J Rheum Dis.  2016 Apr;23(2):82-87. 10.4078/jrd.2016.23.2.82.

NETosis in Autoimmune Diseases

Affiliations
  • 1Department of Microbiology and Immunology, Pusan National University School of Medicine, Yangsan, Korea. dhsohn@pusan.ac.kr

Abstract

Neutrophils are the major antimicrobial cells of the innate immune system, which are recruited rapidly to the sites of infection and provide the primary defense against pathogens. Recent evidence suggests that neutrophils undergo a distinct cell death mechanism called NETosis, which not only contributes to the host defense, but also leads to severe pathological immune responses in cases of dysregulation. Here, we review the general features of NETosis as well as the generation of autoantigens and damage-associated molecular patterns by NETosis in autoimmune diseases. This review discusses the pathogenic role of NETosis in rheumatoid arthritis and systemic lupus erythematosus, where neutrophils may play a key role in the pathogenesis of these diseases, and suggest the possibility of neutrophil extracellular traps as biomarkers and therapeutic targets for the treatment of autoimmune diseases.

Keyword

Neutrophil extracellular traps; NETosis; Citrullination; Damage-associated molecular patterns; Autoimmune diseases

MeSH Terms

Arthritis, Rheumatoid
Autoantigens
Autoimmune Diseases*
Biomarkers
Cell Death
Immune System
Lupus Erythematosus, Systemic
Neutrophils
Autoantigens

Figure

  • Figure 1. NETosis. NETosis is initiated by binding of ligands such as immune complexes and microbes to their receptors on neutrophils, followed by calcium influx and reactive oxygen species (ROS) production. The intracellular signaling induces histone citrullination, chromatin decondensation, and disintegration of nuclear and granular membranes, which enables granular proteins to translocate to the nucleus. Subsequently, granular and cytoplasmic proteins are mixed with the chromatin. Finally, the neutrophil membrane ruptures and fully functional web-like neutrophil extracellular traps (NETs) are released.


Reference

1. Kolaczkowska E, Kubes P. Neutrophil recruitment and function in health and inflammation. Nat Rev Immunol. 2013; 13:159–75.
Article
2. Branzk N, Papayannopoulos V. Molecular mechanisms regulating NETosis in infection and disease. Semin Immunopathol. 2013; 35:513–30.
Article
3. Pruchniak MP, Kotuła I, Manda-Handzlik A. Neutrophil extracellular traps (Nets) impact upon autoimmune disorders. Cent Eur J Immunol. 2015; 40:217–24.
4. Brinkmann V, Reichard U, Goosmann C, Fauler B, Uhlemann Y, Weiss DS, et al. Neutrophil extracellular traps kill bacteria. Science. 2004; 303:1532–5.
Article
5. 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; 176:231–41.
Article
6. Barnado A, Crofford LJ, Oates JC. At the bedside: neutrophil extracellular traps (NETs) as targets for biomarkers and therapies in autoimmune diseases. J Leukoc Biol. 2016; 99:265–78.
Article
7. 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; 5:178ra40.
Article
8. 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:e1000639.
Article
9. Kessenbrock K, Krumbholz M, Schönermarck U, Back W, Gross WL, Werb Z, et al. Netting neutrophils in autoimmune small-vessel vasculitis. Nat Med. 2009; 15:623–5.
Article
10. Remijsen Q, Kuijpers TW, Wirawan E, Lippens S, Vandena-beele P, Vanden Berghe T. Dying for a cause: NETosis, mechanisms behind an antimicrobial cell death modality. Cell Death Differ. 2011; 18:581–8.
Article
11. Papayannopoulos V, Zychlinsky A. NETs: a new strategy for using old weapons. Trends Immunol. 2009; 30:513–21.
Article
12. Cheng OZ, Palaniyar N. NET balancing: a problem in inflammatory lung diseases. Front Immunol. 2013; 4:1.
Article
13. Vorobjeva NV, Pinegin BV. Neutrophil extracellular traps: mechanisms of formation and role in health and disease. Biochemistry (Mosc). 2014; 79:1286–96.
Article
14. Raad H, Paclet MH, 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; 23:1011–22.
Article
15. Hakkim A, Fuchs TA, Martinez NE, 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.
Article
16. Kaplan MJ, Radic M. Neutrophil extracellular traps: dou-ble-edged swords of innate immunity. J Immunol. 2012; 189:2689–95.
Article
17. Wang Y, Li M, Stadler S, Correll S, Li P, Wang D, et al. Histone hypercitrullination mediates chromatin deconden-sation and neutrophil extracellular trap formation. J Cell Biol. 2009; 184:205–13.
Article
18. Wright HL, Moots RJ, Edwards SW. The multifactorial role of neutrophils in rheumatoid arthritis. Nat Rev Rheumatol. 2014; 10:593–601.
Article
19. Neeli I, Khan SN, Radic M. Histone deimination as a response to inflammatory stimuli in neutrophils. J Immunol. 2008; 180:1895–902.
Article
20. 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; 207:1853–62.
Article
21. Hemmers S, Teijaro JR, Arandjelovic S, Mowen KA. PAD4-mediated neutrophil extracellular trap formation is not required for immunity against influenza infection. PLoS One. 2011; 6:e22043.
Article
22. Neeli I, Radic M. Opposition between PKC isoforms regulates histone deimination and neutrophil extracellular chromatin release. Front Immunol. 2013; 4:38.
Article
23. Parker H, Dragunow M, Hampton MB, Kettle AJ, Winter-bourn CC. Requirements for NADPH oxidase and myeloperoxidase in neutrophil extracellular trap formation differ depending on the stimulus. J Leukoc Biol. 2012; 92:841–849.
Article
24. Chen GY, Nuñez G. Sterile inflammation: sensing and reacting to damage. Nat Rev Immunol. 2010; 10:826–37.
Article
25. Piccinini AM, Midwood KS. DAMPening inflammation by modulating TLR signalling. Mediators Inflamm. 2010; 2010; 672395.
Article
26. Takeuchi O, Akira S. Pattern recognition receptors and inflammation. Cell. 2010; 140:805–20.
Article
27. Huang H, Tohme S, Al-Khafaji AB, Tai S, Loughran P, Chen L, et al. Damage-associated molecular pattern-activated neutrophil extracellular trap exacerbates sterile inflammatory liver injury. Hepatology. 2015; 62:600–14.
Article
28. Mitroulis I, Kambas K, Chrysanthopoulou A, Skendros P, Apostolidou E, Kourtzelis I, et al. Neutrophil extracellular trap formation is associated with IL-1β and autophagy-related signaling in gout. PLoS One. 2011; 6:e29318.
Article
29. Garcia-Romo GS, Caielli S, Vega B, Connolly J, Allantaz F, Xu Z, et al. Netting neutrophils are major inducers of type I IFN production in pediatric systemic lupus erythematosus. Sci Transl Med. 2011; 3:73ra20.
Article
30. Sohn DH, Rhodes C, Onuma K, Zhao X, Sharpe O, Gazitt T, et al. Local Joint inflammation and histone citrullination in a murine model of the transition from preclinical autoimmunity to inflammatory arthritis. Arthritis Rheumatol. 2015; 67:2877–87.
Article
31. Firestein GS. Evolving concepts of rheumatoid arthritis. Nature. 2003; 423:356–61.
Article
32. Spengler J, Lugonja B, Ytterberg AJ, Zubarev RA, Creese AJ, Pearson MJ, et al. Release of active peptidyl arginine de-iminases by neutrophils can explain production of extracellular citrullinated autoantigens in rheumatoid arthritis synovial fluid. Arthritis Rheumatol. 2015; 67:3135–45.
Article
33. Tsokos GC. Systemic lupus erythematosus. N Engl J Med. 2011; 365:2110–21.
Article
34. Hakkim A, Fürnrohr BG, Amann K, Laube B, Abed UA, 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.
Article
35. Villanueva E, Yalavarthi S, Berthier CC, Hodgin JB, Khandpur R, Lin AM, et al. Netting neutrophils induce endothelial damage, infiltrate tissues, and expose im-munostimulatory molecules in systemic lupus erythematosus. J Immunol. 2011; 187:538–52.
Article
36. 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; 3:73ra19.
37. 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; 22:146–53.
Article
38. Leffler J, Gullstrand B, Jönsen A, Nilsson JÅ, Martin M, Blom AM, et al. Degradation of neutrophil extracellular traps co-varies with disease activity in patients with systemic lupus erythematosus. Arthritis Res Ther. 2013; 15:R84.
Article
39. Willis VC, Gizinski AM, Banda NK, Causey CP, Knuckley B, Cordova KN, et al. N-α-benzoyl-N5-(2-chloro-1-iminoe-thyl)-L-ornithine amide, a protein arginine deiminase inhibitor, reduces the severity of murine collagen-induced arthritis. J Immunol. 2011; 186:4396–404.
Article
Full Text Links
  • JRD
Actions
Cited
CITED
export Copy
Close
Share
  • Twitter
  • Facebook
Similar articles
Copyright © 2024 by Korean Association of Medical Journal Editors. All rights reserved.     E-mail: koreamed@kamje.or.kr