Hamostaseologie 2021; 41(06): 428-432
DOI: 10.1055/a-1661-0257
Review Article

Thromboinflammation as a Driver of Venous Thromboembolism

Nadine Gauchel
1   Department of Cardiology and Angiology I, University Heart Center Freiburg—Bad Krozingen, Faculty of Medicine, University of Freiburg, Freiburg, Germany
,
Krystin Krauel
1   Department of Cardiology and Angiology I, University Heart Center Freiburg—Bad Krozingen, Faculty of Medicine, University of Freiburg, Freiburg, Germany
,
Muataz Ali Hamad
1   Department of Cardiology and Angiology I, University Heart Center Freiburg—Bad Krozingen, Faculty of Medicine, University of Freiburg, Freiburg, Germany
,
Christoph Bode
1   Department of Cardiology and Angiology I, University Heart Center Freiburg—Bad Krozingen, Faculty of Medicine, University of Freiburg, Freiburg, Germany
,
Daniel Duerschmied
1   Department of Cardiology and Angiology I, University Heart Center Freiburg—Bad Krozingen, Faculty of Medicine, University of Freiburg, Freiburg, Germany
› Author Affiliations
Funding Deutsche Forschungsgemeinschaft, #422681845
 

Abstract

Thrombus formation has been identified as an integral part in innate immunity, termed immunothrombosis. Activation of host defense systems is known to result in a procoagulant environment. In this system, cellular players as well as soluble mediators interact with each other and their dysregulation can lead to the pathological process of thromboinflammation. These mechanisms have been under intensified investigation during the COVID-19 pandemic. In this review, we focus on the underlying mechanisms leading to thromboinflammation as one trigger of venous thromboembolism.


#

Zusammenfassung

Die Thrombusbildung wurde als integraler Bestandteil der angeborenen Immunität identifiziert und als Immunthrombose bezeichnet. Es ist bekannt, dass die Aktivierung von Wirtsabwehrsystemen zu einer pro-thrombotischen Umgebung führt. In diesem System interagieren sowohl zelluläre Bestandteile als auch lösliche Faktoren miteinander, die bei einer Dysregulation den pathologischen Prozess der Thromboinflammation induzieren können. Diese Mechanismen wurden während der COVID-19-Pandemie verstärkt untersucht. In dieser Übersichtsarbeit konzentrieren wir uns auf die zugrunde liegenden Mechanismen, die zur Thromboinflammation führen als ein Auslöser der venösen Thromboembolien.


#

Introduction

Venous thromboembolism (VTE) with its manifestations deep vein thrombosis (DVT) and pulmonary embolism remains a major health care challenge.[1] Besides its obvious role in wound closure, thrombus formation has also been identified as an integral part in innate immunity, termed immunothrombosis.[2] Activation of host defense systems in response to invading pathogens is known to result in a procoagulant environment, which promotes thrombin generation. This cross-link between humoral and cellular amplification pathways as part of the physiological host defense (in this review defined as “immunothrombosis”) should be differentiated from pathophysiological events during “thromboinflammation,”[3] [4] [5] where an overactivation of blood cells, coagulation system, and endothelial cells in response to pathogens or inflammatory triggers results in pathological thrombotic events ([Fig. 1]).

Zoom Image
Fig. 1 Cellular players and soluble mediators driving immunothrombosis and thromboinflammation.

Immunothrombosis mostly occurs in capillaries and venules without a major harm for the host to contain and neutralize foreign pathogens. However, when these mechanisms proceed uncontrolled, it can lead to pathological thrombosis, such as arterial thrombosis or DVT or disseminated intravascular coagulation in sepsis.[6] [7] In this review, we focus on thromboinflammation as a driver of VTE and the novel insights into the underlying mechanisms.


#

Cellular Players and NETs

Mouse studies have revealed that monocytes, neutrophils, and platelets, in concert with the endothelium, are involved in the development of DVT.[6] Different triggers such as ischemic events, infections, or toxins can induce endothelial dysfunction.[8] For example, endothelial activation in endotoxemia culminates in augmented thrombus formation mediated through ICAM-1 (intercellular adhesion molecule-1) and TLR-1 (toll-like receptor 1).[9] In addition, direct pathogen interaction with endothelial cells can stimulate the release of different mediators.[10] Hypoxia is another pathological condition that leads to endothelial dysfunction characterized by increased permeability, a proinflammatory state, and decreased anticoagulant features.[11] Specifically, hypoxia induces upregulation of von Willebrand factor (VWF).[12] Mast cells located in the venous vessel wall release their mediators in response to reduced blood flow and further activate endothelium, which leads to Weibel–Palade body release.[13] Especially VWF release from Weibel–Palade bodies is crucial for deep vein thrombus formation by mediating platelet adhesion via glycoprotein Ibα (GPIbα).[14] [15] Platelet adhesion leads to leukocyte recruitment to the vessel wall and thereby activating innate immunity.[5] Consistently, GPIbα-deficient mice showed impaired platelet and leukocyte accumulation along the endothelium.[6] In addition, platelets themselves sense infection and can be activated by pathogens. Consequently, they form aggregates, trigger the coagulation cascade, and recruit neutrophils and monocytes to prevent the spread of pathogens, thereby actively contributing to thromboinflammation.[16] Furthermore, clinical trials demonstrated that antiplatelet therapy is beneficial in preventing recurrent venous thromboembolic events.[17] [18]

Leukocyte recruitment is essential for the development of DVT.[6] [19] The activation of the coagulation system in venous thrombosis depends on blood-derived tissue factor (TF), which is mainly released by monocytes and locally activated by protein disulfide isomerase.[20] [21] Moreover, neutrophil extracellular traps (NETs) particularly contribute to immunothrombosis by building a scaffold of chromatin and inflammatory and prothrombotic proteins and entrap cells, including activated platelets, enhancing thrombus formation as a positive feedback loop.[22] [23] [24] [25] Direct interaction with pathogens or microbial components, cytokines, and complement factors can induce the release of NETs.[26] Notably, the formation of NETs is promoted by neutrophil interaction with activated platelets.[27] If NETosis is inhibited[28] or NETs are dissolved by DNase,[6] [25] mice are protected from development of DVT in a stenosis model.

Addressing NETosis and cell recruitment in venous thrombosis promises new therapeutic targets in the prophylaxis and therapy of VTE.


#

Complement Factors and Cytokines

Innate immune cells are the main cellular drivers of immunothrombosis as described above. In addition, this process is molecularly regulated by the crosstalk between the coagulation cascade, the complement system, and the cytokines. The complement system can be activated via several pathways and several complement factors can activate platelets, neutrophils, induce endothelial secretion of VWF, and cause endothelial damage.[29] [30] [31] [32] The complement system is an important host defense mechanism that involves a cascade of processes, leading to the formation of the terminal membrane attack complex (MAC) C5b-9. MAC creates a transmembrane channel, triggering cell lysis and death when inserted into the cell membrane of an infected cell or directly onto a pathogen.[31] When these physiological defenses are hyperactivated, they result in excess endothelial damage that can serve as foci for thrombosis. Besides the endothelial damage caused by the complement system, which increases the thrombotic risk, the individual complement components are prothrombotic. Complement component 5a (C5a), for instance, can upregulate the activity of TF and plasminogen activator inhibitor-1 (PAI-1) and can activate neutrophils, resulting in promotion in the formation of NETs.[31] This corresponds to the findings seen in a mouse model where susceptibility to DVT strongly correlates with C5a levels,[33] as well as in humans where high levels of the C3 are associated with a high risk of DVT.[34]

Cytokines on the other hand, which are proteins secreted by various cells including immune cells, serve as an important innate defense mechanism as they recruit adaptive immune cells, and regulate a wide range of processes in the immune system.[35] Cytokines have prothrombotic effects, such as interleukin-6 which increases platelet production and activity, increases the expression of TF on endothelial cells and monocytes, and can also give rise to endothelial dysfunction.[36] [37] Interferon-γ similarly increases platelet production and impairs the vascular endothelium, which in turn increases prothrombotic effects.[37] Interleukin-2 upregulates PAI-1 which can decrease fibrinolysis.[37] It is important to note that not only inflammation causes thrombosis but thrombosis can in turn directly trigger inflammation and a tight, bidirectional connection exists between inflammation and thrombosis.

Intervening in these cross-talks promises future therapeutic options.


#

COVID-19

During the coronavirus disease 2019 (COVID-19) pandemic, increased incidences of thrombotic complications have been observed.[38] The mechanisms contributing to increased thrombosis in COVID-19 involve extensive cross-talk between hemostasis and the immune system.[39] In COVID-19 there are two entities leading to immunothrombosis and thromboinflammation.

One is local immunothrombosis in pulmonary vessels mediated by the infection of alveolar epithelium with the pathogen SARS-CoV-2 (severe acute respiratory syndrome coronavirus 2) via the ACE2 (angiotensin converting enzyme 2) receptor. This leads to a release of inflammatory cytokines such as interleukin-6 and tumor necrosis factor and chemokines such as interleukin-8 and CCL (chemokine [C–C motif] ligand)2 and CCL3[40] [41] which thereby activate epithelial cells, monocytes, and neutrophils. Endothelial cells themselves can also be infected by SARS-CoV-2 via the ACE2 receptor leading to activation and dysfunction. This leads to the activation of the coagulation system and by activating platelets the proinflammatory state is furthermore triggered leading to local coagulation lesions.[42] [43] Interleukin-6 levels show a correlation with fibrinogen levels in COVID-19 patients supporting the theory of thromboinflammation.[44]

In addition to the local immunothrombosis/thromboinflammation in COVID-19 patients, the infection can also lead to a systemic hypercoagulable state, leading to macro- and microvascular thrombosis as a result of thromboinflammation. The overactivation of the complement system leads to the activation of the alternative and lectin pathway which interacts with the coagulation pathway.[45] [46] Furthermore SARS-CoV-2 stimulates the ACE2 receptor and thereby disrupts the renin–angiotensin system, which leads to vasoconstriction and proinflammatory cytokine release,[47] which can trigger a cytokine storm and a systemic inflammatory response. The systemic cytokine release activates endothelial cells in the whole organism leading to endothelial dysfunction.[48] Furthermore, the impact of NETs in COVID-19 has been extensively described to contribute to the procoagulant and proinflammatory state.[49] Autopsy studies revealed the occurrence of NETs in lungs from deceased COVID-19 patients.[50] [51] [52] Moreover, soluble indicators of NETs have been widely detected in the plasma and sera of COVID-19 patients.[53] Most importantly, NETs in cooperation with TF and the complement system were associated with thrombotic events.[54] [55] [56]

Increased levels of antiphospholipid antibodies have been detected in critically ill patients with COVID-19.[57] [58] In the antiphospholipid syndrome, these antibodies remain elevated over time and are known for the development of thromboembolic events. Their role in the development of thromboinflammation in COVID-19 remains controversial, as they are transiently elevated in many acute illnesses and the underlying mechanisms are not yet clearly understood.[59] [60]

Also platelets have been proposed to be prothrombotic players in COVID-19.[61] The majority of studies found hyperactivated platelets during SARS-CoV-2 infection.

Several therapeutic intervention strategies to reduce the risk of developing thrombosis have been proposed to be useful in COVID-19 pathology.[62] They include direct targeting of the coagulation cascade, antiplatelet drugs, inhibitors of NET formation as well as complement and cytokine blockade. However, effective treatment options are still lacking.


#

Conclusion

The mechanisms leading to VTE are complex and closely linked to the innate immune system as well as to inflammatory processes ([Fig. 1]). The most recent and prominent example for these close interactions is the current COVID-19 pandemic with its high incidences of thrombotic complications.

Unraveling these pathomechanisms promises future therapeutic strategies to prevent thromboembolic complications.


#
#

Conflict of Interest

The authors declare that they have no conflict of interest.

Acknowledgments

Nadine Gauchel, Krystin Krauel, Muataz Ali Hamad, and Daniel Duerschmied are members of SFB1425, funded by the Deutsche Forschungsgemeinschaft (DFG, German Research Foundation #422681845).

  • References

  • 1 Raskob GE, Angchaisuksiri P, Blanco AN. et al; ISTH Steering Committee for World Thrombosis Day. Thrombosis: a major contributor to global disease burden. Arterioscler Thromb Vasc Biol 2014; 34 (11) 2363-2371
  • 2 Stark K, Massberg S. Interplay between inflammation and thrombosis in cardiovascular pathology. Nat Rev Cardiol 2021; 18 (09) 666-682
  • 3 Engelmann B, Massberg S. Thrombosis as an intravascular effector of innate immunity. Nat Rev Immunol 2013; 13 (01) 34-45
  • 4 Delabranche X, Helms J, Meziani F. Immunohaemostasis: a new view on haemostasis during sepsis. Ann Intensive Care 2017; 7 (01) 117
  • 5 Jackson SP, Darbousset R, Schoenwaelder SM. Thromboinflammation: challenges of therapeutically targeting coagulation and other host defense mechanisms. Blood 2019; 133 (09) 906-918
  • 6 von Brühl M-L, Stark K, Steinhart A. et al. Monocytes, neutrophils, and platelets cooperate to initiate and propagate venous thrombosis in mice in vivo. J Exp Med 2012; 209 (04) 819-835
  • 7 van der Poll T, de Boer JD, Levi M. The effect of inflammation on coagulation and vice versa. Curr Opin Infect Dis 2011; 24 (03) 273-278
  • 8 Gresele P, Momi S, Migliacci R. Endothelium, venous thromboembolism and ischaemic cardiovascular events. Thromb Haemost 2010; 103 (01) 56-61
  • 9 Obi AT, Andraska E, Kanthi Y. et al. Endotoxaemia-augmented murine venous thrombosis is dependent on TLR-4 and ICAM-1, and potentiated by neutropenia. Thromb Haemost 2017; 117 (02) 339-348
  • 10 Hippenstiel S, Suttorp N. Interaction of pathogens with the endothelium. Thromb Haemost 2003; 89 (01) 18-24
  • 11 Ten VS, Pinsky DJ. Endothelial response to hypoxia: physiologic adaptation and pathologic dysfunction. Curr Opin Crit Care 2002; 8 (03) 242-250
  • 12 Mojiri A, Nakhaii-Nejad M, Phan W-L. et al. Hypoxia results in upregulation and de novo activation of von Willebrand factor expression in lung endothelial cells. Arterioscler Thromb Vasc Biol 2013; 33 (06) 1329-1338
  • 13 Ponomaryov T, Payne H, Fabritz L, Wagner DD, Brill A. Mast cells granular contents are crucial for deep vein thrombosis in mice. Circ Res 2017; 121 (08) 941-950
  • 14 Brill A, Fuchs TA, Chauhan AK. et al. von Willebrand factor-mediated platelet adhesion is critical for deep vein thrombosis in mouse models. Blood 2011; 117 (04) 1400-1407
  • 15 Michels A, Dwyer CN, Mewburn J. et al. von Willebrand factor is a critical mediator of deep vein thrombosis in a mouse model of diet-induced obesity. Arterioscler Thromb Vasc Biol 2020; 40 (12) 2860-2874
  • 16 Guo L, Rondina MT. The era of thromboinflammation: platelets are dynamic sensors and effector cells during infectious diseases. Front Immunol 2019; 10: 2204
  • 17 Becattini C, Agnelli G, Schenone A. et al; WARFASA Investigators. Aspirin for preventing the recurrence of venous thromboembolism. N Engl J Med 2012; 366 (21) 1959-1967
  • 18 Brighton TA, Eikelboom JW, Mann K. et al; ASPIRE Investigators. Low-dose aspirin for preventing recurrent venous thromboembolism. N Engl J Med 2012; 367 (21) 1979-1987
  • 19 Stewart GJ. Neutrophils and deep venous thrombosis. Haemostasis 1993; 23 (Suppl. 01) 127-140
  • 20 Subramaniam S, Jurk K, Hobohm L. et al. Distinct contributions of complement factors to platelet activation and fibrin formation in venous thrombus development. Blood 2017; 129 (16) 2291-2302
  • 21 Hisada Y, Ay C, Auriemma AC, Cooley BC, Mackman N. Human pancreatic tumors grown in mice release tissue factor-positive microvesicles that increase venous clot size. J Thromb Haemost 2017; 15 (11) 2208-2217
  • 22 Fuchs TA, Brill A, Duerschmied D. et al. Extracellular DNA traps promote thrombosis. Proc Natl Acad Sci U S A 2010; 107 (36) 15880-15885
  • 23 Elaskalani O, Abdol Razak NB, Metharom P. Neutrophil extracellular traps induce aggregation of washed human platelets independently of extracellular DNA and histones. Cell Commun Signal 2018; 16 (01) 24
  • 24 Laridan E, Martinod K, De Meyer SF. Neutrophil extracellular traps in arterial and venous thrombosis. Semin Thromb Hemost 2019; 45 (01) 86-93
  • 25 Brill A, Fuchs TA, Savchenko AS. et al. Neutrophil extracellular traps promote deep vein thrombosis in mice. J Thromb Haemost 2012; 10 (01) 136-144
  • 26 Sorvillo N, Cherpokova D, Martinod K, Wagner DD. Extracellular DNA NET-works with dire consequences for health. Circ Res 2019; 125 (04) 470-488
  • 27 Carestia A, Kaufman T, Schattner M. Platelets: new bricks in the building of neutrophil extracellular traps. Front Immunol 2016; 7: 271
  • 28 Martinod K, Demers M, Fuchs TA. et al. Neutrophil histone modification by peptidylarginine deiminase 4 is critical for deep vein thrombosis in mice. Proc Natl Acad Sci U S A 2013; 110 (21) 8674-8679
  • 29 Verschoor A, Langer HF. Crosstalk between platelets and the complement system in immune protection and disease. Thromb Haemost 2013; 110 (05) 910-919
  • 30 Schmidt CQ, Verschoor A. Complement and coagulation: so close, yet so far. Blood 2017; 130 (24) 2581-2582
  • 31 Fletcher-Sandersjöö A, Bellander B-M. Is COVID-19 associated thrombosis caused by overactivation of the complement cascade? A literature review. Thromb Res 2020; 194: 36-41
  • 32 de Bont CM, Boelens WC, Pruijn GJM. NETosis, complement, and coagulation: a triangular relationship. Cell Mol Immunol 2019; 16 (01) 19-27
  • 33 Foley JH, Walton BL, Aleman MM. et al. Complement activation in arterial and venous thrombosis is mediated by plasmin. EBioMedicine 2016; 5: 175-182
  • 34 Nørgaard I, Nielsen SF, Nordestgaard BG. Complement C3 and high risk of venous thromboembolism: 80517 individuals from the Copenhagen General Population Study. Clin Chem 2016; 62 (03) 525-534
  • 35 Borish LC, Steinke JW. 2. Cytokines and chemokines. J Allergy Clin Immunol 2003; 111 (2, Suppl): S460-S475
  • 36 Wang J, Jiang M, Chen X, Montaner LJ. Cytokine storm and leukocyte changes in mild versus severe SARS-CoV-2 infection: review of 3939 COVID-19 patients in China and emerging pathogenesis and therapy concepts. J Leukoc Biol 2020; 108 (01) 17-41
  • 37 Du F, Liu B, Zhang S. COVID-19: the role of excessive cytokine release and potential ACE2 down-regulation in promoting hypercoagulable state associated with severe illness. J Thromb Thrombolysis 2020; 51 (02) 313-329
  • 38 Bikdeli B, Madhavan MV, Jimenez D. et al; Global COVID-19 Thrombosis Collaborative Group, Endorsed by the ISTH, NATF, ESVM, and the IUA, Supported by the ESC Working Group on Pulmonary Circulation and Right Ventricular Function. COVID-19 and thrombotic or thromboembolic disease: implications for prevention, antithrombotic therapy, and follow-up: JACC state-of-the-art review. J Am Coll Cardiol 2020; 75 (23) 2950-2973
  • 39 Loo J, Spittle DA, Newnham M. COVID-19, immunothrombosis and venous thromboembolism: biological mechanisms. Thorax 2021; 76 (04) 412-420
  • 40 Mehta P, McAuley DF, Brown M, Sanchez E, Tattersall RS, Manson JJ. HLH Across Speciality Collaboration, UK. COVID-19: consider cytokine storm syndromes and immunosuppression. Lancet 2020; 395 (10229): 1033-1034
  • 41 Qin C, Zhou L, Hu Z. et al. Dysregulation of immune response in patients with coronavirus 2019 (COVID-19) in Wuhan, China. Clin Infect Dis 2020; 71 (15) 762-768
  • 42 Abou-Ismail MY, Diamond A, Kapoor S, Arafah Y, Nayak L. The hypercoagulable state in COVID-19: Incidence, pathophysiology, and management. Thromb Res 2020; 194: 101-115
  • 43 Levi M, van der Poll T, Büller HR. Bidirectional relation between inflammation and coagulation. Circulation 2004; 109 (22) 2698-2704
  • 44 Ranucci M, Ballotta A, Di Dedda U. et al. The procoagulant pattern of patients with COVID-19 acute respiratory distress syndrome. J Thromb Haemost 2020; 18 (07) 1747-1751
  • 45 Magro C, Mulvey JJ, Berlin D. et al. Complement associated microvascular injury and thrombosis in the pathogenesis of severe COVID-19 infection: a report of five cases. Transl Res 2020; 220: 1-13
  • 46 Carvelli J, Demaria O, Vély F. et al; Explore COVID-19 IPH group, Explore COVID-19 Marseille Immunopole group. Association of COVID-19 inflammation with activation of the C5a-C5aR1 axis. Nature 2020; 588 (7836): 146-150
  • 47 Leisman DE, Deutschman CS, Legrand M. Facing COVID-19 in the ICU: vascular dysfunction, thrombosis, and dysregulated inflammation. Intensive Care Med 2020; 46 (06) 1105-1108
  • 48 Libby P, Lüscher T. COVID-19 is, in the end, an endothelial disease. Eur Heart J 2020; 41 (32) 3038-3044
  • 49 Janiuk K, Jabłońska E, Garley M. Significance of NETs formation in COVID-19. Cells 2021; 10 (01) 151
  • 50 Middleton EA, He X-Y, Denorme F. et al. Neutrophil extracellular traps contribute to immunothrombosis in COVID-19 acute respiratory distress syndrome. Blood 2020; 136 (10) 1169-1179
  • 51 Leppkes M, Knopf J, Naschberger E. et al. Vascular occlusion by neutrophil extracellular traps in COVID-19. EBioMedicine 2020; 58: 102925
  • 52 Nicolai L, Leunig A, Brambs S. et al. Immunothrombotic dysregulation in COVID-19 pneumonia is associated with respiratory failure and coagulopathy. Circulation 2020; 142 (12) 1176-1189
  • 53 Krauel K, Duerschmied D. ICODE: the international COVID-19 thrombosis biomarkers colloquium-novel soluble biomarkers: circulating cell-free nucleic acids and other molecules. J Thromb Thrombolysis 2021 May 12:1–5. doi: 10.1007/s11239-021-02468-6. Epub ahead of print.
  • 54 Zuo Y, Zuo M, Yalavarthi S. et al. Neutrophil extracellular traps and thrombosis in COVID-19. J Thromb Thrombolysis 2021; 51 (02) 446-453
  • 55 Petito E, Falcinelli E, Paliani U. et al; COVIR study investigators. Association of neutrophil activation, more than platelet activation, with thrombotic complications in coronavirus disease 2019. J Infect Dis 2021; 223 (06) 933-944
  • 56 Skendros P, Mitsios A, Chrysanthopoulou A. et al. Complement and tissue factor-enriched neutrophil extracellular traps are key drivers in COVID-19 immunothrombosis. J Clin Invest 2020; 130 (11) 6151-6157
  • 57 Hasan Ali O, Bomze D, Risch L. et al. Severe COVID-19 is associated with elevated serum IgA and antiphospholipid IgA-antibodies. Clin Infect Dis 2020; ciaa1496: ciaa1496
  • 58 Xiao M, Zhang Y, Zhang S. et al. Antiphospholipid antibodies in critically ill patients with COVID-19. Arthritis Rheumatol 2020; 72 (12) 1998-2004
  • 59 Borghi MO, Beltagy A, Garrafa E. et al. Anti-phospholipid antibodies in COVID-19 are different from those detectable in the anti-phospholipid syndrome. Front Immunol 2020; 11: 584241
  • 60 Gkrouzman E, Barbhaiya M, Erkan D, Lockshin MD. Reality check on antiphospholipid antibodies in COVID-19-associated coagulopathy. Arthritis Rheumatol 2021; 73 (01) 173-174
  • 61 Violi F, Cammisotto V, Pignatelli P. Thrombosis in Covid-19 and non-Covid-19 pneumonia: role of platelets. Platelets 2021 Jun 7:1-9. doi: 10.1080/09537104.2021.1936478. Epub ahead of print.
  • 62 Bonaventura A, Vecchié A, Dagna L. et al. Endothelial dysfunction and immunothrombosis as key pathogenic mechanisms in COVID-19. Nat Rev Immunol 2021; 21 (05) 319-329

Address for correspondence

Nadine Gauchel, MD
Department of Cardiology and Angiology I, University Heart Center Freiburg—Bad, Krozingen, Faculty of Medicine, University of Freiburg
Hugstetter Str. 55, 79106 Freiburg
Germany   

Publication History

Received: 30 June 2021

Accepted: 04 October 2021

Article published online:
23 December 2021

© 2021. Thieme. All rights reserved.

Georg Thieme Verlag KG
Rüdigerstraße 14, 70469 Stuttgart, Germany

  • References

  • 1 Raskob GE, Angchaisuksiri P, Blanco AN. et al; ISTH Steering Committee for World Thrombosis Day. Thrombosis: a major contributor to global disease burden. Arterioscler Thromb Vasc Biol 2014; 34 (11) 2363-2371
  • 2 Stark K, Massberg S. Interplay between inflammation and thrombosis in cardiovascular pathology. Nat Rev Cardiol 2021; 18 (09) 666-682
  • 3 Engelmann B, Massberg S. Thrombosis as an intravascular effector of innate immunity. Nat Rev Immunol 2013; 13 (01) 34-45
  • 4 Delabranche X, Helms J, Meziani F. Immunohaemostasis: a new view on haemostasis during sepsis. Ann Intensive Care 2017; 7 (01) 117
  • 5 Jackson SP, Darbousset R, Schoenwaelder SM. Thromboinflammation: challenges of therapeutically targeting coagulation and other host defense mechanisms. Blood 2019; 133 (09) 906-918
  • 6 von Brühl M-L, Stark K, Steinhart A. et al. Monocytes, neutrophils, and platelets cooperate to initiate and propagate venous thrombosis in mice in vivo. J Exp Med 2012; 209 (04) 819-835
  • 7 van der Poll T, de Boer JD, Levi M. The effect of inflammation on coagulation and vice versa. Curr Opin Infect Dis 2011; 24 (03) 273-278
  • 8 Gresele P, Momi S, Migliacci R. Endothelium, venous thromboembolism and ischaemic cardiovascular events. Thromb Haemost 2010; 103 (01) 56-61
  • 9 Obi AT, Andraska E, Kanthi Y. et al. Endotoxaemia-augmented murine venous thrombosis is dependent on TLR-4 and ICAM-1, and potentiated by neutropenia. Thromb Haemost 2017; 117 (02) 339-348
  • 10 Hippenstiel S, Suttorp N. Interaction of pathogens with the endothelium. Thromb Haemost 2003; 89 (01) 18-24
  • 11 Ten VS, Pinsky DJ. Endothelial response to hypoxia: physiologic adaptation and pathologic dysfunction. Curr Opin Crit Care 2002; 8 (03) 242-250
  • 12 Mojiri A, Nakhaii-Nejad M, Phan W-L. et al. Hypoxia results in upregulation and de novo activation of von Willebrand factor expression in lung endothelial cells. Arterioscler Thromb Vasc Biol 2013; 33 (06) 1329-1338
  • 13 Ponomaryov T, Payne H, Fabritz L, Wagner DD, Brill A. Mast cells granular contents are crucial for deep vein thrombosis in mice. Circ Res 2017; 121 (08) 941-950
  • 14 Brill A, Fuchs TA, Chauhan AK. et al. von Willebrand factor-mediated platelet adhesion is critical for deep vein thrombosis in mouse models. Blood 2011; 117 (04) 1400-1407
  • 15 Michels A, Dwyer CN, Mewburn J. et al. von Willebrand factor is a critical mediator of deep vein thrombosis in a mouse model of diet-induced obesity. Arterioscler Thromb Vasc Biol 2020; 40 (12) 2860-2874
  • 16 Guo L, Rondina MT. The era of thromboinflammation: platelets are dynamic sensors and effector cells during infectious diseases. Front Immunol 2019; 10: 2204
  • 17 Becattini C, Agnelli G, Schenone A. et al; WARFASA Investigators. Aspirin for preventing the recurrence of venous thromboembolism. N Engl J Med 2012; 366 (21) 1959-1967
  • 18 Brighton TA, Eikelboom JW, Mann K. et al; ASPIRE Investigators. Low-dose aspirin for preventing recurrent venous thromboembolism. N Engl J Med 2012; 367 (21) 1979-1987
  • 19 Stewart GJ. Neutrophils and deep venous thrombosis. Haemostasis 1993; 23 (Suppl. 01) 127-140
  • 20 Subramaniam S, Jurk K, Hobohm L. et al. Distinct contributions of complement factors to platelet activation and fibrin formation in venous thrombus development. Blood 2017; 129 (16) 2291-2302
  • 21 Hisada Y, Ay C, Auriemma AC, Cooley BC, Mackman N. Human pancreatic tumors grown in mice release tissue factor-positive microvesicles that increase venous clot size. J Thromb Haemost 2017; 15 (11) 2208-2217
  • 22 Fuchs TA, Brill A, Duerschmied D. et al. Extracellular DNA traps promote thrombosis. Proc Natl Acad Sci U S A 2010; 107 (36) 15880-15885
  • 23 Elaskalani O, Abdol Razak NB, Metharom P. Neutrophil extracellular traps induce aggregation of washed human platelets independently of extracellular DNA and histones. Cell Commun Signal 2018; 16 (01) 24
  • 24 Laridan E, Martinod K, De Meyer SF. Neutrophil extracellular traps in arterial and venous thrombosis. Semin Thromb Hemost 2019; 45 (01) 86-93
  • 25 Brill A, Fuchs TA, Savchenko AS. et al. Neutrophil extracellular traps promote deep vein thrombosis in mice. J Thromb Haemost 2012; 10 (01) 136-144
  • 26 Sorvillo N, Cherpokova D, Martinod K, Wagner DD. Extracellular DNA NET-works with dire consequences for health. Circ Res 2019; 125 (04) 470-488
  • 27 Carestia A, Kaufman T, Schattner M. Platelets: new bricks in the building of neutrophil extracellular traps. Front Immunol 2016; 7: 271
  • 28 Martinod K, Demers M, Fuchs TA. et al. Neutrophil histone modification by peptidylarginine deiminase 4 is critical for deep vein thrombosis in mice. Proc Natl Acad Sci U S A 2013; 110 (21) 8674-8679
  • 29 Verschoor A, Langer HF. Crosstalk between platelets and the complement system in immune protection and disease. Thromb Haemost 2013; 110 (05) 910-919
  • 30 Schmidt CQ, Verschoor A. Complement and coagulation: so close, yet so far. Blood 2017; 130 (24) 2581-2582
  • 31 Fletcher-Sandersjöö A, Bellander B-M. Is COVID-19 associated thrombosis caused by overactivation of the complement cascade? A literature review. Thromb Res 2020; 194: 36-41
  • 32 de Bont CM, Boelens WC, Pruijn GJM. NETosis, complement, and coagulation: a triangular relationship. Cell Mol Immunol 2019; 16 (01) 19-27
  • 33 Foley JH, Walton BL, Aleman MM. et al. Complement activation in arterial and venous thrombosis is mediated by plasmin. EBioMedicine 2016; 5: 175-182
  • 34 Nørgaard I, Nielsen SF, Nordestgaard BG. Complement C3 and high risk of venous thromboembolism: 80517 individuals from the Copenhagen General Population Study. Clin Chem 2016; 62 (03) 525-534
  • 35 Borish LC, Steinke JW. 2. Cytokines and chemokines. J Allergy Clin Immunol 2003; 111 (2, Suppl): S460-S475
  • 36 Wang J, Jiang M, Chen X, Montaner LJ. Cytokine storm and leukocyte changes in mild versus severe SARS-CoV-2 infection: review of 3939 COVID-19 patients in China and emerging pathogenesis and therapy concepts. J Leukoc Biol 2020; 108 (01) 17-41
  • 37 Du F, Liu B, Zhang S. COVID-19: the role of excessive cytokine release and potential ACE2 down-regulation in promoting hypercoagulable state associated with severe illness. J Thromb Thrombolysis 2020; 51 (02) 313-329
  • 38 Bikdeli B, Madhavan MV, Jimenez D. et al; Global COVID-19 Thrombosis Collaborative Group, Endorsed by the ISTH, NATF, ESVM, and the IUA, Supported by the ESC Working Group on Pulmonary Circulation and Right Ventricular Function. COVID-19 and thrombotic or thromboembolic disease: implications for prevention, antithrombotic therapy, and follow-up: JACC state-of-the-art review. J Am Coll Cardiol 2020; 75 (23) 2950-2973
  • 39 Loo J, Spittle DA, Newnham M. COVID-19, immunothrombosis and venous thromboembolism: biological mechanisms. Thorax 2021; 76 (04) 412-420
  • 40 Mehta P, McAuley DF, Brown M, Sanchez E, Tattersall RS, Manson JJ. HLH Across Speciality Collaboration, UK. COVID-19: consider cytokine storm syndromes and immunosuppression. Lancet 2020; 395 (10229): 1033-1034
  • 41 Qin C, Zhou L, Hu Z. et al. Dysregulation of immune response in patients with coronavirus 2019 (COVID-19) in Wuhan, China. Clin Infect Dis 2020; 71 (15) 762-768
  • 42 Abou-Ismail MY, Diamond A, Kapoor S, Arafah Y, Nayak L. The hypercoagulable state in COVID-19: Incidence, pathophysiology, and management. Thromb Res 2020; 194: 101-115
  • 43 Levi M, van der Poll T, Büller HR. Bidirectional relation between inflammation and coagulation. Circulation 2004; 109 (22) 2698-2704
  • 44 Ranucci M, Ballotta A, Di Dedda U. et al. The procoagulant pattern of patients with COVID-19 acute respiratory distress syndrome. J Thromb Haemost 2020; 18 (07) 1747-1751
  • 45 Magro C, Mulvey JJ, Berlin D. et al. Complement associated microvascular injury and thrombosis in the pathogenesis of severe COVID-19 infection: a report of five cases. Transl Res 2020; 220: 1-13
  • 46 Carvelli J, Demaria O, Vély F. et al; Explore COVID-19 IPH group, Explore COVID-19 Marseille Immunopole group. Association of COVID-19 inflammation with activation of the C5a-C5aR1 axis. Nature 2020; 588 (7836): 146-150
  • 47 Leisman DE, Deutschman CS, Legrand M. Facing COVID-19 in the ICU: vascular dysfunction, thrombosis, and dysregulated inflammation. Intensive Care Med 2020; 46 (06) 1105-1108
  • 48 Libby P, Lüscher T. COVID-19 is, in the end, an endothelial disease. Eur Heart J 2020; 41 (32) 3038-3044
  • 49 Janiuk K, Jabłońska E, Garley M. Significance of NETs formation in COVID-19. Cells 2021; 10 (01) 151
  • 50 Middleton EA, He X-Y, Denorme F. et al. Neutrophil extracellular traps contribute to immunothrombosis in COVID-19 acute respiratory distress syndrome. Blood 2020; 136 (10) 1169-1179
  • 51 Leppkes M, Knopf J, Naschberger E. et al. Vascular occlusion by neutrophil extracellular traps in COVID-19. EBioMedicine 2020; 58: 102925
  • 52 Nicolai L, Leunig A, Brambs S. et al. Immunothrombotic dysregulation in COVID-19 pneumonia is associated with respiratory failure and coagulopathy. Circulation 2020; 142 (12) 1176-1189
  • 53 Krauel K, Duerschmied D. ICODE: the international COVID-19 thrombosis biomarkers colloquium-novel soluble biomarkers: circulating cell-free nucleic acids and other molecules. J Thromb Thrombolysis 2021 May 12:1–5. doi: 10.1007/s11239-021-02468-6. Epub ahead of print.
  • 54 Zuo Y, Zuo M, Yalavarthi S. et al. Neutrophil extracellular traps and thrombosis in COVID-19. J Thromb Thrombolysis 2021; 51 (02) 446-453
  • 55 Petito E, Falcinelli E, Paliani U. et al; COVIR study investigators. Association of neutrophil activation, more than platelet activation, with thrombotic complications in coronavirus disease 2019. J Infect Dis 2021; 223 (06) 933-944
  • 56 Skendros P, Mitsios A, Chrysanthopoulou A. et al. Complement and tissue factor-enriched neutrophil extracellular traps are key drivers in COVID-19 immunothrombosis. J Clin Invest 2020; 130 (11) 6151-6157
  • 57 Hasan Ali O, Bomze D, Risch L. et al. Severe COVID-19 is associated with elevated serum IgA and antiphospholipid IgA-antibodies. Clin Infect Dis 2020; ciaa1496: ciaa1496
  • 58 Xiao M, Zhang Y, Zhang S. et al. Antiphospholipid antibodies in critically ill patients with COVID-19. Arthritis Rheumatol 2020; 72 (12) 1998-2004
  • 59 Borghi MO, Beltagy A, Garrafa E. et al. Anti-phospholipid antibodies in COVID-19 are different from those detectable in the anti-phospholipid syndrome. Front Immunol 2020; 11: 584241
  • 60 Gkrouzman E, Barbhaiya M, Erkan D, Lockshin MD. Reality check on antiphospholipid antibodies in COVID-19-associated coagulopathy. Arthritis Rheumatol 2021; 73 (01) 173-174
  • 61 Violi F, Cammisotto V, Pignatelli P. Thrombosis in Covid-19 and non-Covid-19 pneumonia: role of platelets. Platelets 2021 Jun 7:1-9. doi: 10.1080/09537104.2021.1936478. Epub ahead of print.
  • 62 Bonaventura A, Vecchié A, Dagna L. et al. Endothelial dysfunction and immunothrombosis as key pathogenic mechanisms in COVID-19. Nat Rev Immunol 2021; 21 (05) 319-329

Zoom Image
Fig. 1 Cellular players and soluble mediators driving immunothrombosis and thromboinflammation.