The vascular endothelium: the cornerstone of organ dysfunction in severe SARS-CoV-2 infection

Stéphanie Pons, Sofiane Fodil, Elie Azoulay, Lara Zafrani, Stéphanie Pons, Sofiane Fodil, Elie Azoulay, Lara Zafrani

Abstract

In severe SARS-CoV-2 infections, emerging data including recent histopathological studies have emphasized the crucial role of endothelial cells (ECs) in vascular dysfunction, immunothrombosis, and inflammation.Histopathological studies have evidenced direct viral infection of ECs, endotheliitis with diffuse endothelial inflammation, and micro- and macrovascular thrombosis both in the venous and arterial circulations. Venous thrombotic events, particularly pulmonary embolism, with elevated D-dimer and coagulation activation are highly prevalent in COVID-19 patients. The pro-inflammatory cytokine storm, with elevated levels of interleukin-6 (IL-6), IL-2 receptor, and tumor necrosis factor-α, could also participate in endothelial dysfunction and leukocyte recruitment in the microvasculature. COVID-19-induced endotheliitis may explain the systemic impaired microcirculatory function in different organs in COVID-19 patients. Ongoing trials directly and indirectly target COVID-19-related endothelial dysfunctions: i.e., a virus-cell entry using recombinant angiotensin-converting enzyme 2 (ACE2) and transmembrane protease serine 2 (TMPRSS-2) blockade, coagulation activation, and immunomodulatory therapies, such as anti-IL-6 strategies. Studies focusing on endothelial dysfunction in COVID-19 patients are warranted as to decipher their precise role in severe SARS-CoV-2 infection and organ dysfunction and to identify targets for further interventions.

Keywords: COVID-19; Cytokines; Endothelial cells; Endothelial dysfunction; SARS-CoV-2; Thrombosis.

Conflict of interest statement

SP received a research grant from the French Intensive Care Society, one from the European Society of Intensive Care Medicine, and one from the Zoll foundation. EA has received fees for lectures from MSD, Pfizer, and Alexion. His institution and research group have received support from Baxter, Jazz Pharma, Fisher&Payckle, Gilead, Alexion, and Ablynx. LZ received a research grant from Jazz Pharma.

Figures

Fig. 1
Fig. 1
The role of endothelial cells in SARS-CoV-2 infection and treatment. a Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) binds with Angiotensin- converting enzyme 2 (ACE2) on the cell membrane of the host cells. Cell invasion also depends on the presence of the protease Transmembrane protease serine 2 (TMPRSS-2) that is able to cleave the viral spike. The recombinant protein of human ACE2 fused with the Fc region of the human immunoglobulin IgG1 (rACE2-IgG1) binds with high affinity to the receptor-binding domain of SARS-CoV-2. Moreover, a protease TMPRSS2 inhibitor is efficient to block SARS-CoV-2 entry into the endothelial cells (ECs). b In patients diagnosed with severe COVID-19, increased levels of pro-inflammatory cytokines, in particular, the soluble interleukin 2-receptor (IL-2R) and interleukin-6 (IL-6) have been observed. ECs express both IL-6 receptor (IL-6R) and IL-2R on their surface. Soluble IL-2R (sIL-2R) is mostly secreted by activated T helper lymphocytes, but might be also secreted by ECs. Binding of IL-6 and IL-2 on their receptors induces a capillary leak. Moreover, IL-6 signaling induces the secretion by ECs of more IL-6 and other cytokines. Tocilizumab, a humanized anti-IL-6 receptor (IL-6R) antibody that inhibits signal transduction by binding sIL-6R and membrane-bound IL-6R, has emerged as a potential immunomodulatory treatment in COVID-19 patients. c During SARS-CoV-2 infection, endothelial dysfunction and microthrombi formation may be secondary to complement activation and membrane attack complexes deposits on ECs. Eculizumab, a human monoclonal antibody designed to bind to the complement protein C5 with high affinity prevents the generation of the terminal membrane attack complex. EC activation also induces a pro-coagulant state by increasing their production of von Willebrand factor (vWF) and factor VIII (FVIII), which participate in clot formation. Heparin from prophylactic to therapeutic doses has been used in COVID-19 patients to inhibit clot formation. Moreover, during SARS-CoV-2 infection, ECs increase their production of plasminogen activator inhibitor 1 (PAI-1), which inhibits the conversion of plasminogen to plasmin and the degradation of clots inducing a hypofibrinolytic state. Tissue plasminogen activator (t-PA) has been tested in COVID-19 patients for its fibrinolytic effect. ACE2: angiotensin-converting enzyme 2; C5: complement C5; FVIII: factor VIII; FDP: fibrin degradation product; IL-2: interleukin-2; IL-6: interleukin-6; IL-2R: interleukin-2 receptor; IL-6-R: interleukin-6 receptor; MAC: membrane attack complex; PAI 1: plasminogen activator inhibitor 1; rACE2-IgG1: recombinant angiotensin-converting enzyme 2 immunoglobulin G1; SARS-Cov-2: severe acute respiratory syndrome coronavirus 2; sIL-2R: soluble interleukin 2 receptor, sIL-6R: soluble interleukin 6 receptor; Th L: T helper lymphocyte; TMPRSS-2: transmembrane protease serine 2 t-PA: tissue plasminogen activator; vWF: von Willebrand factor

References

    1. Zhu J, Ji P, Pang J, Zhong Z, Li H, He C, et al. Clinical characteristics of 3,062 COVID-19 patients: a meta-analysis. J Med Virol. 2020 [cited 2020 May 5]; Available from: .
    1. Marini JJ, Gattinoni L. Management of COVID-19 respiratory distress. JAMA. 2020 [cited 2020 May 5]; Available from: .
    1. Roumenina LT, Rayes J, Frimat M, Fremeaux-Bacchi V. Endothelial cells: source, barrier, and target of defensive mediators. Immunol Rev. 2016;274:307–329.
    1. Daniel AE, van Buul JD. Endothelial junction regulation: a prerequisite for leukocytes crossing the vessel wall. J Innate Immun. 2013;5:324–335.
    1. McKenzie JAG, Ridley AJ. Roles of Rho/ROCK and MLCK in TNF-alpha-induced changes in endothelial morphology and permeability. J Cell Physiol. 2007;213:221–228.
    1. Ley K, Laudanna C, Cybulsky MI, Nourshargh S. Getting to the site of inflammation: the leukocyte adhesion cascade updated. Nat Rev Immunol. 2007;7:678–689.
    1. Taflin C, Favier B, Baudhuin J, Savenay A, Hemon P, Bensussan A, et al. Human endothelial cells generate Th17 and regulatory T cells under inflammatory conditions. Proc Natl Acad Sci U S A. 2011;108:2891–2896.
    1. Teijaro JR, Walsh KB, Cahalan S, Fremgen DM, Roberts E, Scott F, et al. Endothelial cells are central orchestrators of cytokine amplification during influenza virus infection. Cell. 2011;146:980–991.
    1. Pober JS, Merola J, Liu R, Manes TD. Antigen presentation by vascular cells. Front Immunol. 2017;8:1907.
    1. Ma W, Pober JS. Human endothelial cells effectively costimulate cytokine production by, but not differentiation of, naive CD4+ T cells. J Immunol Baltim Md 1950. 1998;161:2158–2167.
    1. Perez VL, Henault L, Lichtman AH. Endothelial antigen presentation: stimulation of previously activated but not naïve TCR-transgenic mouse T cells. Cell Immunol. 1998;189:31–40.
    1. Sage PT, Varghese LM, Martinelli R, Sciuto TE, Kamei M, Dvorak AM, et al. Antigen recognition is facilitated by invadosome-like protrusions formed by memory/effector T cells. J Immunol Baltim Md 1950. 2012;188:3686–3699.
    1. Lin G-L, McGinley JP, Drysdale SB, Pollard AJ. Epidemiology and immune pathogenesis of viral sepsis. Front Immunol. 2018;9:2147.
    1. Varga Z, Flammer AJ, Steiger P, Haberecker M, Andermatt R, Zinkernagel AS, et al. Endothelial cell infection and endotheliitis in COVID-19. Lancet. 2020;395:1417–1418.
    1. CRICS TRIGGERSEP Group (Clinical Research in Intensive Care and Sepsis Trial Group for Global Evaluation and Research in Sepsis), Helms J, Tacquard C, Severac F, Leonard-Lorant I, Ohana M, et al. High risk of thrombosis in patients with severe SARS-CoV-2 infection: a multicenter prospective cohort study. Intensive Care Med. 2020 [cited 2020 May 5]; Available from: .
    1. Monteil V, Kwon H, Prado P, Hagelkrüys A, Wimmer RA, Stahl M, et al. IInhibition of SARS-CoV-2 Infections in Engineered Human Tissues Using Clinical-Grade Soluble Human ACE2. Cell. 2020;181(4):905‐913.e7. 10.1016/j.cell.2020.04.004.
    1. Pons S, Arnaud M, Loiselle M, Arrii E, Azoulay E, Zafrani L. Immune consequences of endothelial cells’ activation and dysfunction during sepsis. Crit Care Clin. 2020;36:401–413.
    1. McGonagle D, O'Donnell JS, Sharif K, Emery P, Bridgewood C. Immune mechanisms of pulmonary intravascular coagulopathy in COVID-19 pneumonia [published online ahead of print, 2020 May 7]. Lancet Rheumatol. 2020. 10.1016/S2665-9913(20)30121-1.
    1. Su H, Yang M, Wan C, Yi L-X, Tang F, Zhu H-Y, et al. Renal histopathological analysis of 26 postmortem findings of patients with COVID-19 in China [published online ahead of print, 2020 Apr 9]. Kidney Int. 2020;S0085-2538(20)30369-0. 10.1016/j.kint.2020.04.003.
    1. Copin MC, Parmentier E, Duburcq T, Poissy J, Mathieu D; Lille COVID-19 ICU and Anatomopathology Group. Time to consider histologic pattern of lung injury to treat critically ill patients with COVID-19 infection. Intensive Care Med. 2020;1–3. 10.1007/s00134-020-06057-8.
    1. Hamming I, Timens W, Bulthuis M, Lely A, Navis G, van Goor H. Tissue distribution of ACE2 protein, the functional receptor for SARS coronavirus. A first step in understanding SARS pathogenesis. J Pathol. 2004;203:631–637.
    1. Ou X, Liu Y, Lei X, Li P, Mi D, Ren L, et al. Characterization of spike glycoprotein of SARS-CoV-2 on virus entry and its immune cross-reactivity with SARS-CoV. Nat Commun. 2020;11:1620.
    1. Hoffmann M, Kleine-Weber H, Schroeder S, Krüger N, Herrler T, Erichsen S, et al. SARS-CoV-2 Cell Entry Depends on ACE2 and TMPRSS2 and Is Blocked by a Clinically Proven Protease Inhibitor. Cell. 2020;181(2):271–280.e8. 10.1016/j.cell.2020.02.052.
    1. Aimes RT, Zijlstra A, Hooper JD, Ogbourne SM, Sit M-L, Fuchs S, et al. Endothelial cell serine proteases expressed during vascular morphogenesis and angiogenesis. Thromb Haemost. 2003;89:561–572.
    1. Pober JS, Sessa WC. Evolving functions of endothelial cells in inflammation. Nat Rev Immunol. 2007;7:803–815.
    1. Betakova T, Kostrabova A, Lachova V, Turianova L. Cytokines induced during influenza virus infection. Curr Pharm Des. 2017 [cited 2020 May 5];23. Available from: .
    1. Channappanavar R, Perlman S. Pathogenic human coronavirus infections: causes and consequences of cytokine storm and immunopathology. Semin Immunopathol. 2017;39:529–539.
    1. Chen G, Wu D, Guo W, Cao Y, Huang D, Wang H, et al. Clinical and immunological features of severe and moderate coronavirus disease 2019. J Clin Invest. 2020;130:2620–2629.
    1. Leisman DE, Deutschman CS, Legrand M. Facing COVID-19 in the ICU: vascular dysfunction, thrombosis, and dysregulated inflammation. Intensive Care Med. 2020 [cited 2020 May 7]; Available from: .
    1. Sinha P, Delucchi KL, McAuley DF, O’Kane CM, Matthay MA, Calfee CS. Development and validation of parsimonious algorithms to classify acute respiratory distress syndrome phenotypes: a secondary analysis of randomised controlled trials. Lancet Respir Med. 2020;8:247–257.
    1. Maude SL, Frey N, Shaw PA, Aplenc R, Barrett DM, Bunin NJ, et al. Chimeric antigen receptor T cells for sustained remissions in leukemia. N Engl J Med. 2014;371:1507–1517.
    1. Jirik FR, Podor TJ, Hirano T, Kishimoto T, Loskutoff DJ, Carson DA, et al. Bacterial lipopolysaccharide and inflammatory mediators augment IL-6 secretion by human endothelial cells. J Immunol Baltim Md 1950. 1989;142:144–147.
    1. Baqui AA, Meiller TF, Chon JJ, Turng BF, Falkler WA. Interleukin-6 production by human monocytes treated with granulocyte-macrophage colony-stimulating factor in the presence of lipopolysaccharide of oral microorganisms. Oral Microbiol Immunol. 1998;13:173–180.
    1. Akira S, Kishimoto+ T. IL-6 and NF-IL6 in acute-phase response and viral infection. Immunol Rev 1992;127:25–50.
    1. Cui R, Tieu B, Recinos A, Tilton RG, Brasier AR. RhoA mediates angiotensin II–induced phospho-Ser536 nuclear factor κB/RelA subunit exchange on the interleukin-6 promoter in VSMCs. Circ Res. 2006;99:723–730.
    1. Hunter CA, Jones SA. IL-6 as a keystone cytokine in health and disease. Nat Immunol. 2015;16:448–457.
    1. Desai TR, Leeper NJ, Hynes KL, Gewertz BL. Interleukin-6 causes endothelial barrier dysfunction via the protein kinase C pathway. J Surg Res. 2002;104:118–123.
    1. Laudes IJ, Chu JC, Huber-Lang M, Guo R-F, Riedemann NC, Sarma JV, et al. Expression and function of C5a receptor in mouse microvascular endothelial cells. J Immunol. 2002;169:5962–5970.
    1. Chen X, Zhao B, Qu Y, Chen Y, Xiong J, Feng Y, et al. Detectable serum SARS-CoV-2 viral load (RNAaemia) is closely correlated with drastically elevated interleukin 6 (IL-6) level in critically ill COVID-19 patients. Clin Infect Dis. 2020;ciaa449.
    1. Ruan Q, Yang K, Wang W, Jiang L, Song J. Clinical predictors of mortality due to COVID-19 based on an analysis of data of 150 patients from Wuhan, China. Intensive Care Med. 2020 [cited 2020 May 5]; Available from: .
    1. Aziz M, Fatima R, Assaly R. Elevated interleukin-6 and severe COVID-19: A meta-analysis [published online ahead of print. J Med Virol. 2020;10.1002/jmv.25948. 10.1002/jmv.25948.
    1. Luo P, Liu Y, Qiu L, Liu X, Liu D, Li J. Tocilizumab treatment in COVID-19: A single center experience. J Med Virol. 2020;92(7):814–18. 10.1002/jmv.25801.
    1. Chen T, Wu D, Chen H, Yan W, Yang D, Chen G, et al. Clinical characteristics of 113 deceased patients with coronavirus disease 2019: retrospective study. BMJ. 2020;368:m1091. 10.1136/bmj.m1091.
    1. Cai Q, Huang D, Ou P, Yu H, Zhu Z, Xia Z, et al. COVID-19 in a designated infectious diseases hospital outside Hubei Province, China [published online ahead of print, 2020 Apr 2]. Allergy. 2020;10.1111/all.14309. 10.1111/all.14309.
    1. Mo P, Xing Y, Xiao Y, Deng L, Zhao Q, Wang H, et al. Clinical characteristics of refractory COVID-19 pneumonia in Wuhan, China [published online ahead of print, 2020 Mar 16]. Clin Infect Dis. 2020;ciaa270. 10.1093/cid/ciaa270.
    1. Wang D, Hu B, Hu C, Zhu F, Liu X, Zhang J, et al. Clinical characteristics of 138 hospitalized patients with 2019 novel coronavirus–infected pneumonia in Wuhan. China JAMA. 2020;323:1061.
    1. Wang F, Hou H, Luo Y, Tang G, Wu S, Huang M, et al. The laboratory tests and host immunity of COVID-19 patients with different severity of illness. JCI Insight. 2020 [cited 2020 May 7]; Available from: .
    1. Hou H, Zhang B, Huang H, Luo Y, Wu S, Tang G, et al. Using IL-2R/lymphocytes for predicting the clinical progression of patients with COVID-19 [published online ahead of print, 2020 May 4]. Clin Exp Immunol. 2020;10.1111/cei.13450. 10.1111/cei.13450.
    1. Mier JW, Gallo RC. Purification and some characteristics of human T-cell growth factor from phytohemagglutinin-stimulated lymphocyte-conditioned media. Proc Natl Acad Sci. 1980;77:6134–6138.
    1. Smith K. Interleukin-2: inception, impact, and implications. Science. 1988;240:1169–1176.
    1. Malek TR, Bayer AL. Tolerance, not immunity, crucially depends on IL-2. Nat Rev Immunol. 2004;4:665–674.
    1. Nelson DL, Rubin LA, Kurman CC, Fritz ME, Boutin B. An analysis of the cellular requirements for the production of soluble interleukin-2 receptors in vitro. J Clin Immunol. 1986;6:114–120.
    1. Minami Y, Kono T, Miyazaki T, Taniguchi T. The IL-2 receptor complex: its structure, function, and target genes. Annu Rev Immunol. 1993;11:245–268.
    1. Rubin L. The molecular basis for the generation of the human soluble interleukin 2 receptor. Cytokine. 1990;2:330–336.
    1. Loughnan MS, Sanderson CJ, Nossal GJ. Soluble interleukin 2 receptors are released from the cell surface of normal murine B lymphocytes stimulated with interleukin 5. Proc Natl Acad Sci. 1988;85:3115–3119.
    1. Eurelings LEM, Miedema JR, Dalm VASH, van Daele PLA, van Hagen PM, van Laar JAM, et al. Sensitivity and specificity of serum soluble interleukin-2 receptor for diagnosing sarcoidosis in a population of patients suspected of sarcoidosis. Mogi M, editor. PLOS ONE. 2019;14:e0223897.
    1. Maeda M, Tanabe-Shibuya J, Miyazato P, Masutani H, Yasunaga J-I, Usami K, et al. IL-2/IL-2 receptor pathway plays a crucial role in the growth and malignant transformation of HTLV-1-infected T cells to develop adult T-cell leukemia. Front Microbiol. 2020;11:356.
    1. Krieg C, Letourneau S, Pantaleo G, Boyman O. Improved IL-2 immunotherapy by selective stimulation of IL-2 receptors on lymphocytes and endothelial cells. Proc Natl Acad Sci. 2010;107:11906–11911.
    1. Lodigiani C, Iapichino G, Carenzo L, Cecconi M, Ferrazzi P, Sebastian T, et al. Venous and arterial thromboembolic complications in COVID-19 patients admitted to an academic hospital in Milan. Italy Thromb Res. 2020;191:9–14.
    1. Klok FA, Kruip MJHA, van der Meer NJM, Arbous MS, Gommers DAMPJ, Kant KM, et al. Incidence of thrombotic complications in critically ill ICU patients with COVID-19. Thromb Res. 2020;191:145–47. 10.1016/j.thromres.2020.04.013.
    1. Dolhnikoff M, Duarte-Neto AN, de Almeida Monteiro RA, Ferraz da Silva LF, Pierre de Oliveira E, Nascimento Saldiva PH, et al. Pathological evidence of pulmonary thrombotic phenomena in severe COVID-19. J Thromb Haemost. 2020 [cited 2020 May 5]; Available from: .
    1. Pernazza A, Mancini M, Rullo E, Bassi M, De Giacomo T, Rocca CD, et al. Early histologic findings of pulmonary SARS-CoV-2 infection detected in a surgical specimen. Virchows Arch. 2020 [cited 2020 May 5]; Available from: .
    1. Wichmann D, Sperhake J-P, Lütgehetmann M, Steurer S, Edler C, Heinemann A, et al. Autopsy findings and venous thromboembolism in patients with COVID-19: a prospective cohort study. Ann Intern Med. 2020:M20–2003.
    1. Tang N, Li D, Wang X, Sun Z. Abnormal coagulation parameters are associated with poor prognosis in patients with novel coronavirus pneumonia. J Thromb Haemost. 2020;18:844–847.
    1. Gralinski LE, Bankhead A, Jeng S, Menachery VD, Proll S, Belisle SE, et al. Mechanisms of severe acute respiratory syndrome coronavirus-induced acute lung injury. Dermody TS, editor mBio. 2013;4:e00271–e00213.
    1. Henry BM, Vikse J, Benoit S, Favaloro EJ, Lippi G. Hyperinflammation and derangement of renin-angiotensin-aldosterone system in COVID-19: a novel hypothesis for clinically suspected hypercoagulopathy and microvascular immunothrombosis. Clin Chim Acta. 2020;507:167–173.
    1. Escher R, Breakey N, Lämmle B. Severe COVID-19 infection associated with endothelial activation. Thromb Res. 2020;190:62.
    1. Magro C, Mulvey JJ, Berlin D, Nuovo G, Salvatore S, Harp J, et al. Complement associated microvascular injury and thrombosis in the pathogenesis of severe COVID-19 infection: a report of five cases [published online ahead of print, 2020 Apr 15]. Transl Res. 2020;220:1–13. 10.1016/j.trsl.2020.04.007.
    1. Kuba K, Imai Y, Rao S, Gao H, Guo F, Guan B, et al. A crucial role of angiotensin converting enzyme 2 (ACE2) in SARS coronavirus–induced lung injury. Nat Med. 2005;11:875–879.
    1. Lei C, Qian K, Li T, Zhang S, Fu W, Ding M, et al. Neutralization of SARS-CoV-2 spike pseudotyped virus by recombinant ACE2-Ig. Nat Commun. 2020;11:2070.
    1. Vaduganathan M, Vardeny O, Michel T, McMurray JJV, Pfeffer MA, Solomon SD. Renin–angiotensin–aldosterone system inhibitors in patients with COVID-19. N Engl J Med. 2020;382:1653–1659.
    1. Zen M, Canova M, Campana C, Bettio S, Nalotto L, Rampudda M, et al. The kaleidoscope of glucorticoid effects on immune system. Autoimmun Rev. 2011;10:305–310.
    1. Zielińska KA, Van Moortel L, Opdenakker G, De Bosscher K, Van den Steen PE. Endothelial response to glucocorticoids in inflammatory diseases. Front Immunol. 2016 [cited 2020 May 10];7. Available from: .
    1. WHO WHO. Clinical management of severe acute respiratory infection when novel coronavirus (nCoV) infection is suspected. 13 March 2020. Available from: .
    1. Russell CD, Millar JE, Baillie JK. Clinical evidence does not support corticosteroid treatment for 2019-nCoV lung injury. Lancet. 2020;395:473–475.
    1. Alhazzani W, Møller MH, Arabi YM, Loeb M, Gong MN, Fan E, et al. Surviving sepsis campaign: guidelines on the management of critically ill adults with coronavirus disease 2019 (COVID-19) Intensive Care Med. 2020;46:854–887.
    1. Zha L, Li S, Pan L, Tefsen B, Li Y, French N, et al. Corticosteroid treatment of patients with coronavirus disease 2019 (COVID-19). Med J Aust. 2020 [cited 2020 May 10]; Available from: .
    1. Wu C, Chen X, Cai Y, Xia J, Zhou X, Xu S, et al. Risk factors associated with acute respiratory distress syndrome and death in patients with coronavirus disease 2019 pneumonia in Wuhan, China. JAMA Intern Med. 2020 [cited 2020 May 10]; Available from: .
    1. Mihara M, Kasutani K, Okazaki M, Nakamura A, Kawai S, Sugimoto M, et al. Tocilizumab inhibits signal transduction mediated by both mIL-6R and sIL-6R, but not by the receptors of other members of IL-6 cytokine family. Int Immunopharmacol. 2005;5:1731–1740.
    1. Xu X, Han M, Li T, Sun W, Wang D, Fu B, et al. Effective treatment of severe COVID-19 patients with tocilizumab. Proc Natl Acad Sci U S A. 2020;117(20):10970–975. 10.1073/pnas.2005615117.
    1. Sciascia S, Aprà F, Baffa A, et al. Pilot prospective open, single-arm multicentre study on off-label use of tocilizumab in patients with severe COVID-19. Clin Exp Rheumatol. 2020;38(3):529–32.
    1. Kollias A, Kyriakoulis KG, Dimakakos E, Poulakou G, Stergiou GS, Syrigos K. Thromboembolic risk and anticoagulant therapy in COVID-19 patients: emerging evidence and call for action. Br J Haematol. 2020;189(5):846–47. 10.1111/bjh.16727.
    1. Thachil J, Tang N, Gando S, Falanga A, Cattaneo M, Levi M, et al. ISTH interim guidance on recognition and management of coagulopathy in COVID-19. J Thromb Haemost. 2020;18:1023–1026.
    1. Tang N, Bai H, Chen X, Gong J, Li D, Sun Z. Anticoagulant treatment is associated with decreased mortality in severe coronavirus disease 2019 patients with coagulopathy. J Thromb Haemost. 2020;18:1094–1099.
    1. Susen S, Tacquard C-A, Godon A, Mansour A, Guarrigue D, Nguyen P, et al. Traitement anticoagulant pour la prévention du risque trombotique chez un patient hospitalisé avec Covid-19 et surveillance de l'hémostase propositions du GIHP et GFHT. 2020.
    1. Bikdeli B, Madhavan MV, Jimenez D, Chuich T, Dreyfus I, Driggin E, et al. COVID-19 and Thrombotic or Thromboembolic Disease: Implications for Prevention, Antithrombotic Therapy, and Follow-up [published online ahead of print, 2020 Apr 15]. J Am Coll Cardiol. 2020;S0735-1097(20)35008-7. 10.1016/j.jacc.2020.04.031.
    1. Young E. The anti-inflammatory effects of heparin and related compounds. Thromb Res. 2008;122:743–752.
    1. Li J, Vlodavsky I. Heparin, heparan sulfate and heparanase in inflammatory reactions. Thromb Haemost. 2009;102:823–828.
    1. Wang J, Hajizadeh N, Moore EE, McIntyre RC, Moore PK, Veress LA, et al. Tissue plasminogen activator (tPA) treatment for COVID-19 associated acute respiratory distress syndrome (ARDS): a case series. J Thromb Haemost. 2020 [cited 2020 May 5]; Available from: .
    1. Jodele S, Medvedovic M, Luebbering N, Chen J, Dandoy CE, Laskin BL, et al. Interferon-complement loop in transplant-associated thrombotic microangiopathy. Blood Adv. 2020;4:1166–1177.
    1. Olson SR, Lu E, Sulpizio E, Shatzel JJ, Rueda JF, DeLoughery TG. When to stop eculizumab in complement-mediated thrombotic microangiopathies. Am J Nephrol. 2018;48:96–107.
    1. Pittock SJ, Berthele A, Fujihara K, Kim HJ, Levy M, Palace J, et al. Eculizumab in aquaporin-4–positive neuromyelitis optica spectrum disorder. N Engl J Med. 2019;381:614–625.
    1. Diurno F, Numis FG, Porta G, Cirillo F, Maddaluno S, Ragozzino A, et al. Eculizumab treatment in patients with COVID-19: preliminary results from real life ASL Napoli 2 Nord experience. Eur Rev Med Pharmacol Sci. 2020;24:4040–4047.

Source: PubMed

3
Sottoscrivi