Review Article
 

Neutrophil Extracellular Traps: Formation and Involvement in Disease Progression

Abstract

Neutrophils are the forerunner in innate immunity by defending the host organisms against infectious pathogens. During such process, neutrophils reach the site of inflammation/infection and eliminate the pathogens by phagocytosis as well as by forming the neutrophil extracellular traps (NETs). NETs trap and eradicate a number of microbes including bacteria, fungi, protozoa, viruses. NETs consist of DNA which is decorated with histones and granular proteins such as neutrophil elastase (NE), gelatinase, myeloperoxidase. NETosis (a process of NETs formation) is also involved in many inflammatory and autoimmune disorders with a major contribution to acute respiratory distress syndrome, sepsis, cystic fibrosis, periodontitis. Hyper NETosis or ineffective clearance of NETs would likely increase the risk of auto-antibody generation against NETs components and contribution in auto-inflammatory diseases. The purpose of this review is intended to highlight the molecular mechanisms of NETosis and its antimicrobial effect. Furthermore, a current status of NETosis in the pathogenesis of inflammatory and autoimmune disorders has been reviewed for better understanding. 

  1. Mayadas TN, Cullere X, Lowell CA. The multifaceted functions of neutrophils. Annual Review of Pathology: Mechanisms of Disease. 2014 Jan 24; 9:181-218.
  2. Nathan C. Neutrophils and immunity: challenges and opportunities. Nat Rev Immunol 2006; 6(3):173–82.
  3. Segal AW. How Neutrophils Kill Microbes. Annu Rev Immunol 2005; 23:197–223.
  4. Robertson CM, Perrone EE, McConnell KW, Dunne WM, Boody B, Brahmbhatt T, et al. Neutrophil depletion causes a fatal defect in murine pulmonary Staphylococcus aureus clearance. J Surg Res 2008; 150(2):278–85.
  5. Swamydas M, Gao J-L, Break TJ, Johnson MD, Jaeger M, Rodriguez CA, et al. CXCR1-mediated neutrophil degranulation and fungal killing promote Candida clearance and host survival. Sci Transl Med 2016; 8(322):322ra10.
  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):312–26.
  7. Selders GS, Fetz AE, Radic MZ, Bowlin GL. An overview of the role of neutrophils in innate immunity, inflammation and host-biomaterial integration. Regen Biomater 2017; 4(1):55-68.
  8. Rosales C, Demaurex N, Lowell CA, Uribe-Querol E. Neutrophils: Their role in innate and adaptive immunity. J Immunol Res 2016; 2016.
  9. Borregaard N, Sørensen OE, Theilgaard-Mönch K. Neutrophil granules: a library of innate immunity proteins. Trends Immunol 2007; 28(8):340–5.
  10. Brinkmann V, Reichard U, Goosmann C, Fauler B, Uhlemann Y, Weiss DS, et al. Neutrophil extracellular traps kill bacteria. Science 2004; 303(5663):1532–5.
  11. Fuchs TA, 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(2):231–41.
  12. Brinkmann V, Zychlinsky A. Beneficial suicide: why neutrophils die to make NETs. Nat Rev Microbiol 2007; 5(8):577–82.
  13. Metzler KD, Fuchs TA, Nauseef WM, Reumaux D, Roesler J, Schulze I, et al. Myeloperoxidase is required for neutrophil extracellular trap formation: implications for innate immunity. Blood 2011; 117(3):953–9.
  14. Wang Y, Li M, Stadler S, Correll S, Li P, Wang D, et al. Histone hypercitrullination mediates chromatin decondensation and neutrophil extracellular trap formation. J Cell Biol 2009; 184(2):205–13.
  15. 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):1853–62.
  16. 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(2):290–304.
  17. Raad H, Paclet M-H, Boussetta T, Kroviarski Y, Morel F, Quinn MT, et al. Regulation of the phagocyte NADPH oxidase activity: phosphorylation of gp91phox/NOX2 by protein kinase C enhances its diaphorase activity and binding to Rac2, p67phox, and p47phox. FASEB J 2009; 23(4):1011–22.
  18. Desai J, Kumar SV, Mulay SR, 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(1):223–9.
  19. DeSouza-Vieira T, Guimarães-Costa A, Rochael NC, Lira MN, Nascimento MT, de Souza Lima-Gomez P, et al. Neutrophil extracellular traps release induced by Leishmania: role of PI3Kγ, ERK, PI3Kσ, PKC, and [Ca2+]. J Leukoc Biol 2016; 100(4):801-10.
  20. Yipp BG, Kubes P. NETosis: how vital is it? Blood 2013; 122(16):2784–94.
  21. Keshari RS, 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(10):e48111.
  22. Patel S, Kumar S, Jyoti A, Srinag BS, Keshari RS, Saluja R, et al. Nitric oxide donors release extracellular traps from human neutrophils by augmenting free radical generation. Nitric Oxide Biol Chem 2010; 22(3):226–34.
  23. Yoo D, Winn M, Pang L, Moskowitz SM, Malech HL, Leto TL, et al. Release of cystic fibrosis airway inflammatory markers from Pseudomonas aeruginosa-stimulated human neutrophils involves NADPH oxidase-dependent extracellular DNA trap formation. J Immunol 1950; 192(10):4728–38.
  24. Menten-Dedoyart C, Faccinetto C, Golovchenko M, Dupiereux I, Van Lerberghe P-B, Dubois S, et al. Neutrophil extracellular traps entrap and kill Borrelia burgdorferi sensu stricto spirochetes and are not affected by Ixodes ricinus tick saliva. J Immunol 2012; 189(11):5393–401.
  25. de Jong HK, Koh GC, Achouiti A, van der Meer AJ, Bulder I, Stephan F, et al. Neutrophil extracellular traps in the host defense against sepsis induced by Burkholderia pseudomallei (melioidosis). Intensive Care Med Exp 2014; 2(1):21.
  26. Mori Y, Yamaguchi M, Terao Y, Hamada S, Ooshima T, Kawabata S. α-Enolase of Streptococcus pneumoniae induces formation of neutrophil extracellular traps. J Biol Chem 2012; 287(13):10472–81.
  27. Lappann M, Danhof S, Guenther F, Olivares-Florez S, Mordhorst IL, Vogel U. In vitro resistance mechanisms of Neisseria meningitidis against neutrophil extracellular traps. Mol Microbiol 2013; 89(3):433–49.
  28. Gunderson CW, Seifert HS. Neisseria gonorrhoeae Elicits Extracellular Traps in Primary Neutrophil Culture While Suppressing the Oxidative Burst. MBio 2015; 6(1):e02452-14.
  29. Seper A, Hosseinzadeh A, Gorkiewicz G, Lichtenegger S, Roier S, Leitner DR, et al. Vibrio cholerae Evades Neutrophil Extracellular Traps by the Activity of Two Extracellular Nucleases. PLOS Pathog 2013; 9(9):e1003614.
  30. Braian C, Hogea V, Stendahl O. Mycobacterium tuberculosis- induced neutrophil extracellular traps activate human macrophages. J Innate Immun 2013; 5(6):591–602.
  31. 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 1950; 190(8):4136–48.
  32. Röhm M, Grimm MJ, D’Auria AC, Almyroudis NG, Segal BH, Urban CF. NADPH oxidase promotes neutrophil extracellular trap formation in pulmonary aspergillosis. Infect Immun 2014; 82(5):1766–77.
  33. Abi Abdallah DS, Lin C, Ball CJ, King MR, Duhamel GE, Denkers EY. Toxoplasma gondii triggers release of human and mouse neutrophil extracellular traps. Infect Immun 2012; 80(2):768–77.
  34. Waisberg M, Molina-Cruz A, Mizurini DM, Gera N, Sousa BC, Ma D, et al. Plasmodium falciparum Infection Induces Expression of a Mosquito Salivary Protein (Agaphelin) That Targets Neutrophil Function and Inhibits Thrombosis without Impairing Hemostasis. PLOS Pathog 2014; 10(9):e1004338.
  35. Kamoshida G, Kikuchi-Ueda T, Tansho-Nagakawa S, Nakano R, Nakano A, Kikuchi H, et al. Acinetobacter baumannii escape from neutrophil extracellular traps (NETs). J Infect Chemother Off J Jpn Soc Chemother 2015; 21(1):43–9.
  36. Rocha JDB, Nascimento MTC, Decote-Ricardo D, Côrte-Real S, Morrot A, Heise N, et al. Capsular polysaccharides from Cryptococcus neoformans modulate production of neutrophil extracellular traps (NETs) by human neutrophils. Sci Rep 2015; 5:8008.
  37. Menegazzi R, Decleva E, Dri P. Killing by neutrophil extracellular traps: fact or folklore? Blood 2012; 119(5):1214–6.
  38. 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(1):109–16.
  39. Netea MG, Brown GD, Kullberg BJ, Gow NAR. An integrated model of the recognition of Candida albicans by the innate immune system. Nat Rev Microbiol 2008; 6(1):67–78.
  40. Urban CF, 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(10):e1000639.
  41. Buchanan JT, Simpson AJ, Aziz RK, Liu GY, Kristian SA, Kotb M, et al. DNase expression allows the pathogen group A Streptococcus to escape killing in neutrophil extracellular traps. Curr Biol CB 2006; 16(4):396–400.
  42. 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 CB 2006; 16(4):401–7.
  43. Juneau RA, Stevens JS, Apicella MA, Criss AK. A thermonuclease of Neisseria gonorrhoeae enhances bacterial escape from killing by neutrophil extracellular traps. J Infect Dis 2015; 212(2):316–24.
  44. de Buhr N, Neumann A, Jerjomiceva N, von Köckritz-Blickwede M, Baums CG. Streptococcus suis DNase SsnA contributes to degradation of neutrophil extracellular traps (NETs) and evasion of NET-mediated antimicrobial activity. Microbiol Read Engl 2014; 160(Pt 2):385–95.
  45. Morita C, Sumioka R, Nakata M, Okahashi N, Wada S, Yamashiro T, et al. Cell Wall-Anchored Nuclease of Streptococcus sanguinis Contributes to Escape from Neutrophil Extracellular Trap-Mediated Bacteriocidal Activity. PLOS ONE 2014; 9(8):e103125.
  46. Guimarães-Costa AB, DeSouza-Vieira TS, Paletta-Silva R, Freitas-Mesquita AL, Meyer-Fernandes JR, Saraiva EM. 3’-nucleotidase/nuclease activity allows Leishmania parasites to escape killing by neutrophil extracellular traps. Infect Immun 2014; 82(4):1732–40.
  47. Glass KA, Longley SJ, Bliss JM, Shaw SK. Protection of Candida parapsilosis from neutrophil killing through internalization by human endothelial cells. Virulence 2015; 6(5):504–14.
  48. Alghamdi AS, Foster DN. Seminal DNase frees spermatozoa entangled in neutrophil extracellular traps. Biol Reprod 2005; 73(6):1174–81.
  49. Fedorka CE, Scoggin KE, Woodward EM, Squires EL, Ball BA, Troedsson MH. The effect of select seminal plasma proteins on endometrial mRNA cytokine expression in mares susceptible to persistent mating‐induced endometritis. Reprod Domest Anim 2017; 52(1):89-96.
  50. Jorch SK, Kubes P. An emerging role for neutrophil extracellular traps in noninfectious disease. Nat Med 2017; 23(3):279-87.
  51. Keshari RS, Jyoti A, Kumar S, Dubey M, Verma A, Srinag BS, et al. Neutrophil extracellular traps contain mitochondrial as well as nuclear DNA and exhibit inflammatory potential. Cytom Part A 2012; 81(3):238–47.
  52. Fattal I, Shental N, Mevorach D, Anaya J-M, Livneh A, Langevitz P, et al. An antibody profile of systemic lupus erythematosus detected by antigen microarray. Immunology 2010; 130(3):337–43.
  53. Papayannopoulos V. Neutrophil extracellular traps in immunity and disease. Nat Rev Immunol 2017.
  54. Morgan WJ, Butler SM, Johnson CA, Colin AA, FitzSimmons SC, Geller DE, et al. Epidemiologic study of cystic fibrosis: design and implementation of a prospective, multicenter, observational study of patients with cystic fibrosis in the US and Canada. Pediatric pulmonology 1999; 28(4):231-41.
  55. Dwyer M, Shan Q, D'ortona S, Maurer R, Mitchell R, Olesen H, 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):765-79.
  56. Skopelja S, Hamilton BJ, Jones JD, Yang M-L, Mamula M, Ashare A, et al. The role for neutrophil extracellular traps in cystic fibrosis autoimmunity. JCI Insight 2016; 1(17):e88912.
  57. Rada B. Neutrophil extracellular trap release driven by bacterial motility: Relevance to cystic fibrosis lung disease. Commun Integr Biol 2017; 10(2):e1296610.
  58. Prandoni P, Lensing AW, Piccioli A, Bernardi E, Simioni P, Girolami B, et al. Recurrent venous thromboembolism and bleeding complications during anticoagulant treatment in patients with cancer and venous thrombosis. Blood 2002; 100(10):3484-8.
  59. Fuchs TA, Brill A, Duerschmied D, Schatzberg D, Monestier M, Myers DD, et al. Extracellular DNA traps promote thrombosis. Proc Natl Acad Sci USA 2010; 107(36):15880–5.
  60. Geddings JE, Mackman N. New players in haemostasis and thrombosis. Thromb Haemost 2014; 111(4):570–4.
  61. McDonald B, Davis RP, Kim S-J, Tse M, Esmon CT, Kolaczkowska E, et al. Platelets and neutrophil extracellular traps collaborate to promote intravascular coagulation during sepsis in mice. Blood 2017; 129(10):1357–67.
  62. Noubouossie DF, Whelihan MF, Yu YB, Sparkenbaugh E, Pawlinski R, Monroe DM, et al. In vitro activation of coagulation by human neutrophil DNA and histone proteins but not neutrophil extracellular traps. Blood 2017; 129(8):1021–9.
  63. Gould TJ, Vu T, Swystun LL, Dwivedi D, Mai S, Weitz JI, Liaw PC. Neutrophil extracellular traps promote thrombin generation through platelet-dependent and platelet-independent mechanisms. Arterioscler Thromb Vasc Biol 2014; 34(9):1977-84.
  64. Stouthard JM, Levi M, Hack CE, Veenhof CH, Romijn HA, Sauerwein HP, et al. Interleukin-6 stimulates coagulation, not fibrinolysis, in humans. Thromb Haemost 1996; 76(5):738-42
  65. Mutlu GM, Green D, Bellmeyer A, Baker CM, Burgess Z, Rajamannan N, et al. Ambient particulate matter accelerates coagulation via an IL-6–dependent pathway. J Clin Invest 2007; 117(10):2952-61.
  66. Haselmayer P, Grosse-Hovest L, von Landenberg P, Schild H, Radsak MP. TREM-1 ligand expression on platelets enhances neutrophil activation. Blood 2007; 110(3):1029-35.
  67. Wohner N, Keresztes Z, Sótonyi P, Szabó L, Komorowicz E, Machovich R, et al. Neutrophil granulocyte‐dependent proteolysis enhances platelet adhesion to the arterial wall under high‐shear flow. J Thromb Haemost 2010; 8(7):1624-31.
  68. Savchenko AS, Martinod K, Seidman MA, Wong SL, Borissoff JI, Piazza G, et al. Neutrophil extracellular traps form predominantly during the organizing stage of human venous thromboembolism development. J Thromb Haemost JTH 2014; 12(6):860–70.
  69. Jiménez Alcázar M, Napirei M, Panda R, Köhler EC, Kremer Hovinga JA, Mannherz HG, et al. Impaired DNase1‐mediated degradation of neutrophil extracellular traps is associated with acute thrombotic microangiopathies. J Thromb Haemost 2015; 13(5):732-42.
  70. Farquharson D, Butcher JP, Culshaw S. Periodontitis, Porphyromonas, and the pathogenesis of rheumatoid arthritis. Mucosal Immunol 2012; 5(2):112–20.
  71. Vitkov L, Klappacher M, Hannig M, Krautgartner WD. Extracellular neutrophil traps in periodontitis. J Periodontal Res 2009; 44(5):664–72.
  72. Palmer LJ, Chapple ILC, Wright HJ, Roberts A, Cooper PR. Extracellular deoxyribonuclease production by periodontal bacteria. J Periodontal Res 2012; 47(4):439–45.
  73. Hirschfeld J, Roberts HM, Chapple IL, Parčina M, Jepsen S, Johansson A, et al. Effects of Aggregatibacter actinomycetemcomitans leukotoxin on neutrophil migration and extracellular trap formation. J Oral Microbiol 2016; 8:83370.
  74. Graves DT, Cochran D. The contribution of interleukin-1 and tumor necrosis factor to periodontal tissue destruction. J Periodontol 2003; 74(3):391-401.
  75. Kida I, Kobayashi S, Takeuchi K, Tsuda H, Hashimoto H, Takasaki Y. Antineutrophil cytoplasmic antibodies against myeloperoxidase, proteinase 3, elastase, cathepsin G and lactoferrin in Japanese patients with rheumatoid arthritis. Mod Rheumatol 2011; 21(1):43–50.
  76. Kusunoki Y, Nakazawa D, Shida H, Hattanda F, Miyoshi A, Masuda S, et al. Peptidylarginine deiminase inhibitor suppresses neutrophil extracellular trap formation and MPO-ANCA production. Front Immunol 2016; 7:227.
  77. Pagnoux C, Mahr A, Cohen P, Guillevin L. Presentation and outcome of gastrointestinal involvement in systemic necrotizing vasculitides: analysis of 62 patients with polyarteritis nodosa, microscopic polyangiitis, Wegener granulomatosis, Churg-Strauss syndrome, or rheumatoid arthritis-associated vasculitis. Medicine 2005; 84(2):115-28.
  78. Wang H, Sha LL, Ma TT, Zhang LX, Chen M, Zhao MH. Circulating level of neutrophil extracellular traps is not a useful biomarker for assessing disease activity in antineutrophil cytoplasmic antibody-associated vasculitis. PloS one 2016; 11(2):e0148197.
  79. Söderberg D, Segelmark M. Neutrophil extracellular Traps in ANCA-Associated vasculitis. Front Immunol 2016; 7:256.
  80. Bartolucci P, Ramanoelina J, Cohen P, Mahr A, Godmer P, Le Hello C, et al. Efficacy of the anti‐TNF‐α antibody infliximab against refractory systemic vasculitides: an open pilot study on 10 patients. Rheumatology 2002; 41(10):1126-32.
  81. Falk RJ, Terrell RS, Charles LA, Jennette JC. Anti-neutrophilcytoplasmicautoantibodiesinduceneutrophils to degranulate and produce oxygen radicals in vitro. Proc Natl Acad Sci U S A 1990; 87(11):4115-9.
  82. Herrmann M, Zoller OM, Hagenhofer M, Voll R, Kalden JR. What triggers anti-dsDNA antibodies? Molecular biology reports 1996; 23(3-4):265-7.
  83. Pieterse E, Hofstra J, Berden J, Herrmann M, Dieker J, van der Vlag J. Acetylated histones contribute to the immunostimulatory potential of neutrophil extracellular traps in systemic lupus erythematosus. Clin Exp Immunol 2015; 179(1):68–74.
  84. Marder W, Knight JS, Kaplan MJ, Somers EC, Zhang X, O’Dell AA, et al. Placental histology and neutrophil extracellular traps in lupus and pre-eclampsia pregnancies. Lupus Sci Med 2016; 3(1):e000134.
  85. Lood C, Blanco LP, Purmalek MM, Carmona-Rivera C, De Ravin SS, Smith CK, et al. Neutrophil extracellular traps enriched in oxidized mitochondrial DNA are interferogenic and contribute to lupus-like disease. Nat Med 2016; 22(2):146–53.
  86. Lande R, Ganguly D, Facchinetti V, Frasca L, Conrad C, Gregorio J, et al. Neutrophils activate plasmacytoid dendritic cells by releasing self-DNA–peptide complexes in systemic lupus erythematosus. Sci Transl Med 2011; 3(73):73ra19.
  87. Dong G, Ye R, Shi W, Liu S, Wang T, Yang X, Yang N, Yu X. IL-17 induces autoantibody overproduction and peripheral blood mononuclear cell overexpression of IL-6 in lupus nephritis patients. Chin Med J (Engl) 2003; 116(4):543-8.
  88. Roussel L, Houle F, Chan C, Yao Y, Bérubé J, Olivenstein R, et al. IL-17 promotes p38 MAPK-dependent endothelial activation enhancing neutrophil recruitment to sites of inflammation. J Immunol 2010; 184(8):4531-7.
  89. Carmona-Rivera C, Zhao W, Yalavarthi S, Kaplan MJ. Neutrophil extracellular traps induce endothelial dysfunction in systemic lupus erythematosus through the activation of matrix metalloproteinase-2. Ann Rheum Dis 2015; 74(7):1417-24.
  90. Pratesi F, Dioni I, Tommasi C, Alcaro MC, Paolini I, Barbetti F, et al. Antibodies from patients with rheumatoid arthritis target citrullinated histone 4 contained in neutrophils extracellular traps. Ann Rheum Dis 2014; 73(7):1414-22.
  91. Scott DL, Farr M, Hawkins CF, Wilkinson R, Bold AM. Serum calcium levels in rheumatoid arthritis. Annals of the rheumatic diseases 1981; 40(6):580-3.
  92. Khandpur R, Carmona-Rivera C, Vivekanandan-Giri A, Gizinski A, Yalavarthi S, Knight JS, et al. NETs are a source of citrullinated autoantigens and stimulate inflammatory responses in rheumatoid arthritis. Sci Transl Med 2013; 5(178):178ra40.
  93. Romero V, Fert-Bober J, Nigrovic PA, Darrah E, Haque UJ, Lee DM, et al. Immune-mediated pore-forming pathways induce cellular hypercitrullination and generate citrullinated autoantigens in rheumatoid arthritis. Sci Transl Med 2013; 5(209):209ra150.
  94. Xu F, Zhao LH, Su JB, Chen T, Wang XQ, Chen JF, et al. The relationship between glycemic variability and diabetic peripheral neuropathy in type 2 diabetes with well-controlled HbA1c. Diabetol Metab Syndr 2014; 6(1):139.
  95. Joshi MB, Lad A, Prasad B, Alevoor S, Balakrishnan A, Ramachandra L, Satyamoorthy K. High glucose modulates IL‐6 mediated immune homeostasis through impeding neutrophil extracellular trap formation. FEBS lett 2013; 587(14):2241-6.
  96. Diana J, Simoni Y, Furio L, Beaudoin L, Agerberth B, Barrat F, et al. Crosstalk between neutrophils, B-1a cells and plasmacytoid dendritic cells initiates autoimmune diabetes. Nat Med 2013; 19(1):65-73.
  97. Dotta F, Censini S, van Halteren AG, Marselli L, Masini M, Dionisi S, et al. Coxsackie B4 virus infection of β cells and natural killer cell insulitis in recent-onset type 1 diabetic patients. Proc Natl Acad Sci U S A 2007; 104(12):5115-20.
  98. Thomas GM, Carbo C, Curtis BRs, Martinod K, Mazo IB, Schatzberg D, et al. Extracellular DNA traps are associated with the pathogenesis of TRALI in humans and mice. Blood 2012; 119(26):6335–43.
  99. Sørensen OE, Borregaard N. Neutrophil extracellular traps - the dark side of neutrophils. J Clin Invest 2016; 126(5):1612–20.
  100. Wong SL, Demers M, Martinod K, Gallant M, Wang Y, Goldfine AB, et al. Diabetes primes neutrophils to undergo NETosis, which impairs wound healing. Nat Med 2015; 21(7):815–9.
  101. Rohrbach AS, Slade DJ, Thompson PR, Mowen KA. Activation of PAD4 in NET formation. Front Immunol 2012; 3.

Files
IssueVol 17, No 3 (2018) QRcode
SectionReview Article(s)
Keywords
Autoimmune disease Inflammation Neutrophils Neutrophil extracellular traps

Rights and permissions
Creative Commons License This work is licensed under a Creative Commons Attribution-NonCommercial 4.0 International License.
How to Cite
1.
Kumar S, Gupta E, Kaushik S, Jyoti A. Neutrophil Extracellular Traps: Formation and Involvement in Disease Progression. Iran J Allergy Asthma Immunol. 2018;17(3):208-220.