Abstract

The COVID-19 pandemic is an unprecedented healthcare emergency causing mortality and illness across the world. Although primarily affecting the lungs, the SARS-CoV-2 virus also affects the cardiovascular system. In addition to cardiac effects, e.g. myocarditis, arrhythmias, and myocardial damage, the vasculature is affected in COVID-19, both directly by the SARS-CoV-2 virus, and indirectly as a result of a systemic inflammatory cytokine storm. This includes the role of the vascular endothelium in the recruitment of inflammatory leucocytes where they contribute to tissue damage and cytokine release, which are key drivers of acute respiratory distress syndrome (ARDS), in disseminated intravascular coagulation, and cardiovascular complications in COVID-19. There is also evidence linking endothelial cells (ECs) to SARS-CoV-2 infection including: (i) the expression and function of its receptor angiotensin-converting enzyme 2 (ACE2) in the vasculature; (ii) the prevalence of a Kawasaki disease-like syndrome (vasculitis) in COVID-19; and (iii) evidence of EC infection with SARS-CoV-2 in patients with fatal COVID-19. Here, the Working Group on Atherosclerosis and Vascular Biology together with the Council of Basic Cardiovascular Science of the European Society of Cardiology provide a Position Statement on the importance of the endothelium in the underlying pathophysiology behind the clinical presentation in COVID-19 and identify key questions for future research to address. We propose that endothelial biomarkers and tests of function (e.g. flow-mediated dilatation) should be evaluated for their usefulness in the risk stratification of COVID-19 patients. A better understanding of the effects of SARS-CoV-2 on endothelial biology in both the micro- and macrovasculature is required, and endothelial function testing should be considered in the follow-up of convalescent COVID-19 patients for early detection of long-term cardiovascular complications.

This paper was handled by Executive Deputy Editor Charalambos Antoniades.

Introduction

Coronavirus disease 2019 (COVID-19) is caused by a single-stranded RNA virus called severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) that probably crossed from bats to humans following a gain-of-function mutation in the spike (S) protein that allows infection of human cells.1 Though primarily known as a disease affecting the respiratory system, cardiovascular complications are common in COVID-19.2,3 These include myocarditis and myocardial injury that may lead to heart failure. Myocardial infarction (MI) and Takotsubo syndrome has been reported in patients with COVID-19.4,5 An increase in troponin levels in COVID-19 is associated with more severe disease and mortality, underlining myocardial injury as a prognostic factor.6 Increased levels of N-terminal probrain natriuretic peptide (NT-proBNP) is also an independent predictor of in-hospital death in COVID-19 patients, but at lower levels than in heart failure,7 suggesting that relatively mild alteration in cardiac function may critically determine outcome in COVID-19. In addition, arrhythmias are remarkably prevalent among patients with COVID-19, with an incidence of 16.7% reported in a Chinese cohort.8 Cardiomyocytes are known to express angiotensin-converting enzyme 2 (ACE2),9 which is the receptor for SARS-CoV-2; however, direct evidence of cardiomyocyte infection is lacking.

In addition, underlying cardiovascular disease may aggravate the clinical course of disease in COVID-19. Meta-analysis of data from Chinese cohorts revealed that fatality rates in hospitalized patients with COVID-19 were elevated in those with cardiovascular disease (10.5%), diabetes (7.3%), or hypertension (6%) compared with patients without comorbidities (0.9%).10 Cohorts of patients from Italy and the USA11 also show that hypertension, diabetes, and obesity are common comorbidities.

Increased D-dimer (fibrin degradation product) levels is a marker of adverse COVID-19 outcomes,12 and disseminated intravascular coagulation with significant risk of venous thrombo-embolism (VTE) and ischaemic stroke has been reported.13–15 A study of systematic duplex ultrasound in 26 severe COVID-19 patients reported a cumulative VTE incidence of 69% despite anticoagulation treatment.16 Ischaemic stroke has been linked to COVID-19 progression because it had a higher incidence in patients with severe disease compared with those with milder symptoms.17 A report of six ischaemic stroke cases from the UK revealed an association with elevated D-dimers, systemic inflammatory changes, and antiphospholipid antibodies, which are indicative of hypercoagulability.18 However, caution should be exercised in attributing stroke to viral infection since there were several potential confounding factors including hypertension and atrial fibrillation, which enhance stroke risk.

We refer the reader to Guzik et al.19 for a more complete description of cardiac effects in COVID-19. The aim of this Position Statement from the Working Group on Atherosclerosis and Vascular Biology together with the Council of Basic Cardiovascular Science of the European Society of Cardiology is to draw attention to the importance of the endothelium in COVID-19 and to encourage research on endothelial dysfunction and biomarkers to tackle the COVID-19 pandemic and its potential long-term cardiovascular complications.

Endothelium and COVID-19

The characteristic hyperinflammatory and procoagulatory state of COVID-19 implies a critical role of the endothelium, both as an effector contributing to inflammation and thrombosis, and as a target organ, whose dysfunction may contribute to poor outcome.20 Of particular note, there is also evidence of SARS-CoV-2 infection of vascular endothelial cells (ECs).21–24

The vascular endothelium forms a critical interface between the circulatory system and underlying tissues, and has vital and ubiquitous roles in cardiovascular homeostasis by regulating the transport of cells, nutrients, and metabolites between the circulation and underlying tissues.25 Several risk factors for cardiovascular disease including diabetes, obesity, dyslipidaemia, smoking, and disturbed blood flow can induce EC dysfunction which is characterized by a spectrum of phenotypes.26–28 These include loss of integrity (e.g. via apoptosis) which is associated with increased permeability; induction of cytokines and adhesion molecules to capture inflammatory cells from the circulation; metabolic changes; a prothrombotic phenotype; and de-differentiation.26,29 This is exemplified in multiple studies showing that quantification of EC function provides a useful marker for early disease detection and stratification of patients with cardiovascular disease.30,31 The European Society of Cardiology recently reviewed this subject and called for further investigation of EC pathophysiological states, optimization and standardization of methodologies for clinical measurement of EC function, as well as larger clinical trials to establish reference values and assess clinical utility.25

Emerging evidence indicates that EC dysfunction is a central feature of COVID-19. This is evidenced by the critical role of the vascular endothelium in inflammation, which is the key driver of cytokine dysregulation in ARDS as well as multiple cardiovascular pathologies. Additionally, the prothrombotic phenotype and disseminated intravascular coagulation observed in COVID-19 reflect dysfunction of ECs, which enhances thrombosis by reduced integrity leading to exposure of prothrombotic subendothelial material, capture of platelets and regulation of clotting cascades, thrombin activation, and fibrin production.32

There are several other lines of evidence that substantiate the role of endothelium in COVID-19 (Figure 1) as detailed below.

Endothelial dysregulation by SARS-CoV-2. Healthy endothelium (left) is characterized by quiescence, intact junctions, anticoagulant anti-inflammatory phenotype, and an intact vasodilation phenotype. The cell in the centre (endothelitis) is infected with SARS-CoV-2, whereas the cells to the right have been activated as a result of cytokine release and activation of prothrombotic pathways. Infection with SARS-CoV-2 is via ACE2 which is subsequently endocytosed, potentially reducing ACE2-mediated regulation of vascular tone. SARS-CoV-2 infection causes endothelial dysfunction at multiple levels including inflammatory activation, cytokine storm, leucocyte infiltration, increased permeability, thrombosis, platelet aggregation, vasoconstriction, production of reactive oxygen species (ROS), and apoptosis.
Figure 1

Endothelial dysregulation by SARS-CoV-2. Healthy endothelium (left) is characterized by quiescence, intact junctions, anticoagulant anti-inflammatory phenotype, and an intact vasodilation phenotype. The cell in the centre (endothelitis) is infected with SARS-CoV-2, whereas the cells to the right have been activated as a result of cytokine release and activation of prothrombotic pathways. Infection with SARS-CoV-2 is via ACE2 which is subsequently endocytosed, potentially reducing ACE2-mediated regulation of vascular tone. SARS-CoV-2 infection causes endothelial dysfunction at multiple levels including inflammatory activation, cytokine storm, leucocyte infiltration, increased permeability, thrombosis, platelet aggregation, vasoconstriction, production of reactive oxygen species (ROS), and apoptosis.

Recruitment of leucocytes, immune response, and tissue injury

Leucocytes play an important role in the pathogenesis of SARS-CoV-2. The importance of the leucocyte–EC axis is exemplified by the observations that patients with severe disease demonstrate a marked increase in blood neutrophils which is associated with lymphopenia, with both CD4+ T cells and CD8+ T cells being lower in severe compared with moderate cases.33 Moreover, histological examination of a severe case who died of SARS-CoV-2 demonstrated lung interstitial mononuclear inflammatory infiltrates, dominated by lymphocytes.34 Through the systemic inflammatory response in COVID-19, referred to as the cytokine storm, or cytokine release syndrome, the endothelium will be directly exposed to proinflammatory cytokines that initiate transcriptional programmes, that in turn induce adhesion molecules and chemokines, driving leucocyte recruitment and inflammation.35 This process can also lead to EC death that contributes to increased vascular permeability and end-organ damage. Through an amplification loop of the inflammatory response, the endothelium may constitute a significant source of proinflammatory cytokines, such as interleukin (IL)-1 and IL-6, that characterize the cytokine storm in COVID-19.36,37 Since the process of lymphocyte trafficking in chronic inflammatory and autoimmune diseases is a major therapeutic target, it is possible that anti-inflammatory therapies developed for these conditions could be repurposed to treat SARS-CoV-2 infection.

It is uncertain whether modulating EC function during the inflammatory response to SARS-CoV-2 would be beneficial, with the dilemma that anti-inflammatory agents might be counterproductive and compromise the immune response against the virus. However, in mouse models of pathogenic influenza and respiratory syncytial virus (RSV) infection, where there is evidence that the EC orchestrate a CD8+ T cell-mediated cytokine storm, agonists of the S1P1 receptor are effective in improving survival.38–40 It is also noteworthy that adiponectin is protective in models of sepsis and ARDS through suppression of EC activation.41 Such observations raise the question of whether the immune response itself may contribute to the chronic inflammatory process in severe SARS-CoV-2 infection. The immunology of SARS-CoV-2 infection has recently been comprehensively reviewed by Vabret et al.42 who noted that in SARS-CoV-1 patients, survival was associated with immunity skewed towards a CD8 cytotoxic response.43 Indeed, there is evidence that expansion of virus-specific CD4 T cells and a robust Th2 response (including increases in plasma IL-4, IL-5, and IL-10) are associated with death in SARS-CoV-1.44 Whether this is the case in SARS-CoV-2 remains to be established. It does, however, appear that the adaptive immune response is an important arbiter of outcomes in SARS-CoV-2, as there is a reported association between plasma IgA titres and severity of disease.45 It is possible that in SARS-CoV-2, antibody-dependent enhancement (ADE) of inflammation plays a role in EC activation and pathology. In this phenomenon, non-neutralizing antibodies facilitate Fc-mediated uptake of virus into macrophages, resulting in their activation and liberation of inflammatory cytokines, leading to the secondary recruitment of inflammatory leucocytes by local ECs. The phenomenon of ADE is readily demonstrable in a number of experimental models of SARS infection, but there is currently no evidence that ADE contributes to pathology in SARS-CoV-2.42 Since the process of EC-dependent leucocyte trafficking in chronic inflammatory and autoimmune diseases is a major therapeutic target, it is possible that anti-inflammatory therapies developed for these conditions could be repurposed to treat SARS-CoV-2 infection. Indeed, the recent success of low dose dexamethasone in the RECOVERY trial, and its adoption as a front-line therapy for SARS-CoV-2 in the UK, is an exemplar of the utility of such an approach (see below).

Endothelium and thrombosis

When dysfunctional, the thrombotic and coagulant properties of the endothelium change.35 In particular, a decreased antiaggregatory prostacyclin production from ECs and an increased proaggregatory thromboxane synthesis from activated platelets46 may skew the homeostatic situation towards a prothrombotic and proinflammatory phenotype. Interestingly, under some inflammatory conditions, ECs express adhesion receptors such as von Willebrand factor on their surface. Both in vitro and in vivo, these conditions have been demonstrated to support the recruitment and activation of platelets to intact endothelial monolayers.47 This in turn can lead to the platelet-dependent secondary recruitment of circulating leucocytes, either by leucocyte interactions with platelets adherent to ECs,48 by the recruitment of circulating heterotypic aggregates of platelets and leucocytes,49 or by the transfer of platelet-borne receptors such as glycoprotein Ib (GPIb) to the leucocyte membrane by platelet-derived microvesicles.50 It is reasonable to assume that such tricellular aggregates (EC–platelet–leucocyte) on the walls of smaller vessels would be sufficient to cause loss of microvascular perfusion in the lungs and other organs. Indeed, compromise of myocardial perfusion could play a role in the elevated levels of troponin evident in many SARS-CoV-2 patients. Intravascular thrombosis and coagulation, in addition, may further damage the endothelium and contribute to endothelial inflammation and dysfunction.35 In some patients with COVID-19, severe microvascular endothelial injury directly mediated by activation of the alternative and lectin complement pathways has been demonstrated and associated with a procoagulant state.51 It remains to be established, however, whether anticoagulation or platelet inhibition in COVID-19 improves endothelial function, and if dampened endothelial inflammation would attenuate the procoagulant state in COVID-19.

ACE2 expression and function in endothelium

ACE2 is intimately linked to cardiovascular physiology as part of the renin–angiotensin–aldosterone system (RAAS), which controls blood pressure by altering vascular tone and function. The related molecule ACE converts angiotensin (Ang) I to Ang II, which promotes vasoconstriction, hypertension, and vascular inflammation. Because of these properties, antihypertensive drugs have been developed to reduce the production [ACE inhibitors (ACEIs)] or downstream effects [Ang II receptor blockers (ARBs)] of Ang II. The effects of ACE are opposed by ACE2, which converts Ang II into Ang 1-7 molecules thereby promoting vasodilatation and reducing hypertension.52 Since ACE2 is expressed in cells of the cardiovascular system,53 there has been considerable interest in the hypothesis that this class of antihypertensive drugs may increase the risk of SARS-CoV-2 infection by increasing the expression of ACE2 in vascular cells.54 However, population-based studies revealed that ACEIs and ARBs do not enhance the risk of COVID-19 or disease severity,55,56 and the European Society of Cardiology and other learned societies recommend that patients should continue with their usual antihypertensive medications during the pandemic [https://www.escardio.org/Councils/Council-on-Hypertension-(CHT)/News/position-statement-of-the-esc-council-on-hypertension-on-ace-inhibitors-and-ang].

The S protein of the coronaviruses mediates viral entry into target cells. Entry depends on physical interactions of the surface unit, S1, of the S protein with a host cell receptor, which facilitates viral attachment to the surface of the target cells. SARS-CoV-2 engages ACE2 as the primary receptor,57–59 and hence it is plausible that COVID-19 may cause reduced bioavailability of ACE2 due to endosomal/lysosomal processing.52 Treatment of COVID-19 patients with recombinant ACE2 is currently under clinical trial because this may act as a decoy receptor,60 hence limiting viral entry. It is notable that recombinant ACE2 treatment may have additional beneficial effects, by increasing its bioavailability at the endothelial surface. The S2 domain of the S protein facilitates membrane fusion, which requires conformational flexibility, achieved by proteolytic cleavages. The cleavage event employs the cellular transmembrane protease, serine 2 (TMPRSS2), and has profound implications on virulence. A TMPRSS2 inhibitor, approved for clinical use, was shown to block entry and might constitute a treatment option.57 It has been reported that TMPRSS2 expression is below the level of detection in microvascular ECs61 and only up-regulated in ECs actively undergoing angiogenic or tubulogenic responses. Further studies are needed to better understand the physiological expression and function of TMPRSS2 in adult ECs. However, the expression of TMPRSS2 alone may not be predictive of its function since serine proteases, such as TMPRSS2, are regulated by nitrosylation.62 Therefore it is plausible that enothelial nitric oxide sytnase (eNOS) activity and subsequent production of nitric oxide (NO) may influence viral infection of ECs by altering TMPRSS2 activity. Taken together, the S protein mediates entry by connecting the virus to the plasma membrane, and by catalysing subsequent virus–cell membrane fusions.

SARS-CoV-2-induced endothelitis

In addition to the respiratory tract, SARS-CoV-2 viral load is detected in the kidneys, liver, heart, and brain,63 which are all highly vascularized tissues. Indeed, Monteil et al. provided an early indication of SARS-CoV-2 tropism for vascularized tissues by demonstrating that this virus can infect human blood vessel and kidney organoids via ACE2.59 It was suggested by Varga et al., by electron microscopy and histology, that SARS-CoV-2 can be detected in ECs of the kidney (glomerular capillaries), small bowel, lung, and myocardium,21 but these data were recently disputed.64 Ackerman et al.22 have shown abnormalities within the pulmonary microvasculature with congestion and microthrombi similar to Menter et al.,23 and by electron microscopy there is endothelial injury and congestion with cell fragments and degenerate organelles in the lumen. Scanning electron microscopy of corrosion casts shows microvasculature of larger diameter with an irregular surface, which may be due to endothelial injury and/or platelet aggregates/fibrin. Intriguingly there was also evidence of intussusceptive angiogenesis, and the authors speculate that this feature may distinguish the pulmonary pathobiology of COVID-19 from other viral infections. SARS-CoV-2 has also been detected in skin ECs,24 and circulating ECs are elevated in patients admitted to hospital with COVID-19.65

Taken together, these studies point to endothelial SARS-CoV-2 infection as a possible direct trigger of endothelial adverse effects in COVID-19. Indeed, SARS-CoV-2-infected endothelium has been associated with EC apoptosis, suggesting a possible mechanism though which the endothelium may become dysfunctional in COVID-19.21 The recent reports of a Kawasaki disease-like syndrome associated with COVID-19 infection in children highlights the importance of the virus on the vasculature.66,67 Kawasaki disease is a systemic vasculitis most commonly seen in children which particularly targets the myocardium and coronary arteries. Although the aetiology of Kawasaki disease is unknown, infectious agents including RNA viruses have been previously postulated as the cause, and the first link to a coronavirus infection was published in 2005.68 The current outbreaks following infection by SARS-CoV-2 are the subject of intense investigation. A recently reported group of 58 hospitalized children were diagnosed with paediatric inflammatory multisystem syndrome temporally associated with severe acute respiratory syndrome coronavirus 2 (PIMS-TS). Of note, important differences were noted when compared with Kawasaki disease.69 Moreover, anecdotal cases with evidence of medium and large vessel vasculitis suggest that similar complications may be seen in some adults with COVID-19.

Pericytes

Pericytes are multifunctional mural cells of the microvasculature and are essential for the maintenance of the integrity of the endothelium.70 Studies suggest that they are involved in COVID-19-related vasculopathy. Recent single-cell or single-nucleus RNA sequencing analyses have shown that ACE2 is highly expressed in pericytes of various organs, such as the heart, both in mouse and in man.71,72 In alveolar capillaries of SARS-CoV-2-infected lung, pericytes are markedly decreased, probably through apoptosis.73 In a genetically modified mouse model with pericyte deficiency (pdgf-bret/ret mouse74) induced by deletion of the PDGF-B (platelet-derived growth factor-B) retention motif, loss of pericyte-induced thrombogenic reactions was seen in ECs.72 Therefore, pericytes, by acting as a direct target for SARS-CoV-2 infection, could play a crucial role in microvascular dysfunction and coagulopathy. It is suggested that a permeable endothelial barrier as observed in hypertension, diabetes, and obesity, which are comorbidity factors in severe cases of COVID-19, allows the virus to reach the pericytes.72

Therapeutic targets

It remains to be determined whether the endothelial dysfunction and injury seen in COVID-19 predominantly reflects direct infection of ECs by SARS-CoV-2 or indirect bystander injury by factors including cytokines, leucocytes, neutrophil nets, and complement activation.75 Nevertheless, an important consideration is the effect of current cardiovascular drugs in this setting (Figure 2). On the one hand, they may offer enhanced endothelial protection, while on the other they may increase endothelial susceptibility. To date, there are only retrospective data to rely upon, and prospective clinical studies in COVID-19 with clearly defined cardiovascular endpoints are required. Drugs including HMG-CoA reductase inhibitors (statins), alpha- and beta-adrenergic blockers, and RAAS antagonists are widely prescribed in those with diabetes mellitus, hypertension, and coronary artery disease, groups known to be at the highest risk from COVID-19. Understanding how these drugs influence outcomes is therefore essential.

Potential interventions to reduce endothelial injury and activation. Endothelial infection with SARS-CoV-2 causes dysregulation of the RAAS, apoptosis, thrombosis, and inflammation (red tones). Several interventions (green tones) can reduce endothelial dysfunction in COVID-19, including modulators of the RAAS (ACE-i, ARBs, ACE2); anti-inflammatory molecules (cytokine inhibitors, dexamethasone, statins); inhibitors of ROS/apoptosis (statins); platelet inhibitors; and anticoagulants. A healthy lifestyle may also reduce endothelial dysfunction in patients with COVID-9.
Figure 2

Potential interventions to reduce endothelial injury and activation. Endothelial infection with SARS-CoV-2 causes dysregulation of the RAAS, apoptosis, thrombosis, and inflammation (red tones). Several interventions (green tones) can reduce endothelial dysfunction in COVID-19, including modulators of the RAAS (ACE-i, ARBs, ACE2); anti-inflammatory molecules (cytokine inhibitors, dexamethasone, statins); inhibitors of ROS/apoptosis (statins); platelet inhibitors; and anticoagulants. A healthy lifestyle may also reduce endothelial dysfunction in patients with COVID-9.

In pre-clinical studies, statins increase expression of ACE2. However, in disease models this may reflect a return to normal levels. Moreover, enhanced expression of ACE2 has cardiovascular benefit.76 Previously observed beneficial effects in influenza and actions of statins including their ability to reduce CD147 expression, optimize lipid raft function, regulate autophagy, minimize endothelial activation, down-regulate prothrombotic pathways, and enhance antithrombotic effects, alongside immunomodulatory actions, suggest that statins may exert important endothelial protective effects both against and during SARS-CoV-2 infection.77–79COVID-19 clinical trials incorporating statins (NCT04333407, NCT04348695, and NCT04380402) have commenced and the results are awaited with interest.

Reported beneficial effects of beta-adrenergic blockers in ARDS and respiratory failure, alongside a potential ability to reduce viral entry by down-regulating ACE2, have resulted in these drugs being suggested as an adjunct therapy for COVID-19, even in those without a primary indication.80 Retrospective studies underway will help inform this hypothesis. An intriguing large, retrospective analysis of alpha-1-adrenergic receptor antagonists in patients suffering from ARDS or pneumonia has revealed that those incidentally prescribed alpha-blockers were less likely to require ventilation or to die following onset of ventilator support. In contrast, beta-adrenergic blockers had no effect. The clinical study was inspired by pre-clinical data, which demonstrated that alpha-blockade can prevent the ARDS-associated cytokine storm and death in mice by interfering with a catecholamine loop.81 These findings suggest that alpha-1-adrenergic receptor antagonists merit retrospective and prospective analysis in COVID-19. The identification of the SARS-CoV-2-associated cytokine storm as a potential therapeutic target is also supported by the early data from the dexamethasone arm of the COVID-19 RECOVERY trial.82

The ability of SARS-CoV-2 to utilize ACE2 as a co-receptor for cellular entry has led to significant interest in the impact of ACEIs and ARBs. Similarly to statins, these commonly used drugs enhance ACE2.76 Although clinical data are sparse, ACEIs and ARBs are reported to improve outcomes in ARDS.83 There has also been speculation that a dysfunctional RAAS is important for COVID-19 disease pathogenesis.84 Initial concern regarding potential susceptibility to SARS-CoV-2 infection and/or exacerbation of its effects in those taking ACEIs or ARBs have been allayed by retrospective clinical studies.85–87 Current advice is to continue taking these drugs prescribed for hypertension, cardiac failure, and chronic renal disease, and for physicians to prescribe them for new clinical indications as normal.84 Prospective studies and clinical trials are now urgently needed. Two clinical trials will study the impact of losartan on COVID-19 (NCT04311177 and NCT04312009). Interest will also focus upon Ang 1-7 peptides, ACE2 itself,76,84 and monoclonal antibodies that prevent SARS-CoV-2 binding to ACE2.88

Position statements

Further research is urgently needed to combat the COVID-19 pandemic, and we emphasize that the role of the vascular endothelium requires close scrutiny. There are today several outstanding questions that need to be addressed to elucidate more precisely the role of EC in COVID-19 and to investigate potential routes to clinical translation.

  • Endothelial biomarkers and tests of function (e.g. flow-mediated dilation, arterial stiffness) should be monitored in studies of COVID-19 outcome and treatment effects. High-quality data collection is needed, with follow-up studies amongst the survivors of acute infection since there are few or no data available on EC function testing in COVID-19. Indeed, collaborative networks have already been established to analyse RNA biomarkers89 and arterial stiffness (CARTESIAN STUDY) to assess vascular consequences of COVID-19. This may help to enable stratification of COVID-19 patients with the highest prothrombotic and cardiovascular risk and allow tailored treatments.

  • The significance of SARS-CoV-2-mediated endocytosis and down-regulation of ACE2 on cardiovascular health is uncertain, but data from ongoing clinical trials to test recombinant ACE2 may be instrumental in addressing this question.

  • The principle effects of SARS-CoV-2 on endothelial function should be determined, including studies of EC activation, leucocyte recruitment, platelet activation, turnover, signalling, etc. ECs from both the micro- and macrovasculature should be investigated. Ageing is an important determinant of COVID-19 outcome, thus the influence of cellular senescence, oxidative stress, and other features of ageing on SARS-CoV-2 infection of the endothelium should also be assessed. The influence of gender on endothelial responses to SARS-CoV-2 and how this relates to the susceptibility and outcome of COVID-19 patients should be investigated.

  • The effects of common cardiovascular drugs such as statins and beta-blockers on endothelial responses to SARS-CoV-19 should be explored, including their influence on ACE2 expression and viral infectivity.

  • The long-term cardiovascular effects following recovery from COVID-19 must be determined during planned patient follow-up so that appropriate preventive measures can be taken in time if needed. Measuring endothelial function in addition to myocardial injury and respiratory function markers in convalescent patients may represent a possible means for early detection of vascular sequelae post-COVID-19.

Authors’ contributions

P.C.E. and M.B. conceived and designed the manuscript, drafted the manuscript, and revised it for important intellectual content. G.E.R., J.C.M., T.J.G., E.O., Z.S., D.N., I.E.H., M.F., J.W., C.W., and M.-L.B.-P. made substantial contributions to the conception and design of the manuscript, and drafted the manuscript or revised it for important intellectual content. All authors approved the final version of the manuscript and agreed to be accountable for the work.

Conflict of interest: none declared.

References

1

Zhou
P
,
Yang
XL
,
Wang
XG
,
Hu
B
,
Zhang
L
,
Zhang
W
,
Si
HR
,
Zhu
Y
,
Li
B
,
Huang
CL
,
Chen
HD
,
Chen
J
,
Luo
Y
,
Guo
H
,
Jiang
RD
,
Liu
MQ
,
Chen
Y
,
Shen
XR
,
Wang
X
,
Zheng
XS
,
Zhao
K
,
Chen
QJ
,
Deng
F
,
Liu
LL
,
Yan
B
,
Zhan
FX
,
Wang
YY
,
Xiao
GF
,
Shi
ZL.
 
A pneumonia outbreak associated with a new coronavirus of probable bat origin
.
Nature
 
2020
;
579
:
270
273
.

2

Guo
T
,
Fan
Y
,
Chen
M
,
Wu
X
,
Zhang
L
,
He
T
,
Wang
H
,
Wan
J
,
Wang
X
,
Lu
Z.
 
Cardiovascular implications of fatal outcomes of patients with coronavirus disease 2019 (COVID-19)
.
JAMA Cardiol
 
2020
;
5
:
1
8
.

3

Driggin
E
,
Madhavan
MV
,
Bikdeli
B
,
Chuich
T
,
Laracy
J
,
Bondi-Zoccai
G
,
Brown
TS
,
Nigoghossian
C
,
Zidar
DA
,
Haythe
J
,
Brodie
D
,
Beckman
JA
,
Kirtane
AJ
,
Stone
GW
,
Krumholz
HM
,
Parikh
SA.
 
Cardiovascular considerations for patients, health care workers, and health systems during the coronavirus disease 2019 (COVID-19) pandemic
.
J Am Coll Cardiol
 
2020
;
75
:
2352
2371
.

4

Stefanini
GG
,
Montorfano
M
,
Trabattoni
D
,
Andreini
D
,
Ferrante
G
,
Ancona
M
,
Metra
M
,
Curello
S
,
Maffeo
D
,
Pero
G
,
Cacucci
M
,
Assanelli
E
,
Bellini
B
,
Russo
F
,
Ielasi
A
,
Tespili
M
,
Danzi
GB
,
Vandoni
P
,
Bollati
M
,
Barbieri
L
,
Oreglia
J
,
Lettieri
C
,
Cremonesi
A
,
Carugo
S
,
Reimers
B
,
Condorelli
G
,
Chieffo
A.
 
ST-elevation myocardial infarction in patients with COVID-19: clinical and angiographic outcomes
.
Circulation
 
2020
;
141
:
2113
2116
.

5

Meyer
P
,
Degrauwe
S
,
Van Delden
C
,
Ghadri
JR
,
Templin
C.
 
Typical takotsubo syndrome triggered by SARS-CoV-2 infection
.
Eur Heart J
 
2020
;
41
:
1860
.

6

Tersalvi
G
,
Vicenzi
M
,
Calabretta
D
,
Biasco
L
,
Pedrazzini
G
,
Winterton
D.
 
Elevated troponin in patients with coronavirus disease 2019: possible mechanisms
.
J Cardiac Fail
 
2020
;
26
:
470
475
.

7

Gao
L
,
Jiang
D
,
Wen
XS
,
Cheng
XC
,
Sun
M
,
He
B
,
You
LN
,
Lei
P
,
Tan
XW
,
Qin
S
,
Cai
GQ
,
Zhang
DY.
 
Prognostic value of NT-proBNP in patients with severe COVID-19
.
Resp Res
 
2020
;
21
:
83
.

8

Wang
D
,
Hu
B
,
Hu
C
,
Zhu
F
,
Liu
X
,
Zhang
J
,
Wang
B
,
Xiang
H
,
Cheng
Z
,
Xiong
Y
,
Zhao
Y
,
Li
Y
,
Wang
X
,
Peng
Z.
 
Clinical characteristics of 138 hospitalized patients with 2019 novel coronavirus-infected pneumonia in Wuhan, China
.
JAMA
 
2020
;
323
:
1061
1069
.

9

Chen
L
,
Li
X
,
Chen
M
,
Feng
Y
,
Xiong
C.
 
The ACE2 expression in human heart indicates new potential mechanism of heart injury among patients infected with SARS-CoV-2
.
Cardiovasc Res
 
2020
;
116
:
1097
1100
.

10

Epidemiology Working Group for NCIP Epidemic Response,

Chinese Center for Disease Control and Prevention. [The epidemiological characteristics of an outbreak of 2019 novel coronavirus diseases (COVID-19) in China
].
Zhonghua Liu Xing Bing Xue Za Zhi
 
2020
;
41
:
145
151
.

11

Richardson
S
,
Hirsch
JS
,
Narasimhan
M
,
Crawford
JM
,
McGinn
T
,
Davidson
KW
,
Barnaby
DP
,
Becker
LB
,
Chelico
JD
,
Cohen
SL
,
Cookingham
J
,
Coppa
K
,
Diefenbach
MA
,
Dominello
AJ,
,
Duer-Hefele
J
,
Falzon
L
,
Gitlin
J
,
Hajizadeh
N
,
Harvin
TG
,
Hirschwerk
DA
,
Kim
EJ
,
Kozel
ZM
,
Marrast
LM
,
Mogavero
JN
,
Osorio
GA
,
Qiu
M
,
Zanos
TP.
 
Presenting characteristics, comorbidities, and outcomes among 5700 patients hospitalized with COVID-19 in the New York City area
.
JAMA
 
2020
;
323
:
2052
2059
.

12

Bikdeli
B
,
Madhavan
MV
,
Jimenez
D
,
Chuich
T
,
Dreyfus
I
,
Driggin
E
,
Nigoghossian
C
,
Ageno
W
,
Madjid
M
,
Guo
Y
,
Tang
LV
,
Hu
Y
,
Giri
J
,
Cushman
M
,
Quere
I
,
Dimakakos
EP,
,
Gibson
CM
,
Lippi
G
,
Favaloro
EJ
,
Fareed
J
,
Caprini
JA
,
Tafur
AJ
,
Burton
JR
,
Francese
DP
,
Wang
EY
,
Falanga
A
,
McLintock
C
,
Hunt
BJ
,
Spyropoulos
AC
,
Barnes
GD
,
Eikelboom
JW
,
Weinberg
I
,
Schulman
S
,
Carrier
M
,
Piazza
G
,
Beckman
JA
,
Steg
PG
,
Stone
GW
,
Rosenkranz
S
,
Goldhaber
SZ
,
Parikh
SA
,
Monreal
M
,
Krumholz
HM
,
Konstantinides
SV
,
Weitz
JI
,
Lip
GYH.
 
COVID-19 and thrombotic or thromboembolic disease: implications for prevention, antithrombotic therapy, and follow-up
.
J Am Coll Cardiol
 
2020
;
75
:
2950
2973
.

13

Gando
S
,
Fujishima
S
,
Saitoh
D
,
Shiraishi
A
,
Yamakawa
K
,
Kushimoto
S
,
Ogura
H
,
Abe
T
,
Mayumi
T
,
Sasaki
J
,
Kotani
J
,
Takeyama
N
,
Tsuruta
R
,
Takuma
K
,
Yamashita
N
,
Shiraishi
SI
,
Ikeda
H
,
Shiino
Y
,
Tarui
T
,
Nakada
TA
,
Hifumi
T
,
Otomo
Y
,
Okamoto
K
,
Sakamoto
Y
,
Hagiwara
A
,
Masuno
T
,
Ueyama
M
,
Fujimi
S
,
Umemura
Y.
 
The significance of disseminated intravascular coagulation on multiple organ dysfunction during the early stage of acute respiratory distress syndrome
.
Thromb Res
 
2020
;
191
:
15
21
.

14

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
.

15

Danzi
GB
,
Loffi
M
,
Galeazzi
G
,
Gherbesi
E.
 
Acute pulmonary embolism and COVID-19 pneumonia: a random association?
 
Eur Heart J
 
2020
;
41
:
1858
.

16

Llitjos
JF
,
Leclerc
M
,
Chochois
C
,
Monsallier
JM
,
Ramakers
M
,
Auvray
M
,
Merouani
K.
 
High incidence of venous thromboembolic events in anticoagulated severe COVID-19 patients
.
J Thromb Haemost
 
2020
;
18
:
1743
1746
.

17

Mao
L
,
Jin
H
,
Wang
M
,
Hu
Y
,
Chen
S
,
He
Q
,
Chang
J
,
Hong
C
,
Zhou
Y
,
Wang
D
,
Miao
X
,
Li
Y
,
Hu
B.
 
Neurologic manifestations of hospitalized patients with coronavirus disease 2019 in Wuhan, China
.
JAMA Neurol
 
2020
;
77
:
1
9
.

18

Beyrouti
R
,
Adams
ME
,
Benjamin
L
,
Cohen
H
,
Farmer
SF
,
Goh
YY
,
Humphries
F
,
Jager
HR
,
Losseff
NA
,
Perry
RJ
,
Shah
S
,
Simister
RJ
,
Turner
D
,
Chandratheva
A
,
Werring
DJ.
 
Characteristics of ischaemic stroke associated with COVID-19
.
J Neurol Neurosurg Psychiatry
 
2020
;
91
:
889
891
.

19

Guzik
TJ
,
Mohiddin
SA
,
Dimarco
A
,
Patel
V
,
Savvatis
K
,
Marelli-Berg
FM
,
Madhur
MS
,
Tomaszewski
M
,
Maffia
P
,
D’Acquisto
F
,
Nicklin
SA
,
Marian
AJ
,
Nosalski
R,
,
Murray
EC
,
Guzik
B
,
Berry
C
,
Touyz
RM
,
Kreutz
R
,
Wang
DW
,
Bhella
D
,
Sagliocco
O
,
Crea
F
,
Thomson
EC
,
McInnes
IB.
 
COVID-19 and the cardiovascular system: implications for risk assessment, diagnosis, and treatment options
.
Cardiovasc Res
 
2020
;
116
:
1666
1687
.

20

Klok
FA
,
Kruip
M
,
van der Meer
NJM
,
Arbous
MS
,
Gommers
D
,
Kant
KM
,
Kaptein
FHJ
,
van Paassen
J
,
Stals
MAM
,
Huisman
MV
,
Endeman
H.
 
Incidence of thrombotic complications in critically ill ICU patients with COVID-19
.
Thromb Res
 
2020
;
191
:
145
147
.

21

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
.

22

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
.

23

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.

24

Colmenero
I
,
Santonja
C
,
Alonso-Riaño
M
,
Noguera-Morel
L
,
Hernández-Martín
A
,
Andina
D
,
Wiesner
T
,
Rodríguez-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.

25

Alexander
Y
,
Osto
E
,
Schmidt-Trucksass
A
,
Shechter
M
,
Trifunovic
D
,
Duncker
DJ
,
Aboyans
V
,
Back
M
,
Badimon
L
,
Cosentino
F
,
De Carlo
M
,
Dorobantu
M
,
Harrison
DG
,
Guzik
TJ
,
Hoefer
I
,
Morris
PD
,
Norata
GD
,
Suades
R
,
Taddei
S
,
Vilahur
G
,
Waltenberger
J
,
Weber
C
,
Wilkinson
F
,
Bochaton-Piallat
ML
,
Evans
PC.
 
Endothelial function in cardiovascular precision medicine: a position paper on behalf of the
 
European Society of Cardiology. Cardiovasc Res
 
2020
;doi: 10.1093/cvr/cvaa085.

26

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

27

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
.

28

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
.

29

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

30

Celermajer
DS
,
Sorensen
KE
,
Gooch
VM
,
Spiegelhalter
DJ
,
Miller
OI
,
Sullivan
ID
,
Lloyd
JK
,
Deanfield
JE.
 
Non-invasive detection of endothelial dysfunction in children and adults at risk of atherosclerosis
.
Lancet
 
1992
;
340
:
1111
1115
.

31

Kitta
Y
,
Obata
JE
,
Nakamura
T
,
Hirano
M
,
Kodama
Y
,
Fujioka
D
,
Saito
Y
,
Kawabata
K
,
Sano
K
,
Kobayashi
T
,
Yano
T
,
Nakamura
K
,
Kugiyama
K.
 
Persistent impairment of endothelial vasomotor function has a negative impact on outcome in patients with coronary artery disease
.
J Am Coll Cardiol
 
2009
;
53
:
323
330
.

32

Wang
M
,
Hao
H
,
Leeper
NJ
,
Zhu
L.
 
Thrombotic regulation from the endothelial cell perspectives
.
Arterioscl Thromb Vasc Biol
 
2018
;
38
:
e90
e95
.

33

Zhang
B
,
Zhou
X
,
Zhu
C
,
Feng
F
,
Qiu
Y
,
Feng
J
,
Jia
Q
,
Song
Q
,
Zhu
B
,
Wang
J.
 
Immune phenotyping based on neutrophil-to-lymphocyte ratio and IgG predicts disease severity and outcome for patients with COVID-19
.
medRxiv
 
2020
:
2020
.
2003
.
2012.20035048
.

34

Xu
Z
,
Shi
L
,
Wang
Y
,
Zhang
J
,
Huang
L
,
Zhang
C
,
Liu
S
,
Zhao
P
,
Liu
H
,
Zhu
L
,
Tai
Y
,
Bai
C
,
Gao
T
,
Song
J
,
Xia
P
,
Dong
J
,
Zhao
J
,
Wang
FS.
 
Pathological findings of COVID-19 associated with acute respiratory distress syndrome
.
Lancet Respi Med
 
2020
;
8
:
420
422
.

35

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

36

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
.

37

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.

38

Teijaro
JR
,
Walsh
KB
,
Cahalan
S
,
Fremgen
DM
,
Roberts
E
,
Scott
F
,
Martinborough
E
,
Peach
R
,
Oldstone
MBA
,
Rosen
H.
 
Endothelial cells are central orchestrators of cytokine amplification during influenza virus infection
.
Cell
 
2011
;
146
:
980
991
.

39

Walsh
KB
,
Teijaro
JR
,
Brock
LG
,
Fremgen
DM
,
Collins
PL
,
Rosen
H
,
Oldstone
MBA.
 
Animal model of respiratory syncytial virus: CD8+ T cells cause a cytokine storm that is chemically tractable by sphingosine-1-phosphate 1 receptor agonist therapy
.
J Virol
 
2014
;
88
:
6281
6293
.

40

Walsh
KB
,
Teijaro
JR
,
Wilker
PR
,
Jatzek
A
,
Fremgen
DM
,
Das
SC
,
Watanabe
T
,
Hatta
M
,
Shinya
K
,
Suresh
M
,
Kawaoka
Y
,
Rosen
H
,
Oldstone
MBA.
 
Suppression of cytokine storm with a sphingosine analog provides protection against pathogenic influenza virus
.
Proc Natl Acad Sci USA
 
2011
;
108
:
12018
12023
.

41

Teoh
H
,
Quan
A
,
Bang
KWA
,
Wang
GL
,
Lovren
F
,
Vu
V
,
Haitsma
JJ
,
Szmitko
PE
,
Al-Omran
M
,
Wang
CH
,
Gupta
M
,
Peterson
MD
,
Zhang
HB
,
Chan
L
,
Freedman
J
,
Sweeney
G
,
Verma
S.
 
Adiponectin deficiency promotes endothelial activation and profoundly exacerbates sepsis-related mortality
.
Am J Physiol Endocrinol Metab
 
2008
;
295
:
E658
E664
.

42

Vabret
N
,
Samstein
R
,
Fernandez
N
,
Merad
M
,
Sinai Immunology Review Project. Advancing scientific knowledge in times of pandemics
.
Nat Rev Immunol
 
2020
;
20
:
338
338
.

43

Ng
OW
,
Chia
A
,
Tan
AT
,
Jadi
RS
,
Leong
HN
,
Bertoletti
A
,
Tan
YJ.
 
Memory T cell responses targeting the SARS coronavirus persist up to 11 years post-infection
.
Vaccine
 
2016
;
34
:
2008
2014
.

44

Li
CKF
,
Wu
H
,
Yan
HP
,
Ma
SW
,
Wang
LL
,
Zhang
MX
,
Tang
XP
,
Temperton
NJ
,
Weiss
RA
,
Brenchley
JM
,
Douek
DC
,
Mongkolsapaya
J
,
Tran
BH
,
Lin
CLS
,
Screaton
GR
,
Hou
JL
,
McMichael
AJ
,
Xu
XN.
 
T cell responses to whole SARS coronavirus in humans
.
J Immunol
 
2008
;
181
:
5490
5500
.

45

Ma
H
,
Zeng
W
,
He
H
,
Zhao
D
,
Yang
Y
,
Jiang
D
,
Zhou
P
,
Qi
Y
,
He
W
,
Zhao
C
,
Yi
R
,
Wang
X
,
Wang
B
,
Xu
Y
,
Yang
Y
,
Kombe Kombe
AJ
,
Ding
C
,
Xie
J
,
Gao
Y
,
Cheng
L
,
Li
Y
,
Ma
X
,
Jin
T.
 
COVID-19 diagnosis and study of serum SARS-CoV-2 specific IgA, IgM and IgG by chemiluminescence immunoanalysis
.
medRxiv
2020:
2020
.
2004
.
2017
.
20064907
.

46

Petri
MH
,
Tellier
C
,
Michiels
C
,
Ellertsen
I
,
Dogne
JM
,
Bäck
M.
 
Effects of the dual TP receptor antagonist and thromboxane synthase inhibitor EV-077 on human endothelial and vascular smooth muscle cells
.
Biochem Biophys Res Commun
 
2013
;
441
:
393
398
.

47

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
.

48

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
.

49

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
.

50

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
.

51

Magro
C
,
Mulvey
JJ
,
Berlin
D
,
Nuovo
G
,
Salvatore
S
,
Harp
J
,
Baxter-Stoltzfus
A
,
Laurence
J.
 
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

Gheblawi
M
,
Wang
K
,
Viveiros
A
,
Nguyen
Q
,
Zhong
JC
,
Turner
AJ
,
Raizada
MK
,
Grant
MB
,
Oudit
GY.
 
Angiotensin-converting enzyme 2: SARS-CoV-2 receptor and regulator of the renin–angiotensin system: celebrating the 20th anniversary of the discovery of ACE2
.
Circ Res
 
2020
;
126
:
1456
1474
.

53

Nicin
L
,
Abplanalp
WT
,
Mellentin
H
,
Kattih
B
,
Tombor
L
,
John
D
,
Schmitto
JD
,
Heineke
J
,
Emrich
F
,
Arsalan
M
,
Holubec
T
,
Walther
T
,
Zeiher
AM
,
Dimmeler
S.
 
Cell type-specific expression of the putative SARS-CoV-2 receptor ACE2 in human hearts
.
Eur Heart J
 
2020
;
41
:
1804
1806
.

54

Fang
L
,
Karakiulakis
G
,
Roth
M.
 
Are patients with hypertension and diabetes mellitus at increased risk for COVID-19 infection?
 
Lancet Resp Med
 
2020
;
8
:
e21
.

55

Mehra
MR
,
Desai
SS
,
Kuy
S
,
Henry
TD
,
Patel
AN.
 
Cardiovascular disease, drug therapy, and mortality in Covid-19
.
N Engl J Med
 
2020
;
382
:
e102
.

56

Mancia
G
,
Rea
F
,
Ludergnani
M
,
Apolone
G
,
Corrao
G.
 
Renin–angiotensin–aldosterone system blockers and the risk of Covid-19
.
N Engl J Med
 
2020
;
382
:
2431
2440
.

57

Hoffmann
M
,
Kleine-Weber
H
,
Schroeder
S
,
Kruger
N,
,
Herrler
T
,
Erichsen
S
,
Schiergens
TS
,
Herrler
G
,
Wu
NH
,
Nitsche
A
,
Muller
MA
,
Drosten
C
,
Pohlmann
S.
 
SARS-CoV-2 cell entry depends on ACE2 and TMPRSS2 and is blocked by a clinically proven protease inhibitor
.
Cell
 
2020
;
181
:
271
280
.e278.

58

Li
W
,
Moore
MJ
,
Vasilieva
N
,
Sui
J
,
Wong
SK
,
Berne
MA
,
Somasundaran
M
,
Sullivan
JL
,
Luzuriaga
K
,
Greenough
TC
,
Choe
H
,
Farzan
M.
 
Angiotensin-converting enzyme 2 is a functional receptor for the SARS coronavirus
.
Nature
 
2003
;
426
:
450
454
.

59

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 SARS-CoV-2 infections in engineered human tissues using clinical-grade soluble human ACE2
.
Cell
 
2020
;
181
:
905
913
.

60

Zhang
H
,
Penninger
JM
,
Li
Y
,
Zhong
N
,
Slutsky
AS.
 
Angiotensin-converting enzyme 2 (ACE2) as a SARS-CoV-2 receptor: molecular mechanisms and potential therapeutic target
.
Intensive Care Med
 
2020
;
46
:
586
590
.

61

Aimes
RT
,
Zijlstra
A
,
Hooper
JD
,
Ogbourne
SM
,
Sit
ML
,
Fuchs
S
,
Gotley
DC
,
Quigley
JP
,
Antalis
TM.
 
Endothelial cell serine proteases expressed during vascular morphogenesis and angiogenesis
.
Thromb Haemost
 
2003
;
89
:
561
572
.

62

Stamler
JS
,
Lamas
S
,
Fang
FC.
 
Nitrosylation. The prototypic redox-based signaling mechanism
.
Cell
 
2001
;
106
:
675
683
.

63

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.

64

Goldsmith
CS
,
Miller
SE
,
Martines
RB
,
Bullock
HA
,
Zaki
SR.
 
Electron microscopy of SARS-CoV-2: a challenging task
.
Lancet
 
2020
;
395
:
e99
.

65

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.

66

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
.

67

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

68

Esper
F
,
Shapiro
ED
,
Weibel
C
,
Ferguson
D
,
Landry
ML
,
Kahn
JS.
 
Association between a novel human coronavirus and Kawasaki disease
.
J Infect Dis
 
2005
;
191
:
499
502
.

69

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.

70

Attwell
D
,
Mishra
A
,
Hall
CN
,
O’Farrell
FM
,
Dalkara
T.
 
What is a pericyte
?
J Cereb Blood Flow Metab
 
2016
;
36
:
451
455
.

71

Chen
L
,
Li
X
,
Chen
M
,
Feng
Y
,
Xiong
C.
 
The ACE2 expression in human heart indicates new potential mechanism of heart injury among patients infected with SARS-CoV-2
.
Cardiovasc Res
 
2020
;
116
:
1097
1100
.

72

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
.

73

Cardot-Leccia
N
,
Hubiche
T
,
Dellamonica
J
,
Burel-Vandenbos
F
,
Passeron
T.
 Pericyte alteration sheds light on micro-vasculopathy in COVID-19 infection.
Intensive Care Med
 
2020
;doi: 10.1007/s00134-020-06147-7.

74

Lindblom
P
,
Gerhardt
H
,
Liebner
S
,
Abramsson
A
,
Enge
M
,
Hellstrom
M
,
Backstrom
G
,
Fredriksson
S
,
Landegren
U
,
Nystrom
HC
,
Bergstrom
G
,
Dejana
E
,
Ostman
A
,
Lindahl
P
,
Betsholtz
C.
 
Endothelial PDGF-B retention is required for proper investment of pericytes in the microvessel wall
.
Genes Dev
 
2003
;
17
:
1835
1840
.

75

Gustafson
D
,
Raju
S
,
Wu
R
,
Ching
C
,
Veitch
S
,
Rathnakumar
K
,
Boudreau
E
,
Howe
KL
,
Fish
JE.
 
Overcoming barriers: the endothelium as a linchpin of coronavirus disease 2019 pathogenesis?
 
Arterioscl Thromb Vasc Biol
 
2020
;
40
:
1818
1829
.

76

South
AM
,
Diz
DI
,
Chappell
MC.
 
COVID-19, ACE2, and the cardiovascular consequences
.
Am J Physiol Heart Circ Physiol
 
2020
;
318
:
H1084
H1090
.

77

Rodrigues-Diez
RR
,
Tejera-Muñoz
A,
,
Marquez-Exposito
L
,
Rayego-Mateos
S
,
Sanchez
LS
,
Marchant
V
,
Santamaria
LT
,
Ramos
AM
,
Ortiz
A
,
Egido
J
,
Ruiz-Ortega
M.
 
Statins: could an old friend help the fight against COVID-19?
 
Br J Pharmacol
 
2020
;doi: 10.1111/bph.15166.

78

Bifulco
M
,
Gazzerro
P.
 
Statin therapy in COVID-19 infection: much more than a single pathway
.
Eur Heart J Cardiovasc Pharmacother
 
2020
;doi: 10.1093/ehjcvp/pvaa055.

79

Castiglione
V
,
Chiriacò
M
,
Emdin
M
,
Taddei
S
,
Vergaro
G.
 
Statin therapy in COVID-19 infection
.
Eur Heart J Cardiovasc Pharmacother
 
2020
;
6
:
258
259
.

80

Vasanthakumar
N.
 
Can beta-adrenergic blockers be used in the treatment of COVID-19?
 
Med Hypotheses
 
2020
;
142
:
109809
.

81

Staedtke
V
,
Bai
RY
,
Kim
K
,
Darvas
M
,
Davila
ML
,
Riggins
GJ
,
Rothman
PB
,
Papadopoulos
N
,
Kinzler
KW
,
Vogelstein
B
,
Zhou
S.
 
Disruption of a self-amplifying catecholamine loop reduces cytokine release syndrome
.
Nature
 
2018
;
564
:
273
277
.

82

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
.
medRxiv
 
2020
:
2020
.
2006
.
2022.20137273
.

83

Karram
T
,
Abbasi
A
,
Keidar
S
,
Golomb
E
,
Hochberg
I
,
Winaver
J
,
Hoffman
A
,
Abassi
Z.
 
Effects of spironolactone and eprosartan on cardiac remodeling and angiotensin-converting enzyme isoforms in rats with experimental heart failure
.
Am J Physiol Heart Circ Physiol
 
2005
;
289
:
H1351
H1358
.

84

Sanchis-Gomar
F
,
Lavie
CJ
,
Perez-Quilis
C
,
Henry
BM
,
Lippi
G.
 
Angiotensin-converting enzyme 2 and antihypertensives (angiotensin receptor blockers and angiotensin-converting enzyme inhibitors) in coronavirus disease 2019
.
Mayo Clin Proc
 
2020
;
95
:
1222
1230
.

85

Reynolds
HR
,
Adhikari
S
,
Pulgarin
C
,
Troxel
AB
,
Iturrate
E
,
Johnson
SB
,
Hausvater
A
,
Newman
JD
,
Berger
JS,
,
Bangalore
S
,
Katz
SD
,
Fishman
GI
,
Kunichoff
D
,
Chen
Y
,
Ogedegbe
G
,
Hochman
JS.
 
Renin–angiotensin–aldosterone system inhibitors and risk of Covid-19
.
N Engl J Med
 
2020
;
382
:
2441
2448
.

86

Mancia
G
,
Rea
F
,
Ludergnani
M
,
Apolone
G
,
Corrao
G.
 
Renin–angiotensin–aldosterone system blockers and the risk of Covid-19
.
N Engl J Med
 
2020
;
382
:
2431
2440
.

87

Bromage
DI
,
Cannata
A
,
Rind
IA
,
Gregorio
C
,
Piper
S
,
Shah
AM
,
McDonagh
TA.
 
The impact of COVID-19 on heart failure hospitalization and management: report from a Heart Failure Unit in London during the peak of the pandemic
.
Eur J Heart Fail
 
2020
;doi: 10.1002/ejhf.1925.

88

Wu
Y
,
Wang
F
,
Shen
C
,
Peng
W
,
Li
D
,
Zhao
C
,
Li
Z
,
Li
S
,
Bi
Y
,
Yang
Y
,
Gong
Y
,
Xiao
H
,
Fan
Z
,
Tan
S
,
Wu
G
,
Tan
W
,
Lu
X
,
Fan
C
,
Wang
Q
,
Liu
Y
,
Zhang
C
,
Qi
J
,
Gao
GF
,
Gao
F
,
Liu
L.
 
A noncompeting pair of human neutralizing antibodies block COVID-19 virus binding to its receptor ACE2
.
Science
 
2020
;
368
:
1274
1278
.

89

Emanueli
C
,
Badimon
L
,
Martelli
F
,
Potočnjak
I
,
Carpusca
I
,
Robinson
EL
,
Devaux Y, the EU-CardioRNA COST Action (CA17129). Call to action for the cardiovascular side of COVID-19: a call for cooperative action from the EU-CardioRNA COST Action
.
Eur Heart J
 
2020
;
41
:
1796
1797
.

This article is published and distributed under the terms of the Oxford University Press, Standard Journals Publication Model (https://academic.oup.com/journals/pages/open_access/funder_policies/chorus/standard_publication_model)