CC BY-NC-ND 4.0 · Thromb Haemost 2022; 122(02): 240-256
DOI: 10.1055/s-0041-1740182
Blood Cells, Inflammation and Infection

Associations between the von Willebrand Factor—ADAMTS13 Axis, Complement Activation, and COVID-19 Severity and Mortality

1   Department of Internal Medicine and Haematology, Semmelweis University, Budapest, Hungary
,
Marienn Réti
2   Department of Haematology and Stem Cell Transplantation, Central Hospital of Southern Pest, Institute of Haematology and Infectious Diseases, Budapest, Hungary
,
Veronika Müller
3   Department of Pulmonology, Semmelweis University, Budapest, Hungary
,
Zsolt Iványi
4   Department of Anaesthesiology and Intensive Therapy, Semmelweis University, Budapest, Hungary
,
János Gál
4   Department of Anaesthesiology and Intensive Therapy, Semmelweis University, Budapest, Hungary
,
László Gopcsa
2   Department of Haematology and Stem Cell Transplantation, Central Hospital of Southern Pest, Institute of Haematology and Infectious Diseases, Budapest, Hungary
,
Péter Reményi
2   Department of Haematology and Stem Cell Transplantation, Central Hospital of Southern Pest, Institute of Haematology and Infectious Diseases, Budapest, Hungary
,
Beáta Szathmáry
5   Department of Infectology, Central Hospital of Southern Pest, Institute of Haematology and Infectious Diseases, Budapest, Hungary
,
Botond Lakatos
5   Department of Infectology, Central Hospital of Southern Pest, Institute of Haematology and Infectious Diseases, Budapest, Hungary
,
János Szlávik
5   Department of Infectology, Central Hospital of Southern Pest, Institute of Haematology and Infectious Diseases, Budapest, Hungary
,
Ilona Bobek
6   Department of Anaesthesiology and Intensive Therapy, Central Hospital of Southern Pest, Institute of Haematology and Infectious Diseases, Budapest, Hungary
,
Zita Z. Prohászka
1   Department of Internal Medicine and Haematology, Semmelweis University, Budapest, Hungary
,
Zsolt Förhécz
1   Department of Internal Medicine and Haematology, Semmelweis University, Budapest, Hungary
,
Blanka Mező
1   Department of Internal Medicine and Haematology, Semmelweis University, Budapest, Hungary
7   Research Group for Immunology and Haematology, Semmelweis University – Eötvös Loránd Research Network (Office for Supported Research Groups), Budapest, Hungary
,
1   Department of Internal Medicine and Haematology, Semmelweis University, Budapest, Hungary
,
1   Department of Internal Medicine and Haematology, Semmelweis University, Budapest, Hungary
,
Erika Kajdácsi
1   Department of Internal Medicine and Haematology, Semmelweis University, Budapest, Hungary
,
László Cervenak
1   Department of Internal Medicine and Haematology, Semmelweis University, Budapest, Hungary
,
Petra Kiszel
7   Research Group for Immunology and Haematology, Semmelweis University – Eötvös Loránd Research Network (Office for Supported Research Groups), Budapest, Hungary
,
Tamás Masszi
1   Department of Internal Medicine and Haematology, Semmelweis University, Budapest, Hungary
,
István Vályi-Nagy*
2   Department of Haematology and Stem Cell Transplantation, Central Hospital of Southern Pest, Institute of Haematology and Infectious Diseases, Budapest, Hungary
,
Zoltán Prohászka*
1   Department of Internal Medicine and Haematology, Semmelweis University, Budapest, Hungary
7   Research Group for Immunology and Haematology, Semmelweis University – Eötvös Loránd Research Network (Office for Supported Research Groups), Budapest, Hungary
› Author Affiliations
Funding The research was financed by the Higher Education Institutional Excellence Programme of the Ministry of Human Capacities in Hungary, within the framework of the molecular biology thematic programme of the Semmelweis University, by the National Office for Innovation and Research (KH130355, and 2020-1.1.6-JOVO-2021-00013 “JOVO” to Z.P.). The study was performed in frame of the Premium Postdoctoral Fellowship Program of the Hungarian Academy of Sciences (PPD2018-016/2018 to D.C.). Z.P. and L.H. are supported by funds of the EU MSCA project CORVOS 860044.


Abstract

Background Endothelial and complement activation were both associated with immunothrombosis, a key determinant of COVID-19 severity, but their interrelation has not yet been investigated.

Objectives We aimed to determine von Willebrand factor (VWF) antigen (VWF:Ag) concentration, VWF collagen binding activity (VWF:CBA), a disintegrin and metalloproteinase with a thrombospondin type 1 motif, member 13 (ADAMTS13) activity (ADAMTS13:Ac), and their ratios in hospitalized COVID-19 patients, and to investigate how these parameters and their constellation with complement activation relate to disease severity and in-hospital mortality in COVID-19.

Methods Samples of 102 hospitalized patients with polymerase chain reaction-confirmed severe acute respiratory syndrome coronavirus 2 positivity were included in our observational cohort study. Patients were stratified according to the peak severity of COVID-19 disease in agreement with the World Health Organization ordinal scale. Twenty-six convalescent plasma donors with previous COVID-19 disease formed the control group. VWF:Ag concentration and VWF:CBA were determined by enzyme-linked immunosorbent assay (ELISA); ADAMTS13:Ac was determined by fluorescence resonance energy transfer. Complement C3 and C3a were measured by turbidimetry and ELISA, respectively. Clinical covariates and markers of inflammation were extracted from hospital records.

Results VWF:Ag and VWF:CBA were elevated in all groups of hospitalized COVID-19 patients and increased in parallel with disease severity. ADAMTS13:Ac was decreased in patients with severe COVID-19, with the lowest values in nonsurvivors. High (> 300%) VWF:Ag concentrations or decreased (< 67%) ADAMTS13:Ac were associated with higher risk of severe COVID-19 disease or in-hospital mortality. The concomitant presence of decreased ADAMTS13:Ac and increased C3a/C3 ratio—indicating complement overactivation and consumption—was a strong independent predictor of in-hospital mortality.

Conclusion Our results suggest that an interaction between the VWF-ADAMTS13 axis and complement overactivation and consumption plays an important role in the pathogenesis of COVID-19.

Author Contributions

G.S., B.M., D.C., L.H., E.K., L.C., and P.K. designed and performed laboratory determinations, interpreted data, and drafted the manuscript. G.S., Z.Z.P., and Z.P. conceptualized research, collected and analyzed clinical information and laboratory data, conducted statistical analysis, interpreted data, and wrote the manuscript. M.R., V.M., Z.F., Z.I., J.G., L.G., P.R., B.S., B.L., J.S., I.B., T.M., I.V.-N. took part in the conceptualization, collected and analyzed clinical information, interpreted and supervised data, and drafted the manuscript. All authors critically revised the final manuscript.


* Shared authorship.


Supplementary Material



Publication History

Received: 30 June 2021

Accepted: 10 October 2021

Article published online:
21 January 2022

© 2022. The Author(s). This is an open access article published by Thieme under the terms of the Creative Commons Attribution-NonDerivative-NonCommercial License, permitting copying and reproduction so long as the original work is given appropriate credit. Contents may not be used for commercial purposes, or adapted, remixed, transformed or built upon. (https://creativecommons.org/licenses/by-nc-nd/4.0/)

Georg Thieme Verlag KG
Rüdigerstraße 14, 70469 Stuttgart, Germany

 
  • References

  • 1 Guan WJ, Ni ZY, Hu Y. et al; China Medical Treatment Expert Group for Covid-19. Clinical characteristics of coronavirus disease 2019 in China. N Engl J Med 2020; 382 (18) 1708-1720
  • 2 Zhou F, Yu T, Du R. et al. Clinical course and risk factors for mortality of adult inpatients with COVID-19 in Wuhan, China: a retrospective cohort study. Lancet 2020; 395 (10229): 1054-1062
  • 3 Chen G, Wu D, Guo W. et al. Clinical and immunological features of severe and moderate coronavirus disease 2019. J Clin Invest 2020; 130 (05) 2620-2629
  • 4 Moore JB, June CH. Cytokine release syndrome in severe COVID-19. Science 2020; 368 (6490): 473-474
  • 5 Helms J, Tacquard C, Severac F. et al; CRICS TRIGGERSEP Group (Clinical Research in Intensive Care and Sepsis Trial Group for Global Evaluation and Research in Sepsis). High risk of thrombosis in patients with severe SARS-CoV-2 infection: a multicenter prospective cohort study. Intensive Care Med 2020; 46 (06) 1089-1098
  • 6 Levi M, Thachil J, Iba T, Levy JH. Coagulation abnormalities and thrombosis in patients with COVID-19. Lancet Haematol 2020; 7 (06) e438-e440
  • 7 Escher R, Breakey N, Lämmle B. Severe COVID-19 infection associated with endothelial activation. Thromb Res 2020; 190: 62
  • 8 Goshua G, Pine AB, Meizlish ML. et al. Endotheliopathy in COVID-19-associated coagulopathy: evidence from a single-centre, cross-sectional study. Lancet Haematol 2020; 7 (08) e575-e582
  • 9 Rauch A, Labreuche J, Lassalle F. et al. Coagulation biomarkers are independent predictors of increased oxygen requirements in COVID-19. J Thromb Haemost 2020; 18 (11) 2942-2953
  • 10 Escher R, Breakey N, Lämmle B. ADAMTS13 activity, von Willebrand factor, factor VIII and D-dimers in COVID-19 inpatients. Thromb Res 2020; 192: 174-175
  • 11 Bazzan M, Montaruli B, Sciascia S, Cosseddu D, Norbiato C, Roccatello D. Low ADAMTS 13 plasma levels are predictors of mortality in COVID-19 patients. Intern Emerg Med 2020; 15 (05) 861-863
  • 12 Huisman A, Beun R, Sikma M, Westerink J, Kusadasi N. Involvement of ADAMTS13 and von Willebrand factor in thromboembolic events in patients infected with SARS-CoV-2. Int J Lab Hematol 2020; 42 (05) e211-e212
  • 13 Martinelli N, Montagnana M, Pizzolo F. et al. A relative ADAMTS13 deficiency supports the presence of a secondary microangiopathy in COVID 19. Thromb Res 2020; 193: 170-172
  • 14 Tiscia GL, Favuzzi G, De Laurenzo A. et al; CSS COVID-19 Group. Reduction of ADAMTS13 levels predicts mortality in SARS-CoV-2 patients. TH Open 2020; 4 (03) e203-e206
  • 15 Delrue M, Siguret V, Neuwirth M. et al. von Willebrand factor/ADAMTS13 axis and venous thromboembolism in moderate-to-severe COVID-19 patients. Br J Haematol 2021; 192 (06) 1097-1100
  • 16 Mancini I, Baronciani L, Artoni A. et al. The ADAMTS13-von Willebrand factor axis in COVID-19 patients. J Thromb Haemost 2021; 19 (02) 513-521
  • 17 Favaloro EJ, Henry BM, Lippi G. Increased VWF and decreased ADAMTS-13 in COVID-19: creating a milieu for (micro)thrombosis. Semin Thromb Hemost 2021; 47 (04) 400-418
  • 18 Philippe A, Chocron R, Gendron N. et al. Circulating Von Willebrand factor and high molecular weight multimers as markers of endothelial injury predict COVID-19 in-hospital mortality. Angiogenesis 2021; 24 (03) 505-517
  • 19 Rodríguez Rodríguez M, Castro Quismondo N, Zafra Torres D, Gil Alos D, Ayala R, Martinez-Lopez J. Increased von Willebrand factor antigen and low ADAMTS13 activity are related to poor prognosis in covid-19 patients. Int J Lab Hematol 2021; 43 (04) O152-O155
  • 20 Sweeney JM, Barouqa M, Krause GJ. et al. Evidence for secondary thrombotic microangiopathy in COVID-19. medRxiv 2020; DOI: 10.1101/2020.10.20.20215608.
  • 21 von Meijenfeldt FA, Havervall S, Adelmeijer J. et al. Prothrombotic changes in patients with COVID-19 are associated with disease severity and mortality. Res Pract Thromb Haemost 2020; 5 (01) 132-141
  • 22 Cugno M, Meroni PL, Gualtierotti R. et al. Complement activation and endothelial perturbation parallel COVID-19 severity and activity. J Autoimmun 2021; 116: 102560
  • 23 Ward SE, Curley GF, Lavin M. et al; Irish COVID-19 Vasculopathy Study (ICVS) Investigators. Von Willebrand factor propeptide in severe coronavirus disease 2019 (COVID-19): evidence of acute and sustained endothelial cell activation. Br J Haematol 2021; 192 (04) 714-719
  • 24 Varga Z, Flammer AJ, Steiger P. et al. Endothelial cell infection and endotheliitis in COVID-19. Lancet 2020; 395 (10234): 1417-1418
  • 25 Neubauer K, Zieger B. Endothelial cells and coagulation. Cell Tissue Res 2021; DOI: 10.1007/s00441-021-03471-2.
  • 26 Ekdahl KN, Teramura Y, Hamad OA. et al. Dangerous liaisons: complement, coagulation, and kallikrein/kinin cross-talk act as a linchpin in the events leading to thromboinflammation. Immunol Rev 2016; 274 (01) 245-269
  • 27 de Nooijer AH, Grondman I, Janssen NAF. et al; RCI-COVID-19 study group. Complement activation in the disease course of coronavirus disease 2019 and its effects on clinical outcomes. J Infect Dis 2021; 223 (02) 214-224
  • 28 Holter JC, Pischke SE, de Boer E. et al. Systemic complement activation is associated with respiratory failure in COVID-19 hospitalized patients. Proc Natl Acad Sci U S A 2020; 117 (40) 25018-25025
  • 29 Sinkovits G, Mező B, Réti M. et al. Complement overactivation and consumption predicts in-hospital mortality in SARS-CoV-2 infection. Front Immunol 2021; 12: 663187
  • 30 Ward PA. The harmful role of c5a on innate immunity in sepsis. J Innate Immun 2010; 2 (05) 439-445
  • 31 Gombos T, Makó V, Cervenak L. et al. Levels of von Willebrand factor antigen and von Willebrand factor cleaving protease (ADAMTS13) activity predict clinical events in chronic heart failure. Thromb Haemost 2009; 102 (03) 573-580
  • 32 Czúcz J, Schaffer G, Csuka D. et al. Endothelial cell function in patients with hereditary angioedema: elevated soluble E-selectin level during inter-attack periods. J Clin Immunol 2012; 32 (01) 61-69
  • 33 Maharaj S, Xue R, Rojan A. Thrombotic thrombocytopenic purpura (TTP) response following COVID-19 infection: Implications for the ADAMTS-13-von Willebrand factor axis. J Thromb Haemost 2021; 19 (04) 1130-1132
  • 34 Blasi A, von Meijenfeldt FA, Adelmeijer J. et al. In vitro hypercoagulability and ongoing in vivo activation of coagulation and fibrinolysis in COVID-19 patients on anticoagulation. J Thromb Haemost 2020; 18 (10) 2646-2653
  • 35 Henry BM, Benoit SW, de Oliveira MHS, Lippi G, Favaloro EJ, Benoit JL. ADAMTS13 activity to von Willebrand factor antigen ratio predicts acute kidney injury in patients with COVID-19: evidence of SARS-CoV-2 induced secondary thrombotic microangiopathy. Int J Lab Hematol 2021; 43 (Suppl. 01) 129-136
  • 36 De Cristofaro R, Liuzzo G, Sacco M, Lancellotti S, Pedicino D, Andreotti F. Marked von Willebrand factor and factor VIII elevations in severe acute respiratory syndrome coronavirus-2-positive, but not severe acute respiratory syndrome coronavirus-2-negative, pneumonia: a case-control study. Blood Coagul Fibrinolysis 2021; 32 (04) 285-289
  • 37 Joly BS, Darmon M, Dekimpe C. et al. Imbalance of von Willebrand factor and ADAMTS13 axis is rather a biomarker of strong inflammation and endothelial damage than a cause of thrombotic process in critically ill COVID-19 patients. J Thromb Haemost 2021; 19 (09) 2193-2198
  • 38 Doevelaar AAN, Bachmann M, Hölzer B. et al. von Willebrand factor multimer formation contributes to immunothrombosis in coronavirus disease 2019. Crit Care Med 2021; 49 (05) e512-e520
  • 39 Pascreau T, Zia-Chahabi S, Zuber B, Tcherakian C, Farfour E, Vasse M. ADAMTS 13 deficiency is associated with abnormal distribution of von Willebrand factor multimers in patients with COVID-19. Thromb Res 2021; 204: 138-140
  • 40 Pinsky DJ, Naka Y, Liao H. et al. Hypoxia-induced exocytosis of endothelial cell Weibel-Palade bodies. A mechanism for rapid neutrophil recruitment after cardiac preservation. J Clin Invest 1996; 97 (02) 493-500
  • 41 Bashir DA, Da Q, Pradhan S. et al. Secretion of von Willebrand factor and suppression of ADAMTS-13 activity by markedly high concentration of ferritin. Clin Appl Thromb Hemost 2021; 27: 1076029621992128
  • 42 Tomar B, Anders HJ, Desai J, Mulay SR. Neutrophils and neutrophil extracellular traps drive necroinflammation in COVID-19. Cells 2020; 9 (06) E1383
  • 43 Zuo Y, Yalavarthi S, Shi H. et al. Neutrophil extracellular traps in COVID-19. JCI Insight 2020; 5 (11) 138999
  • 44 Yang J, Wu Z, Long Q. et al. Insights into immunothrombosis: the interplay among neutrophil extracellular trap, von Willebrand factor, and ADAMTS13. Front Immunol 2020; 11: 610696
  • 45 Sorvillo N, Mizurini DM, Coxon C. et al. Plasma peptidylarginine deiminase IV promotes VWF-platelet string formation and accelerates thrombosis after vessel injury. Circ Res 2019; 125 (05) 507-519
  • 46 Wang Y, Chen J, Ling M, López JA, Chung DW, Fu X. Hypochlorous acid generated by neutrophils inactivates ADAMTS13: an oxidative mechanism for regulating ADAMTS13 proteolytic activity during inflammation. J Biol Chem 2015; 290 (03) 1422-1431
  • 47 Pillai VG, Bao J, Zander CB. et al. Human neutrophil peptides inhibit cleavage of von Willebrand factor by ADAMTS13: a potential link of inflammation to TTP. Blood 2016; 128 (01) 110-119
  • 48 Ward SE, Fogarty H, Karampini E. et al; Irish COVID-19 Vasculopathy Study (iCVS) investigators. ADAMTS13 regulation of VWF multimer distribution in severe COVID-19. J Thromb Haemost 2021; 19 (08) 1914-1921
  • 49 Dong JF, Moake JL, Nolasco L. et al. ADAMTS-13 rapidly cleaves newly secreted ultralarge von Willebrand factor multimers on the endothelial surface under flowing conditions. Blood 2002; 100 (12) 4033-4039
  • 50 Bettoni S, Galbusera M, Gastoldi S. et al. Interaction between multimeric von Willebrand factor and complement: a fresh look to the pathophysiology of microvascular thrombosis. J Immunol 2017; 199 (03) 1021-1040
  • 51 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
  • 52 Cugno M, Meroni PL, Gualtierotti R. et al. Complement activation in patients with COVID-19: a novel therapeutic target. J Allergy Clin Immunol 2020; 146 (01) 215-217
  • 53 Morigi M, Galbusera M, Gastoldi S. et al. Alternative pathway activation of complement by Shiga toxin promotes exuberant C3a formation that triggers microvascular thrombosis. J Immunol 2011; 187 (01) 172-180
  • 54 Ikeda K, Nagasawa K, Horiuchi T, Tsuru T, Nishizaka H, Niho Y. C5a induces tissue factor activity on endothelial cells. Thromb Haemost 1997; 77 (02) 394-398
  • 55 Tedesco F, Pausa M, Nardon E, Introna M, Mantovani A, Dobrina A. The cytolytically inactive terminal complement complex activates endothelial cells to express adhesion molecules and tissue factor procoagulant activity. J Exp Med 1997; 185 (09) 1619-1627
  • 56 Conway EM. Thrombomodulin and its role in inflammation. Semin Immunopathol 2012; 34 (01) 107-125
  • 57 Klos A, Tenner AJ, Johswich KO, Ager RR, Reis ES, Köhl J. The role of the anaphylatoxins in health and disease. Mol Immunol 2009; 46 (14) 2753-2766
  • 58 Kawecki C, Lenting PJ, Denis CV. von Willebrand factor and inflammation. J Thromb Haemost 2017; 15 (07) 1285-1294
  • 59 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
  • 60 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