Platelet activation and platelet-monocyte aggregate formation trigger tissue factor expression in patients with severe COVID-19

Eugenio D Hottz, Isaclaudia G Azevedo-Quintanilha, Lohanna Palhinha, Lívia Teixeira, Ester A Barreto, Camila R R Pão, Cassia Righy, Sérgio Franco, Thiago M L Souza, Pedro Kurtz, Fernando A Bozza, Patrícia T Bozza, Eugenio D Hottz, Isaclaudia G Azevedo-Quintanilha, Lohanna Palhinha, Lívia Teixeira, Ester A Barreto, Camila R R Pão, Cassia Righy, Sérgio Franco, Thiago M L Souza, Pedro Kurtz, Fernando A Bozza, Patrícia T Bozza

Abstract

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is an emergent pathogen responsible for the coronavirus disease 2019 (COVID-19). Since its emergence, the novel coronavirus has rapidly achieved pandemic proportions causing remarkably increased morbidity and mortality around the world. A hypercoagulability state has been reported as a major pathologic event in COVID-19, and thromboembolic complications listed among life-threatening complications of the disease. Platelets are chief effector cells of hemostasis and pathological thrombosis. However, the participation of platelets in the pathogenesis of COVID-19 remains elusive. This report demonstrates that increased platelet activation and platelet-monocyte aggregate formation are observed in severe COVID-19 patients, but not in patients presenting mild COVID-19 syndrome. In addition, exposure to plasma from severe COVID-19 patients increased the activation of control platelets ex vivo. In our cohort of COVID-19 patients admitted to the intensive care unit, platelet-monocyte interaction was strongly associated with tissue factor (TF) expression by the monocytes. Platelet activation and monocyte TF expression were associated with markers of coagulation exacerbation as fibrinogen and D-dimers, and were increased in patients requiring invasive mechanical ventilation or patients who evolved with in-hospital mortality. Finally, platelets from severe COVID-19 patients were able to induce TF expression ex vivo in monocytes from healthy volunteers, a phenomenon that was inhibited by platelet P-selectin neutralization or integrin αIIb/β3 blocking with the aggregation inhibitor abciximab. Altogether, these data shed light on new pathological mechanisms involving platelet activation and platelet-dependent monocyte TF expression, which were associated with COVID-19 severity and mortality.

© 2020 by The American Society of Hematology.

Figures

Graphical abstract
Graphical abstract
Figure 1
Figure 1
Increased platelet activation in critically ill COVID-19 patients. (A-B) The percentage of P-selectin (CD62P) (A) and CD63 (B) surface expression on platelets from SARS-CoV-2− control participants, SARS-CoV-2+ asymptomatic subjects, or symptomatic patients presenting mild to severe COVID-19 syndrome. (C) The concentration of TXB2 in plasma from control subjects or patients with COVID-19 presenting mild to severe syndrome. (D) Quantification of TXB2, PF4, and PDGF in tracheal aspirates from severe COVID-19 patients under mechanical ventilation (n = 19). The horizontal lines in the box plots represent the median, the box edges represent the interquartile ranges, and the whiskers indicate the minimal and maximal value in each group. *P < .05 between selected groups. ns, nonsignificant.
Figure 2
Figure 2
Inflammatory mediators generated in severe COVID-19 contribute to platelet activation. (A-B) The percentage of P-selectin (CD62P) (A) and CD63 (B) expression on platelets was plotted against the concentration of fibrinogen in plasma. Linear regression and Spearman correlation were calculated according to the distribution of the dots. (C-D) Platelets from healthy volunteers were incubated with plasma from severe COVID-19 patients (severe, n = 16) or from SARS-CoV2− subjects (control, n = 11) for 2 hours. The percentages of P-selectin (C) and CD63 (D) are shown. All experiments were repeated with platelets from 3 independent healthy volunteers with similar results, and representative data from 1 of the platelet donors are shown. The horizontal lines in the box plots represent the median, the box edges represent the interquartile ranges, and the whiskers indicate the minimal and maximal value in each group. *P < .05 between selected groups.
Figure 3
Figure 3
Increased platelet-monocyte aggregates in severe COVID-19 patients. (A) Gating strategy for analysis of platelet-monocyte aggregates in patients with COVID-19 or control subjects. (B) Percentage of platelet-monocyte complexes (CD14+CD41+) among monocytes from SARS-CoV-2− control volunteers, SARS-CoV-2+ asymptomatic subjects, or symptomatic patients presenting mild to severe COVID-19 syndrome. The horizontal lines in the box plots represent the median, the box edges represent the interquartile ranges, and the whiskers indicate the minimal and maximal value in each group. *P < .05 between selected groups. (C) The percentage of platelet P-selectin (CD62P) surface expression was plotted against the percentage of platelet-monocyte complexes in the same infected subjects. Linear regression and Pearson correlation were calculated according to the distribution of the dots. (D) Phalloidin-labeled cytospin blood samples from control subjects or COVID-19 patients. Platelets and leukocytes obtained from whole blood after lysis of erythrocytes were labeled with Alexa 488–phalloidin (F actin, green) and DAPI (nuclei, blue). Scale bar, 10 μm; inset, 5 μm. Representative images of platelet-monocyte aggregates are shown. The fourth panel shows orthogonal views from different planes of the confocal image. The green line generated a 2-dimensional (2D) image that is shown in the top green rectangle whereas the red line–generated 2D image is shown in the red rectangle on the right. SSC-H, side scatter height.
Figure 4
Figure 4
Platelet-monocyte complexes express TF in severe COVID-19 patients. (A-B) The percentage of TF surface expression on monocytes (A) or platelets (B) from SARS-CoV2− control volunteers, SARS-Cov2+ asymptomatic subjects, or symptomatic patients presenting mild to severe COVID-19 syndrome. (C) The percentage of TF surface expression on monocytes that were complexed with platelets (CD14+CD41+) or circulating freely (CD14+CD41−) in severe COVID-19 patients. The horizontal lines in the box plots represent the median, the box edges represent the interquartile ranges, and the whiskers indicate the minimal and maximal value in each group. *P < .05 between selected groups.
Figure 5
Figure 5
Increased platelet activation and TF expression by monocytes associate with severity and mortality in COVID-19. (A-E) Severe COVID-19 patients admitted to the ICU were stratified between those requiring invasive mechanical ventilation or noninvasive O2 supplementation. The percentages of P-selectin (CD62P) (A) and CD63 (B) expression on platelets, the concentration of TXB2 in plasma (C), the percentage of monocytes forming aggregates with platelets (D), and TF expression on monocytes (E) are shown. (F-K) Severe COVID-19 patients were stratified according to the 28-day mortality outcome as survivors or nonsurvivors. The percentages of P-selectin (CD62P) (F) and CD63 (G) expression on platelets, the concentration of TXB2 in plasma (H), the quantification of PDGF-BB in tracheal aspirates (I), and the percentage of monocytes forming aggregates with platelets (J) and expressing TF (K) are shown. The horizontal lines in the box plots represent the median, the box edges represent the interquartile ranges, and the whiskers indicate the minimal and maximal value in each group. The gray box indicates the interquartile ranges and the dotted line represents the median of the SARS-CoV2− control group. *P < .05 between selected groups.
Figure 6
Figure 6
Platelets from COVID-19 patients induce TF expression in monocytes through mechanisms depending on P-selectin and integrin αIIb3. (A) Monocytes from healthy volunteers were incubated with platelets from severe COVID-19 patients (COVID-19 platelets) or from heterologous healthy volunteers (control platelets) for the indicated time points. The percentage of TF-expressing monocytes is shown. (B-D) Control monocytes were exposed to platelets from severe COVID-19 patients for 2 hours in the presence of anti–P-selectin (anti-CD62P) neutralizing antibody, the anti-αIIb/β3 antibody abciximab, or isotype-matched IgG. The percentage of TF-expressing monocytes (B), platelet-monocyte complexes (C), and the percentage and the mean fluorescence intensity (MFI) of P-selectin expression on CD14+CD41+ monocytes (D) are shown in each condition. (E-F) Control monocytes were exposed to platelets from COVID-19 patients for 2 hours in the presence of aspirin (100 µM) or clopidogrel (300 µM). Bars represent mean plus or minus standard error of the mean of monocytes exposed to platelets from 6 independent COVID-19 patients. All experiments were repeated with monocytes from 2 (B-D), 3 (E-F), or 4 (A) independent healthy volunteers with similar results, and representative data from 1 of the donors are shown. *P < .05 between selected groups.

References

    1. Zhou P, Yang XL, Wang XG, et al. A pneumonia outbreak associated with a new coronavirus of probable bat origin. Nature. 2020;579(7798):270–273.
    1. World Health Organization,. Coronavirus Disease (COVID-19) Situation Report-122. . Accessed 22 May 2020.
    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(4):844–847.
    1. Klok FA, Kruip MJHA, van der Meer NJM, et al. Incidence of thrombotic complications in critically ill ICU patients with COVID-19. Thromb Res. 2020;191:145–147.
    1. Middeldorp S, Coppens M, van Haaps TF, et al. Incidence of venous thromboembolism in hospitalized patients with COVID‐19. J Thromb Haemost. 2020;18(8):1995-2002.
    1. Giamarellos-Bourboulis EJ, Netea MG, Rovina N, et al. Complex immune dysregulation in COVID-19 patients with severe respiratory failure. Cell Host Microbe. 2020;27(6):992–1000.e3.
    1. Lippi G, Plebani M, Henry BM. Thrombocytopenia is associated with severe coronavirus disease 2019 (COVID-19) infections: a meta-analysis. Clin Chim Acta. 2020;506:145–148.
    1. Yang X, Yang Q, Wang Y, et al. Thrombocytopenia and its association with mortality in patients with COVID‐19. J Thromb Haemost. 2020;18(6):1469–1472.
    1. Jiang S-Q, Huang Q-F, Xie W-M, Lv C, Quan X-Q. The association between severe COVID‐19 and low platelet count: evidence from 31 observational studies involving 7613 participants. Br J Haematol. 2020;190(1):e29–e33.
    1. Zhang L, Yan X, Fan Q, et al. D‐dimer levels on admission to predict in‐hospital mortality in patients with Covid‐19. J Thromb Haemost. 2020;18(6):1324–1329.
    1. 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.
    1. Stevens H, McFadyen JD. Platelets as central actors in thrombosis-reprising an old role and defining a new character. Semin Thromb Hemost. 2019;45(8):802–809.
    1. Koupenova M, Clancy L, Corkrey HA, Freedman JE. Circulating platelets as mediators of immunity, inflammation, and thrombosis. Circ Res. 2018;122(2):337–351.
    1. Middleton EA, Rondina MT, Schwertz H, Zimmerman GA. Amicus or adversary revisited: platelets in acute lung injury and acute respiratory distress syndrome. Am J Respir Cell Mol Biol. 2018;59(1):18–35.
    1. Kapur R, Zufferey A, Boilard E, Semple JW. Nouvelle cuisine: platelets served with inflammation. J Immunol. 2015;194(12):5579–5587.
    1. Semple JW, Italiano JE, Jr., Freedman J. Platelets and the immune continuum. Nat Rev Immunol. 2011;11(4):264–274.
    1. Popa M, Tahir S, Elrod J, et al. Role of CD40 and ADAMTS13 in von Willebrand factor-mediated endothelial cell-platelet-monocyte interaction. Proc Natl Acad Sci USA. 2018;115(24):E5556–E5565.
    1. Pan D, Amison RT, Riffo-Vasquez Y, et al. P-Rex and Vav Rac-GEFs in platelets control leukocyte recruitment to sites of inflammation. Blood. 2015;125(7):1146–1158.
    1. Weyrich AS, Elstad MR, McEver RP, et al. Activated platelets signal chemokine synthesis by human monocytes. J Clin Invest. 1996;97(6):1525–1534.
    1. Hottz ED, Medeiros-de-Moraes IM, Vieira-de-Abreu A, et al. Platelet activation and apoptosis modulate monocyte inflammatory responses in dengue. J Immunol. 2014;193(4):1864–1872.
    1. Clark SR, Ma AC, Tavener SA, et al. Platelet TLR4 activates neutrophil extracellular traps to ensnare bacteria in septic blood. Nat Med. 2007;13(4):463–469.
    1. Swystun LL, Liaw PC. The role of leukocytes in thrombosis. Blood. 2016;128(6):753–762.
    1. Celi A, Pellegrini G, Lorenzet R, et al. P-selectin induces the expression of tissue factor on monocytes. Proc Natl Acad Sci USA. 1994;91(19):8767–8771.
    1. Ivanov II, Apta BHR, Bonna AM, Harper MT. Platelet P-selectin triggers rapid surface exposure of tissue factor in monocytes. Sci Rep. 2019;9(1):13397.
    1. Lindmark E, Tenno T, Siegbahn A. Role of platelet P-selectin and CD40 ligand in the induction of monocytic tissue factor expression. Arterioscler Thromb Vasc Biol. 2000;20(10):2322–2328.
    1. Grover SP, Mackman N. Tissue factor: an essential mediator of hemostasis and trigger of thrombosis. Arterioscler Thromb Vasc Biol. 2018;38(4):709–725.
    1. Martinod K, Wagner DD. Thrombosis: tangled up in NETs. Blood. 2014;123(18):2768–2776.
    1. Franks ZG, Campbell RA, Weyrich AS, Rondina MT. Platelet-leukocyte interactions link inflammatory and thromboembolic events in ischemic stroke. Ann N Y Acad Sci. 2010;1207:11–17.
    1. Hottz ED, Bozza FA, Bozza PT. Platelets in immune response to virus and immunopathology of viral infections. Front Med (Lausanne) 2018;5:121.
    1. Sung PS, Huang TF, Hsieh SL. Extracellular vesicles from CLEC2-activated platelets enhance dengue virus-induced lethality via CLEC5A/TLR2. Nat Commun. 2019;10(1):2402.
    1. Hottz ED, Quirino-Teixeira AC, Valls-de-Souza R, Zimmerman GA, Bozza FA, Bozza PT. Platelet function in HIV plus dengue coinfection associates with reduced inflammation and milder dengue illness. Sci Rep. 2019;9(1):7096.
    1. Mesquita EC, Hottz ED, Amancio RT, et al. Persistent platelet activation and apoptosis in virologically suppressed HIV-infected individuals. Sci Rep. 2018;8(1):14999.
    1. Koupenova M, Corkrey HA, Vitseva O, et al. The role of platelets in mediating a response to human influenza infection. Nat Commun. 2019;10(1):1780.
    1. Hottz ED, Lopes JF, Freitas C, et al. Platelets mediate increased endothelium permeability in dengue through NLRP3-inflammasome activation. Blood. 2013;122(20):3405–3414.
    1. Quirino-Teixeira AC, Rozini SV, Barbosa-Lima G, et al. Inflammatory signaling in dengue-infected platelets requires translation and secretion of nonstructural protein 1. Blood Adv. 2020;4(9):2018–2031.
    1. Guo T, Fan Y, Chen M, et al. Cardiovascular implications of fatal outcomes of patients with coronavirus disease 2019 (COVID-19) JAMA Cardiol. 2020;5(7):1–8.
    1. Wu C, Chen X, Cai Y, 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;180(7):1–11.
    1. Shi S, Qin M, Shen B, et al. Association of cardiac injury with mortality in hospitalized patients with COVID-19 in Wuhan, China. JAMA Cardiol. 2020;5(7):802.
    1. Akhvlediani T, Ali SM, Angus DC, et al. ISARIC Clinical Characterisation Group Global outbreak research: harmony not hegemony. Lancet Infect Dis. 2020;20(7):770–772.
    1. Vabret N, Britton GJ, Gruber C, et al. Sinai Immunology Review Project Immunology of COVID-19: current state of the science. Immunity. 2020;52(6):910–941.
    1. Chen G, Wu D, Guo W, et al. Clinical and immunological features of severe and moderate coronavirus disease 2019. J Clin Invest. 2020;130(5):2620–2629.
    1. 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.
    1. Jose RJ, Manuel A. COVID-19 cytokine storm: the interplay between inflammation and coagulation. Lancet Respir Med. 2020;8(6):e46–e47.
    1. Larsen E, Celi A, Gilbert GE, et al. PADGEM protein: a receptor that mediates the interaction of activated platelets with neutrophils and monocytes. Cell. 1989;59(2):305–312.
    1. Guo L, Rondina MT. The era of thromboinflammation: platelets are dynamic sensors and effector cells during infectious diseases. Front Immunol. 2019;10:2204.
    1. Durrant TN, van den Bosch MT, Hers I. Integrin αIIbβ3 outside-in signaling. Blood. 2017;130(14):1607–1619.
    1. Vardakas KZ, Matthaiou DK, Falagas ME. Incidence, characteristics and outcomes of patients with severe community acquired-MRSA pneumonia. Eur Respir J. 2009;34(5):1148–1158.
    1. Boussekey N, Leroy O, Georges H, Devos P, d'Escrivan T, Guery B. Diagnostic and prognostic values of admission procalcitonin levels in community-acquired pneumonia in an intensive care unit. Infection. 2005;33(4):257–263.
    1. Wong RSM, Wu A, To KF, et al. Haematological manifestations in patients with severe acute respiratory syndrome: retrospective analysis. BMJ. 2003;326(7403):1358–1362.
    1. Nguyen T, Kyle UG, Jaimon N, et al. Coinfection with Staphylococcus aureus increases risk of severe coagulopathy in critically ill children with influenza A (H1N1) virus infection. Crit Care Med. 2012;40(12):3246–3250.
    1. Ackermann M, Verleden SE, Kuehnel M, et al. Pulmonary vascular endothelialitis, thrombosis, and angiogenesis in Covid-19. N Engl J Med. 2020;383(2):120–128.
    1. Dolhnikoff M, Duarte-Neto AN, de Almeida Monteiro RA, et al. Pathological evidence of pulmonary thrombotic phenomena in severe COVID-19. J Thromb Haemost. 2020;18(6):1517–1519.
    1. Connors JM, Levy JH. COVID-19 and its implications for thrombosis and anticoagulation. Blood. 2020;135(23):2033–2040.
    1. Schwertz H, Tolley ND, Foulks JM, et al. Signal-dependent splicing of tissue factor pre-mRNA modulates the thrombogenicity of human platelets. J Exp Med. 2006;203(11):2433–2440.
    1. Müller I, Klocke A, Alex M, et al. Intravascular tissue factor initiates coagulation via circulating microvesicles and platelets. FASEB J. 2003;17(3):476–478.
    1. Rondina MT, Schwertz H, Harris ES, et al. The septic milieu triggers expression of spliced tissue factor mRNA in human platelets. J Thromb Haemost. 2011;9(4):748–758.
    1. Franks Z, Campbell RA, Vieira de Abreu A, et al. Methicillin-resistant Staphylococcus aureus-induced thrombo-inflammatory response is reduced with timely antibiotic administration. Thromb Haemost. 2013;109(4):684–695.
    1. Mayne E, Funderburg NT, Sieg SF, et al. Increased platelet and microparticle activation in HIV infection: upregulation of P-selectin and tissue factor expression. J Acquir Immune Defic Syndr. 2012;59(4):340–346.
    1. Li H, Liu L, Zhang D, et al. SARS-CoV-2 and viral sepsis: observations and hypotheses. Lancet. 2020;395(10235):1517–1520.
    1. Schechter ME, Andrade BB, He T, et al. Inflammatory monocytes expressing tissue factor drive SIV and HIV coagulopathy. Sci Transl Med. 2017;9(405):eaam5441.
    1. Christersson C, Johnell M, Siegbahn A. Tissue factor and IL8 production by P-selectin-dependent platelet-monocyte aggregates in whole blood involves phosphorylation of Lyn and is inhibited by IL10. J Thromb Haemost. 2008;6(6):986–994.
    1. Christersson C, Johnell M, Siegbahn A. The influence of direct thrombin inhibitors on the formation of platelet-leukocyte aggregates and tissue factor expression. Thromb Res. 2010;126(4):e327–e333.
    1. Dann R, Hadi T, Montenont E, et al. Platelet-derived MRP-14 induces monocyte activation in patients with symptomatic peripheral artery disease. J Am Coll Cardiol. 2018;71(1):53–65.
    1. Campbell RA, Franks Z, Bhatnagar A, et al. Granzyme A in human platelets regulates the synthesis of proinflammatory cytokines by monocytes in aging. J Immunol. 2018;200(1):295–304.

Source: PubMed

3
Suscribir