease therapies and their effects on NETs
-TocilizumabAntibody against IL-6 receptorIn vitro IL-6 and PMA-induced NETsBlocked NET formationRAUnknown[119]
Rituximab and belimumabBlocking IC formationPhase IIa studyReduced NET formationRADecreased lupus disease activity[120]
IVIG-SMechanisms unknownMPO-AAV rat modelReduced NET formation and ANCA titersAAVUnknown[121]
Abbreviations: AAV: Anti-neutrophil cytoplasmic antibodies associated vasculitis, ANCA: Anti-neutrophil cytoplasmic antibodies, AP: acute pancreatitis, APS: antiphospholipid syndrome, CatC: cathepsin C, CI-amidine: chloramidine, dsDNA: double strand DNA, EA: elastase-alpha1-antitrypsin, GSK199: hydrochloride, HSC: hematopoietic stem cells, IC: immune complex, IcatC: inhibitor cathepsin C, IFN: interferon, IVIG-S: sulfo-immunoglobulins, MPO: myeloperoxidase, NAC: n-acetylcysteine, NETs: neutrophil extracellular traps, PAD4: peptidylarginine deiminase 4, pGIA: glucose 6-phosphate isomerase induced arthritis, PMA: phorbol myristate acetate, PNH: paroxysmal nocturnal haemoglobinuria, PR3: proteinase 3, PRAK: p38-regulated/activated protein kinase, RA: rheumatoid arthritis, SLE: systemic lupus erythematosus, SLEDAI: systemic lupus erythematosus disease activity index, β2-GP1: β2-glycoprotein.

Share and Cite

MDPI and ACS Style

Fousert, E.; Toes, R.; Desai, J. Neutrophil Extracellular Traps (NETs) Take the Central Stage in Driving Autoimmune Responses. Cells 2020, 9, 915. https://doi.org/10.3390/cells9040915

AMA Style

Fousert E, Toes R, Desai J. Neutrophil Extracellular Traps (NETs) Take the Central Stage in Driving Autoimmune Responses. Cells. 2020; 9(4):915. https://doi.org/10.3390/cells9040915

Chicago/Turabian Style

Fousert, Esther, René Toes, and Jyaysi Desai. 2020. "Neutrophil Extracellular Traps (NETs) Take the Central Stage in Driving Autoimmune Responses" Cells 9, no. 4: 915. https://doi.org/10.3390/cells9040915

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop