Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Review Article
  • Published:

Neutrophils and neutrophil extracellular traps in the liver and gastrointestinal system

Key Points

  • Neutrophil extracellular traps (NETs) are extracellular structures composed of chromatin coated with histones, proteases and granular and cytosolic proteins that help catch and kill microorganisms

  • Kupffer cells and neutrophils in the liver cooperate to eliminate pathogens in circulation

  • NETs have a potential role in gastrointestinal infection and sepsis, whereas several pathogenic bacteria are capable of escaping or hijacking NET-mediated capturing and killing

  • Excess NET formation is associated with the pathology of inflammatory liver and gastrointestinal diseases

  • Exposure to components of NETs might generate autoantibodies in gastrointestinal autoimmune diseases and facilitate the inappropriate immune response

  • Therapies that target key pathways in NET formation, with or without other treatments, might improve the treatment for gastrointestinal inflammatory diseases, cancer and thrombosis

Abstract

Neutrophil extracellular traps (NETs) have an important role during infection by helping neutrophils to capture and kill pathogens. However, evidence is accumulating that uncontrolled or excessive production of NETs is related to the exacerbation of inflammation and the development of autoimmunity, cancer metastasis and inappropriate thrombosis. In this Review, we focus on the role of NETs in the liver and gastrointestinal system, outlining their protective and pathological effects. The latest mechanistic insights in NET formation, interactions between microorganisms and NETs and the relationship between neutrophil subtypes and their functions are also discussed. Additionally, we describe the potential importance of NET-related molecules, including cell-free DNA and hypercitrullinated histones, as biomarkers and targets for therapeutic intervention in gastrointestinal diseases.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Figure 1: Mechanisms of neutrophil extracellular trap formation.
Figure 2: Kupffer cells and neutrophils cooperate to capture bacteria in the liver vasculature.
Figure 3: Excessive neutrophil extracellular trap formation induced by inflammation causes tissue injury.
Figure 4: Tumour-induced neutrophil extracellular trap formation facilitates tumorigenesis and thrombus formation.

Similar content being viewed by others

References

  1. Kolaczkowska, E. & Kubes, P. Neutrophil recruitment and function in health and inflammation. Nat. Rev. Immunol. 13, 159–175 (2013).

    Article  CAS  PubMed  Google Scholar 

  2. Brinkmann, V. et al. Neutrophil extracellular traps kill bacteria. Science 303, 1532–1535 (2004).

    Article  CAS  PubMed  Google Scholar 

  3. Li, P. et al. PAD4 is essential for antibacterial innate immunity mediated by neutrophil extracellular traps. J. Exp. Med. 207, 1853–1862 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  4. Clark, S. R. et al. Platelet TLR4 activates neutrophil extracellular traps to ensnare bacteria in septic blood. Nat. Med. 13, 463–469 (2007).

    CAS  PubMed  Google Scholar 

  5. Pilsczek, F. H. et al. A novel mechanism of rapid nuclear neutrophil extracellular trap formation in response to Staphylococcus aureus. J. Immunol. 185, 7413–7425 (2010).

    CAS  PubMed  Google Scholar 

  6. Bianchi, M. et al. Restoration of NET formation by gene therapy in CGD controls aspergillosis. Blood 114, 2619–2622 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  7. Jorch, S. K. & Kubes, P. An emerging role for neutrophil extracellular traps in noninfectious disease. Nat. Med. 23, 279–287 (2017).

    CAS  PubMed  Google Scholar 

  8. Lood, C. et al. Neutrophil extracellular traps enriched in oxidized mitochondrial DNA are interferogenic and contribute to lupus-like disease. Nat. Med. 22, 146–153 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  9. Wong, S. L. et al. Diabetes primes neutrophils to undergo NETosis, which impairs wound healing. Nat. Med. 21, 815–819 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  10. Fuchs, T. A. et al. Extracellular DNA traps promote thrombosis. Proc. Natl Acad. Sci. USA 107, 15880–15885 (2010).

    CAS  PubMed  Google Scholar 

  11. Caudrillier, A. et al. Platelets induce neutrophil extracellular traps in transfusion-related acute lung injury. J. Clin. Invest. 122, 2661–2671 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  12. Bennike, T. B. et al. Neutrophil extracellular traps in ulcerative colitis: a proteome analysis of intestinal biopsies. Inflamm. Bowel Dis. 21, 2052–2067 (2015).

    PubMed  PubMed Central  Google Scholar 

  13. Cools-Lartigue, J. et al. Neutrophil extracellular traps sequester circulating tumor cells and promote metastasis. J. Clin. Invest. 123, 3446–3458 (2013).

    CAS  PubMed Central  Google Scholar 

  14. Takei, H., Araki, A., Watanabe, H., Ichinose, A. & Sendo, F. Rapid killing of human neutrophils by the potent activator phorbol 12-myristate 13-acetate (PMA) accompanied by changes different from typical apoptosis or necrosis. J. Leukoc. Biol. 59, 229–240 (1996).

    CAS  PubMed  Google Scholar 

  15. Urban, C. F. et al. Neutrophil extracellular traps contain calprotectin, a cytosolic protein complex involved in host defense against Candida albicans. PLoS Pathog. 5, e1000639 (2009).

    PubMed  PubMed Central  Google Scholar 

  16. Jaillon, S. et al. The humoral pattern recognition receptor PTX3 is stored in neutrophil granules and localizes in extracellular traps. J. Exp. Med. 204, 793–804 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  17. Fuchs, T. A. et al. Novel cell death program leads to neutrophil extracellular traps. J. Cell Biol. 176, 231–241 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  18. Kobayashi, S. D. & DeLeo, F. R. Role of neutrophils in innate immunity: a systems biology-level approach. Wiley Interdiscip. Rev. Syst. Biol. Med. 1, 309–333 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  19. Gupta, A. K., Giaglis, S., Hasler, P. & Hahn, S. Efficient neutrophil extracellular trap induction requires mobilization of both intracellular and extracellular calcium pools and is modulated by cyclosporine A. PLoS ONE 9, e97088 (2014).

    PubMed  PubMed Central  Google Scholar 

  20. Hakkim, A. et al. Activation of the Raf-MEK-ERK pathway is required for neutrophil extracellular trap formation. Nat. Chem. Biol. 7, 75–77 (2011).

    CAS  PubMed  Google Scholar 

  21. Van Avondt, K., van der Linden, M., Naccache, P. H., Egan, D. A. & Meyaard, L. Signal inhibitory receptor on leukocytes-1 limits the formation of neutrophil extracellular traps, but preserves intracellular bacterial killing. J. Immunol. 196, 3686–3694 (2016).

    CAS  PubMed  Google Scholar 

  22. Wang, Y. et al. Histone hypercitrullination mediates chromatin decondensation and neutrophil extracellular trap formation. J. Cell Biol. 184, 205–213 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  23. Palmer, L. J. et al. Hypochlorous acid regulates neutrophil extracellular trap release in humans. Clin. Exp. Immunol. 167, 261–268 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  24. Metzler, K. D., Goosmann, C., Lubojemska, A., Zychlinsky, A. & Papayannopoulos, V. A myeloperoxidase-containing complex regulates neutrophil elastase release and actin dynamics during NETosis. Cell Rep. 8, 883–896 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  25. Papayannopoulos, V., Metzler, K. D., Hakkim, A. & Zychlinsky, A. Neutrophil elastase and myeloperoxidase regulate the formation of neutrophil extracellular traps. J. Cell Biol. 191, 677–691 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  26. Amulic, B. et al. Cell-cycle proteins control production of neutrophil extracellular traps. Dev. Cell 43, 449–462.e5 (2017).

    CAS  PubMed  Google Scholar 

  27. Remijsen, Q. et al. Neutrophil extracellular trap cell death requires both autophagy and superoxide generation. Cell Res. 21, 290–304 (2011).

    CAS  PubMed  Google Scholar 

  28. Desai, J., Mulay, S. R., Nakazawa, D. & Anders, H. J. Matters of life and death. How neutrophils die or survive along NET release and is “NETosis” = necroptosis? Cell. Mol. Life Sci. 73, 2211–2219 (2016).

    CAS  PubMed  Google Scholar 

  29. Yipp, B. G. et al. Infection-induced NETosis is a dynamic process involving neutrophil multitasking in vivo. Nat. Med. 18, 1386–1393 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  30. Yipp, B. G. & Kubes, P. NETosis: how vital is it? Blood 122, 2784–2794 (2013).

    CAS  PubMed  Google Scholar 

  31. Byrd, A. S., O'Brien, X. M., Johnson, C. M., Lavigne, L. M. & Reichner, J. S. An extracellular matrix-based mechanism of rapid neutrophil extracellular trap formation in response to Candida albicans. J. Immunol. 190, 4136–4148 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  32. McDonald, B., Urrutia, R., Yipp, B. G., Jenne, C. N. & Kubes, P. Intravascular neutrophil extracellular traps capture bacteria from the bloodstream during sepsis. Cell Host Microbe 12, 324–333 (2012).

    CAS  PubMed  Google Scholar 

  33. Deppermann, C. & Kubes, P. Platelets and infection. Semin. Immunol. 28, 536–545 (2016).

    CAS  PubMed  Google Scholar 

  34. Rochael, N. C. et al. Classical ROS-dependent and early/rapid ROS-independent release of neutrophil extracellular traps triggered by Leishmania parasites. Sci. Rep. 5, 18302 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  35. Leppkes, M. et al. Externalized decondensed neutrophil chromatin occludes pancreatic ducts and drives pancreatitis. Nat. Commun. 7, 10973 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  36. Yousefi, S., Mihalache, C., Kozlowski, E., Schmid, I. & Simon, H. U. Viable neutrophils release mitochondrial DNA to form neutrophil extracellular traps. Cell Death Differ. 16, 1438–1444 (2009).

    CAS  PubMed  Google Scholar 

  37. Yousefi, S. et al. Catapult-like release of mitochondrial DNA by eosinophils contributes to antibacterial defense. Nat. Med. 14, 949–953 (2008).

    CAS  PubMed  Google Scholar 

  38. Konig, M. F. & Andrade, F. A. Critical reappraisal of neutrophil extracellular traps and NETosis mimics based on differential requirements for protein citrullination. Front. Immunol. 7, 461 (2016).

    PubMed  PubMed Central  Google Scholar 

  39. Hoppenbrouwers, T. et al. In vitro induction of NETosis: comprehensive live imaging comparison and systematic review. PLoS ONE 12, e0176472 (2017).

    PubMed  PubMed Central  Google Scholar 

  40. Zhang, Q. et al. Circulating mitochondrial DAMPs cause inflammatory responses to injury. Nature 464, 104–107 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  41. Marques, P. E. et al. Hepatic DNA deposition drives drug-induced liver injury and inflammation in mice. Hepatology 61, 348–360 (2015).

    CAS  PubMed  Google Scholar 

  42. Nauseef, W. M. & Kubes, P. Pondering neutrophil extracellular traps with healthy skepticism. Cell. Microbiol. 18, 1349–1357 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  43. Belorgey, D. & Bieth, J. G. Effect of polynucleotides on the inhibition of neutrophil elastase by mucus proteinase inhibitor and alpha 1-proteinase inhibitor. Biochemistry 37, 16416–16422 (1998).

    CAS  PubMed  Google Scholar 

  44. Kolaczkowska, E. et al. Molecular mechanisms of NET formation and degradation revealed by intravital imaging in the liver vasculature. Nat. Commun. 6, 6673 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  45. Yan, J., Li, S. & Li, S. The role of the liver in sepsis. Int. Rev. Immunol. 33, 498–510 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  46. McDonald, B. & Kubes, P. Neutrophils and intravascular immunity in the liver during infection and sterile inflammation. Toxicol. Pathol. 40, 157–165 (2012).

    CAS  PubMed  Google Scholar 

  47. Surewaard, B. G. et al. Identification and treatment of the Staphylococcus aureus reservoir in vivo. J. Exp. Med. 213, 1141–1151 (2016).

    PubMed  PubMed Central  Google Scholar 

  48. David, B. A. et al. Combination of mass cytometry and imaging analysis reveals origin, location, and functional repopulation of liver myeloid cells in mice. Gastroenterology 151, 1176–1191 (2016).

    CAS  PubMed  Google Scholar 

  49. Krenkel, O. & Tacke, F. Liver macrophages in tissue homeostasis and disease. Nat. Rev. Immunol. 17, 306–321 (2017).

    CAS  PubMed  Google Scholar 

  50. Helmy, K. Y. et al. CRIg: a macrophage complement receptor required for phagocytosis of circulating pathogens. Cell 124, 915–927 (2006).

    CAS  PubMed  Google Scholar 

  51. Holers, V. M. Complement and its receptors: new insights into human disease. Annu. Rev. Immunol. 32, 433–459 (2014).

    CAS  PubMed  Google Scholar 

  52. Zeng, Z. et al. CRIg functions as a macrophage pattern recognition receptor to directly bind and capture blood-borne Gram-positive bacteria. Cell Host Microbe 20, 99–106 (2016).

    CAS  PubMed  Google Scholar 

  53. Ravetch, J. V. & Bolland, S. IgG Fc receptors. Annu. Rev. Immunol. 19, 275–290 (2001).

    CAS  PubMed  Google Scholar 

  54. Wong, C. H., Jenne, C. N., Petri, B., Chrobok, N. L. & Kubes, P. Nucleation of platelets with blood-borne pathogens on Kupffer cells precedes other innate immunity and contributes to bacterial clearance. Nat. Immunol. 14, 785–792 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  55. Bleriot, C. et al. Liver-resident macrophage necroptosis orchestrates type 1 microbicidal inflammation and type-2-mediated tissue repair during bacterial infection. Immunity 42, 145–158 (2015).

    CAS  PubMed  Google Scholar 

  56. McDonald, B., Jenne, C. N., Zhuo, L., Kimata, K. & Kubes, P. Kupffer cells and activation of endothelial TLR4 coordinate neutrophil adhesion within liver sinusoids during endotoxemia. Am. J. Physiol. Gastrointest. Liver Physiol. 305, G797–806 (2013).

    CAS  PubMed  Google Scholar 

  57. Jenne, C. N. et al. Neutrophils recruited to sites of infection protect from virus challenge by releasing neutrophil extracellular traps. Cell Host Microbe 13, 169–180 (2013).

    CAS  PubMed  Google Scholar 

  58. Saitoh, T. et al. Neutrophil extracellular traps mediate a host defense response to human immunodeficiency virus-1. Cell Host Microbe 12, 109–116 (2012).

    CAS  PubMed  Google Scholar 

  59. Ward, C. M., Tetaz, T. J., Andrews, R. K. & Berndt, M. C. Binding of the von Willebrand factor A1 domain to histone. Thromb. Res. 86, 469–477 (1997).

    CAS  PubMed  Google Scholar 

  60. Averhoff, P., Kolbe, M., Zychlinsky, A. & Weinrauch, Y. Single residue determines the specificity of neutrophil elastase for Shigella virulence factors. J. Mol. Biol. 377, 1053–1066 (2008).

    CAS  PubMed  Google Scholar 

  61. Buchanan, J. T. et al. DNase expression allows the pathogen group A Streptococcus to escape killing in neutrophil extracellular traps. Curr. Biol. 16, 396–400 (2006).

    CAS  PubMed  Google Scholar 

  62. Derre-Bobillot, A. et al. Nuclease A (Gbs0661), an extracellular nuclease of Streptococcus agalactiae, attacks the neutrophil extracellular traps and is needed for full virulence. Mol. Microbiol. 89, 518–531 (2013).

    CAS  PubMed  Google Scholar 

  63. Mollerherm, H. et al. Yersinia enterocolitica-mediated degradation of neutrophil extracellular traps (NETs). FEMS Microbiol. Lett. 362, fnv192 (2015).

    PubMed  Google Scholar 

  64. Carestia, A. et al. Mediators and molecular pathways involved in the regulation of neutrophil extracellular trap formation mediated by activated platelets. J. Leukoc. Biol. 99, 153–162 (2016).

    CAS  PubMed  Google Scholar 

  65. Khoruts, A. & Sadowsky, M. J. Understanding the mechanisms of faecal microbiota transplantation. Nat. Rev. Gastroenterol. Hepatol. 13, 508–516 (2016).

    PubMed  PubMed Central  Google Scholar 

  66. Seper, A. et al. Vibrio cholerae evades neutrophil extracellular traps by the activity of two extracellular nucleases. PLoS Pathog. 9, e1003614 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  67. Juneau, R. A., Stevens, J. S., Apicella, M. A. & Criss, A. K. A thermonuclease of Neisseria gonorrhoeae enhances bacterial escape from killing by neutrophil extracellular traps. J. Infect. Dis. 212, 316–324 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  68. Abi Abdallah, D. S. et al. Toxoplasma gondii triggers release of human and mouse neutrophil extracellular traps. Infect. Immun. 80, 768–777 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  69. Konstantinidis, T. et al. Immunomodulatory role of clarithromycin in Acinetobacter baumannii infection via formation of neutrophil extracellular traps. Antimicrob. Agents Chemother. 60, 1040–1048 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  70. Brogden, G. et al. beta-Glucan protects neutrophil extracellular traps against degradation by Aeromonas hydrophila in carp (Cyprinus carpio). Fish Shellfish Immunol. 33, 1060–1064 (2012).

    CAS  PubMed  Google Scholar 

  71. Bruns, S. 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. 6, e1000873 (2010).

    PubMed  PubMed Central  Google Scholar 

  72. Marin-Esteban, V. et al. Afa/Dr diffusely adhering Escherichia coli strain C1845 induces neutrophil extracellular traps that kill bacteria and damage human enterocyte-like cells. Infect. Immun. 80, 1891–1899 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  73. Crane, J. K., Broome, J. E. & Lis, A. Biological activities of uric acid in infection due to enteropathogenic and Shiga-toxigenic Escherichia coli. Infect. Immun. 84, 976–988 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  74. Berends, E. T. et al. Nuclease expression by Staphylococcus aureus facilitates escape from neutrophil extracellular traps. J. Innate Immun. 2, 576–586 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  75. Liechti, G. W. & Goldberg, J. B. Helicobacter pylori salvages purines from extracellular host cell DNA utilizing the outer membrane-associated nuclease NucT. J. Bacteriol. 195, 4387–4398 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  76. Schilcher, K. et al. Increased neutrophil extracellular trap-mediated Staphylococcus aureus clearance through inhibition of nuclease activity by clindamycin and immunoglobulin. J. Infect. Dis. 210, 473–482 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  77. Neumann, A. et al. Novel role of the antimicrobial peptide LL-37 in the protection of neutrophil extracellular traps against degradation by bacterial nucleases. J. Innate Immun. 6, 860–868 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  78. Saha, P. et al. Bacterial siderophores hijack neutrophil functions. J. Immunol. 198, 4293–4303 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  79. Halverson, T. W., Wilton, M., Poon, K. K., Petri, B. & Lewenza, S. DNA is an antimicrobial component of neutrophil extracellular traps. PLoS Pathog. 11, e1004593 (2015).

    PubMed  PubMed Central  Google Scholar 

  80. Kalliomaki, M., Salminen, S., Poussa, T., Arvilommi, H. & Isolauri, E. Probiotics and prevention of atopic disease: 4-year follow-up of a randomised placebo-controlled trial. Lancet 361, 1869–1871 (2003).

    PubMed  Google Scholar 

  81. Yan, F. et al. Soluble proteins produced by probiotic bacteria regulate intestinal epithelial cell survival and growth. Gastroenterology 132, 562–575 (2007).

    CAS  PubMed  Google Scholar 

  82. Vong, L., Lorentz, R. J., Assa, A., Glogauer, M. & Sherman, P. M. Probiotic Lactobacillus rhamnosus inhibits the formation of neutrophil extracellular traps. J. Immunol. 192, 1870–1877 (2014).

    CAS  PubMed  Google Scholar 

  83. Vong, L. et al. Selective enrichment of commensal gut bacteria protects against Citrobacter rodentium-induced colitis. Am. J. Physiol. Gastrointest. Liver Physiol. 309, G181–G192 (2015).

    CAS  PubMed  Google Scholar 

  84. Vong, L., Yeung, C. W., Pinnell, L. J. & Sherman, P. M. Adherent-invasive Escherichia coli exacerbates antibiotic-associated intestinal dysbiosis and neutrophil extracellular trap activation. Inflamm. Bowel Dis. 22, 42–54 (2016).

    PubMed  Google Scholar 

  85. Dicker, A. J. et al. Neutrophil extracellular traps are associated with disease severity and microbiota diversity in chronic obstructive pulmonary disease. J. Allergy Clin. Immunol. http://dx.doi.org/10.1016/j.jaci.2017.04.022 (2017).

  86. Kubes, P. & Mehal, W. Z. Sterile inflammation in the liver. Gastroenterology 143, 1158–1172 (2012).

    CAS  PubMed  Google Scholar 

  87. Lowe, P. P. et al. Alcohol-related changes in the intestinal microbiome influence neutrophil infiltration, inflammation and steatosis in early alcoholic hepatitis in mice. PLoS ONE 12, e0174544 (2017).

    PubMed  PubMed Central  Google Scholar 

  88. Rensen, S. S. et al. Neutrophil-derived myeloperoxidase aggravates non-alcoholic steatohepatitis in low-density lipoprotein receptor-deficient mice. PLoS ONE 7, e52411 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  89. Talukdar, S. et al. Neutrophils mediate insulin resistance in mice fed a high-fat diet through secreted elastase. Nat. Med. 18, 1407–1412 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  90. Mansuy-Aubert, V. et al. Imbalance between neutrophil elastase and its inhibitor alpha1-antitrypsin in obesity alters insulin sensitivity, inflammation, and energy expenditure. Cell Metab. 17, 534–548 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  91. Merza, M. et al. Neutrophil extracellular traps induce trypsin activation, inflammation, and tissue damage in mice with severe acute pancreatitis. Gastroenterology 149, 1920–1931.e8 (2015).

    CAS  PubMed  Google Scholar 

  92. Bilyy, R. et al. Neutrophil extracellular traps form a barrier between necrotic and viable areas in acute abdominal inflammation. Front. Immunol. 7, 424 (2016).

    PubMed  PubMed Central  Google Scholar 

  93. Singer, M. et al. The Third International Consensus Definitions for Sepsis and Septic Shock (Sepsis-3). JAMA 315, 801–810 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  94. Tanaka, K. et al. In vivo characterization of neutrophil extracellular traps in various organs of a murine sepsis model. PLoS ONE 9, e111888 (2014).

    PubMed  PubMed Central  Google Scholar 

  95. Fattahi, F. et al. Organ distribution of histones after intravenous infusion of FITC histones or after sepsis. Immunol. Res. 61, 177–186 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  96. Dwivedi, D. J. et al. Prognostic utility and characterization of cell-free DNA in patients with severe sepsis. Crit. Care 16, R151 (2012).

    PubMed  PubMed Central  Google Scholar 

  97. Hampson, P. et al. Neutrophil dysfunction, immature granulocytes, and cell-free DNA are early biomarkers of sepsis in burn-injured patients: a prospective observational cohort study. Ann. Surg. 265, 1241–1249 (2017).

    PubMed  Google Scholar 

  98. Hirsch, J. G. Bactericidal action of histone. J. Exp. Med. 108, 925–944 (1958).

    CAS  PubMed  PubMed Central  Google Scholar 

  99. Saffarzadeh, M. et al. Neutrophil extracellular traps directly induce epithelial and endothelial cell death: a predominant role of histones. PLoS ONE 7, e32366 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  100. Xu, J. et al. Extracellular histones are major mediators of death in sepsis. Nat. Med. 15, 1318–1321 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  101. Czaikoski, P. G. et al. Neutrophil extracellular traps induce organ damage during experimental and clinical sepsis. PLoS ONE 11, e0148142 (2016).

    PubMed  PubMed Central  Google Scholar 

  102. McDonald, B. et al. Platelets and neutrophil extracellular traps collaborate to promote intravascular coagulation during sepsis in mice. Blood 129, 1357–1367 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  103. Martinod, K. et al. PAD4-deficiency does not affect bacteremia in polymicrobial sepsis and ameliorates endotoxemic shock. Blood 125, 1948–1956 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  104. Meng, W. et al. Depletion of neutrophil extracellular traps in vivo results in hypersusceptibility to polymicrobial sepsis in mice. Crit. Care 16, R137 (2012).

    PubMed  PubMed Central  Google Scholar 

  105. Derikx, J. P., Poeze, M., van Bijnen, A. A., Buurman, W. A. & Heineman, E. Evidence for intestinal and liver epithelial cell injury in the early phase of sepsis. Shock 28, 544–548 (2007).

    CAS  PubMed  Google Scholar 

  106. Gao, X. et al. Neutrophil extracellular traps contribute to the intestine damage in endotoxemic rats. J. Surg. Res. 195, 211–218 (2015).

    CAS  PubMed  Google Scholar 

  107. Kimball, A. S., Obi, A. T., Diaz, J. A. & Henke, P. K. The emerging role of NETs in venous thrombosis and immunothrombosis. Front. Immunol. 7, 236 (2016).

    PubMed  PubMed Central  Google Scholar 

  108. Gould, T. J. et al. Neutrophil extracellular traps promote thrombin generation through platelet-dependent and platelet-independent mechanisms. Arterioscler. Thromb. Vasc. Biol. 34, 1977–1984 (2014).

    CAS  PubMed  Google Scholar 

  109. Ammollo, C. T., Semeraro, F., Xu, J., Esmon, N. L. & Esmon, C. T. Extracellular histones increase plasma thrombin generation by impairing thrombomodulin-dependent protein C activation. J. Thromb. Haemost. 9, 1795–1803 (2011).

    CAS  PubMed  Google Scholar 

  110. Demers, M. et al. Cancers predispose neutrophils to release extracellular DNA traps that contribute to cancer-associated thrombosis. Proc. Natl Acad. Sci. USA 109, 13076–13081 (2012).

    CAS  PubMed  Google Scholar 

  111. Demers, M. & Wagner, D. D. Neutrophil extracellular traps: a new link to cancer-associated thrombosis and potential implications for tumor progression. Oncoimmunology 2, e22946 (2013).

    PubMed  PubMed Central  Google Scholar 

  112. Massberg, S. et al. Reciprocal coupling of coagulation and innate immunity via neutrophil serine proteases. Nat. Med. 16, 887–896 (2010).

    CAS  PubMed  Google Scholar 

  113. Semeraro, F. et al. Extracellular histones promote thrombin generation through platelet-dependent mechanisms: involvement of platelet TLR2 and TLR4. Blood 118, 1952–1961 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  114. Alazawi, W., Pirmadjid, N., Lahiri, R. & Bhattacharya, S. Inflammatory and immune responses to surgery and their clinical impact. Ann. Surg. 264, 73–80 (2016).

    PubMed  Google Scholar 

  115. Lord, J. M. et al. The systemic immune response to trauma: an overview of pathophysiology and treatment. Lancet 384, 1455–1465 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  116. Itagaki, K. et al. Mitochondrial DNA released by trauma induces neutrophil extracellular traps. PLoS ONE 10, e0120549 (2015).

    PubMed  PubMed Central  Google Scholar 

  117. Slaba, I. et al. Imaging the dynamic platelet-neutrophil response in sterile liver injury and repair in mice. Hepatology 62, 1593–1605 (2015).

    CAS  PubMed  Google Scholar 

  118. Honda, M. et al. Intravital imaging of neutrophil recruitment reveals the efficacy of FPR1 blockade in hepatic ischemia-reperfusion injury. J. Immunol. 198, 1718–1728 (2017).

    CAS  PubMed  Google Scholar 

  119. Eltzschig, H. K. & Eckle, T. Ischemia and reperfusion — from mechanism to translation. Nat. Med. 17, 1391–1401 (2011).

    CAS  PubMed  Google Scholar 

  120. Zhai, Y., Petrowsky, H., Hong, J. C., Busuttil, R. W. & Kupiec-Weglinski, J. W. Ischaemia-reperfusion injury in liver transplantation — from bench to bedside. Nat. Rev. Gastroenterol. Hepatol. 10, 79–89 (2013).

    CAS  PubMed  Google Scholar 

  121. Oklu, R., Albadawi, H., Jones, J. E., Yoo, H. J. & Watkins, M. T. Reduced hind limb ischemia-reperfusion injury in Toll-like receptor-4 mutant mice is associated with decreased neutrophil extracellular traps. J. Vasc. Surg. 58, 1627–1636 (2013).

    PubMed  Google Scholar 

  122. Albadawi, H. et al. Effect of DNase I treatment and neutrophil depletion on acute limb ischemia-reperfusion injury in mice. J. Vasc. Surg. 64, 484–493 (2016).

    PubMed  Google Scholar 

  123. Savchenko, A. S. et al. VWF-mediated leukocyte recruitment with chromatin decondensation by PAD4 increases myocardial ischemia/reperfusion injury in mice. Blood 123, 141–148 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  124. Ge, L. et al. Neutrophil extracellular traps in ischemia-reperfusion injury-induced myocardial no-reflow: therapeutic potential of DNase-based reperfusion strategy. Am. J. Physiol. Heart Circ. Physiol. 308, H500–509 (2015).

    CAS  PubMed  Google Scholar 

  125. Huang, H. et al. Damage-associated molecular pattern-activated neutrophil extracellular trap exacerbates sterile inflammatory liver injury. Hepatology 62, 600–614 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  126. Al-Khafaji, A. B. et al. Superoxide induces neutrophil extracellular trap formation in a TLR-4 and NOX-dependent mechanism. Mol. Med. 22, 621–631 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  127. Nakazawa, D. et al. Histones and neutrophil extracellular traps enhance tubular necrosis and remote organ injury in ischemic AKI. J. Am. Soc. Nephrol. 28, 1753–1768 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  128. Carter, M. B., Wilson, M. A., Wead, W. B. & Garrison, R. N. Pulmonary subpleural arteriolar diameters during intestinal ischemia/reperfusion. J. Surg. Res. 59, 51–58 (1995).

    CAS  PubMed  Google Scholar 

  129. Gupta, S. & Kaplan, M. J. The role of neutrophils and NETosis in autoimmune and renal diseases. Nat. Rev. Nephrol. 12, 402–413 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  130. Rosenberg, L. et al. Histologic markers of inflammation in patients with ulcerative colitis in clinical remission. Clin. Gastroenterol. Hepatol. 11, 991–996 (2013).

    PubMed  PubMed Central  Google Scholar 

  131. de Souza, H. S. & Fiocchi, C. Immunopathogenesis of IBD: current state of the art. Nat. Rev. Gastroenterol. Hepatol. 13, 13–27 (2016).

    CAS  PubMed  Google Scholar 

  132. Uchiyama, K. et al. Serpin B1 protects colonic epithelial cell via blockage of neutrophil elastase activity and its expression is enhanced in patients with ulcerative colitis. Am. J. Physiol. Gastrointest. Liver Physiol. 302, G1163–G1170 (2012).

    CAS  PubMed  Google Scholar 

  133. Kato, S. et al. Increased expression of long pentraxin PTX3 in inflammatory bowel diseases. Dig. Dis. Sci. 53, 1910–1916 (2008).

    CAS  PubMed  Google Scholar 

  134. Darrah, E. & Andrade, F. NETs: the missing link between cell death and systemic autoimmune diseases? Front. Immunol. 3, 428 (2012).

    PubMed  Google Scholar 

  135. He, Z. et al. Phosphotidylserine exposure and neutrophil extracellular traps enhance procoagulant activity in patients with inflammatory bowel disease. Thromb. Haemost. 115, 738–751 (2016).

    PubMed  Google Scholar 

  136. Zhou, G. et al. CD177+ neutrophils as functionally activated neutrophils negatively regulate IBD. Gut http://dx.doi.org/10.1136/gutjnl-2016-313535 (2017).

  137. Schauer, C. et al. Aggregated neutrophil extracellular traps limit inflammation by degrading cytokines and chemokines. Nat. Med. 20, 511–517 (2014).

    CAS  PubMed  Google Scholar 

  138. Ruemmele, F. M. et al. Diagnostic accuracy of serological assays in pediatric inflammatory bowel disease. Gastroenterology 115, 822–829 (1998).

    CAS  PubMed  Google Scholar 

  139. Zhou, G. et al. ASCA, ANCA, ALCA and many more: are they useful in the diagnosis of inflammatory bowel disease? Dig. Dis. 34, 90–97 (2016).

    PubMed  Google Scholar 

  140. Jarrot, P. A. & Kaplanski, G. Pathogenesis of ANCA-associated vasculitis: an update. Autoimmun. Rev. 15, 704–713 (2016).

    CAS  PubMed  Google Scholar 

  141. Kessenbrock, K. et al. Netting neutrophils in autoimmune small-vessel vasculitis. Nat. Med. 15, 623–625 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  142. Sangaletti, S. et al. Neutrophil extracellular traps mediate transfer of cytoplasmic neutrophil antigens to myeloid dendritic cells toward ANCA induction and associated autoimmunity. Blood 120, 3007–3018 (2012).

    CAS  PubMed  Google Scholar 

  143. Sugi, K. et al. Antineutrophil cytoplasmic antibodies in Japanese patients with inflammatory bowel disease: prevalence and recognition of putative antigens. Am. J. Gastroenterol. 94, 1304–1312 (1999).

    CAS  PubMed  Google Scholar 

  144. Mahler, M. et al. PR3-ANCA: a promising biomarker for ulcerative colitis with extensive disease. Clin. Chim. Acta 424, 267–273 (2013).

    CAS  PubMed  Google Scholar 

  145. Mieli-Vergani, G. & Vergani, D. Autoimmune hepatitis. Nat. Rev. Gastroenterol. Hepatol. 8, 320–329 (2011).

    CAS  PubMed  Google Scholar 

  146. Burlingame, R. W., Rubin, R. L. & Rosenberg, A. M. Autoantibodies to chromatin components in juvenile rheumatoid arthritis. Arthritis Rheum. 36, 836–841 (1993).

    CAS  PubMed  Google Scholar 

  147. Czaja, A. J., Nishioka, M., Morshed, S. A. & Hachiya, T. Patterns of nuclear immunofluorescence and reactivities to recombinant nuclear antigens in autoimmune hepatitis. Gastroenterology 107, 200–207 (1994).

    CAS  PubMed  Google Scholar 

  148. Selmi, C., Bowlus, C. L., Gershwin, M. E. & Coppel, R. L. Primary biliary cirrhosis. Lancet 377, 1600–1609 (2011).

    CAS  PubMed  Google Scholar 

  149. Kaplan, M. M. & Gershwin, M. E. Primary biliary cirrhosis. N. Engl. J. Med. 353, 1261–1273 (2005).

    CAS  PubMed  Google Scholar 

  150. Bambha, K. et al. Incidence, clinical spectrum, and outcomes of primary sclerosing cholangitis in a United States community. Gastroenterology 125, 1364–1369 (2003).

    PubMed  Google Scholar 

  151. Mendes, F. & Lindor, K. D. Primary sclerosing cholangitis: overview and update. Nat. Rev. Gastroenterol. Hepatol. 7, 611–619 (2010).

    PubMed  Google Scholar 

  152. Stinton, L. M. et al. PR3-ANCA: a promising biomarker in primary sclerosing cholangitis (PSC). PLoS ONE 9, e112877 (2014).

    PubMed  PubMed Central  Google Scholar 

  153. Kerkar, N. et al. De-novo autoimmune hepatitis after liver transplantation. Lancet 351, 409–413 (1998).

    CAS  PubMed  Google Scholar 

  154. Kerkar, N. & Yanni, G. 'De novo' and 'recurrent' autoimmune hepatitis after liver transplantation: a comprehensive review. J. Autoimmun. 66, 17–24 (2016).

    PubMed  Google Scholar 

  155. Dubel, L., Farges, O., Johanet, C., Sebagh, M. & Bismuth, H. High incidence of antitissue antibodies in patients experiencing chronic liver allograft rejection. Transplantation 65, 1072–1075 (1998).

    CAS  PubMed  Google Scholar 

  156. Eksteen, B., Afford, S. C., Wigmore, S. J., Holt, A. P. & Adams, D. H. Immune-mediated liver injury. Semin. Liver Dis. 27, 351–366 (2007).

    CAS  PubMed  Google Scholar 

  157. Mano, Y. et al. Preoperative neutrophil-to-lymphocyte ratio is a predictor of survival after hepatectomy for hepatocellular carcinoma: a retrospective analysis. Ann. Surg. 258, 301–305 (2013).

    PubMed  Google Scholar 

  158. Asaoka, T. et al. Prognostic impact of preoperative NLR and CA19-9 in pancreatic cancer. Pancreatology 16, 434–440 (2016).

    CAS  PubMed  Google Scholar 

  159. Malik, H. Z. et al. Preoperative prognostic score for predicting survival after hepatic resection for colorectal liver metastases. Ann. Surg. 246, 806–814 (2007).

    PubMed  Google Scholar 

  160. Fridlender, Z. G. et al. Polarization of tumor-associated neutrophil phenotype by TGF-beta: “N1” versus “N2” TAN. Cancer Cell 16, 183–194 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  161. Fridlender, Z. G. & Albelda, S. M. Tumor-associated neutrophils: friend or foe? Carcinogenesis 33, 949–955 (2012).

    CAS  PubMed  Google Scholar 

  162. Hubert, P. et al. Antibody-dependent cell cytotoxicity synapses form in mice during tumor-specific antibody immunotherapy. Cancer Res. 71, 5134–5143 (2011).

    CAS  PubMed  Google Scholar 

  163. van Gisbergen, K. P., Geijtenbeek, T. B. & van Kooyk, Y. Close encounters of neutrophils and DCs. Trends Immunol. 26, 626–631 (2005).

    CAS  PubMed  Google Scholar 

  164. Beauvillain, C. et al. Neutrophils efficiently cross-prime naive T cells in vivo. Blood 110, 2965–2973 (2007).

    CAS  PubMed  Google Scholar 

  165. Jablonska, J., Leschner, S., Westphal, K., Lienenklaus, S. & Weiss, S. Neutrophils responsive to endogenous IFN-beta regulate tumor angiogenesis and growth in a mouse tumor model. J. Clin. Invest. 120, 1151–1164 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  166. Gabrilovich, D. I. et al. The terminology issue for myeloid-derived suppressor cells. Cancer Res. 67, 425–426. (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  167. Brandau, S., Moses, K. & Lang, S. The kinship of neutrophils and granulocytic myeloid-derived suppressor cells in cancer: cousins, siblings or twins? Semin. Cancer Biol. 23, 171–182 (2013).

    CAS  PubMed  Google Scholar 

  168. Moses, K. & Brandau, S. Human neutrophils: their role in cancer and relation to myeloid-derived suppressor cells. Semin. Immunol. 28, 187–196 (2016).

    CAS  PubMed  Google Scholar 

  169. Toor, S. M. et al. Increased levels of circulating and tumor-infiltrating granulocytic myeloid cells in colorectal cancer patients. Front. Immunol. 7, 560 (2016).

    PubMed  PubMed Central  Google Scholar 

  170. Kalathil, S., Lugade, A. A., Miller, A., Iyer, R. & Thanavala, Y. Higher frequencies of GARP+CTLA-4+Foxp3+ T regulatory cells and myeloid-derived suppressor cells in hepatocellular carcinoma patients are associated with impaired T-cell functionality. Cancer Res. 73, 2435–2444 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  171. Sagiv, J. Y. et al. Phenotypic diversity and plasticity in circulating neutrophil subpopulations in cancer. Cell Rep. 10, 562–573 (2015).

    CAS  PubMed  Google Scholar 

  172. Millrud, C. R. et al. NET-producing CD16high CD62Ldim neutrophils migrate to tumor sites and predict improved survival in patients with HNSCC. Int. J. Cancer 140, 2557–2567 (2017).

    CAS  PubMed  Google Scholar 

  173. Tohme, S. et al. Neutrophil extracellular traps promote the development and progression of liver metastases after surgical stress. Cancer Res. 76, 1367–1380 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  174. Pieterse, E. et al. Neutrophil extracellular traps drive endothelial-to-mesenchymal transition. Arterioscler. Thromb. Vasc. Biol. 37, 1371–1379 (2017).

    CAS  PubMed  Google Scholar 

  175. Najmeh, S. et al. Neutrophil extracellular traps sequester circulating tumor cells via beta1-integrin mediated interactions. Int. J. Cancer 140, 2321–2330 (2017).

    CAS  PubMed  Google Scholar 

  176. Park, J. et al. Cancer cells induce metastasis-supporting neutrophil extracellular DNA traps. Sci. Transl Med. 8, 361ra138 (2016).

    PubMed  PubMed Central  Google Scholar 

  177. Guglietta, S. & Rescigno, M. Hypercoagulation and complement: connected players in tumor development and metastases. Semin. Immunol. 28, 578–586 (2016).

    CAS  PubMed  Google Scholar 

  178. Boone, B. A. et al. The receptor for advanced glycation end products (RAGE) enhances autophagy and neutrophil extracellular traps in pancreatic cancer. Cancer Gene Ther. 22, 326–334 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  179. Yang, C. et al. Procoagulant role of neutrophil extracellular traps in patients with gastric cancer. Int. J. Clin. Exp. Pathol. 8, 14075–14086 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  180. Arelaki, S. et al. Gradient infiltration of neutrophil extracellular traps in colon cancer and evidence for their involvement in tumour growth. PLoS ONE 11, e0154484 (2016).

    PubMed  PubMed Central  Google Scholar 

  181. Guglietta, S. et al. Coagulation induced by C3aR-dependent NETosis drives protumorigenic neutrophils during small intestinal tumorigenesis. Nat. Commun. 7, 11037 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  182. Barkin, J. S. & Goldstein, J. A. Diagnostic and therapeutic approach to pancreatic cancer. Biomed. Pharmacother. 54, 400–409 (2000).

    CAS  PubMed  Google Scholar 

  183. Wen, F., Shen, A., Choi, A., Gerner, E. W. & Shi, J. Extracellular DNA in pancreatic cancer promotes cell invasion and metastasis. Cancer Res. 73, 4256–4266 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  184. Abdol Razak, N., Elaskalani, O. & Metharom, P. Pancreatic cancer-induced neutrophil extracellular traps: a potential contributor to cancer-associated thrombosis. Int. J. Mol. Sci. 18, 487 (2017).

    PubMed Central  Google Scholar 

  185. Hecht, S. S. Tobacco carcinogens, their biomarkers and tobacco-induced cancer. Nat. Rev. Cancer 3, 733–744 (2003).

    CAS  PubMed  Google Scholar 

  186. Hosseinzadeh, A., Thompson, P. R., Segal, B. H. & Urban, C. F. Nicotine induces neutrophil extracellular traps. J. Leukoc. Biol. 100, 1105–1112 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  187. Donnellan, E., Kevane, B., Bird, B. R. & Ainle, F. N. Cancer and venous thromboembolic disease: from molecular mechanisms to clinical management. Curr. Oncol. 21, 134–143 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  188. Blom, J. W., Doggen, C. J., Osanto, S. & Rosendaal, F. R. Malignancies, prothrombotic mutations, and the risk of venous thrombosis. JAMA 293, 715–722 (2005).

    CAS  PubMed  Google Scholar 

  189. Brill, A. et al. Neutrophil extracellular traps promote deep vein thrombosis in mice. J. Thromb. Haemost. 10, 136–144 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  190. von Bruhl, M. L. et al. Monocytes, neutrophils, and platelets cooperate to initiate and propagate venous thrombosis in mice in vivo. J. Exp. Med. 209, 819–835 (2012).

    PubMed  PubMed Central  Google Scholar 

  191. Kambas, K. et al. Tissue factor expression in neutrophil extracellular traps and neutrophil derived microparticles in antineutrophil cytoplasmic antibody associated vasculitis may promote thromboinflammation and the thrombophilic state associated with the disease. Ann. Rheum. Dis. 73, 1854–1863 (2014).

    CAS  PubMed  Google Scholar 

  192. Cedervall, J. et al. Neutrophil extracellular traps accumulate in peripheral blood vessels and compromise organ function in tumor-bearing animals. Cancer Res. 75, 2653–2662 (2015).

    CAS  PubMed  Google Scholar 

  193. Thomson, A. H. Human recombinant DNase in cystic fibrosis. J. R. Soc. Med. 88 (Suppl. 25), 24–29 (1995).

    PubMed  PubMed Central  Google Scholar 

  194. Sayah, D. M. et al. Neutrophil extracellular traps are pathogenic in primary graft dysfunction after lung transplantation. Am. J. Respir. Crit. Care Med. 191, 455–463 (2015).

    PubMed  PubMed Central  Google Scholar 

  195. Macanovic, M. et al. The treatment of systemic lupus erythematosus (SLE) in NZB/W F1 hybrid mice; studies with recombinant murine DNase and with dexamethasone. Clin. Exp. Immunol. 106, 243–252 (1996).

    CAS  PubMed  PubMed Central  Google Scholar 

  196. Davis, J. C. Jr et al. Recombinant human Dnase I (rhDNase) in patients with lupus nephritis. Lupus 8, 68–76 (1999).

    PubMed  Google Scholar 

  197. Neeli, I., Dwivedi, N., Khan, S. & Radic, M. Regulation of extracellular chromatin release from neutrophils. J. Innate Immun. 1, 194–201 (2009).

    CAS  PubMed  Google Scholar 

  198. Bjornsdottir, H. et al. Neutrophil NET formation is regulated from the inside by myeloperoxidase-processed reactive oxygen species. Free Radic. Biol. Med. 89, 1024–1035 (2015).

    PubMed  Google Scholar 

  199. Campbell, A. M., Kashgarian, M. & Shlomchik, M. J. NADPH oxidase inhibits the pathogenesis of systemic lupus erythematosus. Sci. Transl Med. 4, 157ra141 (2012).

    PubMed  PubMed Central  Google Scholar 

  200. Maicas, N. et al. Deficiency of Nrf2 accelerates the effector phase of arthritis and aggravates joint disease. Antioxid. Redox Signal 15, 889–901 (2011).

    CAS  PubMed  Google Scholar 

  201. Winkelstein, J. A. et al. Chronic granulomatous disease. Report on a national registry of 368 patients. Medicine 79, 155–169 (2000).

    CAS  PubMed  Google Scholar 

  202. Lai, Z. W. et al. N-Acetylcysteine reduces disease activity by blocking mammalian target of rapamycin in T cells from systemic lupus erythematosus patients: a randomized, double-blind, placebo-controlled trial. Arthritis Rheum. 64, 2937–2946 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  203. Stravitz, R. T. et al. Effects of N-acetylcysteine on cytokines in non-acetaminophen acute liver failure: potential mechanism of improvement in transplant-free survival. Liver Int. 33, 1324–1331 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  204. D'Amico, F. et al. Use of N-acetylcysteine during liver procurement: a prospective randomized controlled study. Liver Transpl. 19, 135–144 (2013).

    PubMed  Google Scholar 

  205. Orban, J. C. et al. Effect of N-acetylcysteine pretreatment of deceased organ donors on renal allograft function: a randomized controlled trial. Transplantation 99, 746–753 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  206. Khandpur, R. et al. NETs are a source of citrullinated autoantigens and stimulate inflammatory responses in rheumatoid arthritis. Sci. Transl Med. 5, 178ra140 (2013).

    Google Scholar 

  207. Taylor, P. C. & Feldmann, M. Anti-TNF biologic agents: still the therapy of choice for rheumatoid arthritis. Nat. Rev. Rheumatol 5, 578–582 (2009).

    CAS  PubMed  Google Scholar 

  208. Kunwar, S., Dahal, K. & Sharma, S. Anti-IL-17 therapy in treatment of rheumatoid arthritis: a systematic literature review and meta-analysis of randomized controlled trials. Rheumatol Int. 36, 1065–1075 (2016).

    CAS  PubMed  Google Scholar 

  209. Danese, S., Vuitton, L. & Peyrin-Biroulet, L. Biologic agents for IBD: practical insights. Nat. Rev. Gastroenterol. Hepatol. 12, 537–545 (2015).

    CAS  PubMed  Google Scholar 

  210. Wang, Y. et al. Amelioration of lupus-like autoimmune disease in NZB/WF1 mice after treatment with a blocking monoclonal antibody specific for complement component C5. Proc. Natl Acad. Sci. USA 93, 8563–8568 (1996).

    CAS  PubMed  Google Scholar 

  211. Jayne, D. R. et al. Randomized Trial of C5a Receptor Inhibitor Avacopan in ANCA-Associated Vasculitis. J. Am. Soc. Nephrol. 28, 2756–2767 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  212. Meier-Kriesche, H. U. et al. Immunosuppression: evolution in practice and trends, 1994–2004. Am. J. Transplant. 6, 1111–1131 (2006).

    CAS  PubMed  Google Scholar 

  213. Zavada, J. et al. Cyclosporine A or intravenous cyclophosphamide for lupus nephritis: the Cyclofa-Lune study. Lupus 19, 1281–1289 (2010).

    CAS  PubMed  Google Scholar 

  214. Lee, Y. H., Lee, H. S., Choi, S. J., Dai Ji, J. & Song, G. G. Efficacy and safety of tacrolimus therapy for lupus nephritis: a systematic review of clinical trials. Lupus 20, 636–640 (2011).

    CAS  PubMed  Google Scholar 

  215. Zheng, W. et al. PF-1355, a mechanism-based myeloperoxidase inhibitor, prevents immune complex vasculitis and anti-glomerular basement membrane glomerulonephritis. J. Pharmacol. Exp. Ther. 353, 288–298 (2015).

    CAS  PubMed  Google Scholar 

  216. Knight, J. S. et al. Peptidylarginine deiminase inhibition is immunomodulatory and vasculoprotective in murine lupus. J. Clin. Invest. 123, 2981–2993 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  217. Tsuda, Y. et al. Three different neutrophil subsets exhibited in mice with different susceptibilities to infection by methicillin-resistant Staphylococcus aureus. Immunity 21, 215–226 (2004).

    CAS  PubMed  Google Scholar 

  218. Denny, M. F. et al. A distinct subset of proinflammatory neutrophils isolated from patients with systemic lupus erythematosus induces vascular damage and synthesizes type I IFNs. J. Immunol. 184, 3284–3297 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  219. Clemmensen, S. N. et al. Olfactomedin 4 defines a subset of human neutrophils. J. Leukoc. Biol. 91, 495–500 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  220. Welin, A. 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 8, e69575 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  221. Christoffersson, G. et al. VEGF-A recruits a proangiogenic MMP-9-delivering neutrophil subset that induces angiogenesis in transplanted hypoxic tissue. Blood 120, 4653–4662 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  222. Zhang, D. et al. Neutrophil ageing is regulated by the microbiome. Nature 525, 528–532 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

The authors thank Servier for providing Servier Medical Art, which was used for creation of original figures. The work in the authors' laboratories is supported by grants from the Canadian Institutes of Health Research, Alberta Innovates Health Solutions, the Heart and Stroke Foundation of Canada and the Canada Research Chairs programme.

Author information

Authors and Affiliations

Authors

Contributions

M.H. contributed to all aspects of this manuscript. P.K. provided substantial contributions to the discussion of content and reviewed and edited the manuscript before submission.

Corresponding author

Correspondence to Paul Kubes.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

PowerPoint slides

Glossary

Chromatin

A structure in the eukaryotic cell nucleus that contains primarily DNA and nuclear proteins such as histones.

Necrosis

Unprogrammed death of cells and living tissue induced by external or internal factors.

Neutrophil elastase

A serine protease secreted by neutrophils during inflammation.

Myeloperoxidase

The most abundant protein of neutrophils; catalyses the conversion of H2O2 and chloride into hypochlorous acid.

NADPH oxidase complex

A membrane-bound enzyme complex that produces reactive oxygen species when activated.

Hypercitrullination

The hyperactive conversion of arginine to citrulline.

Chemotax

The movement of organisms responding to chemical stimuli.

Phagocytose

The process by which phagocytes ingest or engulf other cells or particles.

Mitophagy

The specific process for the autophagic elimination of damaged mitochondria.

Transfection

The introduction of a segment of DNA or RNA into a eukaryotic cell by use of various physical or chemical methods or through viral infection.

Kupffer cells

Self-sustaining, liver-resident macrophages found in the liver sinusoids.

Liver sinusoids

Sinusoidal blood vessels that are lined with endothelial cells and that receive blood from terminal branches of the hepatic artery and portal vein and deliver it into central veins.

Complement receptor of immunoglobulin superfamily

A macrophage complement receptor that recognizes the activated form of complement C3, which opsonizes pathogens, apoptotic cells and foreign antigens.

von Willebrand factor

A large multimeric glycoprotein circulating in blood plasma that binds coagulation factor VIII and platelets and that mediates platelet adhesion to collagen at sites of vascular injury.

Damage-associated molecular patterns

Host biomolecules released by cellular injury that act as endogenous danger signals to activate the inflammatory response.

Cytokine storm

Uncontrolled excessive cytokine release that leads to detrimental effects, including leakage from capillaries, tissue oedema, organ failure and shock.

Anti-neutrophil cytoplasmic autoantibodies

Autoantibodies that bind to enzymes from neutrophil cytoplasmic granules and are biomarkers for a number of autoimmune diseases.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Honda, M., Kubes, P. Neutrophils and neutrophil extracellular traps in the liver and gastrointestinal system. Nat Rev Gastroenterol Hepatol 15, 206–221 (2018). https://doi.org/10.1038/nrgastro.2017.183

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nrgastro.2017.183

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing