Platelets Promote Thromboinflammation in SARS-CoV-2 Pneumonia

Francesco Taus, Gianluca Salvagno, Stefania Canè, Cristiano Fava, Fulvia Mazzaferri, Elena Carrara, Varvara Petrova, Roza Maria Barouni, Francesco Dima, Andrea Dalbeni, Simone Romano, Giovanni Poli, Marco Benati, Simone De Nitto, Giancarlo Mansueto, Manuela Iezzi, Evelina Tacconelli, Giuseppe Lippi, Vincenzo Bronte, Pietro Minuz, Francesco Taus, Gianluca Salvagno, Stefania Canè, Cristiano Fava, Fulvia Mazzaferri, Elena Carrara, Varvara Petrova, Roza Maria Barouni, Francesco Dima, Andrea Dalbeni, Simone Romano, Giovanni Poli, Marco Benati, Simone De Nitto, Giancarlo Mansueto, Manuela Iezzi, Evelina Tacconelli, Giuseppe Lippi, Vincenzo Bronte, Pietro Minuz

Abstract

Objective: Pulmonary thrombosis is observed in severe acute respiratory syndrome coronavirus 2 pneumonia. Aim was to investigate whether subpopulations of platelets were programmed to procoagulant and inflammatory activities in coronavirus disease 2019 (COVID-19) patients with pneumonia, without comorbidities predisposing to thromboembolism. Approach and Results: Overall, 37 patients and 28 healthy subjects were studied. Platelet-leukocyte aggregates, platelet-derived microvesicles, the expression of P-selectin, and active fibrinogen receptor on platelets were quantified by flow cytometry. The profile of 45 cytokines, chemokines, and growth factors released by platelets was defined by immunoassay. The contribution of platelets to coagulation factor activity was selectively measured. Numerous platelet-monocyte (mean±SE, 67.9±4.9%, n=17 versus 19.4±3.0%, n=22; P<0.0001) and platelet-granulocyte conjugates (34.2±4.04% versus 8.6±0.7%; P<0.0001) were detected in patients. Resting patient platelets had similar levels of P-selectin (10.9±2.6%, n=12) to collagen-activated control platelets (8.7±1.5%), which was not further increased by collagen activation on patient platelets (12.4±2.5%, P=nonsignificant). The agonist-stimulated expression of the active fibrinogen receptor was reduced by 60% in patients (P<0.0001 versus controls). Cytokines (IL [interleukin]-1α, IL-1β, IL-1RA, IL-4, IL-10, IL-13, IL, 17, IL-27, IFN [interferon]-α, and IFN-γ), chemokines (MCP-1/CCL2 [monocyte chemoattractant protein 1]), and growth factors (VEGF [vascular endothelial growth factor]-A/D) were released in significantly larger amounts upon stimulation of COVID-19 platelets. Platelets contributed to increased fibrinogen, VWF (von Willebrand factor), and factor XII in COVID-19 patients. Patients (28.5±0.7 s, n=32), unlike controls (31.6±0.5 s, n=28; P<0.001), showed accelerated factor XII-dependent coagulation.

Conclusions: Platelets in COVID-19 pneumonia are primed to spread proinflammatory and procoagulant activities in systemic circulation.

Keywords: blood platelets; inflammation; interferons; monocytes; thrombosis.

Conflict of interest statement

None.

Figures

Figure 1.
Figure 1.
Computed tomography perfusion angiography (CTPA) scans. Axial CTPA images with mediastinal (A) and lung (B) window showing filling defects involving the proximal tract of left pulmonary artery (arrow). Diffuse ground glass opacifications together with diffuse thickening of interlobular septa are visible (B). MIP (maximum intensity projection) reformatted images on coronal plane (C) show filling defects both in some segmental and subsegmental branches of left pulmonary artery (small arrow) and in corresponding next venous branches (large arrow).
Figure 2.
Figure 2.
Representative blood smear from patients with severe acute respiratory syndrome coronavirus 2 pneumonia showing platelet anisopoichilocytosis (A–D), platelet satellitism (E–H), and platelet engulfment by lymphocytes (F).
Figure 3.
Figure 3.
Platelet phenotype. Whole-blood analysis of monocytes and neutrophil-platelet aggregates shows higher percentage of plateletmonocyte aggregates (A) and platelet-neutrophil aggregates (B) in citrated whole blood from coronavirus disease 2019 (COVID-19) patients (n=17) than healthy controls (n=22). The percentage of resting platelets expressing P-selectin in COVID-19 patients (n=12) is similar to that observed in platelets from healthy controls (n=22) stimulated with collagen (C). P-selectin expression does not further increase when platelets are stimulated with collagen (C). The expression of the active form of fibrinogen receptor αIIbβ3, as detected by the monoclonal antibody PAC-1, is similar under resting conditions in patients and healthy controls and lower in patients (n=16) in platelets stimulated with collagen (D). The number of platelet-derived microvesicles (PMV) is slightly higher in patients (n=15) than in controls (n=22; E) and correlates with the surface expression of P-selectin in COVID-19 patients (F). CD62P (P-selectin) indicates cluster of differentiation 62P; and PTL, platelets.
Figure 4.
Figure 4.
Coagulation and coagulation factors assays. Activated partial thromboplastin time (APTT) was tested using plasma and platelet-rich plasma (PRP) from coronavirus disease 2019 (COVID-19) patients (n=32) and healthy controls (n=28; A). The activity of the coagulation factor VIII is similarly higher in plasma and PRP in COVID-19 patients, correlates with APTT (B and C), and is not stored in platelets, as demonstrated by the effects of platelets from patients added to control plasma (B). Factor XII activity does not differ in patients (n=20) and controls (n=20; D) but correlates with APTT only in patients (F) and increases when platelets from patients were suspended in control plasma (n=12; D and E). Plasma VWF (von Willebrand factor) antigen (Ag), collagen binding (CB), and ristocetin cofactor (RCo) is increased in COVID-19 patients (n=9) compared with controls (n=20; G). Fibrinogen activity is higher in plasma and PRP from patients (n=20) than controls (n=20; H). PTL indicates platelets.

References

    1. Chen N, Zhou M, Dong X, Qu J, Gong F, Han Y, Qiu Y, Wang J, Liu Y, Wei Y, et al. Epidemiological and clinical characteristics of 99 cases of 2019 novel coronavirus pneumonia in Wuhan, China: a descriptive study. Lancet. 2020;395:507–513. doi: 10.1016/S0140-6736(20)30211-7
    1. Bhatraju PK, Ghassemieh BJ, Nichols M, Kim R, Jerome KR, Nalla AK, Greninger AL, Pipavath S, Wurfel MM, Evans L, et al. COVID-19 in critically ill patients in the Seattle Region — case series. N Engl J Med. 2020;382:2012–2022. doi: 10.1056/nejmoa2004500
    1. Wichmann D, Sperhake J-P, Lütgehetmann M, Steurer S, Edler C, Heinemann A, Heinrich F, Mushumba H, Kniep I, Schröder AS, et al. Autopsy findings and venous thromboembolism in patients with COVID-19: a prospective cohort study. Ann Intern Med. 2020;173:268–277. doi: 10.7326/M20-2003
    1. Cattaneo M, Bertinato EM, Birocchi S, Brizio C, Malavolta D, Manzoni M, Muscarella G, Orlandi M. Pulmonary embolism or pulmonary thrombosis in COVID-19? Is the recommendation to use high-dose heparin for thromboprophylaxis justified? Thromb Haemost. 2020;120:1230–1232. doi: 10.1055/s-0040-1712097
    1. Ackermann M, Verleden S, Kuehnel M, Haverich A, Welte T, Laenger F, Vanstapel A, Werlein C, Stark H, Tzankov A, et al. Pulmonary vascular endothelialitis, thrombosis, and angiogenesis in COVID-19. N Engl J Med. 2020;383:120–128. doi: 10.1056/NEJMoa2015432
    1. Nunes Duarte-Neto A, de Almeida Monteiro RA, da Silva LFF, Malheiros DMAC, de Oliveira EP, Theodoro Filho J, Pinho JRR, Soares Gomes-Gouvêa M, Salles APM, de Oliveira IRS, et al. Pulmonary and systemic involvement of COVID-19 assessed by ultrasound-guided minimally invasive autopsy [published online May 22, 2020]. Histopathology. doi: 10.1111/his.14160
    1. Thachil J, Srivastava A. SARS-2 coronavirus-associated hemostatic lung abnormality in COVID-19: is it pulmonary thrombosis or pulmonary embolism? [published online May 12, 2020] Semin Thromb Hemost. doi: 10.1055/s-0040-1712155
    1. Bompard F, Monnier H, Saab I, Tordjman M, Abdoul H, Fournier L, Sanchez O, Lorut C, Chassagnon G, Revel M. Pulmonary embolism in patients with COVID-19 pneumonia. Eur Respir J. 2020;18:1233–1234. doi: 10.1017/CBO9781107415324.004
    1. Connors JM, Levy JH. COVID-19 and its implications for thrombosis and anticoagulation. Blood. 2020;135:2033–2040. doi: 10.1182/blood.2020006000
    1. Huang C, Wang Y, Li X, Ren L, Zhao J, Hu Y, Zhang L, Fan G, Xu J, Gu X, et al. Clinical features of patients infected with 2019 novel coronavirus in Wuhan, China. Lancet. 2020;365:497–506. doi: 10.1016/S0140-6736:30183-5
    1. Pavoni V, Gianesello L, Pazzi M, Stera C, Meconi T, Frigieri FC. Evaluation of coagulation function by rotation thromboelastometry in critically ill patients with severe COVID-19 pneumonia. J Thromb Thrombolysis. 2020;50:281–286. doi: 10.1007/s11239-020-02130-7
    1. Wang T, Chen R, Liu C, Liang W, Guan W, Tang R, Tang C, Zhang N, Zhong N, Li S. Attention should be paid to venous thromboembolism prophylaxis in the management of COVID-19. Lancet Haematol. 2020;7:e362–e363. doi: 10.1016/S2352-3026(20)30109-5
    1. Hottz ED, Azevedo-Quintanilha IG, Palhinha L, Teixeira L, Barreto EA, Pão CRR, Righy C, Franco S, Souza TML, Kurtz P, et al. Platelet activation and platelet-monocyte aggregate formation trigger tissue factor expression in patients with severe COVID-19. Blood. 2020;136:1330–1341. doi: 10.1182/blood.2020007252
    1. Chen G, Wu D, Guo W, Cao Y, Huang D, Wang H, Wang T, Zhang X, Chen H, Yu H, et al. Clinical and immunologic features in severe and moderate Coronavirus Disease 2019. J Clin Invest. 2020;130:2620–2629. doi: 10.1172/JCI137244
    1. Varga Z, Flammer AJ, Steiger P, Haberecker M, Andermatt R, Zinkernagel AS, Mehra MR, Schuepbach RA, Ruschitzka F, Moch H. Endothelial cell infection and endotheliitis in COVID-19. Lancet. 2020;395:1417–1418. doi: 10.1016/S0140-6736(20)30937-5
    1. Gaertner F, Massberg S. Patrolling the vascular borders: platelets in immunity to infection and cancer. Nat Rev Immunol. 2019;19:747–760. doi: 10.1038/s41577-019-0202-z
    1. Nicolai L, Leunig A, Brambs S, Kaiser R, Weinberger T, Weigand M, Muenchhoff M, Hellmuth JC, Ledderose S, Schulz H, et al. Immunothrombotic dysregulation in COVID-19 pneumonia is associated with respiratory failure and coagulopathy. Circulation. 2020;142:1176–1189. doi: 10.1161/CIRCULATIONAHA.120.048488
    1. Jackson SP, Darbousset R, Schoenwaelder SM. Thromboinflammation: challenges of therapeutically targeting coagulation and other host defense mechanisms. Blood. 2019;133:906–918. doi: 10.1182/blood-2018-11-882993
    1. Agbani EO, Poole AW. Procoagulant platelets: generation, function, and therapeutic targeting in thrombosis. Blood. 2017;130:2171–2179. doi: 10.1182/blood-2017-05-787259
    1. van der Meijden PEJ, Heemskerk JWM. Platelet biology and functions: new concepts and clinical perspectives. Nat Rev Cardiol. 2019;16:166–179. doi: 10.1038/s41569-018-0110-0
    1. Minuz P, Mansueto G, Mazzaferri F, Fava C, Dalbeni A, Ambrosetti MC, Sibani M, Tacconelli E. High rate of pulmonary thromboembolism in patients with SARS-CoV-2 pneumonia [published online June 18, 2020]. Clin Microbiol Infect. doi: 10.1016/j.cmi.2020.06.011
    1. Borghesi A, Maroldi R. COVID-19 outbreak in Italy: experimental chest X-ray scoring system for quantifying and monitoring disease progression. Radiol Med. 2020;125:509–513. doi: 10.1007/s11547-020-01200-3
    1. Buoro S, Moioli V, Seghezzi M, Previtali G, Alessio MG, Simon Lopez R, Ortolani C, Ottomano C, Lippi G. Evaluation and comparison of automated hematology analyzer, flow cytometry, and digital morphology analyzer for monocyte counting. Int J Lab Hematol. 2018;40:577–585. doi: 10.1111/ijlh.12868
    1. Furman MI, Barnard MR, Krueger LA, Fox ML, Shilale EA, Lessard DM, Marchese P, Frelinger AL, Goldberg RJ, Michelson AD. Circulating monocyte-platelet aggregates are an early marker of acute myocardial infarction. J Am Coll Cardiol. 2001;105:2130–2132. doi: 10.1016/S0735-1097(01)01485-1
    1. Giacomazzi A, Degan M, Calabria S, Meneguzzi A, Minuz P. Antiplatelet agents inhibit the generation of platelet-derived microparticles. Front Pharmacol. 2016;7:314 doi: 10.3389/fphar.2016.00314
    1. Mezger M, Nording H, Sauter R, Graf T, Heim C, von Bubnoff N, Ensminger SM, Langer HF. Platelets and immune responses during thromboinflammation. Front Immunol. 2019;10:1731 doi: 10.3389/fimmu.2019.01731
    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. doi: 10.1111/jth.14768
    1. Koupenova M, Corkrey HA, Vitseva O, Manni G, Pang CJ, Clancy L, Yao C, Rade J, Levy D, Wang JP, et al. The role of platelets in mediating a response to human influenza infection. Nat Commun. 2019;10:1780 doi: 10.1038/s41467-019-09607-x
    1. Bronte V, Ugel S, Tinazzi E, Vella A, De Sanctis F, Canè S, Batani V, Trovato R, Fiore A, Petrova V, et al. Baricitinib restrains the immune dysregulation in severe COVID-19 patients [published online August 18, 2020]. J Clin Invest. doi: 10.1172/jci141772
    1. Rondina MT, Brewster BA, Grissom CK, Zimmerman GA, Kastendieck DH, Harris ES, Weyrich AS. In vivo platelet activation in critically ill patients with primary 2009 influenza A(H1N1). Chest. 2012;141:1490–1495. doi: 10.1378/chest.11-2860
    1. Pulavendran S, Rudd JM, Maram P, Thomas PG, Akhilesh R, Malayer JR, Chow VTK, Teluguakula N. Combination therapy targeting platelet activation and virus replication protects mice against lethal influenza pneumonia. Am J Respir Cell Mol Biol. 2019;61:689–701. doi: 10.1165/rcmb.2018-0196OC
    1. Middleton EA, He X-Y, Denorme F, Campbell RA, Ng D, Salvatore SP, Mostyka M, Baxter-Stoltzfus A, Borczuk AC, Loda M, et al. Neutrophil extracellular traps (NETs) contribute to immunothrombosis in COVID-19 acute respiratory distress syndrome. Blood. 2020;136:1169–1179. doi: 10.1182/blood.2020007008
    1. Barnes BJ, Adrover JM, Baxter-Stoltzfus A, Borczuk A, Cools-Lartigue J, Crawford JM, Daßler-Plenker J, Guerci P, Huynh C, Knight JS, et al. Targeting potential drivers of COVID-19: neutrophil extracellular traps. J Exp Med. 2020;217:e20200652 doi: 10.1084/jem.20200652
    1. Sung PS, Hsieh SL. CLEC2 and CLEC5A: pathogenic host factors in acute viral infections. Front Immunol. 2019;10:2867 doi: 10.3389/fimmu.2019.02867
    1. Zellweger RM, Eddy WE, Tang WW, Miller R, Shresta S. CD8 + T cells prevent antigen-induced antibody-dependent enhancement of dengue disease in mice. J Immunol. 2014;193:4117–4124. doi: 10.4049/jimmunol.1401597
    1. Cunin P, Nigrovic PA. Megakaryocytes as immune cells. J Leukoc Biol. 2019;105:1111–1121. doi: 10.1002/JLB.MR0718-261RR
    1. Zhou F, Yu T, Du R, Fan G, Liu Y, Liu Z, Xiang J, Wang Y, Song B, Gu X, et al. Clinical course and risk factors for mortality of adult inpatients with COVID-19 in Wuhan, China: a retrospective cohort study. Lancet. 2020;395:1054–1062. doi: 10.1016/S0140-6736(20)30566-3
    1. Liu K, Fang YY, Deng Y, Liu W, Wang MF, Ma JP, Xiao W, Wang YN, Zhong MH, Li CH, et al. Clinical characteristics of novel coronavirus cases in tertiary hospitals in Hubei Province. Chin Med J (Engl). 2020;133:1025–1031. doi: 10.1097/CM9.0000000000000744
    1. Zhang W, Zhao Y, Zhang F, Wang Q, Li T, Liu Z, Wang J, Qin Y, Zhang X, Yan X, et al. The use of anti-inflammatory drugs in the treatment of people with severe coronavirus disease 2019 (COVID-19): the experience of clinical immunologists from China. Clin Immunol. 2020;214:108393 doi: 10.1016/j.clim.2020.108393
    1. Battinelli EM, Thon JN, Okazaki R, Peters CG, Vijey P, Wilkie AR, Noetzli LJ, Flaumenhaft R, Italiano JE. Megakaryocytes package contents into separate a-granules that are differentially distributed in platelets. Blood Adv. 2019;3:3092–3098. doi: 10.1182/bloodadvances.2018020834
    1. Podolnikova NP, Yakovlev S, Yakubenko VP, Wang X, Gorkun OV, Ugarova TP. The interaction of integrin αIIb β3 with fibrin occurs through multiple binding sites in the α IIb β-propeller domain. J Biol Chem. 2014;289:2371–2383. doi: 10.1074/jbc.M113.518126
    1. Mattheij NJ, Gilio K, van Kruchten R, Jobe SM, Wieschhaus AJ, Chishti AH, Collins P, Heemskerk JW, Cosemans JM. Dual mechanism of integrin αIIbβ3 closure in procoagulant platelets. J Biol Chem. 2013;288:13325–13336. doi: 10.1074/jbc.M112.428359
    1. Raghunathan V, Zilberman-Rudenko J, Olson SR, Lupu F, McCarty OJT, Shatzel JJ. The contact pathway and sepsis. Res Pract Thromb Haemost. 2019;3:331–339. doi: 10.1002/rth2.12217
    1. Renné T, Pozgajová M, Grüner S, Schuh K, Pauer HU, Burfeind P, Gailani D, Nieswandt B. Defective thrombus formation in mice lacking coagulation factor XII. J Exp Med. 2005;202:271–281. doi: 10.1084/jem.20050664
    1. Nechipurenko DY, Receveur N, Yakimenko AO, Shepelyuk TO, Yakusheva AA, Kerimov RR, Obydennyy SI, Eckly A, Léon C, Gachet C, et al. Clot contraction drives the translocation of procoagulant platelets to thrombus surface. Arterioscler Thromb Vasc Biol. 2019;39:37–47. doi: 10.1161/ATVBAHA.118.311390
    1. Ruiz FA, Lea CR, Oldfield E, Docampo R. Human platelet dense granules contain polyphosphate and are similar to acidocalcisomes of bacteria and unicellular eukaryotes. J Biol Chem. 2004;279:44250–44257. doi: 10.1074/jbc.M406261200
    1. Verhoef JJ, Barendrecht AD, Nickel KF, Dijkxhoorn K, Kenne E, Labberton L, McCarty OJ, Schiffelers R, Heijnen HF, Hendrickx AP, et al. Polyphosphate nanoparticles on the platelet surface trigger contact system activation. Blood. 2017;129:1707–1717. doi: 10.1182/blood-s2016-08-734988
    1. Zilberman-Rudenko J, Reitsma SE, Puy C, Rigg RA, Smith SA, Tucker EI, Silasi R, Merkulova A, McCrae KR, Maas C, et al. Factor XII activation promotes platelet consumption in the presence of bacterial-type long-chain polyphosphate in vitro and in vivo. Arterioscler Thromb Vasc Biol. 2018;38:1748–1760. doi: 10.1161/ATVBAHA.118.311193
    1. Lefrançais E, Ortiz-Muñoz G, Caudrillier A, Mallavia B, Liu F, Sayah DM, Thornton EE, Headley MB, David T, Coughlin SR, et al. The lung is a site of platelet biogenesis and a reservoir for haematopoietic progenitors. Nature. 2017;544:105–109. doi: 10.1038/nature21706
    1. Coenen DM, Mastenbroek TG, Cosemans JMEM. Platelet interaction with activated endothelium: mechanistic insights from microfluidics. Blood. 2017;130:2819–2828. doi: 10.1182/blood-2017-04-780825
    1. Gros A, Syvannarath V, Lamrani L, Ollivier V, Loyau S, Goerge T, Nieswandt B, Jandrot-Perrus M, Ho-Tin-Noé B. Single platelets seal neutrophil-induced vascular breaches via GPVI during immune-complex-mediated inflammation in mice. Blood. 2015;126:1017–1026. doi: 10.1182/blood-2014-12-617159
    1. Antoniak S, Owens AP, III, Baunacke M, Williams JC, Lee RD, Weithäuser A, Sheridan PA, Malz R, Luyendyk JP, Esserman DA, et al. PAR-1 contributes to the innate immune response during viral infection. J Clin Invest. 2013;123:1310–1322. doi: 10.1172/JCI66125
    1. Sexton TR, Zhang G, Macaulay TE, Callahan LA, Charnigo R, Vsevolozhskaya OA, Li Z, Smyth S. Ticagrelor reduces thromboinflammatory markers in patients with pneumonia. JACC Basic Transl Sci. 2018;3:435–449. doi: 10.1016/j.jacbts.2018.05.005
    1. Nickel KF, Long AT, Fuchs TA, Butler LM, Renné T. Factor XII as a therapeutic target in thromboembolic and inflammatory diseases. Arterioscler Thromb Vasc Biol. 2017;37:13–20. doi: 10.1161/ATVBAHA.116.308595

Source: PubMed

3
Suscribir