Neutrophil extracellular traps and thrombosis in COVID-19 ========================================================= * Yu Zuo * Melanie Zuo * Srilakshmi Yalavarthi * Kelsey Gockman * Jacqueline A. Madison * Hui Shi * Jason S. Knight * Yogendra Kanthi ## ABSTRACT Here, we report on four patients whose hospitalizations for COVID-19 were complicated by venous thromboembolism (**VTE**). All demonstrated high levels of D-dimer as well as high neutrophil-to-lymphocyte ratios. For three patients, we were able to test sera for neutrophil extracellular trap (**NET**) remnants and found significantly elevated levels of cell-free DNA, myeloperoxidase-DNA complexes, and citrullinated histone H3. Neutrophil-derived S100A8/A9 (calprotectin) was also elevated. Given strong links between hyperactive neutrophils, NET release, and thrombosis in many inflammatory diseases, the potential relationship between NETs and VTE should be further investigated in COVID-19. ## INTRODUCTION Severe acute respiratory syndrome coronavirus 2 (**SARS-CoV-2**) causes the disease known as coronavirus disease 2019 (**COVID-19**). It most commonly presents with influenza-like illness and viral pneumonia, but in its most severe manifestation progresses to acute respiratory distress syndrome (**ARDS**) and multi-organ failure1. To date the viral pandemic has resulted in more than two million infections worldwide ([https://coronavirus.jhu.edu/map.html](https://coronavirus.jhu.edu/map.html)). In COVID-19, elevated levels of blood neutrophils predict severe respiratory disease and unfavorable outcomes2, 3. Neutrophil-derived neutrophil extracellular traps (**NETs**) play a pathogenic role in many thrombo-inflammatory states including sepsis4, 5, thrombosis6–8, and respiratory failure9, 10. NETs are extracellular webs of chromatin and microbicidal proteins that are an evolutionarily conserved aspect of innate immune host-defense11; however, NETs also have potential to initiate and propagate inflammation and thrombosis12, 13. NETs deliver a variety of oxidant enzymes to the extracellular space, including myeloperoxidase, NADPH oxidase, and nitric oxide synthase14, while also serving as a source of extracellular histones that carry significant cytotoxic potential15, 16. NETs are drivers of cardiovascular disease by propagating inflammation in vessel walls17. Furthermore, when formed intravascularly, NETs can occlude arteries18, veins19, and microscopic vessels20. Early studies of COVID-19 suggest a high risk of morbid arterial events21, and the risk of venous thromboembolism (**VTE**) is increasingly revealing itself as more data become available22. Descriptive and mechanistic studies to date that examine COVID-19 pathophysiology have focused on monocytes and lymphocytes more so than neutrophils and their effector products— including NETs. Here, we describe four cases of VTE in patients hospitalized with COVID-19 and provide evidence for neutrophil hyperactivity. ## RESULTS We identified four patients admitted to a large academic medical center with COVID-19 who also developed VTE (either deep vein thrombosis or pulmonary embolism) despite immediate initiation of prophylactic-dose heparin. Three of the patients were diagnosed with VTE within 48 hours of admission. All three had markedly elevated D-dimers and neutrophil-to-lymphocyte ratios (**Table 1**). For two of the early VTE patients, we were able to access sera for measurement of NET remnants. Three different NET-associated markers (cell-free DNA, myeloperoxidase-DNA complexes, and citrullinated-histone H3) were elevated in both patients (**Table 1**). We also detected elevated levels of the classic marker of neutrophil activation, S100A8/A9 (also known as calprotectin). Neither patient had positive testing for antiphospholipid antibodies23. View this table: [Table 1:](http://medrxiv.org/content/early/2020/05/05/2020.04.30.20086736/T1) Table 1: Characteristics of three patients diagnosed with VTE soon after admission We identified a fourth patient who developed VTE several weeks into his hospitalization. This 66-year-old man’s course was complicated by respiratory failure requiring mechanical ventilation beginning on day 6. He received prophylactic-dose heparin throughout his hospitalization, but was nevertheless diagnosed on day 20 with extensive right-lower-extremity deep vein thrombosis. We were able to test serum from day 8 of his hospitalization for NET remnants (**Table 2**). Interestingly, markers of neutrophil activation including NET remnants were already significantly elevated on day 8 (**Table 2**); this is in contrast to D-dimer and neutrophil count, which were only mildly elevated. Both D-dimer and neutrophil count were elevated by the time the patient was diagnosed with deep vein thrombosis on day 20 (**Table 2**). The patient did not have elevated levels of antiphospholipid antibodies23. View this table: [Table 2:](http://medrxiv.org/content/early/2020/05/05/2020.04.30.20086736/T2) Table 2: Characteristics of a fourth patient diagnosed with VTE during hospitalization ## DISCUSSION Hyperactivity of the coagulation system is a common finding of severe COVID-1924. Indeed, many patients have a profile to suggest a prothrombotic diathesis including high levels of fibrin degradation products (D-dimer), elevated fibrinogen levels, and low antithrombin levels24, 25. Here, we report four cases of COVID-19-associated VTE. We measured three markers commonly used to detect NET remnants in blood (cell-free DNA, myeloperoxidase-DNA complexes, and citrullinated-histone H3), as well as a fourth marker, S100A8/A9 (calprotectin), which is commonly used to track neutrophil activation. All tests were elevated in patients diagnosed with VTE, including as early as 12 days prior to detection of VTE in one case. Given the known link between NETs and venous thrombosis in many inflammatory diseases, these data suggest that the role of NETs in COVID-19-associated thrombophilia warrants systematic investigation. While a recent report suggested antiphospholipid antibodies may be drivers of thrombosis in some COVID-19 patients23, such antibodies were not detected here. Examples of NETs as drivers of thrombosis are myriad, as intravascular NET release is responsible for initiation and accretion of thrombotic events in arteries, veins, and microvessels, where thrombotic disease can drive end-organ damage in lungs, heart, kidneys, and other organs26, 27. Mechanistically, DNA in NETs may directly activate the extrinsic pathway of coagulation28, while NETs also present tissue factor to initiate the intrinsic pathway29. Serine proteases in NETs such as neutrophil elastase dismantle brakes on coagulation such as tissue factor pathway inhibitor30. Bidirectional interplay between NETs and platelets might also be critical for COVID-19-associated thrombosis as has been characterized in a variety of disease models27, 28. Approaches to combatting NETs31, 32 include the dismantling of NETs with deoxyribonucleases and strategies that prevent initiation of NET release such as neutrophil elastase inhibitors and peptidylarginine deiminase 4 inhibitors. As we await definitive antiviral and immunologic solutions to the current pandemic, we posit that anti-neutrophil therapies may be part of a personalized strategy for some individuals affected by COVID-19. Furthermore, those patients with hyperactive neutrophils may be at particularly high risk for VTE and might therefore benefit from more aggressive anticoagulation while hospitalized. ## METHODS ### Human samples Blood was collected into serum separator tubes by a trained hospital phlebotomist. After completion of biochemical testing ordered by the clinician, the remaining serum was stored at 4°C for up to 48 hours before it was deemed “discarded” and released to the research laboratory. Serum samples were immediately divided into small aliquots and stored at −80°C until the time of testing. This study complied with all relevant ethical regulations, and was approved by the University of Michigan Institutional Review Board (HUM00179409), which waived the requirement for informed consent given the discarded nature of the samples. Healthy volunteers were recruited through a posted flyer; exclusion criteria for these controls included history of a systemic autoimmune disease, active infection, and pregnancy. For preparation of control serum, blood was collected in a similar manner as the COVID-19 patient samples. These serum samples were divided into small aliquots and stored at −80°C until the time of testing. ### Quantification of cell-free DNA Cell-free DNA was quantified in sera using the Quant-iT PicoGreen dsDNA Assay Kit (Invitrogen, P11496) according to the manufacturer’s instructions. ### Quantification of citrullinated-histone H3 Citrullinated-histone H3 was quantified in sera using the Citrullinated Histone H3 (Clone 11D3) ELISA Kit (Cayman, 501620) according to the manufacturer’s instructions. ### Quantification of myeloperoxidase-DNA complexes Myeloperoxidase-DNA complexes were quantified similarly to what has been previously described 33. This protocol used several reagents from the Cell Death Detection ELISA kit (Roche). First, a high-binding EIA/RIA 96-well plate (Costar) was coated overnight at 4°C with anti-human myeloperoxidase antibody (Bio-Rad 0400-0002), diluted to a concentration of 1 μg/ml in coating buffer (Cell Death kit). The plate was washed two times with wash buffer (0.05% Tween 20 in PBS), and then blocked with 4% bovine serum albumin in PBS (supplemented with 0.05% Tween 20) for 2 hours at room temperature. The plate was again washed five times, before incubating for 90 minutes at room temperature with 10% serum or plasma in the aforementioned blocking buffer (without Tween 20). The plate was washed five times, and then incubated for 90 minutes at room temperature with 10x anti-DNA antibody (HRP-conjugated; from the Cell Death kit) diluted 1:100 in blocking buffer. After five more washes, the plate was developed with 3,3’,5,5’-Tetramethylbenzidine (TMB) substrate (Invitrogen) followed by a 2N sulfuric acid stop solution. Absorbance was measured at a wavelength of 450 nm using a Cytation 5 Cell Imaging Multi-Mode Reader (BioTek). Data were normalized to *in vitro-prepared* NET standards included on every plate, which were quantified based on their DNA content. ### Quantification of antiphospholipid antibodies Antiphospholipid antibodies were quantified in sera using the Quanta Lite® ACA IgG, ACA IgM, ACA IgA, (β2GPI IgG, (β2GPI IgM, (β2GPI IgA, aPS/PT IgG, aPS/PT IgM Kits (Inova Diagnostics) according to the manufacturer’s instructions. Manufacturer-recommended cut off was used to determine positive values. ## Data Availability Once published in a peer-reviewed journal, all primary data will be available upon request to the corresponding authors. ## AUTHORSHIP YZ, MZ, SY, KG, JM, and HS conducted experiments and analyzed data. YZ, MZ, YK, and JSK conceived the study and analyzed data. All authors participated in writing the manuscript and gave approval before submission. ## ACKNOWLEDGEMENTS The work was supported by a COVID-19 Cardiovascular Impact Research Ignitor Grant from the Michigan Medicine Frankel Cardiovascular Center as well as by the A. Alfred Taubman Medical Research Institute. YZ was supported by career development grants from the Rheumatology Research Foundation and APS ACTION. JAM was partially supported by the VA Healthcare System. YK was supported by the NIH (K08HL131993, R01HL150392), Falk Medical Research Trust Catalyst Award, and the JOBST-American Venous Forum Award. JSK was supported by grants from the NIH (R01HL115138), Lupus Research Alliance, and Burroughs Wellcome Fund. YZ and JSK also thank all members of the “COVID-19 NETwork” for their helpful advice and encouragement. ## Footnotes * ‡ Jason S. Knight and Yogendra Kanthi are co-corresponding authors * **Competing interests:** The authors have no financial conflicts to disclose. * Received April 30, 2020. * Revision received April 30, 2020. * Accepted May 5, 2020. * © 2020, Posted by Cold Spring Harbor Laboratory The copyright holder for this pre-print is the author. All rights reserved. The material may not be redistributed, re-used or adapted without the author's permission. ## REFERENCES 1. 1.Pedersen SF and Ho YC. SARS-CoV-2: A Storm is Raging. J Clin Invest. 2020. 2. 2.Zhang B, Zhou X, Zhu C, Feng F, Qiu Y, Feng J, Jia Q, Song Q, Zhu B and Wang J. Immune phenotyping based on neutrophil-to-lymphocyte ratio and IgG predicts disease severity and outcome for patients with COVID-19. 2020:2020.03.12.20035048. 3. 3.Song C-Y, Xu J, He J-Q and Lu Y-Q. COVID-19 early warning score: a multi-parameter screening tool to identify highly suspected patients. 2020:2020.03.05.20031906. 4. 4.Iba T, Levy JH, Raj A and Warkentin TE. Advance in the Management of Sepsis-Induced Coagulopathy and Disseminated Intravascular Coagulation. J Clin Med. 2019;8. 5. 5.Ward PA and Fattahi F. New strategies for treatment of infectious sepsis. J Leukoc Biol. 2019;106:187–192. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1002/JLB.4MIR1118-425R&link_type=DOI) 6. 6.Ali RA, Gandhi AA, Meng H, Yalavarthi S, Vreede AP, Estes SK, Palmer OR, Bockenstedt PL, Pinsky DJ, Greve JM, Diaz JA, Kanthi Y and Knight JS. Adenosine receptor agonism protects against NETosis and thrombosis in antiphospholipid syndrome. Nat Commun. 2019;10:1916. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/s41467-019-09801-x&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=31015489&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2020%2F05%2F05%2F2020.04.30.20086736.atom) 7. 7.Meng H, Yalavarthi S, Kanthi Y, Mazza LF, Elfline MA, Luke CE, Pinsky DJ, Henke PK and Knight JS. In Vivo Role of Neutrophil Extracellular Traps in Antiphospholipid Antibody-Mediated Venous Thrombosis. Arthritis Rheumatol. 2017;69:655–667. 8. 8.Yadav V, Chi L, Zhao R, Tourdot BE, Yalavarthi S, Jacobs BN, Banka A, Liao H, Koonse S, Anyanwu AC, Visovatti SH, Holinstat MA, Kahlenberg JM, Knight JS, Pinsky DJ and Kanthi Y. Ectonucleotidase tri(di)phosphohydrolase-1 (ENTPD-1) disrupts inflammasome/interleukin 1beta-driven venous thrombosis. J Clin Invest. 2019;129:2872–2877. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1172/JCI124804&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=30990798&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2020%2F05%2F05%2F2020.04.30.20086736.atom) 9. 9.Potey PM, Rossi AG, Lucas CD and Dorward DA. Neutrophils in the initiation and resolution of acute pulmonary inflammation: understanding biological function and therapeutic potential. J Pathol. 2019;247:672–685. 10. 10.Frantzeskaki F, Armaganidis A and Orfanos SE. Immunothrombosis in Acute Respiratory Distress Syndrome: Cross Talks between Inflammation and Coagulation. Respiration. 2017;93:212–225. [PubMed](http://medrxiv.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2020%2F05%2F05%2F2020.04.30.20086736.atom) 11. 11.Brinkmann V, Reichard U, Goosmann C, Fauler B, Uhlemann Y, Weiss DS, Weinrauch Y and Zychlinsky A. Neutrophil extracellular traps kill bacteria. Science. 2004;303:1532–5. [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6Mzoic2NpIjtzOjU6InJlc2lkIjtzOjEzOiIzMDMvNTY2My8xNTMyIjtzOjQ6ImF0b20iO3M6NTA6Ii9tZWRyeGl2L2Vhcmx5LzIwMjAvMDUvMDUvMjAyMC4wNC4zMC4yMDA4NjczNi5hdG9tIjt9czo4OiJmcmFnbWVudCI7czowOiIiO30=) 12. 12.Twaddell SH, Baines KJ, Grainge C and Gibson PG. The Emerging Role of Neutrophil Extracellular Traps in Respiratory Disease. Chest. 2019;156:774–782. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.chest.2019.06.012&link_type=DOI) 13. 13.Porto BN and Stein RT. Neutrophil Extracellular Traps in Pulmonary Diseases: Too Much of a Good Thing? Front Immunol. 2016;7:311. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.3389/fimmu.2016.00311&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=27574522&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2020%2F05%2F05%2F2020.04.30.20086736.atom) 14. 14.Smith CK, Vivekanandan-Giri A, Tang C, Knight JS, Mathew A, Padilla RL, Gillespie BW, Carmona-Rivera C, Liu X, Subramanian V, Hasni S, Thompson PR, Heinecke JW, Saran R, Pennathur S and Kaplan MJ. Neutrophil extracellular trap-derived enzymes oxidize high-density lipoprotein: an additional proatherogenic mechanism in systemic lupus erythematosus. Arthritis Rheumatol. 2014;66:2532–2544. 15. 15.Silvestre-Roig C, Braster Q, Wichapong K, Lee EY, Teulon JM, Berrebeh N, Winter J, Adrover JM, Santos GS, Froese A, Lemnitzer P, Ortega-Gomez A, Chevre R, Marschner J, Schumski A, Winter C, Perez-Olivares L, Pan C, Paulin N, Schoufour T, Hartwig H, Gonzalez-Ramos S, Kamp F, Megens RTA, Mowen KA, Gunzer M, Maegdefessel L, Hackeng T, Lutgens E, Daemen M, von Blume J, Anders HJ, Nikolaev VO, Pellequer JL, Weber C, Hidalgo A, Nicolaes GAF, Wong GCL and Soehnlein O. Externalized histone H4 orchestrates chronic inflammation by inducing lytic cell death. Nature. 2019;569:236–240. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/s41586-019-1167-6&link_type=DOI) 16. 16.Saffarzadeh M, Juenemann C, Queisser MA, Lochnit G, Barreto G, Galuska SP, Lohmeyer J and Preissner KT. Neutrophil extracellular traps directly induce epithelial and endothelial cell death: a predominant role of histones. PLoS One. 2012;7:e32366. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1371/journal.pone.0032366&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=22389696&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2020%2F05%2F05%2F2020.04.30.20086736.atom) 17. 17.Warnatsch A, Ioannou M, Wang Q and Papayannopoulos V. Inflammation. Neutrophil extracellular traps license macrophages for cytokine production in atherosclerosis. Science. 2015;349:316–20. [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6Mzoic2NpIjtzOjU6InJlc2lkIjtzOjEyOiIzNDkvNjI0NS8zMTYiO3M6NDoiYXRvbSI7czo1MDoiL21lZHJ4aXYvZWFybHkvMjAyMC8wNS8wNS8yMDIwLjA0LjMwLjIwMDg2NzM2LmF0b20iO31zOjg6ImZyYWdtZW50IjtzOjA6IiI7fQ==) 18. 18.Doring Y, Soehnlein O and Weber C. Neutrophil Extracellular Traps in Atherosclerosis and Atherothrombosis. Circ Res. 2017;120:736–743. [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6MTA6ImNpcmNyZXNhaGEiO3M6NToicmVzaWQiO3M6OToiMTIwLzQvNzM2IjtzOjQ6ImF0b20iO3M6NTA6Ii9tZWRyeGl2L2Vhcmx5LzIwMjAvMDUvMDUvMjAyMC4wNC4zMC4yMDA4NjczNi5hdG9tIjt9czo4OiJmcmFnbWVudCI7czowOiIiO30=) 19. 19.Brill A, Fuchs TA, Savchenko AS, Thomas GM, Martinod K, De Meyer SF, Bhandari AA and Wagner DD. Neutrophil extracellular traps promote deep vein thrombosis in mice. J Thromb Haemost. 2012;10:136–44. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1111/j.1538-7836.2011.04544.x&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=22044575&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2020%2F05%2F05%2F2020.04.30.20086736.atom) 20. 20.Tanaka K, Koike Y, Shimura T, Okigami M, Ide S, Toiyama Y, Okugawa Y, Inoue Y, Araki T, Uchida K, Mohri Y, Mizoguchi A and Kusunoki M. In vivo characterization of neutrophil extracellular traps in various organs of a murine sepsis model. PLoS One. 2014;9:e111888. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1371/journal.pone.0111888&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=25372699&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2020%2F05%2F05%2F2020.04.30.20086736.atom) 21. 21.Zhou F, Yu T, Du R, Fan G, Liu Y, Liu Z, Xiang J, Wang Y, Song B, Gu X, Guan L, Wei Y, Li H, Wu X, Xu J, Tu S, Zhang Y, Chen H and Cao B. Clinical course and risk factors for mortality of adult inpatients with COVID-19 in Wuhan, China: a retrospective cohort study. Lancet. 2020;395:1054–1062. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/S0140-6736(20)30566-3&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2020%2F05%2F05%2F2020.04.30.20086736.atom) 22. 22.Bunce PE, High SM, Nadjafi M, Stanley K, Liles WC and Christian MD. Pandemic H1N1 influenza infection and vascular thrombosis. Clin Infect Dis. 2011;52:e14–7. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1093/cid/ciq125&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=21288835&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2020%2F05%2F05%2F2020.04.30.20086736.atom) 23. 23.Zhang Y, Xiao M, Zhang S, Xia P, Cao W, Jiang W, Chen H, Ding X, Zhao H, Zhang H, Wang C, Zhao J, Sun X, Tian R, Wu W, Wu D, Ma J, Chen Y, Zhang D, Xie J, Yan X, Zhou X, Liu Z, Wang J, Du B, Qin Y, Gao P, Qin X, Xu Y, Zhang W, Li T, Zhang F, Zhao Y, Li Y and Zhang S. Coagulopathy and Antiphospholipid Antibodies in Patients with Covid-19. N Engl J Med. 2020. 24. 24.Tang N, Li D, Wang X and Sun Z. Abnormal coagulation parameters are associated with poor prognosis in patients with novel coronavirus pneumonia. J Thromb Haemost. 2020;18:844–847. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1111/jth.14768&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2020%2F05%2F05%2F2020.04.30.20086736.atom) 25. 25.Han H, Yang L, Liu R, Liu F, Wu KL, Li J, Liu XH and Zhu CL. Prominent changes in blood coagulation of patients with SARS-CoV-2 infection. Clin Chem Lab Med. 2020. 26. 26.Pfeiler S, Massberg S and Engelmann B. Biological basis and pathological relevance of microvascular thrombosis. Thromb Res. 2014;133 Suppl 1:S35–7. 27. 27.Ma AC and Kubes P. Platelets, neutrophils, and neutrophil extracellular traps (NETs) in sepsis. J Thromb Haemost. 2008;6:415–20. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1111/j.1538-7836.2007.02865.x&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=18088344&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2020%2F05%2F05%2F2020.04.30.20086736.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000253480900002&link_type=ISI) 28. 28.Gould TJ, Vu TT, Swystun LL, Dwivedi DJ, Mai SH, Weitz JI and Liaw PC. Neutrophil extracellular traps promote thrombin generation through platelet-dependent and platelet-independent mechanisms. Arterioscler Thromb Vasc Biol. 2014;34:1977–84. [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6NzoiYXR2YmFoYSI7czo1OiJyZXNpZCI7czo5OiIzNC85LzE5NzciO3M6NDoiYXRvbSI7czo1MDoiL21lZHJ4aXYvZWFybHkvMjAyMC8wNS8wNS8yMDIwLjA0LjMwLjIwMDg2NzM2LmF0b20iO31zOjg6ImZyYWdtZW50IjtzOjA6IiI7fQ==) 29. 29.Kambas K, Chrysanthopoulou A, Vassilopoulos D, Apostolidou E, Skendros P, Girod A, Arelaki S, Froudarakis M, Nakopoulou L, Giatromanolaki A, Sidiropoulos P, Koffa M, Boumpas DT, Ritis K and Mitroulis I. 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. 2014;73:1854–63. [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6MTE6ImFubnJoZXVtZGlzIjtzOjU6InJlc2lkIjtzOjEwOiI3My8xMC8xODU0IjtzOjQ6ImF0b20iO3M6NTA6Ii9tZWRyeGl2L2Vhcmx5LzIwMjAvMDUvMDUvMjAyMC4wNC4zMC4yMDA4NjczNi5hdG9tIjt9czo4OiJmcmFnbWVudCI7czowOiIiO30=) 30. 30.Massberg S, Grahl L, von Bruehl ML, Manukyan D, Pfeiler S, Goosmann C, Brinkmann V, Lorenz M, Bidzhekov K, Khandagale AB, Konrad I, Kennerknecht E, Reges K, Holdenrieder S, Braun S, Reinhardt C, Spannagl M, Preissner KT and Engelmann B. Reciprocal coupling of coagulation and innate immunity via neutrophil serine proteases. Nat Med. 2010;16:887–96. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/nm.2184&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=20676107&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2020%2F05%2F05%2F2020.04.30.20086736.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000280649200028&link_type=ISI) 31. 31.Thalin C, Hisada Y, Lundstrom S, Mackman N and Wallen H. Neutrophil Extracellular Traps: Villains and Targets in Arterial, Venous, and Cancer-Associated Thrombosis. Arterioscler Thromb Vasc Biol. 2019;39:1724–1738. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1161/ATVBAHA.119.312463&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=31315434&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2020%2F05%2F05%2F2020.04.30.20086736.atom) 32. 32.Barnado A, Crofford LJ and 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. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1189/jlb.5BT0615-234R&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=26658004&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2020%2F05%2F05%2F2020.04.30.20086736.atom) 33. 33.Kessenbrock K, Krumbholz M, Schonermarck U, Back W, Gross WL, Werb Z, Grone HJ, Brinkmann V and Jenne DE. Netting neutrophils in autoimmune small-vessel vasculitis. Nat Med. 2009;15:623–5. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/nm.1959&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=19448636&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2020%2F05%2F05%2F2020.04.30.20086736.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000266731600020&link_type=ISI)