Endothelial injury and dysfunction in COVID-19

Endothelial dysfunction in COVID-19: a position paper of the ESC Working Group for Atherosclerosis and Vascular Biology, and the ESC Council of Basic Cardiovascular Science

Literature - Evans PC, Rainger GE, Mason JC et al., - Cardiovasc Res. 2020 Dec 1;116(14):2177-2184. doi: 10.1093/cvr/cvaa230.

The position paper of the ESC Working Group for Atherosclerosis and Vascular Biology, and the ESC Council of Basic Cardiovascular Science draws attention towards the role of endothelium in the pathophysiology of COVID-19. The article discusses effects of a systemic inflammatory cytokine storm in COVID-19 on endothelial phenotype and function. Possible direct effects of SARS-CoV-2 infection of endothelial cells (ECs) on endothelial dysfunction are addressed as well. Moreover, the authors discuss outstanding questions that need to be addressed to improve our understanding of the role of the vascular endothelium in COVID-19.

Endothelial dysfunction in COVID-19 and its role in inflammation and thrombosis

The hyperinflammatory and procoagulatory state which has been observed in COVID-19 patients suggests that the endothelium plays an important role in this disease. SARS-CoV-2 infection can lead to dysfunctional or injured endothelium. Endothelial dysfunction can have an impact on multiple levels, including reduced integrity, increased permeability, activation of an immune response and thrombosis [1-4].

The endothelium plays a crucial role in an amplification loop of the inflammatory response in COVID-19 [5,6]. The endothelium is exposed to proinflammatory cytokines due to a systemic inflammatory cytokine storm. This induces -via a cascade of processes- leucocyte recruitment and inflammation [7]. This process can also cause death of ECs, which in turn may lead to increased vascular permeability and end-organ damage. This raises the question whether anti-inflammatory therapies that are currently used in the treatment of chronic inflammatory and autoimmune diseases may be beneficial in the treatment of COVID-19. On the other hand, it can be argued that anti-inflammatory agents can be counterproductive in the immune response against the virus. The RECOVERY trial showed positive effects of the use of low dose dexamethasone in hospitalized COVID-19 patients [8].

The thrombotic and coagulant properties of endothelium change when it becomes dysfunctional [7]. A prothrombotic and proinflammatory state can cause the recruitment and activation of platelets to intact endothelial monolayers [9]. This can subsequently induce secondary recruitment of circulating leucocytes [10-12]. The authors argue that it is reasonable to assume that aggregates of ECs, platelets and leucocytes on the walls of smaller vessels can lead to loss of microvascular perfusion in organs.

SARS-CoV-2 infection in endothelial cells

Besides indirect effects of COVID-19 on endothelial function through a systemic inflammatory state, evidence from several studies point to SARS-CoV-2 infection of ECs as a possible direct trigger of endothelial adverse effects in COVID-19 [13-18]. SARS-CoV-2 infection of ECs has been associated with EC apoptosis, which may point to a direct mechanism of how COVID-19 can cause endothelial dysfunction [19]. In addition, an inflammatory multisystem syndrome has been associated with COVID-19 in children [20,21]. This syndrome shows similarities to Kawasaki disease, a systemic vasculitis which particularly targets coronary arteries and the myocardium. However, important differences to Kawasaki disease were reported [22].

The authors also refer to a study that suggests that pericytes may act as a direct target for SARS-CoV-2 infection [23]. Pericytes are essential for the maintenance of the integrity of endothelium. Apoptosis of pericytes could therefore play an important role in microvascular dysfunction and coagulopathy.

Therapeutic targets and position statements

The authors emphasize the need for prospective clinical studies with clearly defined CV endpoints in COVID-19 patients. Such studies should also investigate interventions with current CV drugs which could potentially reduce endothelial dysfunction in COVID-19. Potential candidates include modulators of RAAS (ACEi, ARBs, ACE2), anti-inflammatory drugs (cytokine inhibitors, dexamethasone, statins), inhibitors of reactive oxygen species (ROS) and apoptosis (statins), platelet inhibitors and anticoagulants.

Finally, the authors provide five position statements on role of endothelium in the pathophysiology of COVID-19. The detailed statements can be found in the original position paper. In short, these statements are: i) endothelial biomarkers and EC function should be monitored in studies to assess vascular consequences of COVID-19; ii) the significance of SARS-CoV-2-mediated endocytosis and down-regulation of ACE1 on CV health needs to be clarified; iii) studies of EC activation, leucocyte recruitment, platelet activation, turnover and signaling in COVID-19 are needed to reveal the principle effects of SARS-CoV-2 on endothelial function. Since ageing is an established determinant of COVID-19 outcome, features of aging on SARS-CoV-2 infection of ECs should be investigated; iv) effects of common CV drugs on endothelial responses in COVID-19 should be studied; v) long-term CV effects of COVID-19 after recovery should be determined during patient follow-up. Endothelial function testing can be valuable for early detection of vascular consequences of COVID-19.

References

1. Souilhol C, Harmsen MC, Evans PC, Krenning G. Endothelial–mesenchymal transition in atherosclerosis. Cardiovasc Res 2018;114:565–577.

2. Souilhol C, Serbanovic-Canic J, Fragiadaki M, Chico TJ, Ridger V, Roddie H, Evans PC. Endothelial responses to shear stress in atherosclerosis: a novel role for developmental genes. Nat Rev Cardiol 2020;17:52–63.

3. Mundi S, Massaro M, Scoditti E, Carluccio MA, van Hinsbergh VWM, Iruela-Arispe ML, De Caterina R. Endothelial permeability, LDL deposition, and cardiovascular risk factors—a review. Cardiovasc Res 2018;114:35–52.

4. Li X, Sun X, Carmeliet P. Hallmarks of endothelial cell metabolism in health and disease. Cell Metab 2019;30:414–433.

5. Huang C, Wang Y, Li X, Ren L, Zhao J, Hu Y, Zhang L, Fan G, Xu J, Gu X, Cheng Z, Yu T, Xia J, Wei Y, Wu W, Xie X, Yin W, Li H, Liu M, Xiao Y, Gao H, Guo L, Xie J, Wang G, Jiang R, Gao Z, Jin Q, Wang J, Cao B. Clinical features of patients infected with 2019 novel coronavirus in Wuhan, China. Lancet 2020;395:497–506.

6. Chen X, Zhao B, Qu Y, Chen Y, Xiong J, Feng Y, Men D, Huang Q, Liu Y, Yang B, Ding J, Li F. 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;doi: 10.1093/cid/ciaa449.

7. Chousterman BG, Swirski FK, Weber GF. Cytokine storm and sepsis disease pathogenesis. Semin Immunopathol 2017;39:517–528.

8. Horby P, Lim WS, Emberson J, Mafham M, Bell J, Linsell L, Staplin N, Brightling C, Ustianowski A, Elmahi E, Prudon B, Green C, Felton T, Chadwick D, Rege K, Fegan C, Chappell LC, Faust SN, Jaki T, Jeffery K, Montgomery A, Rowan K, Juszczak E, Baillie JK, Haynes R, Landray MJ. Effect of dexamethasone in hospitalized patients with COVID-19: preliminary report. N Engl J Med. 2020 Jul 17;NEJMoa2021436.doi: 10.1056/NEJMoa2021436.

9. Tull SP, Anderson SI, Hughan SC, Watson SP, Nash GB, Rainger GE. Cellular pathology of atherosclerosis: smooth muscle cells promote adhesion of platelets to cocultured endothelial cells. Circ Res 2006;98:98–104.

10. Kuckleburg CJ, Yates CM, Kalia N, Zhao Y, Nash GB, Watson SP, Rainger GE. Endothelial cell-borne platelet bridges selectively recruit monocytes in human and mouse models of vascular inflammation. Cardiovasc Res 2011;91:134–141.

11. Alharbi A, Thompson JP, Brindle NP, Stover CM. Ex vivo modelling of the formation of inflammatory platelet–leucocyte aggregates and their adhesion on endothelial cells, an early event in sepsis. Clin Exp Med 2019;19:321–337.

12. Chimen M, Evryviadou A, Box CL, Harrison MJ, Hazeldine J, Dib LH, Kuravi SJ, Payne H, Price JMJ, Kavanagh D, Iqbal AJ, Lax S, Kalia N, Brill A, Thomas SG, Belli A, Crombie N, Adams RA, Evans SA, Deckmyn H, Lord JM, Harrison P, Watson SP, Nash GB, Rainger GE. Appropriation of GPlb alpha from platelet-derived extracellular vesicles supports monocyte recruitment in systemic inflammation. Haematologica 2020;105:1248–1261.

13. Puelles VG, Lutgehetmann M, Lindenmeyer MT, Sperhake JP, Wong MN, Allweiss L, Chilla S, Heinemann A, Wanner N, Liu S, Braun F, Lu S, Pfefferle S, Schroder AS, Edler C, Gross O, Glatzel M, Wichmann D, Wiech T, Kluge S, Pueschel K, Aepfelbacher M, Huber TB. Multiorgan and renal tropism of SARS-CoV-2. N Engl J Med 2020;doi: 10.1056/NEJMc2011400.

14. Monteil V, Kwon H, Prado P, Hagelkruys A, Wimmer RA, Stahl M, Leopoldi A, Garreta E, Hurtado Del Pozo C, Prosper F, Romero JP, Wirnsberger G, Zhang H, Slutsky AS, Conder R, Montserrat N, Mirazimi A, Penninger JM. Inhibition of SARSCoV-2 infections in engineered human tissues using clinical-grade soluble human ACE2. Cell 2020;181:905–913.

15. Ackermann M, Verleden SE, Kuehnel M, Haverich A, Welte T, Laenger F, Vanstapel A, Werlein C, Stark H, Tzankov A, Li WW, Li VW, Mentzer SJ, Jonigk D. Pulmonary vascular endothelialitis, thrombosis, and angiogenesis in Covid-19. N Engl J Med 2020;383:120–128.

16. Menter T, Haslbauer JD, Nienhold R, Savic S, Hopfer H, Deigendesch N, Frank S, Turek D, Willi N, Pargger H, Bassetti S, Leuppi JD, Cathomas G, Tolnay M, Mertz KD, Tzankov A. Post-mortem examination of COVID19 patients reveals diffuse alveolar damage with severe capillary congestion and variegated findings of lungs and other organs suggesting vascular dysfunction. Histopathology 2020;doi: 10.1111/his.14134.

17. Colmenero I, Santonja C, Alonso-Ria~no M, Noguera-Morel L, Herna´ndez-Martı´n A, Andina D, Wiesner T, Rodrı´guez-Peralto JL, Requena L, Torrelo A. SARS-CoV-2 endothelial infection causes COVID-19 chilblains: histopathological, immunohistochemical and ultrastructural study of 7 paediatric cases. Br J Dermatol 2020;doi: 10.1111/bjd.19327.

18. Khider L, Gendron N, Goudot G, Chocron R, Hauw-Berlemont C, Cheng C, Rivet N, Pere H, Roffe A, Clerc S, Lebeaux D, Debuc B, Veyer D, Rance B, Gaussem P, Bertil S, Badoual C, Juvin P, Planquette B, Messas E, Sanchez O, Hulot JS, Diehl JL, Mirault T, Smadja DM. Curative anticoagulation prevents endothelial lesion in COVID-19 patients. J Thromb Haemost 2020;doi: 10.1111/jth.14968.

19. 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.

20. Verdoni L, Mazza A, Gervasoni A, Martelli L, Ruggeri M, Ciuffreda M, Bonanomi E, D’Antiga L. An outbreak of severe Kawasaki-like disease at the Italian epicentre of the SARS-CoV-2 epidemic: an observational cohort study. Lancet 2020;395:1771–1778.

21. Riphagen S, Gomez X, Gonzalez-Martinez C, Wilkinson N, Theocharis P. Hyperinflammatory shock in children during COVID-19 pandemic. Lancet 2020;395:1607–1608.

22. Whittaker E, Bamford A, Kenny J, Kaforou M, Jones CE, Shah P, Ramnarayan P, Fraisse A, Miller O, Davies P, Kucera F, Brierley J, McDougall M, Carter M, Tremoulet A, Shimizu C, Herberg J, Burns JC, Lyall H, Levin M, PIMS-TS Study Group and EUCLIDS and PERFORM Consortia. Clinical characteristics of 58 children with a pediatric inflammatory multisystem syndrome temporally associated with SARS-CoV-2. JAMA 2020;doi: 10.1001/jama.2020.10369.

23. He L, Mäe MA, Sun Y, Muhl L, Nahar K, Liébanas EV, Fagerlund MJ, Oldner A, Liu J, Genové G, Pietilä R, Zhang L, Xie Y, Leptidis S, Mocci G, Stritt S, Osman A, Anisimov A, Hemanthakumar KA, Räsenen M, Björkegren J, Vanlandewijck M, Blomgren K, Hansson E, Mäkinen T, Peng X-R, Arnold TD, Alitalo K, Eriksson LI, Lendahl U, Betsholtz C. Pericyte-specific vascular expression of SARS-CoV-2 receptor ACE2—implications for microvascular inflammation and hypercoagulopathy in COVID-19 patients. bioRxiv 2020: 2020.2005.2011.088500.

Find this article online at Cardiovascular Research

Facebook Comments

Register

We’re glad to see you’re enjoying PACE-CME…
but how about a more personalized experience?

Register for free