Neutrophils are an absolutely essential part of the innate immune system, playing an essential role in the control of infectious diseases but more recently are also being viewed as important players in tissue repair. Neutrophils are able to counteract an infection through phagocytosis and/or the release of neutrophil extracellular traps (NETs). By contrast, neutrophils help repair damaged tissues, limiting NET production but still phagocytosing debris. However, when inflammation is recurrent, or the inciting agent persists, neutrophils through a frustrated inability to resolve the problem can release NETs to exacerbate tissue damage during inappropriate inflammation. In this review, we discuss the mechanisms of NET formation, as well as the apparent paradoxical role of neutrophils and NETs in host defense, chronic inflammation, and tissue disrepair.

Neutrophils are the predominant leukocyte population in human blood and among the first cells recruited to an inflammatory site. During an infection or tissue damage, pathogen-associated molecular patterns and/or damage-associated molecular patterns (DAMPs) are sensed by pattern recognition receptors as well as DAMP receptors, which can activate resident cells to produce inflammatory mediators, such as chemokines CXCL1 and CXCL2, which bind to and activate G-protein–coupled receptors on neutrophils.1  In addition, DAMPs and pathogen-associated molecular patterns may also directly activate receptors on neutrophils to induce recruitment.2  Neutrophils display a wide range of effector mechanisms to counteract pathogens that include phagocytosis and the production of reactive oxygen species (ROS), proteases, and neutrophil extracellular traps (NETs).3  Thus, impairment of neutrophil recruitment to an infectious nidus facilitates pathogen dissemination into blood and vital organs, development of systemic infection, and eventual death.4,5  This is best exemplified by the fact that neutropenia is the biggest risk factor for infection.6 

During sterile tissue injuries, neutrophils are also essential to participate in the clearance of cellular debris, returning tissue to homeostasis.7  From an evolutionary standpoint, it would seem reasonable to conclude that the system has been optimized to allow for bacterial clearance while minimizing surrounding collateral damage. However, pathogens have also evolved mechanisms to subvert the immune system, causing neutrophils to respond in a more aggressive manner, leading to untoward tissue injury.8  Moreover, lack of resolution or persistence of sterile injury may also lead to inappropriate inflammation.9  This type of persistent injury is very different from normal repair and involves either the presence of foreign substances for which our immune system has not evolved an appropriate response (eg, recurrent smoke inhalation, prolonged alcohol and drug overdose, inappropriate high-fat or high-cholesterol diet) and surgical procedures, including transplantation, ischemia reperfusion, or foreign body implantation. We would also include among these nonevolutionary situations, experimental models created by investigators to study inappropriate inflammation, including exposure of animals to carbon tetrachloride, bleomycin, acetaminophen (Tylenol), and many other experimental substances. It is under these different conditions that the neutrophils, our greatest allies, in an attempt to eradicate what they perceive as danger, respond overexuberantly or inappropriately, causing bystander tissue damage and contributing to immunopathology. In this review, we describe the mechanisms underlying NET formation in response to natural stimuli like bacteria and also highlight the paradoxical role of neutrophils and NETs in unnatural situations.

Until the year 2004, phagocytosis and subsequent oxidant- and protease-dependent killing of pathogens inside of phagolyososomes were thought to be the major neutrophil effector mechanisms. This concept was changed by Brinkmann and colleagues,10  who showed that neutrophil stimulation with phorbol myristate acetate (PMA) released weblike structures of DNA coated with histones, elastase, myeloperoxidase (MPO), and cathepsin G. These structures were given the name neutrophil extracellular traps (NETs), which were initially proposed to immobilize or trap and kill bacteria extracellularly.10  Although in that first publication there was no mention of neutrophil death during NET release, the preface to that paper suggested the name NETosis, implying neutrophil death.11  The same investigators in subsequent work avidly dissected the role of NETosis using PMA and showed NET release was associated with the rupture of the neutrophil.12  As such, NETosis was further characterized as a cell death program distinct from apoptosis and necrosis and dependent on the generation of ROS by reduced nicotinamide adenine dinucleotide phosphate (NADPH) oxidase.11,12  PMA-induced NETosis was dependent on plasma membrane lysis, which followed chromatin decondensation and chromatin mixing with cytoplasmic granule proteins.12  As such, NETosis was retained and was identified by extracellular DNA decorated with various neutrophil-derived granular proteins, including MPO and elastase. Although many groups have reproduced NETosis with PMA in vitro, to our knowledge, no one has been able to visualize exploding or NETosing neutrophils in vivo (despite sincere attempts).

By contrast, in parallel studies in vitro, it was demonstrated that NET formation induced by natural stimuli can occur entirely independent of cell lysis and subsequent cell death.13,14  Although the composition of NETs is assumed to be similar regardless of stimulus, proteomics have only been done on PMA-induced NETs. However, 1 group showed that nonlytic NET release was mitochondrial15  not nuclear, and by definition, these NETs would lack histones, a major antimicrobial and host cell toxin. A second group using primarily bacteria or bacterial products with or without platelets reported that nonlytic NETs were made of nuclear not mitochondrial DNA, which had reduced proteolytic activity.13,14  It is our personal view that PMA likely activates many pathways in neutrophils and en masse causes NET formation, oxidant production as well as a necrotic cell death that may complicate the study of NET release. In fact, NETs can be detected at 30 minutes with PMA, whereas cell death occurs at 3 hours, at which point an unregulated release of all intracellular contents is seen and perhaps makes NETs much more toxic. Alternatively, PMA-induced NET release and PMA-induced lysis are causally unrelated events. As a final point, mainly for clarity, rather than using the term lytic and nonlytic NETosis (the latter being a contradiction in terms), we will use the term lytic and nonlytic NET formation.

Lytic NET formation

NET formation followed by cell death or lytic NET release was the first to be described (Figure 1A). Stimulation of neutrophils with PMA resulted in the activation of NADPH oxidase, via PKC and Raf-MEK-ERK signaling pathway and consequent ROS generation.16  This activated protein-arginine deiminase 4 (PAD4), which hypercitrullinated histones, causing chromatin decondensation,17,18  while simultaneously MPO and neutrophil elastase (NE) were released from cytoplasmic azurophilic granules.19  Overall, 24 different proteins have been described in the NETome.20  MPO was reported to bind to chromatin and activate NE in small azurosome structures that can be seen in vitro as well as in vivo.21  NE degraded actin filaments in the cytoplasm, translocated to the nucleus, and cleaved histones.21  Subsequently, the nuclear envelope broke down via cell-cycle proteins,22  releasing the chromatin in the cytosol, which mixed with cytosolic proteins.12  The mechanism involved in cell lysis and NET release involves gasdermin D (GSDMD),23,24  a recently identified factor that mediates pyroptosis in macrophages.25  During PMA-inducing NET release, NE cleaves GSDMD to its active form (GSDMD-NT),26  which forms pores in the plasma membrane and granule membranes, enhancing NE and other granule content release.23 

Figure 1.

Mechanisms of NET formation. (A) PMA and other stimuli induce lytic-NET formation. Stimulation of neutrophils with PMA resulted in the activation of NADPH oxidase, via PKC and Raf-MEK-ERK signaling pathway and consequent ROS generation. Afterward, PAD4 is activated and citrullinates arginine on histones causing chromatin decondensation. MPO and NE are released from cytoplasmic azurophilic granules and then translocated to the nucleus contributing to unfolding of chromatin. Subsequently, the nuclear envelope broke down, releasing the chromatin in the cytosol, which mixed with cytosolic proteins. NE also cleaves GSDMD in the cytosol to its active form (GSDMD-NT), which, besides forming pores in the plasma membrane, also mediates pore formation in nuclear and granule membranes, enhancing NE and other granular content release. Finally, NETs are released, and the neutrophil dies. (B) Nonlytic NET formation is induced by the recognition of stimuli through Toll-like receptor 2 (TLR2), TLR4, or complement receptors independent of NAPDH oxidase activation. S aureus and C albicans activate TLR2 and complement receptors, respectively, and E coli or LPS-activated platelets activate TLR4. Along with PAD4 activation and NE translocation to the nucleus, chromatin decondensation proceeds and protein-decorated chromatin is expelled via vesicles without plasma membrane disruption. After the release of NETs, neutrophils are still alive for further functions.

Figure 1.

Mechanisms of NET formation. (A) PMA and other stimuli induce lytic-NET formation. Stimulation of neutrophils with PMA resulted in the activation of NADPH oxidase, via PKC and Raf-MEK-ERK signaling pathway and consequent ROS generation. Afterward, PAD4 is activated and citrullinates arginine on histones causing chromatin decondensation. MPO and NE are released from cytoplasmic azurophilic granules and then translocated to the nucleus contributing to unfolding of chromatin. Subsequently, the nuclear envelope broke down, releasing the chromatin in the cytosol, which mixed with cytosolic proteins. NE also cleaves GSDMD in the cytosol to its active form (GSDMD-NT), which, besides forming pores in the plasma membrane, also mediates pore formation in nuclear and granule membranes, enhancing NE and other granular content release. Finally, NETs are released, and the neutrophil dies. (B) Nonlytic NET formation is induced by the recognition of stimuli through Toll-like receptor 2 (TLR2), TLR4, or complement receptors independent of NAPDH oxidase activation. S aureus and C albicans activate TLR2 and complement receptors, respectively, and E coli or LPS-activated platelets activate TLR4. Along with PAD4 activation and NE translocation to the nucleus, chromatin decondensation proceeds and protein-decorated chromatin is expelled via vesicles without plasma membrane disruption. After the release of NETs, neutrophils are still alive for further functions.

Close modal

Studies demonstrated that the mechanism of NET formation could vary depending on the initial stimulus that activates neutrophils. Interestingly, NET formation is inhibited in the absence of NADPH oxidase in both patients with chronic granulomatous diseases27  and in knockout mice28  stimulated with Aspergillus nidulans or PMA, respectively. However, during neutrophil stimulation with Staphylococcus aureus,14  NET formation was independent of oxidant production. This was subsequently shown to also be the case in Candida albicans29 ; ionomycin30,31  and nicotine32  induced NET release where the process occurred independent of NADPH oxidase. However, the mechanism proposed for NET formation by nicotine32  and ionomycin31  was dependent on AKT signaling and calcium-induced mitochondrial ROS-release (ie, oxidant dependent but via a different mechanism). Numerous studies have also demonstrated that the pharmacological or genetic blockade of MPO and PAD4 reduce or impair NET release.19,33,34  In contrast, NET formation in response to Pseudomonas aeruginosa seems to be independent of MPO activity,30  whereas C albicans,35 Klebsiella pneumoniae,36  and cholesterol crystals37  induce NET release independently of PAD4. Recent data indicate that histone citrullination is not enough to promote chromatin decondensation. The inhibition of NE blocked chromatin decondensation without interfering with histone citrullination.38  PMA required PKC to induce NETs, while Helicobacter pylori–induced NETs were independent of PKC.16  Altogether, the mechanisms by which NETs were formed depended on the stimulus. However, such huge discrepancies beg the question whether different processes are being studied.

Nonlytic NET release

The time required for lytic NET formation has been reported to occur primarily at 3 to 4 hours. By contrast, neutrophils can also release NETs in a very rapid (5-60 minutes) and cell death–independent manner (Figure 1B). Indeed, neutrophils bound by lipopolysaccharide (LPS)-activated platelets in vivo or in vitro formed NETs within a few minutes but restricted Sytox green entry, a live cell-impermeant nucleic acid staining dye.13  During human sepsis, nonlytic NET release occurred via TLR4 activation of platelets that then bound to the neutrophils. Similar results were seen when Escherichia coli was administered in vivo, with platelets immediately tethering to neutrophils to induce NET formation while lysis was not observed.39  Other bacteria also induced nonlytic NET release. Mechanistically, it was demonstrated that NADPH oxidase was not required during nonlytic NET formation in response to S aureus,14  and neutrophils were still able to migrate and phagocytose.40,41  In addition, nonlytic NET formation required specific receptors, including activation of TLRs and complement receptors during infection with S aureus40  and C albicans.29  Moreover, human neutrophils primed with granulocyte macrophage colony–stimulating factor and subsequently stimulated with LPS or complement factor 5a for a short period also released NETs, but in a process dependent on mitochondrial, instead of nuclear, DNA.15  Interestingly, optic atrophy 1, a mitochondrial inner membrane protein that is important for mitochondrial biogenesis, was also important for NETosis through the stimulation of microtubule network formation and subsequent DNA release.42  In a recent study, that in our opinion provides important insight regarding NET formation, Branzk and colleagues38  demonstrated that neutrophils were able to sense microbe size and selectively released NETs in response to large pathogens, such as C albicans hyphae, but not in response to small yeast or single bacterium. The mechanism reported in this study was that phagocytosis prevented NET release through downregulation of NE translocation to the nucleus.38  This would likely extend to bacteria like S aureus that in vivo are found as clumps and to biofilms,43  which would both induce NET release rather than phagocytosis. It is also worth mentioning that NET release can be prevented by phagocytosis of platelets,44  suggesting that regardless of the particulate matter that is phagocytosed, NET release will be blocked. It also explains why some but not other neutrophils make NETs in infections and why neutrophils that phagocytose bacteria do not subsequently release bacteria during lytic NET formation.

The work with PMA has informed studies on nonlytic NET formation. For example, NE is also translocated to the nucleus during nonlytic NET formation, and PAD4 is activated, inducing chromatin decondensation.45  However, instead of plasma membrane disruption for NET release, protein-decorated chromatin is secreted via vesicles, allowing neutrophils to stay alive for further functions.40  Because neutrophils have very low to nonexistent transcriptional activity, loss of the nucleus did not impair processes such as phagocytosis, release of cytotoxic molecules, or motility, although the latter was altered because neutrophils use the nucleus as a fulcrum during crawling.40  Nevertheless, much like their close relatives red blood cells and platelets, neutrophils devoid of nuclei, known as cytoplasts, still performed important functions.46  Recently, it was demonstrated that cytoplasts derived from neutrophils that had released NETs were able to activate lung dendritic cells to differentiate naive CD4+ T cells to antigen-specific T-helper 17 effectors in a murine model of severe asthma, identifying a potential pathogenic role for these cellular remnants.47  Using sandwich enzyme-linked immunosorbent assays employing antibodies against MPO and N-terminal histone tails as a measure of nonlytic NETs, it was demonstrated that circulating NETs from septic patients are derived from an NADPH oxidase–independent nonlytic pathway,48  corroborating previous findings from experimental sepsis.13,39  However, nonlytic NET formation has been less studied and (1) how vesicles of DNA get formed and released, (2) what happens to anuclear neutrophils/cytoplasts, and (3) what are the signaling pathways need to be determined.

There has been a large effort to identify neutrophil subsets and delineate which ones release NETs. Indeed, not all neutrophils release NETs; only 20% to 25% of neutrophils release NETs after S aureus stimulation.41  Circulating neutrophils can be easily separated from peripheral blood mononuclear cells by density gradient differences after centrifugation. However, during an inflammatory process, a population of neutrophils with altered density colocalizes with peripheral blood mononuclear cells density fractions, which have been called low-density neutrophils (LDNs).49  LDNs are a heterogeneous population containing both immature and mature neutrophils, and their functions differ depending on the inflammatory stimulus. Interestingly, it has been demonstrated that LDNs have an increased capacity to generate NETs in autoimmune diseases50,51  and in a model of spontaneous small intestinal tumors,52  raising the possibility that these are a distinct proinflammatory neutrophil subset.53  Numerous molecular markers have been proposed to delineate different neutrophil populations with respect to NET production. CD177 neutrophils stimulated with LPS were not able to release NETs.54,55  Olfactomedin 4 (OLFM4), a matrix glycoprotein predominantly found within specific granules, is also differentially expressed on 10% to 30% of neutrophils,56  aligning nicely with the percentage of neutrophils that make NETs. NET formation leads to OLFM4 secretion,57  and a recent study demonstrated that septic patients with a high percentage of OLFM4+ neutrophils were at higher risk of organ failure and death.58  However, direct evidence linking OLFM4-positive neutrophils and NET releasing neutrophils has yet to be confirmed. As such, molecular markers that designate NET-producing neutrophils are not available, begging the question whether NET-producing neutrophils are simply older, more mature, or more primed neutrophils, but all 1 population.59,60 

During infection, NETs mediate host defense by trapping and killing microorganisms.41  Indeed, the first study that described NETs demonstrated that NETs were important for the sequestration and killing of bacteria by delivering a high local concentration of antimicrobial molecules.10  Subsequent studies confirmed that NETs were also formed in vivo, mediating the trapping of bacteria. Using in vivo intravital microscopy, it was possible to visualize NET trapping of E coli in hepatic sinusoids, and it was demonstrated that the disruption of NETs resulted in the spread of the bacteria systemically.13,39  In addition, the systemic treatment of S aureus–infected mice with DNase also resulted in the spread of bacteria from the infection site to the circulation.40  Likewise, fungi61,62  and virus63  were visualized being trapped by NETs in vivo and in vitro. C albicans61  and Aspergillus fumigatus62  were trapped by human neutrophil-released NETs and by NETs formed in the lung of infected mice, respectively; PMA-activated human neutrophils were also able to capture HIV-1 in vitro, an event inhibited after DNase treatment.63  However, the molecular mechanisms by which NETs bind and trap microorganisms are poorly understood, although charge has been proposed.12  Once in a NET, there is growing evidence that this structure is capable of killing bacteria. Several studies have demonstrated that NETs kill microorganisms in vitro through the action of microbicidal components. Indeed, histones have a potent antimicrobial capacity.64  Moreover, NE actively targets bacterial virulence factors from Shigella flexneri, culminating in bacteria killing in vitro.65  Calprotectin is also present in NETs, and its inhibition reduced the antifungal activity of NETs in vitro.20 

Not surprisingly, as is the case with any important mechanism, pathogens have learned to counter and/or subvert NET production to favor their survival. Indeed, some of the earliest studies on NETs revealed that microbial DNase was a virulence factor that could help pathogens escape NETs, making them more apt to disseminate.66  Moreover, pathogens have learned to express virulence factors that suppress or induce NET release. Recently, it was demonstrated that pore-forming leukocidins PVL and HlgAB, toxins released from S aureus biofilms, induce both neutrophil death and NET formation. Whether this was a form of lytic NET formation or just neutrophil lysis due to these pore-forming molecules was difficult to delineate. Nevertheless, the NET release was implicated in the persistence of S aureus biofilms in vitro and in a chronic model of skin infection in vivo.8 

Acute inflammation

However, like all processes, too many NETs in infection can be detrimental to the host. One of the earliest studies on NETs showed that the interaction between neutrophils and activated platelets induced NET formation, and this led to endothelial cell damage and organ injury after E coli infection.13  Other studies have also demonstrated NET-induced tissue injury.39,67  Acute respiratory distress syndrome (ARDS) is a life-threatening disorder characterized by widespread inflammatory lung injury often caused by neutrophil-pathogen interactions. The partial reduction of NETs by DNase I treatment or partial PAD4-deficiency (Pad4+/−) reduced acute lung injury induced by bacteria and improved survival, whereas complete NET inhibition by PAD4 deficiency (Pad4−/−) reduced lung injury, but was counterbalanced by increased bacterial load and inflammation.68  There is growing evidence that histones in NETs may be the cytotoxic component that harms the endothelium and epithelium. Interestingly, the cytotoxic activity of NETs on lung epithelial cells was suppressed after histone, but not DNA inhibition.69  NETs contribute to hepatic damage during bloodstream infection with methicillin-resistant S aureus.70  However, DNase treatment only partially reduced the injury, because DNase failed to remove histones that were attached to the vessel wall via von Willebrand factor. Indeed, S aureus–induced tissue damage was almost abolished in PAD-4−/− and Elane−/−(NE−/−) mice, whose neutrophils are unable to release NETs.70  Corroborating these data, it was demonstrated that during NADPH oxidase-dependent NET formation, elastase degraded the N-terminal histone tail within NETs, which only happened during lytic NET release.48  Moreover, nonlytic NET release exhibited increased immunostimulatory effects on endothelial cells compared with lytic NETs, suggesting that the N-terminal histone tail could be responsible for cytotoxicity to host cells.48  Histones are also involved with the formation of microaggregates in the circulation. The activation of TLR by extracellular histone proteins leads to the generation of thrombin and activation of platelets, resulting in microaggregates that contribute to organ damage.71  Importantly, NETs form clots in the circulation of patients and mice with sepsis, an emerging noncanonical mechanism for vascular occlusion and organ damage.72 

The role of neutrophils and NETs during sepsis remains enigmatic. It is well known that the failure of neutrophil migration to the infectious nidus is associated with dissemination and sepsis. However, the systemic activation of neutrophils results in their accumulation in secondary organs, such as lung, leading to bystander organ damage by mechanisms that include NETs.5,39  Therefore, the inhibition of NETs may lead to beneficial and detrimental outcomes. The depletion of NETs by recombinant human DNase delayed bacterial clearance and aggravated the pathology during polymicrobial sepsis in mice.73 Pad4−/− mice exhibited some protection from LPS-induced endotoxemia, suggesting that NETs do cause damage in this model74 ; however, this is not a live microbial infection. Moreover, no protection and no effect on bacteremia were noted from PAD4-deficient mice exposed to polymicrobial sepsis.75  The treatment of mice with DNase or histone-neutralizing antibodies, in association with antibiotics, reduced organ damage and improved survival of mice submitted to polymicrobial sepsis.76,77  In addition, NET-containing HMGB-1 induced peritoneal macrophage pyroptosis (a form of cell death) through the activation of caspase-1.78  Although these studies indicate that the combination of microbicidal agents with inhibitors of NET activity could be a possible strategy to minimize the detrimental tissue damage caused by NETs during sepsis, with so many different toxic products it seems inconceivable that DNase alone would be sufficient to reduce tissue damage.

Little is known about NET clearance, and it is proposed that endogenous DNase simply chops up NETs. However, this could potentially release the remaining toxic NET proteins. Perhaps other mechanisms of clearance exist. Monocyte-derived macrophages from healthy but not ARDS patients efficiently phagocytose NETs and apoptotic neutrophils.78  In addition, activation of AMP-activated protein kinase, which is a metabolic sensor that regulates cellular energy production in macrophages, or neutralization of HMGB1 in bronchial-alveolar lavage fluids improved efferocytosis and NET clearance. This represents an important strategy to limit the exacerbated inflammatory response and organ damage induced by NETs during ARDS.

Chronic inflammation

Several chronic inflammatory diseases are also characterized by a sustained influx of neutrophils, and persistent NET release. During respiratory chronic diseases, such as cystic fibrosis (CF) and chronic obstructive pulmonary disease (COPD),80  neutrophils and NETs contribute to the reduction of pulmonary function by blocking airways.81  Chronic infection of the lungs is associated with sputum that is rich in neutrophil proteins and DNA, which are thought to arise from NETs.82  Indeed, persistent NET formation has been found in patients with CF and COPD, which is associated with inflammation and disease severity.80,83-85  Although there is a prevalence of recurrent infection in patients with CF and COPD, whether NETs are detrimental or beneficial remains unclear.86  During CF, P aeruginosa is often present in the lung of patients and can induce NET formation. However, a number of clinical isolates of P aeruginosa have been shown to be resistant to NET-mediated killing, and the NETs appear to contribute to the generation of fibrotic areas in the lung, which may become a replicative niche for the bacteria.87  Neutrophils from CF subjects live longer due to decreased apoptosis and form more NETs.88  Apart from causing direct damage, NETs also provide proinflammatory stimuli to macrophages, boosting inflammation in CF subjects.88  Interestingly, inhaled recombinant DNase treatment helped to solubilize sputum from CF patients and improved lung function in CF mice.89  However, DNase treatment carries the risk of liberating highly active enzymes and toxic molecules like histones, which can damage the lung epithelium.90  Therefore, despite its widespread use to help clear airways, other benefits or detrimental side effects need to be further assessed.

Neutrophils and NETs may also play critical roles during autoimmune diseases.45  These inflammatory diseases are defined as pathological conditions in which the immune system is intolerant to autoantigens, leading to effector mechanisms, such as autoantibodies and autoreactive lymphocytes. This deteriorates tissues progressively and culminates in organ failure and death. NETs may play significant roles in the initiation phase of autoimmune disorders by exposing intracellular endogenous components to the immune system, which exacerbates inflammation or even results in the production of autoantibodies.45  Patients with systemic lupus erythematosus (SLE) and rheumatoid arthritis exhibited elevated levels of NETs in the serum and in the synovial fluid, respectively.51,91,92  Interestingly, those patients but not healthy controls have a distinct low-density granulocyte population in the circulation with enhanced capacity to produce NETs spontaneously. Accordingly, SLE patients have high levels of anti-ribonucleoprotein and anti-DNA antibodies in their serum, and rheumatoid arthritis patients present autoantibodies directed against citrullinated proteins, such as histones.93,94 

The role of NETs during SLE is complicated. Blocking mitochondrial ROS production blocked NET formation in a mouse model of lupus and reduced disease severity.53  However, mice that do not express NADPH oxidase exhibit more severe disease.95  This apparent contradiction can be explained by a study showing that circulating NETs from patients with lupus erythematosus are derived from nonlytic NET formation, that form independent of NADPH oxidase.48  This may explain why individuals with chronic granulomatous disease, who fail to produce ROS through NADPH oxidase, might still make NETs and have a propensity for developing SLE. In addition, a subset of patients with lupus has an accumulation of NETs due to a DNase impairment and a reduction in DNA clearance.91  Although some of these diseases clearly suggest neutrophil-derived autoantigens as potential mediators, there are studies with opposing results, and it certainly is unclear whether targeting NETs once the disease is established will be a viable medical intervention.

It is always worth identifying situations where a process does not occur. Indeed, during healthy wound repair, neutrophils are involved but do not make NETs.96  After the release of DAMPs by damaged cells, neutrophils are recruited to the site of injury, where they remove cellular debris.7  They also harbor enzymatic activity (matrix metalloproteinases that activate vascular endothelial growth factor) that is important for the revascularization of damaged tissues and/or for the recruitment or activation of repair promoting cells. After executing their functions, neutrophils must be cleared either by macrophages that leads to the release of anti-inflammatory cytokines7  or by reentering the vasculature, in a process called reverse migration, homing to the bone marrow, where they are thought to die by apoptosis.97  It is likely critical that neutrophils are not left to linger in these inflammatory sites because this has been shown to lead to poor healing.97  During a normal healing process, neutrophils release very few if any NETs.96  In diabetes, NETs may delay wound healing,98  whereas NET aggregates may actually contribute to the resolution of inflammation by degrading cytokines and chemokines in a murine model of gout.99  This mechanism could account for the spontaneous remission of acute inflammatory attacks elicited by monosodium urate crystals in individuals with gout.99 

As such, inhibiting NETs may benefit in some diseases, but we have evolved this system to help us survive, suggesting according to Darwin’s theory of evolution that NET production has been selected for, and as such, the good properties of NETs likely outweigh the bad. Of course, many of the proposed detrimental effects of NETs occur in diseases caused by bad behavior practiced by the past 2 to 3 generations (fat rich diets, smoking, alcohol, etc), making these “nonevolutionary diseases.” Even so, we will need to identify intelligent ways of preventing NET release and not just getting rid of DNA and hoping that magically histones and proteases will also disappear. Finally, it is our view that the host has evolved to make NETs to help trap and kill pathogens, and it is sufficiently important that pathogens have evolved the production of DNases in an attempt to thwart this important antimicrobial defense mechanism. Interfering with this pathway could help pathogens evade host immunity leading to dire consequences.

The authors thank Luiz Gustavo Gardinassi and Gustavo Quirino for proofreading the manuscript and Bruna Araujo David for providing assistance with the figure.

This work is supported by grants from the Canadian Institutes of Health Research, the Heart and Stroke Foundation of Canada, and the Canada Research Chairs program.

Contribution: F.V.S.C. and P.K. wrote, discussed, and reviewed the manuscript.

Conflict-of-interest disclosure: The authors declare no competing financial interests.

Correspondence: Paul Kubes, Calvin, Phoebe, and Joan Snyder Institute for Chronic Diseases, University of Calgary, HRIC 3280 Hospital Dr NW, Calgary, AB T2N 4N1, Canada; e-mail: pkubes@ucalgary.ca.

1.
Kolaczkowska
E
,
Kubes
P
.
Neutrophil recruitment and function in health and inflammation
.
Nat Rev Immunol
.
2013
;
13
(
3
):
159
-
175
.
2.
Pittman
K
,
Kubes
P
.
Damage-associated molecular patterns control neutrophil recruitment
.
J Innate Immun
.
2013
;
5
(
4
):
315
-
323
.
3.
Nathan
C
.
Neutrophils and immunity: challenges and opportunities
.
Nat Rev Immunol
.
2006
;
6
(
3
):
173
-
182
.
4.
Alves-Filho
JC
,
Spiller
F
,
Cunha
FQ
.
Neutrophil paralysis in sepsis
.
Shock
.
2010
;
34
(
suppl 1
):
15
-
21
.
5.
Sônego
F
,
Castanheira
FVeS
,
Ferreira
RG
, et al
.
Paradoxical roles of the neutrophil in sepsis: protective and deleterious
.
Front Immunol
.
2016
;
7
(
APR
):
155
.
6.
Bouma
G
,
Ancliff
PJ
,
Thrasher
AJ
,
Burns
SO
.
Recent advances in the understanding of genetic defects of neutrophil number and function
.
Br J Haematol
.
2010
;
151
(
4
):
312
-
326
.
7.
Wang
J
.
Neutrophils in tissue injury and repair
.
Cell Tissue Res
.
2018
;
371
(
3
):
531
-
539
.
8.
Bhattacharya
M
,
Berends
ETM
,
Chan
R
, et al
.
Staphylococcus aureus biofilms release leukocidins to elicit extracellular trap formation and evade neutrophil-mediated killing
.
Proc Natl Acad Sci USA
.
2018
;
115
(
28
):
7416
-
7421
.
9.
Kovtun
A
,
Messerer
DAC
,
Scharffetter-Kochanek
K
,
Huber-Lang
M
,
Ignatius
A
.
Neutrophils in tissue trauma of the skin, bone, and lung: two sides of the same coin
.
J Immunol Res
.
2018
;
2018
:
1
-
12
.
10.
Brinkmann
V
,
Reichard
U
,
Goosmann
C
, et al
.
Neutrophil extracellular traps kill bacteria
.
Science
.
2004
;
303
(
5663
):
1532
-
1535
.
11.
Steinberg
BE
,
Grinstein
S
.
Unconventional roles of the NADPH oxidase: signaling, ion homeostasis, and cell death
.
Sci STKE
.
2007
;
2007
(
379
):
pe11
.
12.
Fuchs
TA
,
Abed
U
,
Goosmann
C
, et al
.
Novel cell death program leads to neutrophil extracellular traps
.
J Cell Biol
.
2007
;
176
(
2
):
231
-
241
.
13.
Clark
SR
,
Ma
AC
,
Tavener
SA
, et al
.
Platelet TLR4 activates neutrophil extracellular traps to ensnare bacteria in septic blood
.
Nat Med
.
2007
;
13
(
4
):
463
-
469
.
14.
Pilsczek
FH
,
Salina
D
,
Poon
KKH
, et al
.
A novel mechanism of rapid nuclear neutrophil extracellular trap formation in response to Staphylococcus aureus
.
J Immunol
.
2010
;
185
(
12
):
7413
-
7425
.
15.
Yousefi
S
,
Mihalache
C
,
Kozlowski
E
,
Schmid
I
,
Simon
HU
.
Viable neutrophils release mitochondrial DNA to form neutrophil extracellular traps
.
Cell Death Differ
.
2009
;
16
(
11
):
1438
-
1444
.
16.
Hakkim
A
,
Fuchs
TA
,
Martinez
NE
, et al
.
Activation of the Raf-MEK-ERK pathway is required for neutrophil extracellular trap formation
.
Nat Chem Biol
.
2011
;
7
(
2
):
75
-
77
.
17.
Wang
Y
,
Li
M
,
Stadler
S
, et al
.
Histone hypercitrullination mediates chromatin decondensation and neutrophil extracellular trap formation
.
J Cell Biol
.
2009
;
184
(
2
):
205
-
213
.
18.
Li
P
,
Li
M
,
Lindberg
MR
,
Kennett
MJ
,
Xiong
N
,
Wang
Y
.
PAD4 is essential for antibacterial innate immunity mediated by neutrophil extracellular traps
.
J Exp Med
.
2010
;
207
(
9
):
1853
-
1862
.
19.
Papayannopoulos
V
,
Metzler
KD
,
Hakkim
A
,
Zychlinsky
A
.
Neutrophil elastase and myeloperoxidase regulate the formation of neutrophil extracellular traps
.
J Cell Biol
.
2010
;
191
(
3
):
677
-
691
.
20.
Urban
CF
,
Ermert
D
,
Schmid
M
, et al
.
Neutrophil extracellular traps contain calprotectin, a cytosolic protein complex involved in host defense against Candida albicans
.
PLoS Pathog
.
2009
;
5
(
10
):
e1000639
.
21.
Metzler
KD
,
Goosmann
C
,
Lubojemska
A
,
Zychlinsky
A
,
Papayannopoulos
V
.
A myeloperoxidase-containing complex regulates neutrophil elastase release and actin dynamics during NETosis
.
Cell Reports
.
2014
;
8
(
3
):
883
-
896
.
22.
Amulic
B
,
Knackstedt
SL
,
Abu Abed
U
, et al
.
Cell-Cycle proteins control production of neutrophil extracellular traps
.
Dev Cell
.
2017
;
43
(
4
):
449
-
462.e5
.
23.
Sollberger
G
,
Choidas
A
,
Burn
GL
, et al
.
Gasdermin D plays a vital role in the generation of neutrophil extracellular traps
.
Sci Immunol
.
2018
;
3
(
26
):
eaar6689
.
24.
Chen
KW
,
Monteleone
M
,
Boucher
D
, et al
.
Noncanonical inflammasome signaling elicits gasdermin D–dependent neutrophil extracellular traps
.
Sci Immunol
.
2018
;
3
(
26
):
eaar6676
.
25.
Shi
J
,
Zhao
Y
,
Wang
K
, et al
.
Cleavage of GSDMD by inflammatory caspases determines pyroptotic cell death
.
Nature
.
2015
;
526
(
7575
):
660
-
665
.
26.
Kambara
H
,
Liu
F
,
Zhang
X
, et al
.
Gasdermin D exerts anti-inflammatory effects by promoting neutrophil death
.
Cell Reports
.
2018
;
22
(
11
):
2924
-
2936
.
27.
Bianchi
M
,
Hakkim
A
,
Brinkmann
V
, et al
.
Restoration of NET formation by gene therapy in CGD controls aspergillosis
.
Blood
.
2009
;
114
(
13
):
2619
-
2622
.
28.
Ermert
D
,
Urban
CF
,
Laube
B
,
Goosmann
C
,
Zychlinsky
A
,
Brinkmann
V
.
Mouse neutrophil extracellular traps in microbial infections
.
J Innate Immun
.
2009
;
1
(
3
):
181
-
193
.
29.
Byrd
AS
,
O’Brien
XM
,
Johnson
CM
,
Lavigne
LM
,
Reichner
JS
.
An extracellular matrix-based mechanism of rapid neutrophil extracellular trap formation in response to Candida albicans
.
J Immunol
.
2013
;
190
(
8
):
4136
-
4148
.
30.
Parker
H
,
Dragunow
M
,
Hampton
MB
,
Kettle
AJ
,
Winterbourn
CC
.
Requirements for NADPH oxidase and myeloperoxidase in neutrophil extracellular trap formation differ depending on the stimulus
.
J Leukoc Biol
.
2012
;
92
(
4
):
841
-
849
.
31.
Douda
DN
,
Khan
MA
,
Grasemann
H
,
Palaniyar
N
.
SK3 channel and mitochondrial ROS mediate NADPH oxidase-independent NETosis induced by calcium influx
.
Proc Natl Acad Sci USA
.
2015
;
112
(
9
):
2817
-
2822
.
32.
Hosseinzadeh
A
,
Thompson
PR
,
Segal
BH
,
Urban
CF
.
Nicotine induces neutrophil extracellular traps
.
J Leukoc Biol
.
2016
;
100
(
5
):
1105
-
1112
.
33.
Metzler
KD
,
Fuchs
TA
,
Nauseef
WM
, et al
.
Myeloperoxidase is required for neutrophil extracellular trap formation: implications for innate immunity
.
Blood
.
2011
;
117
(
3
):
953
-
959
.
34.
Knight
JS
,
Zhao
W
,
Luo
W
, et al
.
Peptidylarginine deiminase inhibition is immunomodulatory and vasculoprotective in murine lupus
.
J Clin Invest
.
2013
;
123
(
7
):
2981
-
2993
.
35.
Guiducci
E
,
Lemberg
C
,
Küng
N
,
Schraner
E
,
Theocharides
APA
,
LeibundGut-Landmann
S
.
Candida albicans-induced NETosis is independent of peptidylarginine deiminase 4
.
Front Immunol
.
2018
;
9
:
1573
.
36.
Claushuis
TAM
,
van der Donk
LEH
,
Luitse
AL
, et al
.
Role of peptidylarginine deiminase 4 in neutrophil extracellular trap formation and host defense during Klebsiella pneumoniae-induced pneumonia-derived sepsis
.
J Immunol
.
2018
;
201
(
4
):
1241
-
1252
.
37.
Warnatsch
A
,
Ioannou
M
,
Wang
Q
,
Papayannopoulos
V
.
Inflammation. Neutrophil extracellular traps license macrophages for cytokine production in atherosclerosis
.
Science
.
2015
;
349
(
6245
):
316
-
320
.
38.
Branzk
N
,
Lubojemska
A
,
Hardison
SE
, et al
.
Neutrophils sense microbe size and selectively release neutrophil extracellular traps in response to large pathogens
.
Nat Immunol
.
2014
;
15
(
11
):
1017
-
1025
.
39.
McDonald
B
,
Urrutia
R
,
Yipp
BG
,
Jenne
CN
,
Kubes
P
.
Intravascular neutrophil extracellular traps capture bacteria from the bloodstream during sepsis
.
Cell Host Microbe
.
2012
;
12
(
3
):
324
-
333
.
40.
Yipp
BG
,
Petri
B
,
Salina
D
, et al
.
Infection-induced NETosis is a dynamic process involving neutrophil multitasking in vivo
.
Nat Med
.
2012
;
18
(
9
):
1386
-
1393
.
41.
Yipp
BG
,
Kubes
P
,
Dc
W
,
Yipp
BG
,
Kubes
P
.
NETosis : how vital is it? 
Blood
.
2013
;
122
(
16
):
2784
-
2794
.
42.
Amini
P
,
Stojkov
D
,
Felser
A
, et al
.
Neutrophil extracellular trap formation requires OPA1-dependent glycolytic ATP production
.
Nat Commun
.
2018
;
9
(
1
):
2958
.
43.
Thanabalasuriar
A
,
Scott
BNV
,
Peiseler
M
, et al
.
Neutrophil extracellular traps confine Pseudomonas aeruginosa ocular biofilms and restrict brain invasion
.
Cell Host Microbe
.
In press
.
44.
Maugeri
N
,
Rovere-Querini
P
,
Evangelista
V
, et al
.
Neutrophils phagocytose activated platelets in vivo: a phosphatidylserine, P-selectin, and β2 integrin-dependent cell clearance program
.
Blood
.
2009
;
113
(
21
):
5254
-
5265
.
45.
Jorch
SK
,
Kubes
P
.
An emerging role for neutrophil extracellular traps in noninfectious disease
.
Nat Med
.
2017
;
23
(
3
):
279
-
287
.
46.
Roos
D
,
Voetman
AA
,
Meerhof
LJ
.
Functional activity of enucleated human polymorphonuclear leukocytes
.
J Cell Biol
.
1983
;
97
(
2
):
368
-
377
.
47.
Krishnamoorthy
N
,
Douda
DN
,
Brüggemann
TR
, et al
.
Neutrophil cytoplasts induce T H 17 differentiation and skew inflammation toward neutrophilia in severe asthma
.
Sci Immunol
.
2018
;
3
(
26
):
eaao4747
.
48.
Pieterse
E
,
Rother
N
,
Yanginlar
C
, et al
.
Cleaved N-terminal histone tails distinguish between NADPH oxidase (NOX)-dependent and NOX-independent pathways of neutrophil extracellular trap formation
.
Ann. Rheum. Dis
.
2018
;
77
(
12
):
1790
-
1798
.
49.
Deniset
JF
,
Kubes
P
.
Neutrophil heterogeneity: bona fide subsets or polarization states?
J Leukoc Biol
.
2018
;
103
(
5
):
829
-
838
.
50.
Garcia-Romo
GS
,
Caielli
S
,
Vega
B
, et al
.
Netting neutrophils are major inducers of type I IFN production in pediatric systemic lupus erythematosus
.
Sci Transl Med
.
2011
;
3
(
73
):
73ra20
.
51.
Khandpur
R
,
Carmona-Rivera
C
,
Vivekanandan-Giri
A
, et al
.
NETs are a source of citrullinated autoantigens and stimulate inflammatory responses in rheumatoid arthritis
.
Sci Transl Med
.
2013
;
5
(
178
):
178ra40
.
52.
Guglietta
S
,
Chiavelli
A
,
Zagato
E
, et al
.
Coagulation induced by C3aR-dependent NETosis drives protumorigenic neutrophils during small intestinal tumorigenesis
.
Nat Commun
.
2016
;
7
(
1
):
11037
.
53.
Lood
C
,
Blanco
LP
,
Purmalek
MM
, et al
.
Neutrophil extracellular traps enriched in oxidized mitochondrial DNA are interferogenic and contribute to lupus-like disease
.
Nat Med
.
2016
;
22
(
2
):
146
-
153
.
54.
Zhou
GX
,
Liu
ZJ
.
Potential roles of neutrophils in regulating intestinal mucosal inflammation of inflammatory bowel disease
.
J Dig Dis
.
2017
;
18
(
9
):
495
-
503
.
55.
Zhou
G
,
Yu
L
,
Fang
L
, et al
.
CD177+ neutrophils as functionally activated neutrophils negatively regulate IBD
.
Gut
.
2018
;
67
(
6
):
1052
-
1063
.
56.
Clemmensen
SN
,
Bohr
CT
,
Rørvig
S
, et al
.
Olfactomedin 4 defines a subset of human neutrophils
.
J Leukoc Biol
.
2012
;
91
(
3
):
495
-
500
.
57.
Welin
A
,
Amirbeagi
F
,
Christenson
K
, et al
.
The human neutrophil subsets defined by the presence or absence of OLFM4 both transmigrate into tissue in vivo and give rise to distinct NETs in vitro
.
PLoS One
.
2013
;
8
(
7
):
e69575
.
58.
Alder
MN
,
Opoka
AM
,
Lahni
P
,
Hildeman
DA
,
Wong
HR
.
Olfactomedin-4 Is a Candidate Marker for a Pathogenic Neutrophil Subset in Septic Shock
.
Crit Care Med
.
2017
;
45
(
4
):
e426
-
e432
.
59.
Deniset
JF
,
Kubes
P
.
Recent advances in understanding neutrophils
.
F1000 Res
.
2016
;
5
(
0
):
2912
.
60.
Kubes
P
.
The enigmatic neutrophil: what we do not know
.
Cell Tissue Res
.
2018
;
371
(
3
):
399
-
406
.
61.
Urban
CF
,
Reichard
U
,
Brinkmann
V
,
Zychlinsky
A
.
Neutrophil extracellular traps capture and kill Candida albicans yeast and hyphal forms
.
Cell Microbiol
.
2006
;
8
(
4
):
668
-
676
.
62.
Bruns
S
,
Kniemeyer
O
,
Hasenberg
M
, et al
.
Production of extracellular traps against Aspergillus fumigatus in vitro and in infected lung tissue is dependent on invading neutrophils and influenced by hydrophobin RodA
.
PLoS Pathog
.
2010
;
6
(
4
):
e1000873
.
63.
Saitoh
T
,
Komano
J
,
Saitoh
Y
, et al
.
Neutrophil extracellular traps mediate a host defense response to human immunodeficiency virus-1
.
Cell Host Microbe
.
2012
;
12
(
1
):
109
-
116
.
64.
Hirsch
JG
.
Bactericidal action of histone
.
J Exp Med
.
1958
;
108
(
6
):
925
-
944
.
65.
Weinrauch
Y
,
Drujan
D
,
Shapiro
SD
,
Weiss
J
,
Zychlinsky
A
.
Neutrophil elastase targets virulence factors of enterobacteria
.
Nature
.
2002
;
417
(
6884
):
91
-
94
.
66.
Buchanan
JT
,
Simpson
AJ
,
Aziz
RK
, et al
.
DNase expression allows the pathogen group A Streptococcus to escape killing in neutrophil extracellular traps
.
Curr Biol
.
2006
;
16
(
4
):
396
-
400
.
67.
Tanaka
K
,
Koike
Y
,
Shimura
T
, et al
.
In vivo characterization of neutrophil extracellular traps in various organs of a murine sepsis model
.
PLoS One
.
2014
;
9
(
11
):
e111888
.
68.
Lefrançais
E
,
Mallavia
B
,
Zhuo
H
,
Calfee
CS
,
Looney
MR
.
Maladaptive role of neutrophil extracellular traps in pathogen-induced lung injury
.
JCI Insight
.
2018
;
3
(
3
):
e98178
.
69.
Saffarzadeh
M
,
Juenemann
C
,
Queisser
MA
, et al
.
Neutrophil extracellular traps directly induce epithelial and endothelial cell death: a predominant role of histones
.
PLoS One
.
2012
;
7
(
2
):
e32366
.
70.
Kolaczkowska
E
,
Jenne
CN
,
Surewaard
BGJ
, et al
.
Molecular mechanisms of NET formation and degradation revealed by intravital imaging in the liver vasculature
.
Nat Commun
.
2015
;
6
(
1
):
6673
.
71.
Semeraro
F
,
Ammollo
CT
,
Morrissey
JH
, et al
.
Extracellular histones promote thrombin generation through platelet-dependent mechanisms: involvement of platelet TLR2 and TLR4
.
Blood
.
2011
;
118
(
7
):
1952
-
1961
.
72.
Jiménez-Alcázar
M
,
Rangaswamy
C
,
Panda
R
, et al
.
Host DNases prevent vascular occlusion by neutrophil extracellular traps
.
Science
.
2017
;
358
(
6367
):
1202
-
1206
.
73.
Meng
W
,
Paunel-Görgülü
A
,
Flohé
S
, et al
.
Depletion of neutrophil extracellular traps in vivo results in hypersusceptibility to polymicrobial sepsis in mice
.
Crit Care
.
2012
;
16
(
4
):
R137
.
74.
McDonald
B
,
Davis
RP
,
Kim
SJ
, et al
.
Platelets and neutrophil extracellular traps collaborate to promote intravascular coagulation during sepsis in mice
.
Blood
.
2017
;
129
(
10
):
1357
-
1367
.
75.
Martinod
K
,
Fuchs
TA
,
Zitomersky
NL
, et al
.
PAD4-deficiency does not affect bacteremia in polymicrobial sepsis and ameliorates endotoxemic shock
.
Blood
.
2015
;
125
(
12
):
1948
-
1956
.
76.
Czaikoski
PG
,
Mota
JMSC
,
Nascimento
DC
, et al
.
Neutrophil extracellular traps induce organ damage during experimental and clinical sepsis
.
PLoS One
.
2016
;
11
(
2
):
e0148142
.
77.
Xu
J
,
Zhang
X
,
Pelayo
R
, et al
.
Extracellular histones are major mediators of death in sepsis
.
Nat Med
.
2009
;
15
(
11
):
1318
-
1321
.
78.
Chen
L
,
Zhao
Y
,
Lai
D
, et al
.
Neutrophil extracellular traps promote macrophage pyroptosis in sepsis
.
Cell Death Dis
.
2018
;
9
(
6
):
597
.
79.
Grégoire
M
,
Uhel
F
,
Lesouhaitier
M
, et al
.
Impaired efferocytosis and neutrophil extracellular trap clearance by macrophages in ARDS
.
Eur Respir J
.
2018
;
52
(
2
):
1702590
.
80.
Grabcanovic-Musija
F
,
Obermayer
A
,
Stoiber
W
, et al
.
Neutrophil extracellular trap (NET) formation characterises stable and exacerbated COPD and correlates with airflow limitation
.
Respir Res
.
2015
;
16
(
1
):
59
.
81.
Sollberger
G
,
Tilley
DO
,
Zychlinsky
A
.
Neutrophil extracellular traps: the biology of chromatin externalization
.
Dev Cell
.
2018
;
44
(
5
):
542
-
553
.
82.
Dwyer
M
,
Shan
Q
,
D’Ortona
S
, et al
.
Cystic fibrosis sputum DNA has NETosis characteristics and neutrophil extracellular trap release is regulated by macrophage migration-inhibitory factor
.
J Innate Immun
.
2014
;
6
(
6
):
765
-
779
.
83.
Pedersen
F
,
Marwitz
S
,
Holz
O
, et al
.
Neutrophil extracellular trap formation and extracellular DNA in sputum of stable COPD patients
.
Respir Med
.
2015
;
109
(
10
):
1360
-
1362
.
84.
Wright
TK
,
Gibson
PG
,
Simpson
JL
,
McDonald
VM
,
Wood
LG
,
Baines
KJ
.
Neutrophil extracellular traps are associated with inflammation in chronic airway disease
.
Respirology
.
2016
;
21
(
3
):
467
-
475
.
85.
Dicker
AJ
,
Crichton
ML
,
Pumphrey
EG
, et al
.
Neutrophil extracellular traps are associated with disease severity and microbiota diversity in patients with chronic obstructive pulmonary disease
.
J Allergy Clin Immunol
.
2018
;
141
(
1
):
117
-
127
.
86.
Roesch
EA
,
Nichols
DP
,
Chmiel
JF
.
Inflammation in cystic fibrosis: an update
.
Pediatr Pulmonol
.
2018
;
53
(
S3
):
S30
-
S50
.
87.
Papayannopoulos
V
.
Neutrophil extracellular traps in immunity and disease
.
Nat Rev Immunol
.
2018
;
18
(
2
):
134
-
147
.
88.
Gray
RD
,
Hardisty
G
,
Regan
KH
, et al
.
Delayed neutrophil apoptosis enhances NET formation in cystic fibrosis
.
Thorax
.
2018
;
73
(
2
):
134
-
144
.
89.
Papayannopoulos
V
,
Staab
D
,
Zychlinsky
A
.
Neutrophil elastase enhances sputum solubilization in cystic fibrosis patients receiving DNase therapy
.
PLoS One
.
2011
;
6
(
12
):
e28526
.
90.
Brinkmann
V
.
Neutrophil extracellular traps in the second decade
.
J Innate Immun
.
2018
;
10
(
5-6
):
414
-
421
.
91.
Hakkim
A
,
Fürnrohr
BG
,
Amann
K
, et al
.
Impairment of neutrophil extracellular trap degradation is associated with lupus nephritis
.
Proc Natl Acad Sci USA
.
2010
;
107
(
21
):
9813
-
9818
.
92.
Wang
W
,
Jian
Z
,
Guo
J
,
Ning
X
.
Increased levels of serum myeloperoxidase in patients with active rheumatoid arthritis
.
Life Sci
.
2014
;
117
(
1
):
19
-
23
.
93.
Gupta
S
,
Kaplan
MJ
.
The role of neutrophils and NETosis in autoimmune and renal diseases
.
Nat Rev Nephrol
.
2016
;
12
(
7
):
402
-
413
.
94.
Sur Chowdhury
C
,
Giaglis
S
,
Walker
UA
,
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
(
3
):
R122
.
95.
Campbell
AM
,
Kashgarian
M
,
Shlomchik
MJ
.
NADPH oxidase inhibits the pathogenesis of systemic lupus erythematosus
.
Sci Transl Med
.
2012
;
4
(
157
):
157ra141-157ra141
.
96.
Slaba
I
,
Wang
J
,
Kolaczkowska
E
,
McDonald
B
,
Lee
WY
,
Kubes
P
.
Imaging the dynamic platelet-neutrophil response in sterile liver injury and repair in mice
.
Hepatology
.
2015
;
62
(
5
):
1593
-
1605
.
97.
Wang
J
,
Hossain
M
,
Thanabalasuriar
A
, et al
.
Visualizing the function and fate of neutrophils in sterile injury and repair
.
Science
.
2017
;
358
(
6359
):
111
-
116
.
98.
Wong
SL
,
Demers
M
,
Martinod
K
, et al
.
Diabetes primes neutrophils to undergo NETosis, which impairs wound healing
.
Nat Med
.
2015
;
21
(
7
):
815
-
819
.
99.
Schauer
C
,
Janko
C
,
Munoz
LE
, et al
.
Aggregated neutrophil extracellular traps limit inflammation by degrading cytokines and chemokines
.
Nat Med
.
2014
;
20
(
5
):
511
-
517
.
Sign in via your Institution