Submitted Successfully!
To reward your contribution, here is a gift for you: A free trial for our video production service.
Thank you for your contribution! You can also upload a video entry or images related to this topic.
Version Summary Created by Modification Content Size Created at Operation
1 -- 966 2023-12-06 16:29:35 |
2 format Meta information modification 966 2023-12-07 02:35:56 |

Video Upload Options

Do you have a full video?

Confirm

Are you sure to Delete?
Cite
If you have any further questions, please contact Encyclopedia Editorial Office.
Kumar, R.; Patil, G.; Dayal, S. NETosis in Disease Condition. Encyclopedia. Available online: https://encyclopedia.pub/entry/52455 (accessed on 02 May 2024).
Kumar R, Patil G, Dayal S. NETosis in Disease Condition. Encyclopedia. Available at: https://encyclopedia.pub/entry/52455. Accessed May 02, 2024.
Kumar, Rahul, Gokul Patil, Sanjana Dayal. "NETosis in Disease Condition" Encyclopedia, https://encyclopedia.pub/entry/52455 (accessed May 02, 2024).
Kumar, R., Patil, G., & Dayal, S. (2023, December 06). NETosis in Disease Condition. In Encyclopedia. https://encyclopedia.pub/entry/52455
Kumar, Rahul, et al. "NETosis in Disease Condition." Encyclopedia. Web. 06 December, 2023.
NETosis in Disease Condition
Edit

Ischemic thrombotic disease, characterized by the formation of obstructive blood clots within arteries or veins, is a condition associated with life-threatening events, such as stroke, myocardial infarction, deep vein thrombosis, and pulmonary embolism. The conventional therapeutic strategy relies on treatments with anticoagulants that unfortunately pose an inherent risk of bleeding complications. These anticoagulants primarily target clotting factors, often overlooking upstream events, including the release of neutrophil extracellular traps (NETs). Neutrophils are integral components of the innate immune system, traditionally known for their role in combating pathogens through NET formation. Emerging evidence has now revealed that NETs contribute to a prothrombotic milieu by promoting platelet activation, increasing thrombin generation, and providing a scaffold for clot formation.

thrombosis NLRP3 inflammasome NETs

1. Introduction

In recent years, NETosis has emerged as a critical player in the context of aging and age-associated diseases [1]. Neutrophils from aged individuals or aged mice exhibit altered NETosis kinetics, with some reports suggesting an enhanced NET formation with age [2][3], or in aged individuals with severe vasculitis [4][5], while others indicate minimal or no change in NETosis in otherwise healthy aged mice [6]. The age-related changes in NETosis may contribute to chronic inflammation, a hallmark of aging, and may also impact the immune response to infections in the elderly. Therefore, understanding the mechanisms underlying the age-related alterations in NETosis is crucial for unraveling the complexities of aging.

2. NETosis in Acute Ischemic Conditions

Elevated plasma levels of extracellular histones have been reported in ischemic or thrombotic conditions, such as myocardial infarction [7], stroke [8][9], ischemic outcomes after angioplasty [10], and DVT [11][12]. Stakos et al. observed that neutrophils isolated from IRA aspiration in STEMI patients were more prone to the release of NETs in comparison to non-infarct-related coronary arteries and control individuals [13]. NETs were found to be constitutively present in the thrombi retrieved during endovascular therapy in patients with acute ischemic stroke [14]. However, the authors could not find any significant association between the circulating markers of NETs and the final thrombolysis in cerebral infarction (TICI) score [14]. Borissoff et al. reported that the circulating markers of NETosis were independently associated with the severity of coronary atherosclerosis, the occurrence of major adverse cardiac events, and the presence of a prothrombotic state [15]. Surprisingly, a deficiency of PAD-4 in hematopoietic cells did not display a significant impact on the progression of atheromatous plaques in hypercholesterolemic mice [16], despite detecting the presence of NETs in atherosclerotic lesions from human carotid endarterectomy tissue [16]. This may indicate that the mechanisms may differ between mice and humans. An alternative explanation could be that the process of inducing disease conditions in mice may not necessarily phenocopy human disease pathology. Retinal vein occlusion (RVO), which refers to the obstruction of the retinal venous system due to thrombus formation, has emerged as the second most common retinal vascular disorders [17][18]. Recently, Wan et al. reported that the elevated plasma levels of NETs are associated with RVO and can be exploited as a potential biomarker [19]. In summary, NETs have been identified in several ischemic conditions but their mechanistic contributions to pathological ischemia is not fully understood in most of the disease states.

3. NETosis in Cancer-Associated Thrombosis

Several tumors and cancer cells secrete a cytokine called the granulocyte colony-stimulating factor (G-CSF) [20][21][22][23][24], which exhibits the potential to induce NETosis and promote thrombosis [25]. Higher levels of circulating NET markers predicted the occurrence of DVT in cancer patients and was associated with a poor prognosis in a large cohort in the Vienna Cancer and Thrombosis Study (CATS) [26]. This was an important study since it included patients with malignancy at varied sites, such as brain, breast, bronchus, stomach, prostate etc. [26]. The elevated circulating levels of MPO-DNA was also associated with thrombosis in patients with myeloproliferative neoplasms (MPN) [27]. Similarly, histone–DNA complexes have been demonstrated in the thrombi in patients with different cancer types [28][29] and circulating H3Cit has been associated with the markers of thrombosis [28].
There was also in vivo evidence for NETs promoting venous thrombosis in mouse cancer models. Demers et al. [25], using murine models of lung and breast carcinoma and chronic myelogenous leukemia, demonstrated an increased sensitivity of neutrophils undergoing NETosis. Hisada et al. observed elevations of H3Cit and cfDNA in the plasma and thrombi from mice bearing human pancreatic tumors, and the administration of DNase 1 or the depletion of neutrophils reduced the thrombus size in mice bearing human tumors [30]. Similar observations have been made by others in mammary cancer [31][32][33]. Wolach et al. further extended the findings on the role of NETs in animal models of cancer-associated thrombosis to MPN represented by the Jak2V617F mutation [34]. A Jak2WT mouse transplanted with Jak2V617F bone marrow had elevated NETs compared to the Jak2WT mice and developed spontaneous pulmonary thrombosis, which was absent when the mice were engrafted with PAD-4-deficient Jak2V617F bone marrow. The Jak2V617F mice treated with DNase 1 or the JAK inhibitor Ruxolitinib had a reduced thrombus size when subjected to the IVC stenosis model for DVT. Taken together, these findings implicated a mechanistic role of NETs in cancer-associated ischemic thrombosis.

4. NETosis and Age in the Context of COVID-19

NETosis is by far the major underlying pathway observed in COVID-19-associated thrombosis [35][36][37][38][39][40]. Age may exert a profound influence on the intricate dynamics of the immune response during COVID-19 [41][42]. Understanding this relationship is vital because it offers valuable insights into why older individuals often experience more severe forms of COVID-19. As individuals age, their immune systems undergo several changes, including alterations in neutrophil function and a predisposition to chronic inflammation, creating an environment that may prime the immune response for heightened NETosis [43]. These age-related changes can result in a more robust NETosis response when confronted with viral infections like SARS-CoV-2. Consequently, the amplification of NETosis in older individuals can contribute to increased inflammation, tissue damage, and a more severe clinical course of COVID-19 [44].

References

  1. Sabbatini, M.; Bona, E.; Novello, G.; Migliario, M.; Renò, F. Aging hampers neutrophil extracellular traps (NETs) efficacy. Aging Clin. Exp. Res. 2022, 34, 2345–2353.
  2. Martinod, K.; Witsch, T.; Erpenbeck, L.; Savchenko, A.; Hayashi, H.; Cherpokova, D.; Gallant, M.; Mauler, M.; Cifuni, S.M.; Wagner, D.D. Peptidylarginine deiminase 4 promotes age-related organ fibrosis. J. Exp. Med. 2017, 214, 439–458.
  3. Ortmann, W.; Kolaczkowska, E. Age is the work of art? Impact of neutrophil and organism age on neutrophil extracellular trap formation. Cell Tissue Res. 2018, 371, 473–488.
  4. Matsuda, Y.; Itabashi, M.; Tachibana, Y.; Sugihara, T.; Sakashita, Y.; Matsubara, T.; Murayama, S.; Yumura, W.; Shimizu, A.; Takei, T.; et al. Citrullinated histone H3 expression in anti-neutrophil cytoplasmic antibody-associated vasculitis in older Japanese autopsy patients. Geriatr. Gerontol. Int. 2019, 19, 259–264.
  5. Matsuda, Y.; Hamayasu, H.; Seki, A.; Nonaka, K.; Wang, T.; Matsumoto, T.; Hamano, Y.; Sumikura, H.; Kumasaka, T.; Murayama, S.; et al. Presence of Citrullinated Histone H3-Positive Neutrophils in Microscopic Polyangiitis from the Early Phase: An Autopsy Proven Case. Pathol. Int. 2016, 66, 466–471.
  6. Kumar, R.; Sonkar, V.K.; Swamy, J.; Ahmed, A.; Sharathkumar, A.A.; Pierce, G.L.; Dayal, S. DNase 1 Protects From Increased Thrombin Generation and Venous Thrombosis During Aging: Cross-Sectional Study in Mice and Humans. J. Am. Heart Assoc. 2022, 11, e021188.
  7. Shah, M.; He, Z.; Rauf, A.; Beikoghli Kalkhoran, S.; Heiestad, C.M.; Stenslokken, K.O.; Parish, C.R.; Soehnlein, O.; Arjun, S.; Davidson, S.M.; et al. Extracellular histones are a target in myocardial ischaemia-reperfusion injury. Cardiovasc. Res. 2022, 118, 1115–1125.
  8. De Meyer, S.F.; Suidan, G.L.; Fuchs, T.A.; Monestier, M.; Wagner, D.D. Extracellular chromatin is an important mediator of ischemic stroke in mice. Arter. Thromb. Vasc. Biol. 2012, 32, 1884–1891.
  9. Valles, J.; Lago, A.; Santos, M.T.; Latorre, A.M.; Tembl, J.I.; Salom, J.B.; Nieves, C.; Moscardo, A. Neutrophil extracellular traps are increased in patients with acute ischemic stroke: Prognostic significance. Thromb. Haemost. 2017, 117, 1919–1929.
  10. Demyanets, S.; Stojkovic, S.; Mauracher, L.M.; Kopp, C.W.; Wojta, J.; Thaler, J.; Panzer, S.; Gremmel, T. Surrogate Markers of Neutrophil Extracellular Trap Formation are Associated with Ischemic Outcomes and Platelet Activation after Peripheral Angioplasty and Stenting. J. Clin. Med. 2020, 9, 304.
  11. Brill, A.; Fuchs, T.A.; Savchenko, A.S.; Thomas, G.M.; Martinod, K.; De Meyer, S.F.; Bhandari, A.A.; Wagner, D.D. Neutrophil extracellular traps promote deep vein thrombosis in mice. J. Thromb. Haemost. 2012, 10, 136–144.
  12. Fuchs, T.A.; Brill, A.; Wagner, D.D. Neutrophil extracellular trap (NET) impact on deep vein thrombosis. Arter. Thromb. Vasc. Biol. 2012, 32, 1777–1783.
  13. Stakos, D.A.; Kambas, K.; Konstantinidis, T.; Mitroulis, I.; Apostolidou, E.; Arelaki, S.; Tsironidou, V.; Giatromanolaki, A.; Skendros, P.; Konstantinides, S.; et al. Expression of functional tissue factor by neutrophil extracellular traps in culprit artery of acute myocardial infarction. Eur. Heart J. 2015, 36, 1405–1414.
  14. Ducroux, C.; Di Meglio, L.; Loyau, S.; Delbosc, S.; Boisseau, W.; Deschildre, C.; Ben Maacha, M.; Blanc, R.; Redjem, H.; Ciccio, G.; et al. Thrombus Neutrophil Extracellular Traps Content Impair tPA-Induced Thrombolysis in Acute Ischemic Stroke. Stroke 2018, 49, 754–757.
  15. Borissoff, J.I.; Joosen, I.A.; Versteylen, M.O.; Brill, A.; Fuchs, T.A.; Savchenko, A.S.; Gallant, M.; Martinod, K.; Ten Cate, H.; Hofstra, L.; et al. Elevated levels of circulating DNA and chromatin are independently associated with severe coronary atherosclerosis and a prothrombotic state. Arter. Thromb. Vasc. Biol. 2013, 33, 2032–2040.
  16. Franck, G.; Mawson, T.L.; Folco, E.J.; Molinaro, R.; Ruvkun, V.; Engelbertsen, D.; Liu, X.; Tesmenitsky, Y.; Shvartz, E.; Sukhova, G.K.; et al. Roles of PAD4 and NETosis in Experimental Atherosclerosis and Arterial Injury: Implications for Superficial Erosion. Circ. Res. 2018, 123, 33–42.
  17. Wan, W.; Liu, H.; Long, Y.; Wan, W.; Li, Q.; Zhu, W.; Wu, Y. The association between circulating neutrophil extracellular trap related biomarkers and retinal vein occlusion incidence: A case-control pilot study. Exp. Eye Res. 2021, 210, 108702.
  18. Fukui, Y.; Kawashima, M.; Kawaguchi, K.; Takeuchi, M.; Hirata, M.; Kataoka, T.R.; Sakurai, T.; Kataoka, M.; Kanao, S.; Nakamoto, Y.; et al. Granulocyte-colony-stimulating factor-producing metaplastic carcinoma of the breast with significant elevation of serum interleukin-17 and vascular endothelial growth factor levels. Int. Cancer Conf. J. 2018, 7, 107–113.
  19. Yang, X.; Liu, F.; Xu, Z.; Chen, C.; Wu, X.; Li, G.; Li, J. Expression of granulocyte colony stimulating factor receptor in human colorectal cancer. Postgrad. Med. J. 2005, 81, 333–337.
  20. Uematsu, T.; Tsuchie, K.; Ukai, K.; Kimoto, E.; Funakawa, T.; Mizuno, R. Granulocyte-colony stimulating factor produced by pancreatic carcinoma. Int. J. Pancreatol. 1996, 19, 135–139.
  21. Kowanetz, M.; Wu, X.; Lee, J.; Tan, M.; Hagenbeek, T.; Qu, X.; Yu, L.; Ross, J.; Korsisaari, N.; Cao, T.; et al. Granulocyte-colony stimulating factor promotes lung metastasis through mobilization of Ly6G+Ly6C+ granulocytes. Proc. Natl. Acad. Sci. USA 2010, 107, 21248–21255.
  22. Jiang, X.; Lopez, A.; Holyoake, T.; Eaves, A.; Eaves, C. Autocrine production and action of IL-3 and granulocyte colony-stimulating factor in chronic myeloid leukemia. Proc. Natl. Acad. Sci. USA 1999, 96, 12804–12809.
  23. Demers, M.; Krause, D.S.; Schatzberg, D.; Martinod, K.; Voorhees, J.R.; Fuchs, T.A.; Scadden, D.T.; Wagner, D.D. Cancers predispose neutrophils to release extracellular DNA traps that contribute to cancer-associated thrombosis. Proc. Natl. Acad. Sci. USA 2012, 109, 13076–13081.
  24. Mauracher, L.M.; Posch, F.; Martinod, K.; Grilz, E.; Daullary, T.; Hell, L.; Brostjan, C.; Zielinski, C.; Ay, C.; Wagner, D.D.; et al. Citrullinated histone H3, a biomarker of neutrophil extracellular trap formation, predicts the risk of venous thromboembolism in cancer patients. J. Thromb. Haemost. 2018, 16, 508–518.
  25. Guy, A.; Favre, S.; Labrouche-Colomer, S.; Deloison, L.; Gourdou-Latyszenok, V.; Renault, M.A.; Riviere, E.; James, C. High circulating levels of MPO-DNA are associated with thrombosis in patients with MPN. Leukemia 2019, 33, 2544–2548.
  26. Thalin, C.; Demers, M.; Blomgren, B.; Wong, S.L.; von Arbin, M.; von Heijne, A.; Laska, A.C.; Wallen, H.; Wagner, D.D.; Aspberg, S. NETosis promotes cancer-associated arterial microthrombosis presenting as ischemic stroke with troponin elevation. Thromb. Res. 2016, 139, 56–64.
  27. Oklu, R.; Sheth, R.A.; Wong, K.H.K.; Jahromi, A.H.; Albadawi, H. Neutrophil extracellular traps are increased in cancer patients but does not associate with venous thrombosis. Cardiovasc. Diagn. Ther. 2017, 7, S140–S149.
  28. Kumar, R.; Katare, P.B.; Lentz, S.R.; Modi, A.J.; Sharathkumar, A.A.; Dayal, S. Thrombotic potential during pediatric acute lymphoblastic leukemia induction: Role of cell-free DNA. Res. Pr. Thromb. Haemost. 2021, 5, e12557.
  29. Hisada, Y.; Grover, S.P.; Maqsood, A.; Houston, R.; Ay, C.; Noubouossie, D.F.; Cooley, B.C.; Wallen, H.; Key, N.S.; Thalin, C.; et al. Neutrophils and neutrophil extracellular traps enhance venous thrombosis in mice bearing human pancreatic tumors. Haematologica 2019, 105, 218–225.
  30. Varady, C.B.S.; Oliveira, A.C.; Monteiro, R.Q.; Gomes, T. Recombinant human DNase I for the treatment of cancer-associated thrombosis: A pre-clinical study. Thromb. Res. 2021, 203, 131–137.
  31. Gomes, T.; Varady, C.B.S.; Lourenco, A.L.; Mizurini, D.M.; Rondon, A.M.R.; Leal, A.C.; Goncalves, B.S.; Bou-Habib, D.C.; Medei, E.; Monteiro, R.Q. IL-1beta Blockade Attenuates Thrombosis in a Neutrophil Extracellular Trap-Dependent Breast Cancer Model. Front. Immunol. 2019, 10, 2088.
  32. Wolach, O.; Sellar, R.S.; Martinod, K.; Cherpokova, D.; McConkey, M.; Chappell, R.J.; Silver, A.J.; Adams, D.; Castellano, C.A.; Schneider, R.K.; et al. Increased neutrophil extracellular trap formation promotes thrombosis in myeloproliferative neoplasms. Sci. Transl. Med. 2018, 10, eaan8292.
  33. Zuo, Y.; Estes, S.K.; Ali, R.A.; Gandhi, A.A.; Yalavarthi, S.; Shi, H.; Sule, G.; Gockman, K.; Madison, J.A.; Zuo, M.; et al. Prothrombotic autoantibodies in serum from patients hospitalized with COVID-19. Sci. Transl. Med. 2020, 12, eabd3876.
  34. Zuo, Y.; Yalavarthi, S.; Shi, H.; Gockman, K.; Zuo, M.; Madison, J.A.; Blair, C.; Weber, A.; Barnes, B.J.; Egeblad, M.; et al. Neutrophil extracellular traps in COVID-19. JCI Insight 2020, 5, e138999.
  35. Skendros, P.; Mitsios, A.; Chrysanthopoulou, A.; Mastellos, D.C.; Metallidis, S.; Rafailidis, P.; Ntinopoulou, M.; Sertaridou, E.; Tsironidou, V.; Tsigalou, C.; et al. Complement and tissue factor-enriched neutrophil extracellular traps are key drivers in COVID-19 immunothrombosis. J. Clin. Invest. 2020, 130, 6151–6157.
  36. Zuo, Y.; Zuo, M.; Yalavarthi, S.; Gockman, K.; Madison, J.A.; Shi, H.; Woodard, W.; Lezak, S.P.; Lugogo, N.L.; Knight, J.S.; et al. Neutrophil extracellular traps and thrombosis in COVID-19. J. Thromb. Thrombolysis 2021, 51, 446–453.
  37. Middleton, E.A.; He, X.Y.; Denorme, F.; Campbell, R.A.; Ng, D.; Salvatore, S.P.; Mostyka, M.; Baxter-Stoltzfus, A.; Borczuk, A.C.; Loda, M.; et al. Neutrophil extracellular traps contribute to immunothrombosis in COVID-19 acute respiratory distress syndrome. Blood 2020, 136, 1169–1179.
  38. Ng, H.; Havervall, S.; Rosell, A.; Aguilera, K.; Parv, K.; von Meijenfeldt, F.A.; Lisman, T.; Mackman, N.; Thalin, C.; Phillipson, M. Circulating Markers of Neutrophil Extracellular Traps Are of Prognostic Value in Patients With COVID-19. Arter. Thromb. Vasc. Biol. 2021, 41, 988–994.
  39. Janiuk, K.; Jabłońska, E.; Garley, M. Significance of NETs Formation in COVID-19. Cells 2021, 10, 151.
  40. Yaqinuddin, A.; Kvietys, P.; Kashir, J. COVID-19: Role of neutrophil extracellular traps in acute lung injury. Respir. Investig. 2020, 58, 419–420.
  41. Li, X.; Li, C.; Zhang, W.; Wang, Y.; Qian, P.; Huang, H. Inflammation and aging: Signaling pathways and intervention therapies. Signal Transduct. Target. Ther. 2023, 8, 239.
  42. Zhu, Y.; Chen, X.; Liu, X. NETosis and Neutrophil Extracellular Traps in COVID-19: Immunothrombosis and Beyond. Front. Immunol. 2022, 13, 838011.
  43. Hanna, E.B.; Rossen, J.; Eustes, A.S.; Dayal, S. Heavy lone coronary artery thrombosis treated by stent retriever, in the setting of COVID-19 infection. Catheter. Cardiovasc. Interv. 2022, 99, 457–461.
  44. Schurink, B.; Roos, E.; Radonic, T.; Barbe, E.; Bouman, C.S.C.; de Boer, H.H.; de Bree, G.J.; Bulle, E.B.; Aronica, E.M.; Florquin, S.; et al. Viral presence and immunopathology in patients with lethal COVID-19: A prospective autopsy cohort study. Lancet Microbe 2020, 1, e290–e299.
More
Information
Contributors MDPI registered users' name will be linked to their SciProfiles pages. To register with us, please refer to https://encyclopedia.pub/register : , ,
View Times: 140
Entry Collection: COVID-19
Revisions: 2 times (View History)
Update Date: 07 Dec 2023
1000/1000