Extracellular Traps – NETosis and METosis
Zacharoula Konsoula (zkons664 at yahoo dot com)
Georgetown University Medical Center, United States
DOI
//dx.doi.org/10.13070/mm.en.9.2714
Date
last modified : 2022-11-04; original version : 2019-03-04
Cite as
MATER METHODS 2019;9:2714
Abstract

An overview of extracellular traps, including NETosis and METosis.

Introduction

Neutrophil granulocytes play a pivotal role in innate immunity by defending the host against invading pathogens through approaches such as phagocytosis, the formation of reactive oxygen species (ROS), degranulation, and the generation of neutrophil extracellular traps (NETs), a process known as NETosis. NETosis is a type of programmed cell death distinct from apoptosis and necrosis. NETs were firstly described in 2004 as a novel preventive mechanism [5]. Morphological transformations occurring during NETosis consist of the disintegration of nuclear and granule membranes and the combining of nuclear, granular, and cytoplasmic components. A disruption in the plasma membrane incites the release of extracellular chromatin traps. Upon in vitro activation with phorbol myristate acetate (PMA), interleukin 8 (IL-8) or lipopolysaccharide (LPS), neutrophils release granule proteins and chromatin to form NETs through an active process [6]. Thrombosis induced by NETosis is likely the cause of adverse effects of ChAdOx1 nCoV-19 adenoviral vector vaccine against corona virus disease 2019 (COVID-19) in some patients [7].

Extracellular Traps – NETosis and METosis figure 1
Figure 1. Schematic representation of the cellular processes involved in the formation of ETs. From [1].

Similarly, other cell types, such as eosinophils, mast cells, and macrophages, can also cause death by this mechanism; therefore, the process was renamed as ETosis, referring to cell death with the release of extracellular traps (ETs). The underlying mechanism behind the ET formation remains mostly unknown, and the biological importance of ETs is just beginning to emerge. This review will focus on the formation and function of ETs by neutrophils and monocytes / macrophages. We will also discuss the mechanisms that regulate the release of ETs during infection and other disease processes. Figure 1 presents schematically the cellular processes involved in the formation of ETs [1]. Table 1 lists the proteins commonly studied in extracellular traps and most cited antibodies against them among the over 60,000 formal publications Labome has surveyed for Validated Antibody Database.

Protein Detail Top three suppliers
ELANEelastaseDako M0752 (13), Santa Cruz Biotechnology sc-53388 (1), Invitrogen MA1-10608 (1)
FCGR2AFc fragment of IgG receptor IIaBioLegend 303202 (10), BD Biosciences 557333 (8), Bio-Rad MCA1075 (6)
LRP1LDL receptor related protein 1Abcam ab92544 (10), Invitrogen 37-7600 (3), MilliporeSigma L2420 (2)
MPOmyeloperoxidaseInvitrogen MA1-20074 (10), Abcam ab25989 (8), Dako F071401-1 (3)
NOX3NADPH oxidase 3ProSci 7925 (1)
NOX4NADPH oxidase 4Abcam ab133303 (16), ProSci 7927 (2), Invitrogen MA5-32090 (1)
PADI4peptidyl arginine deiminase 4Abcam ab128086 (8), BioLegend 684202 (1)
Table 1. Proteins commonly studied in extracellular traps and their number of citations with antibody applications of immunohistochemistry, immunocytochemistry, flow cytometry, and ELISA, among the publications Labome has surveyed for Validated Antibody Database. The most cited monoclonal antibody from each supplier is listed.
Structure and Composition

NETosis is neutrophil-related cell death characterized by the secretion of large web-like structures described as NETs. NETs are composed of chromatin fibers with diameters of 15–17 nm that are made up of DNA and histones H1, H2A, H2B, H3, and H4. In addition, NETs consist of proteins from azurophilic granules (i.e., neutrophil elastase (NE) cathepsin G, and myeloperoxidase (MPO)), specific granules (lactoferrin) and tertiary granules (gelatinase) [5]. Figure 2 presents scanning electron microscope images of NETs in an infected mouse lung model [2].

Extracellular Traps – NETosis and METosis figure 2
Figure 2. Scanning electron microscope images of NETs induced by Candida albicans in an infected mouse lung model. From [2].

NE is a serine protease that eliminates bacteria [8], and MPO activates the oxidation of halides by hydrogen peroxide [9]. NE and MPO knockout mice are predisposed to bacterial and fungal infections [10]. NE plays a critical role in instigating NET formation and interacts with MPO to initiate chromatin decondensation [11]. Histones are the most abundant NET constituent, and several histones have shown to possess the antimicrobial potential [2]. In particular, the histones H2A and H2B have demonstrated antimicrobial activities against Gram-positive and Gram-negative bacteria and fungi [12].

NETosis Pathway

NETs generation is an active and distinctive process from neutrophil apoptosis and necrosis [3]. NETosis mainly involves reactive oxygen and nitrogen species (ROS/RNS) and activation requires nicotinamide adenine dinucleotide phosphate (NADPH) oxidase and MPO [3, 13]. NADPH-oxidase induces superoxide radicals, prompting the generation of hydrogen peroxide, which is employed by neutrophils to generate hypochlorite that kills bacteria and might further cause lipid peroxidation and membrane damage [14].

There is an accumulating scientific body of evidence that NADPH-mediated ROS formation is essential to drive the NETs formation. For instance, neutrophils derived from patients with chronic granulomatous disease with mutations in the NADPH oxidase that disrupted ROS generation [15] were not capable of inducing the formation of NETs [3]. Besides, ROS scavengers (i.e., N-acetylcysteine, diphenyleneiodonium, or Trolox) prevented NETosis [3] in human neutrophils upon stimulation with PMA and have confirmed NADPH oxidase-dependent NET formation. The exact role of ROS in NETosis formation has not been fully understood, but it is believed to work through two separate mechanisms. Some studies suggest ROS promote the morphological changes noted during NETosis [16]. ROS may alternatively inactivate caspases, prevent apoptosis and advance autophagy. This action leads to the dissolution of cellular membranes [17]. Scientific evidence as to whether there are pathways of NETosis that employ a ROS-independent pathway appears insufficient and contradictory [18].

NETosis Apoptosis Necrosis
Programmed cell deathProgrammed cell deathCell damage releasing intracellular contents and leading to death
Nuclear chromatin decondensation with disintegration of the nuclear membrane into numerous small vesiclesNuclear chromatin condensation without disintegration of the nuclear membraneCellular swelling and bursting
VacuolizationMembrane blebbingMembrane and organelle disintegration
Table 2. Differences between NETosis, apoptosis, and necrosis.

NETosis is a new type of cell death, with distinctive features from apoptosis and necrosis. NETosis appears to be a pathway not mediated by caspases and kinases (e.g., RIP-1) since neither inhibition of caspases by zVAD-fmk nor inhibition of RIP1 kinases by necrostatin-1 impacted NETosis. Moreover, the process does not seem to involve DNA fragmentation or phosphatidylserine (PS) exposure of the cellular membrane [2, 17]. However, in NETosis, both the nuclear as well as the granular membranes are subject to fragmentation. Table 2 lists the morphological features of NETosis in comparison with apoptosis and necrosis.

Extracellular Traps – NETosis and METosis figure 3
Figure 3. Transmission electron microscopy (a–h) and confocal immunofluorescence (i–l) images of the disintegration of the nucleus and granules during NETosis. From [3].

Morphological transformations occurring during NETosis consist of the disintegration of nuclear and granular membranes and integration of nuclear, granular, and cytoplasmic components. Lastly, disruption of the plasma membrane occurs, and DNA combined with the granular substances is secreted into the extracellular environment [3]. Therefore, NETosis should be regarded as a distinct cell death program and is different from both apoptosis and necrosis. Figure 3 shows transmission electron microscopy and confocal immunofluorescence images of the disintegration of the nucleus and granules during NETosis [3].

Suicidal versus vital NETosis

Two major NET release mechanisms have been reported, suicidal and vital NETosis.

Suicidal NETosis involves the triggering of the Raf-MEK-ERK pathway, NADPH oxidase-dependent pathways, the generation of ROS, and receptor-interacting protein kinase/mixed lineage kinase domain-like-mediated signals [19, 20]. This initiating effect appears to be the primary path for NET release and is an irreversible process. Different receptors, such as Toll-like receptors (TLRs), Fc receptors, or complement receptors, have been reported to be involved in NETosis [5]. Briefly, receptor activation leads to endoplasmic reticulum calcium store release and increased cytoplasmic calcium. Elevated calcium ions stimulate the activity of protein kinase C (PKC), the phosphorylation of gp91phox, and the functional assembly of cytosolic and membrane-bound subunits of NADPH oxidase, leading to the formation of ROS. Under the activation of ROS, the granules, and the nuclear envelope break. Subsequently, the secreted nuclear, granular, and cytoplasmic substances combine (Figure 1).

In this pathway, peptidylarginine deiminase 4 (PAD4)-dependent citrullination of histones promotes decondensation of DNA generating a mixture of DNA and bactericidal proteins, encompassing MPO and NE, which are initially found in intracytoplasmic granules [21]. Afterward, these materials are released from the ruptured plasma membrane. The prerequisite of PAD4 in NET formation is still under exploration. It was reported that PAD4-deficient mice presented decreased NET formation during group A Streptococcus pyogenes infection [21], but they maintained the potential to generate NETs against influenza infection [22].

The majority of the mechanistic understanding on lytic NETosis comes from in vitro studies examining NETosis by treating isolated neutrophils with PMA, for example, [23, 24]. NET secretion by cell death is a slow process (120-240 min) and may allow a window of time for microbes to cause an infection. In contrast, an alternative rapid process (5-60 min) for NET formation has been reported called vital NETosis. This latter process is ROS-independent in response to Staphylococcus aureus [25] and Candida albicans [26, 27]. Moreover, vital NETosis entails vesicular movement of DNA from within the nucleus to the extracellular space [25]. As a result, this pathway preserves the integrity of the plasma membranes, and it does not involve lytic death of the neutrophils [28].

Method Detection marker Advantage Disadvantage Reference
Immunostaining and microscopyCo-localization of neutrophil-derived proteins and extracellular DNAEasy to performArtificial NET staining [3, 6, 11, 29, 30]
Citrullinated histonesEasy to performOnly PAD4-dependent NETosis measured [31, 32]
Flow cytometryMPO-DNA complex; MPO and citrullinated histonesHigh specificityPrevalence needed [33, 34]
ELISAMPO/NE-DNA complex; PAD4/DNA-complex detectionHigh specificityStandardization required [35, 36]
Picogreen kitCell-free DNAAvailable kitOther cell death-derived DNA can be measured [23, 32]
Table 3. Comparison of methods for measuring NETosis.
Measurement of NET formation

Several methods are developed to analyze NETs [37] including transmission electron microscopy, scanning electron microscopy, and immunofluorescence [25, 27]. During NETosis, the nuclei of neutrophils lose its shape, and the nuclear envelope and the granule membranes disintegrate leading to the mixing of the NET components [6, 38]. Table 3 compares different methods for measuring NETosis.

Co-localization of extracellular DNA and neutrophil-derived proteins, including MPO and NE, was noted by microscopic observation [32, 39]. Enzyme-linked immunosorbent assay (ELISA) was also used to detect the complexes of DNA and neutrophil-derived proteins, including MPO [32], NE, and PAD4 from different patients [29, 35, 40].

Fluorescence-activated cell sorting (FACS) was used to determine NETosis by the detection of MPO and citrullinated histones [33]. SYTOX® Green is a fluorescent dye that is used to label DNA, but it does not permeate the plasma membrane [32]. SYTOX Green showed co-localization of MPO and plasma membrane-appendant DNA of the PMA-treated neutrophils [34]. Cell-free DNA (quantitatively using a kit, Picogreen) was used to detect soluble NET remnants in fluid samples, such as in sera [32, 41].

The above approaches are often combined to address the NET formation. Ali RA et al combined the SYTOX Green assay, the NET-associated myeloperoxidase (MPO) assay, neutrophil elastase staining, and citrullinated histone H3 staining to investigate the effect of antiphospholipid antibodies on NET and its alleviation by adenosine receptor agonism [42]. A Constantinescu-Bercu et al visualize NETosis through co-labeling of neutrophils with cell permeable Hoechst dye and cell impermeable Sytox Green [31], an approach similarly used by LM Silva et al [43].

Inducers of NET Formation

NETs present broad-ranging potency against a diversity of pathogens including Gram-positive and Gram-negative bacteria, fungi, parasites, and viruses. Furthermore, intrinsic mediators such as hydrogen peroxide [3], cytokines [44], chemokines [45], cholesterol [46] and autoantibodies [29] promote the NET generation. Pro-inflammatory factors, including tumor necrosis factor (TNF)-α, interferon (IFN)-γ, IL-17 and IL-8 [47], can trigger NETosis.

PMA is the most commonly employed stimulus for activating NETosis [31]. However, it is not physiologically applicable, since it does not activate physiological processes in vivo [5]. Several other inducers have been presented, but their NETosis potential has not been consistent. For example, some studies, NETosis was observed after 30 min in the presence of 100 ng/ml LPS [48, 49], whereas in other studies, 10 µg/ml LPS failed to ead to NETosis formation [17]. The differences in experimental setting, timing, and dosing might contribute to such variability.

Gram–positive bacteria Gram–negative bacteria Fungi Parasites Viruses
Staphylococcus aureus [3, 25, 50]
Streptococcus pneumoniae [51, 52]
Streptococcus pyogenes [53]
Escherichia coli [54] Candida albicans [2, 26]
Aspergillus fumigatus [55, 56]
Plasmodium falciparum [57] Feline leukemia virus [58]
Human immunodeficiency virus-1 [59]
Influenza [22]
Table 4. Microbial organisms that elicit a NET response.
Microbicidal Activity

NET formation is limited to particular microbes. The characteristics that determine effective NET stimulants are not well defined. Several bacteria and fungi were reported to potently induce NET formation, such as Staphylococcus aureus [3, 25, 50], Streptococcus pneumoniae [51, 52], Streptococcus pyogenes [53], Escherichia coli [54], Candida albicans [2, 26], and Aspergillus fumigatus [55, 56]. Table 4 outlines microbial inducers of NETs.

Several elements of NETs are instrumental in microbicidal activity. The enzymatic activity of MPO on NETs is a requisite to kill Staphylococcus aureus [18] and the enzymatic activity of NE is essential to generate NETs in a pulmonary model of Klebsiella pneumoniae infection [11]. Antibodies against histones can abolish the NET-moderated microbicidal activity [5]. The fungicidal activity of NETs has been attributed to calprotectin (a complex of S100A8/S100A9), which chelates divalent metal ions. Calprotectin plays a part in the NETs-associated antifungal activity against Candida albicans in vivo infection models [2]. Interestingly, calprotectin is released during Covid-19 infection and its plasma level was found to distinguish between mild and severe forms of COVID-19 [60].

However, some studies challenge the microbicidal capacity of NETs since NET formation appears to be induced by particular pathogens and experimental conditions. For instance, in the presence of DNase treatment in the medium which dismantles NETs, viable Staphylococcus aureus and Candida albicans blastospores were secreted from NETs [61].

Notably, several bacteria can degenerate NETs by nucleases and get away from NET-mediated entrapment and elimination. These involve the Gram-negative pathogen Vibrium cholera [62] and the Gram-positive bacteria Streptococcus pneumoniae [51], Group A Streptococcus [53], Staphylococcus aureus [50], and Streptococcus agalactiae [63]. This degeneration highlights the significant role nucleases play as pathogenic factors.

In the presence of two extracellular nucleases Dns and Xds, Vibrium cholera could break down the DNA component of the NETs [62] and nuclease formation by Staphylococcus aureus promoted resistance against NET-associated antimicrobial activity of neutrophils and lead to lung disease pathogenesis in vivo [50].

Overall, NETs have been shown to kill or inhibit the growth of several bacterial, fungal, and parasite species. However, the general importance of NET-mediated killing of microbes may vary depending on the type of pathogen and the techniques employed to assess the microbial killing.

Pathophysiological condition Reference
COVID-19 [64]
Gallstone formation [65]
Thrombosis [66, 67]
Systemic lupus erythematosus [30]
Rheumatoid arthritis [35, 44]
Antiphospholipid antibody syndrome [66, 67]
Atherosclerosis [46]
Vasculitis [29]
Pancreatitis [68]
Type I and type II diabetes [36, 69]
Pre-eclamsia [70]
Cardiovascular [24, 71]
Sepsis [28]
Malaria [57]
Table 5. NET-associated involvement in diseases.
NET-associated Host Tissue Damage

The role of NETosis is unclear between antimicrobial defense and host tissue damage. Several components are toxic to the host cell and trigger autoimmunity. NETs have been implicated in thrombosis [66, 67, 72, 73] as well as diseases like systemic lupus erythematosus (SLE) [30], rheumatoid arthritis [35, 44], antiphospholipid antibody syndrome [66], vasculitis [29], pancreatitis [68], type I [36], and type II diabetes [69], pre-eclamsia [70], cardiovascular problems [71], sepsis [28], and malaria [57]. Table 5 summarizes the pathophysiological conditions in which NETs have been formed.

For example, NETs are elevated in rheumatoid arthritis [16], antiphospholipid antibody syndrome [66], vasculitis [29, 74], SLE [30] and diabetic [75] patients compared to healthy control groups. Neutrophils from rheumatoid arthritis patients demonstrated elevated spontaneous NET production in vitro, accompanied by increased ROS generation, augmented NE and MPO expression, nuclear translocation of PAD4, and modified nuclear morphology [35]. Besides, NET formation promoted the autoimmune response against neutrophil components in individuals with small-vessel vasculitis [74, 76], rheumatoid arthritis [35] and SLE patients [30]. Therefore, inhibiting or cleaving NETs may improve some of the disorders mentioned above.

Also, NETs were shown to be important for thrombosis in murine models of both antiphospholipid antibody syndrome and deep vein thrombosis [66, 67, 73] and NETs elicit pancreatitis by ductal occlusion [68]. Furthermore, secretion of NETs in the vascular space elicited a pro-coagulant condition and triggered activation of platelets causing thrombosis [77].

Aberrant production of NETs and scarcity of DNases to dismantle NETs might contribute to tissue damage and autoimmune diseases in patients [74]. Therefore, the timely removal of NETs may be essential for tissue homeostasis to block the presentation of self-antigens.

Extracellular Traps – NETosis and METosis figure 4
Figure 4. Scanning electron microscopy images of uninfected J774A.1 cells (A-B), METs-LS induced by Escherichia coli (C–D) and METs-LS induced by Candida albicans (E-F). Fluorescence images (G-H) of DNA (blue) and FITC-labeled Escherichia coli (Green). From [4].
Extracellular Traps and Macrophages

NETosis was first described in neutrophils, but other cell types including monocytes and macrophages are capable of releasing ETs composed of DNA and antimicrobial proteins.

Monocytes/macrophages have been shown to release ETs in a process called METosis [4, 39, 78-80]. Figure 4 presents scanning electron microscope images of METs induced by Escherichia coli and Candida albicans. Treatment with DNase I or nucleases degrade METs. Studies staining well-characterized structures of ETs confirmed the METs features.

Cell Type MET components Reference
Human alveolar macrophagesmatrix metalloproteinase 12 [81]
Human glomerular macrophagesMyeloperoxidase [82]
Human monocyte-derived macrophageshistone H4 [83, 84]
Human peripheral-blood monocyteshistones H2 and H3; elastase; myeloperoxidase; [85, 86]
Mouse macrophages (RAW 264.7 cells)histones H2 and H4 [78, 79]
Mouse monocyte; macrophage (J774A.1 cells)histone H2; myeloperoxidase [4, 79]
Rat macrophageshistone H2; myeloperoxidase [87]
Bovine monocyteshistone H3; myeloperoxidase [88]
Table 6. Types of monocytes and macrophages that generate METs.

NE has been found in the METs of human peripheral-blood monocytes [89]. In the same way as NETs, MPO has been shown to be part of METs of various macrophages including human glomerular macrophages, human peripheral-blood monocytes, THP-1 macrophage-like cells, murine J774A.1 macrophage-like cells, bovine monocytes, and caprine monocytes [4, 82, 88-90]. Table 6 lists types of monocytes and macrophages reported to generate METs and the protein components associated with METosis.

Macrophage ET generation has been reported to be enhanced by statins, which are inhibitors of the rate-limiting enzyme within the cholesterol biosynthesis 3-hydroxy 3-methyglutaryl coenzyme A (HMG-CoA) reductase. Furthermore, the rise in the formation of METs was noted after preventing the activity of HMG-CoA reductase using small interfering Ribonucleic Acid (siRNA) or after treatment of macrophages with the downstream HMG-CoA reductase product mevalonate [78]. However, the extent to which the molecular mechanism leading to the formation of METs is similar to the process already attributed to NETs remains to be further clarified.

The limited data available shows that METosis is a cell death pathway involving NADPH oxidase dependency, similar to the process in neutrophils. Traditional features of ETs were corroborated in METosis by the co-localization of extracellular DNA with histones H3 or MPO in parasite-entrapping structures. Monocyte-derived ETs were eliminated by DNase I treatment and notably decreased by inhibitors of MPO and NADPH oxidase [88]. Also, treatment of caprine monocyte ET structures with NADPH oxidase inhibitor diphenylene iodondium (DPI) remarkably decreased Etosis [90]. These findings provide evidence for the essential role of ROS and MPO in METosis formation. Treatment of alveolar macrophages with the ROS inhibitor apocinin prevented MET release [81]. It was also shown using the fluorescent dihydrorhodamine 123 that human alveolar macrophages forming METs had a 2-fold increase in ROS fluorescence compared to cells not forming METs [81].

Gram–positive bacteria Gram–negative bacteria Fungi Parasites
Staphylococcus aureus [78, 80]
Streptococcus agalactiae [79]
Klebsiella pneumoniae [85]
Mannheimia haemolytica [91]
Escherichia coli [89, 92]
Candida albicans [4, 89] Toxoplasma gondii [93]
Besnoitia besnoiti [88]
Table 7. Microbial organisms that elicit a MET response.

Several studies have shown that METs have microbicidal activity against different microorganisms. Several bacteria and fungi were reported to potently induce MET formation, such as Staphylococcus aureus [78, 80], Streptococcus agalactiae [79], Haemophilus influenzae [81], Klebsiella pneumoniae [85], Mannheimia haemolytica [91], Escherichia coli [4, 89] and Candida albicans [4, 89]. Microbial inducers of METs are outlined in Table 7.

Conclusion

ETs display broad-ranging effectiveness against a diversity of pathogens including Gram-positive and Gram-negative bacteria, fungi, parasites, and viruses. Nevertheless, experimental data indicate that ET formation appear to be limited to particular microbes and the characteristics that define effective ET stimulants are not well defined. NETosis especially seems to be strictly governed, and dysregulation has been associated with severe autoimmune conditions. Therefore, as the biological importance of ETs is beginning to be unraveled, the molecular mechanisms regulating the ETs formation and the downstream pathways need to be further explored. Future work characterizing ET properties associated with particular disease states such as kidney transplant rejection [94], or microbial infections will advance the understanding of the role of ETs in diseases and open potential avenues for therapeutic modulation.

References
  1. Goldmann O, Medina E. The expanding world of extracellular traps: not only neutrophils but much more. Front Immunol. 2012;3:420 pubmed publisher
  2. Urban C, Ermert D, Schmid M, Abu Abed U, Goosmann C, Nacken W, et al. Neutrophil extracellular traps contain calprotectin, a cytosolic protein complex involved in host defense against Candida albicans. PLoS Pathog. 2009;5:e1000639 pubmed publisher
  3. Fuchs T, Abed U, Goosmann C, Hurwitz R, Schulze I, Wahn V, et al. Novel cell death program leads to neutrophil extracellular traps. J Cell Biol. 2007;176:231-41 pubmed
  4. Liu P, Wu X, Liao C, Liu X, Du J, Shi H, et al. Escherichia coli and Candida albicans induced macrophage extracellular trap-like structures with limited microbicidal activity. PLoS ONE. 2014;9:e90042 pubmed publisher
  5. Brinkmann V, Reichard U, Goosmann C, Fauler B, Uhlemann Y, Weiss D, et al. Neutrophil extracellular traps kill bacteria. Science. 2004;303:1532-5 pubmed
  6. Brinkmann V, Laube B, Abu Abed U, Goosmann C, Zychlinsky A. Neutrophil extracellular traps: how to generate and visualize them. J Vis Exp. 2010;: pubmed publisher
  7. Holm S, Kared H, Michelsen A, Kong X, Dahl T, Schultz N, et al. Immune complexes, innate immunity, and NETosis in ChAdOx1 vaccine-induced thrombocytopenia. Eur Heart J. 2021;: pubmed publisher
  8. Weinrauch Y, Drujan D, Shapiro S, Weiss J, Zychlinsky A. Neutrophil elastase targets virulence factors of enterobacteria. Nature. 2002;417:91-4 pubmed
  9. Nauseef W. How human neutrophils kill and degrade microbes: an integrated view. Immunol Rev. 2007;219:88-102 pubmed
  10. Belaaouaj A, Kim K, Shapiro S. Degradation of outer membrane protein A in Escherichia coli killing by neutrophil elastase. Science. 2000;289:1185-8 pubmed
  11. Papayannopoulos V, Metzler K, Hakkim A, Zychlinsky A. Neutrophil elastase and myeloperoxidase regulate the formation of neutrophil extracellular traps. J Cell Biol. 2010;191:677-91 pubmed publisher
  12. Li G, Mine Y, Hincke M, Nys Y. Isolation and characterization of antimicrobial proteins and peptide from chicken liver. J Pept Sci. 2007;13:368-78 pubmed
  13. Bartholeyns J, Baudhuin P. [Proceedings: Cytostatic properties of pancreatic ribonuclease A dimer]. Arch Int Physiol Biochim. 1976;84:139-40 pubmed
  14. Pattison D, Davies M. Reactions of myeloperoxidase-derived oxidants with biological substrates: gaining chemical insight into human inflammatory diseases. Curr Med Chem. 2006;13:3271-90 pubmed
  15. Clark F, Klebanoff S. Chronic granulomatous disease: studies of a family with impaired neutrophil chemotactic, metabolic and bactericidal function. Am J Med. 1978;65:941-8 pubmed
  16. Kaplan M, Radic M. Neutrophil extracellular traps: double-edged swords of innate immunity. J Immunol. 2012;189:2689-95 pubmed publisher
  17. Remijsen Q, Vanden Berghe T, Wirawan E, Asselbergh B, Parthoens E, De Rycke R, et al. Neutrophil extracellular trap cell death requires both autophagy and superoxide generation. Cell Res. 2011;21:290-304 pubmed publisher
  18. Parker H, Albrett A, Kettle A, Winterbourn C. Myeloperoxidase associated with neutrophil extracellular traps is active and mediates bacterial killing in the presence of hydrogen peroxide. J Leukoc Biol. 2012;91:369-76 pubmed publisher
  19. Hakkim A, Fuchs T, Martinez N, Hess S, Prinz H, Zychlinsky A, et al. Activation of the Raf-MEK-ERK pathway is required for neutrophil extracellular trap formation. Nat Chem Biol. 2011;7:75-7 pubmed publisher
  20. Desai J, Kumar S, Mulay S, Konrad L, Romoli S, Schauer C, et al. PMA and crystal-induced neutrophil extracellular trap formation involves RIPK1-RIPK3-MLKL signaling. Eur J Immunol. 2016;46:223-9 pubmed publisher
  21. Li P, Li M, Lindberg M, Kennett M, Xiong N, Wang Y. PAD4 is essential for antibacterial innate immunity mediated by neutrophil extracellular traps. J Exp Med. 2010;207:1853-62 pubmed publisher
  22. Hemmers S, Teijaro J, Arandjelovic S, Mowen K. PAD4-mediated neutrophil extracellular trap formation is not required for immunity against influenza infection. PLoS ONE. 2011;6:e22043 pubmed publisher
  23. Silvestre Roig C, Braster Q, Wichapong K, LEE E, Teulon J, Berrebeh N, et al. Externalized histone H4 orchestrates chronic inflammation by inducing lytic cell death. Nature. 2019;569:236-240 pubmed publisher
  24. Weckbach L, Grabmaier U, Uhl A, Gess S, Boehm F, Zehrer A, et al. Midkine drives cardiac inflammation by promoting neutrophil trafficking and NETosis in myocarditis. J Exp Med. 2019;216:350-368 pubmed publisher
  25. Pilsczek F, Salina D, Poon K, Fahey C, Yipp B, Sibley C, et al. A novel mechanism of rapid nuclear neutrophil extracellular trap formation in response to Staphylococcus aureus. J Immunol. 2010;185:7413-25 pubmed publisher
  26. Byrd A, O Brien X, Johnson C, Lavigne L, Reichner J. An extracellular matrix-based mechanism of rapid neutrophil extracellular trap formation in response to Candida albicans. J Immunol. 2013;190:4136-48 pubmed publisher
  27. Rochael N, Guimarães Costa A, Nascimento M, DeSouza Vieira T, Oliveira M, Garcia E Souza L, et al. Classical ROS-dependent and early/rapid ROS-independent release of Neutrophil Extracellular Traps triggered by Leishmania parasites. Sci Rep. 2015;5:18302 pubmed publisher
  28. Clark S, Ma A, Tavener S, McDonald B, Goodarzi Z, Kelly M, et al. Platelet TLR4 activates neutrophil extracellular traps to ensnare bacteria in septic blood. Nat Med. 2007;13:463-9 pubmed
  29. Kessenbrock K, Krumbholz M, Schönermarck U, Back W, Gross W, Werb Z, et al. Netting neutrophils in autoimmune small-vessel vasculitis. Nat Med. 2009;15:623-5 pubmed publisher
  30. Hakkim A, Fürnrohr B, Amann K, Laube B, Abed U, Brinkmann V, et al. Impairment of neutrophil extracellular trap degradation is associated with lupus nephritis. Proc Natl Acad Sci U S A. 2010;107:9813-8 pubmed publisher
  31. Constantinescu Bercu A, Grassi L, Frontini M, Salles Crawley I, Woollard K, Crawley J. Activated αIIbβ3 on platelets mediates flow-dependent NETosis via SLC44A2. elife. 2020;9: pubmed publisher
  32. Binet F, Cagnone G, Crespo Garcia S, Hata M, Neault M, Dejda A, et al. Neutrophil extracellular traps target senescent vasculature for tissue remodeling in retinopathy. Science. 2020;369: pubmed publisher
  33. Gavillet M, Martinod K, Renella R, Harris C, Shapiro N, Wagner D, et al. Flow cytometric assay for direct quantification of neutrophil extracellular traps in blood samples. Am J Hematol. 2015;90:1155-8 pubmed publisher
  34. Masuda S, Shimizu S, Matsuo J, Nishibata Y, Kusunoki Y, Hattanda F, et al. Measurement of NET formation in vitro and in vivo by flow cytometry. Cytometry A. 2017;91:822-829 pubmed publisher
  35. Sur Chowdhury C, Giaglis S, Walker U, 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 pubmed publisher
  36. Wang Y, Xiao Y, Zhong L, Ye D, Zhang J, Tu Y, et al. Increased neutrophil elastase and proteinase 3 and augmented NETosis are closely associated with ?-cell autoimmunity in patients with type 1 diabetes. Diabetes. 2014;63:4239-48 pubmed publisher
  37. Masuda S, Nakazawa D, Shida H, Miyoshi A, Kusunoki Y, Tomaru U, et al. NETosis markers: Quest for specific, objective, and quantitative markers. Clin Chim Acta. 2016;459:89-93 pubmed publisher
  38. Patel S, Kumar S, Jyoti A, Srinag B, Keshari R, Saluja R, et al. Nitric oxide donors release extracellular traps from human neutrophils by augmenting free radical generation. Nitric Oxide. 2010;22:226-34 pubmed publisher
  39. Nakazawa D, Shida H, Kusunoki Y, Miyoshi A, Nishio S, Tomaru U, et al. The responses of macrophages in interaction with neutrophils that undergo NETosis. J Autoimmun. 2016;67:19-28 pubmed publisher
  40. Nakazawa D, Tomaru U, Suzuki A, Masuda S, Hasegawa R, Kobayashi T, et al. Abnormal conformation and impaired degradation of propylthiouracil-induced neutrophil extracellular traps: implications of disordered neutrophil extracellular traps in a rat model of myeloperoxidase antineutrophil cytoplasmic antibody-associated vasc. Arthritis Rheum. 2012;64:3779-87 pubmed publisher
  41. Zhang S, Lu X, Shu X, Tian X, Yang H, Yang W, et al. Elevated plasma cfDNA may be associated with active lupus nephritis and partially attributed to abnormal regulation of neutrophil extracellular traps (NETs) in patients with systemic lupus erythematosus. Intern Med. 2014;53:2763-71 pubmed
  42. Ali R, Gandhi A, Meng H, Yalavarthi S, Vreede A, Estes S, et al. Adenosine receptor agonism protects against NETosis and thrombosis in antiphospholipid syndrome. Nat Commun. 2019;10:1916 pubmed publisher
  43. Silva L, Doyle A, Greenwell Wild T, Dutzan N, Tran C, Abusleme L, et al. Fibrin is a critical regulator of neutrophil effector function at the oral mucosal barrier. Science. 2021;374:eabl5450 pubmed publisher
  44. Khandpur R, Carmona Rivera C, Vivekanandan Giri A, Gizinski A, Yalavarthi S, Knight J, et al. NETs are a source of citrullinated autoantigens and stimulate inflammatory responses in rheumatoid arthritis. Sci Transl Med. 2013;5:178ra40 pubmed publisher
  45. Rossaint J, Herter J, Van Aken H, Napirei M, Döring Y, Weber C, et al. Synchronized integrin engagement and chemokine activation is crucial in neutrophil extracellular trap-mediated sterile inflammation. Blood. 2014;123:2573-84 pubmed publisher
  46. Warnatsch A, Ioannou M, Wang Q, Papayannopoulos V. Inflammation. Neutrophil extracellular traps license macrophages for cytokine production in atherosclerosis. Science. 2015;349:316-20 pubmed publisher
  47. Keshari R, Jyoti A, Dubey M, Kothari N, Kohli M, Bogra J, et al. Cytokines induced neutrophil extracellular traps formation: implication for the inflammatory disease condition. PLoS ONE. 2012;7:e48111 pubmed publisher
  48. Alfaro C, Teijeira A, Oñate C, Perez G, Sanmamed M, Andueza M, et al. Tumor-Produced Interleukin-8 Attracts Human Myeloid-Derived Suppressor Cells and Elicits Extrusion of Neutrophil Extracellular Traps (NETs). Clin Cancer Res. 2016;22:3924-36 pubmed publisher
  49. Cacciotto C, Cubeddu T, Addis M, Anfossi A, Tedde V, Tore G, et al. Mycoplasma lipoproteins are major determinants of neutrophil extracellular trap formation. Cell Microbiol. 2016;18:1751-1762 pubmed publisher
  50. Berends E, Horswill A, Haste N, Monestier M, Nizet V, von Köckritz Blickwede M. Nuclease expression by Staphylococcus aureus facilitates escape from neutrophil extracellular traps. J Innate Immun. 2010;2:576-86 pubmed publisher
  51. Beiter K, Wartha F, Albiger B, Normark S, Zychlinsky A, Henriques Normark B. An endonuclease allows Streptococcus pneumoniae to escape from neutrophil extracellular traps. Curr Biol. 2006;16:401-7 pubmed
  52. Mori Y, Yamaguchi M, Terao Y, Hamada S, Ooshima T, Kawabata S. ?-Enolase of Streptococcus pneumoniae induces formation of neutrophil extracellular traps. J Biol Chem. 2012;287:10472-81 pubmed publisher
  53. Buchanan J, Simpson A, Aziz R, Liu G, Kristian S, Kotb M, et al. DNase expression allows the pathogen group A Streptococcus to escape killing in neutrophil extracellular traps. Curr Biol. 2006;16:396-400 pubmed
  54. Grinberg N, Elazar S, Rosenshine I, Shpigel N. Beta-hydroxybutyrate abrogates formation of bovine neutrophil extracellular traps and bactericidal activity against mammary pathogenic Escherichia coli. Infect Immun. 2008;76:2802-7 pubmed publisher
  55. Bruns S, Kniemeyer O, Hasenberg M, Aimanianda V, Nietzsche S, Thywissen A, et al. Production of extracellular traps against Aspergillus fumigatus in vitro and in infected lung tissue is dependent on invading neutrophils and influenced by hydrophobin RodA. PLoS Pathog. 2010;6:e1000873 pubmed publisher
  56. McCormick A, Heesemann L, Wagener J, Marcos V, Hartl D, Loeffler J, et al. NETs formed by human neutrophils inhibit growth of the pathogenic mold Aspergillus fumigatus. Microbes Infect. 2010;12:928-36 pubmed publisher
  57. Baker V, Imade G, Molta N, Tawde P, Pam S, Obadofin M, et al. Cytokine-associated neutrophil extracellular traps and antinuclear antibodies in Plasmodium falciparum infected children under six years of age. Malar J. 2008;7:41 pubmed publisher
  58. Wardini A, Guimarães Costa A, Nascimento M, Nadaes N, Danelli M, Mazur C, et al. Characterization of neutrophil extracellular traps in cats naturally infected with feline leukemia virus. J Gen Virol. 2010;91:259-64 pubmed publisher
  59. Saitoh T, Komano J, Saitoh Y, Misawa T, Takahama M, Kozaki T, et al. Neutrophil extracellular traps mediate a host defense response to human immunodeficiency virus-1. Cell Host Microbe. 2012;12:109-16 pubmed publisher
  60. Silvin A, Chapuis N, Dunsmore G, Goubet A, Dubuisson A, Derosa L, et al. Elevated Calprotectin and Abnormal Myeloid Cell Subsets Discriminate Severe from Mild COVID-19. Cell. 2020;: pubmed publisher
  61. Menegazzi R, Decleva E, Dri P. Killing by neutrophil extracellular traps: fact or folklore?. Blood. 2012;119:1214-6 pubmed publisher
  62. Seper A, Hosseinzadeh A, Gorkiewicz G, Lichtenegger S, Roier S, Leitner D, et al. Vibrio cholerae evades neutrophil extracellular traps by the activity of two extracellular nucleases. PLoS Pathog. 2013;9:e1003614 pubmed publisher
  63. Derré Bobillot A, Cortes Perez N, Yamamoto Y, Kharrat P, Couvé E, Da Cunha V, et al. Nuclease A (Gbs0661), an extracellular nuclease of Streptococcus agalactiae, attacks the neutrophil extracellular traps and is needed for full virulence. Mol Microbiol. 2013;89:518-31 pubmed publisher
  64. Zhu Y, Chen X, Liu X. NETosis and Neutrophil Extracellular Traps in COVID-19: Immunothrombosis and Beyond. Front Immunol. 2022;13:838011 pubmed publisher
  65. Munoz L, Boeltz S, Bilyy R, Schauer C, Mahajan A, Widulin N, et al. Neutrophil Extracellular Traps Initiate Gallstone Formation. Immunity. 2019;: pubmed publisher
  66. Yalavarthi S, Gould T, Rao A, Mazza L, Morris A, Nuñez Alvarez C, 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 pubmed publisher
  67. Meng H, Yalavarthi S, Kanthi Y, Mazza L, Elfline M, Luke C, et al. In Vivo Role of Neutrophil Extracellular Traps in Antiphospholipid Antibody-Mediated Venous Thrombosis. Arthritis Rheumatol. 2017;69:655-667 pubmed publisher
  68. Leppkes M, Maueröder C, Hirth S, Nowecki S, Günther C, Billmeier U, et al. Externalized decondensed neutrophil chromatin occludes pancreatic ducts and drives pancreatitis. Nat Commun. 2016;7:10973 pubmed publisher
  69. Menegazzo L, Ciciliot S, Poncina N, Mazzucato M, Persano M, Bonora B, et al. NETosis is induced by high glucose and associated with type 2 diabetes. Acta Diabetol. 2015;52:497-503 pubmed publisher
  70. Gupta A, Hasler P, Gebhardt S, Holzgreve W, Hahn S. Occurrence of neutrophil extracellular DNA traps (NETs) in pre-eclampsia: a link with elevated levels of cell-free DNA?. Ann N Y Acad Sci. 2006;1075:118-22 pubmed
  71. Mangold A, Alias S, Scherz T, Hofbauer T, Jakowitsch J, Panzenböck A, et al. Coronary neutrophil extracellular trap burden and deoxyribonuclease activity in ST-elevation acute coronary syndrome are predictors of ST-segment resolution and infarct size. Circ Res. 2015;116:1182-92 pubmed publisher
  72. Fuchs T, Brill A, Duerschmied D, Schatzberg D, Monestier M, Myers D, et al. Extracellular DNA traps promote thrombosis. Proc Natl Acad Sci U S A. 2010;107:15880-5 pubmed publisher
  73. Martinod K, Demers M, Fuchs T, Wong S, Brill A, Gallant M, et al. Neutrophil histone modification by peptidylarginine deiminase 4 is critical for deep vein thrombosis in mice. Proc Natl Acad Sci U S A. 2013;110:8674-9 pubmed publisher
  74. Lögters T, Margraf S, Altrichter J, Cinatl J, Mitzner S, Windolf J, et al. The clinical value of neutrophil extracellular traps. Med Microbiol Immunol. 2009;198:211-9 pubmed publisher
  75. Wong S, Demers M, Martinod K, Gallant M, Wang Y, Goldfine A, et al. Diabetes primes neutrophils to undergo NETosis, which impairs wound healing. Nat Med. 2015;21:815-9 pubmed publisher
  76. Ramos Kichik V, Mondragón Flores R, Mondragón Castelán M, Gonzalez Pozos S, Muñiz Hernández S, Rojas Espinosa O, et al. Neutrophil extracellular traps are induced by Mycobacterium tuberculosis. Tuberculosis (Edinb). 2009;89:29-37 pubmed publisher
  77. Demers M, Wagner D. NETosis: a new factor in tumor progression and cancer-associated thrombosis. Semin Thromb Hemost. 2014;40:277-83 pubmed publisher
  78. Chow O, von Köckritz Blickwede M, Bright A, Hensler M, Zinkernagel A, Cogen A, et al. Statins enhance formation of phagocyte extracellular traps. Cell Host Microbe. 2010;8:445-54 pubmed publisher
  79. Vega V, Crotty Alexander L, Charles W, Hwang J, Nizet V, De Maio A. Activation of the stress response in macrophages alters the M1/M2 balance by enhancing bacterial killing and IL-10 expression. J Mol Med (Berl). 2014;92:1305-17 pubmed publisher
  80. Shen F, Tang X, Cheng W, Wang Y, Wang C, Shi X, et al. Fosfomycin enhances phagocyte-mediated killing of Staphylococcus aureus by extracellular traps and reactive oxygen species. Sci Rep. 2016;6:19262 pubmed publisher
  81. King P, Sharma R, O Sullivan K, Selemidis S, Lim S, Radhakrishna N, et al. Nontypeable Haemophilus influenzae induces sustained lung oxidative stress and protease expression. PLoS ONE. 2015;10:e0120371 pubmed publisher
  82. O Sullivan K, Lo C, Summers S, Elgass K, McMillan P, Longano A, et al. Renal participation of myeloperoxidase in antineutrophil cytoplasmic antibody (ANCA)-associated glomerulonephritis. Kidney Int. 2015;88:1030-46 pubmed publisher
  83. Wong K, Jacobs W. Mycobacterium tuberculosis exploits human interferon ? to stimulate macrophage extracellular trap formation and necrosis. J Infect Dis. 2013;208:109-19 pubmed publisher
  84. Bonne Année S, Kerepesi L, Hess J, Wesolowski J, Paumet F, Lok J, et al. Extracellular traps are associated with human and mouse neutrophil and macrophage mediated killing of larval Strongyloides stercoralis. Microbes Infect. 2014;16:502-11 pubmed publisher
  85. Webster S, Daigneault M, Bewley M, Preston J, Marriott H, Walmsley S, et al. Distinct cell death programs in monocytes regulate innate responses following challenge with common causes of invasive bacterial disease. J Immunol. 2010;185:2968-79 pubmed publisher
  86. Jönsson B, Bylund J, Johansson B, Telemo E, Wold A. Cord-forming mycobacteria induce DNA meshwork formation by human peripheral blood mononuclear cells. Pathog Dis. 2013;67:54-66 pubmed publisher
  87. Bryukhin G, Shopova A. Characteristics of Mononuclear Extracellular Traps in the Offspring of Female Rats with Drug-Induced Hepatitis. Bull Exp Biol Med. 2015;159:435-7 pubmed publisher
  88. Muñoz Caro T, Silva L, Ritter C, Taubert A, Hermosilla C. Besnoitia besnoiti tachyzoites induce monocyte extracellular trap formation. Parasitol Res. 2014;113:4189-97 pubmed publisher
  89. Halder L, Abdelfatah M, Jo E, Jacobsen I, Westermann M, Beyersdorf N, et al. Factor H Binds to Extracellular DNA Traps Released from Human Blood Monocytes in Response to Candida albicans. Front Immunol. 2016;7:671 pubmed publisher
  90. Perez D, Munoz M, Molina J, Muñoz Caro T, Silva L, Taubert A, et al. Eimeria ninakohlyakimovae induces NADPH oxidase-dependent monocyte extracellular trap formation and upregulates IL-12 and TNF-?, IL-6 and CCL2 gene transcription. Vet Parasitol. 2016;227:143-50 pubmed publisher
  91. Aulik N, Hellenbrand K, Czuprynski C. Mannheimia haemolytica and its leukotoxin cause macrophage extracellular trap formation by bovine macrophages. Infect Immun. 2012;80:1923-33 pubmed publisher
  92. Hortin G, Gibson B, Fok K. Alpha 2-antiplasmin's carboxy-terminal lysine residue is a major site of interaction with plasmin. Biochem Biophys Res Commun. 1988;155:591-6 pubmed
  93. Reichel M, Muñoz Caro T, Sanchez Contreras G, Rubio García A, Magdowski G, Gärtner U, et al. Harbour seal (Phoca vitulina) PMN and monocytes release extracellular traps to capture the apicomplexan parasite Toxoplasma gondii. Dev Comp Immunol. 2015;50:106-15 pubmed publisher
  94. Verhoeven J, Baan C, Peeters A, Clahsen van Groningen M, Nieboer D, Herzog M, et al. Circulating cell-free nucleosomes as biomarker for kidney transplant rejection: a pilot study. Clin Epigenetics. 2021;13:32 pubmed publisher
ISSN : 2329-5139