49
views
0
recommends
+1 Recommend
2 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: not found

      Lack of Evidence of Angiotensin-Converting Enzyme 2 Expression and Replicative Infection by SARS-CoV-2 in Human Endothelial Cells

      letter
      , BSc(Hons) 1 , * , , PhD 3 , * , , PhD 4 , 8 , * , , PhD 3 , * , , BSc(Hons) 2 , , , PhD 5 , , , PhD 3 , , PhD 3 , , PhD 4 , , PhD 6 , , PhD 5 , , PhD 2 , , MD, PhD 2 , , PhD 5 , , PhD 6 , 9 , , PhD 3 , , , PhD 4 , 6 , 7 , , , MD, PhD 3 , , , PhD 1 , , , , MD, PhD 3 , ,
      Circulation
      Lippincott Williams & Wilkins
      angiotensin-converting enzyme 2, COVID-19, endothelial cells, endothelial injury, SARS-CoV-2

      Read this article at

      ScienceOpenPublisherPMC
      Bookmark
          There is no author summary for this article yet. Authors can add summaries to their articles on ScienceOpen to make them more accessible to a non-specialist audience.

          Abstract

          A striking feature of severe forms of coronavirus disease 2019 (COVID-19), the current pandemic caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), is severe endothelial injury with micro- and macrothrombotic disease in the lung and other organs, including the heart. This has led to speculation that viral infection may damage the endothelium through 2 mechanisms: indirectly, via neighborhood effects, circulating mediators, and immune mechanisms or directly by viral infection of endothelial cells (ECs). To support the hypothesis of direct viral damage of ECs via virus-induced infection, the cells should express the main receptor for SARS-CoV-2, ACE2 (angiotensin-converting enzyme 2), a metalloprotease component of the renin–angiotensin hormone system and a critical regulator of cardiovascular homeostasis. 1 Indeed, several recent review articles propose that SARS-CoV-2 binding to ACE2 on ECs is the mechanism through which the virus may cause direct endothelial damage and endothelialitis. 1 However, expression of ACE2 in ECs has not been convincingly demonstrated to support this assumption, nor has there been sufficient evidence to support a direct infection of ECs by SARS-CoV-2. To address the questions of ACE2 expression in human ECs and of the ability of SARS-CoV-2 to infect the endothelium, we interrogated transcriptomic and epigenomic data on human ECs and studied the interaction and replication of SARS-Cov-2 and its viral proteins with ECs in vitro. The data, analytic methods, and study materials will be maintained by the corresponding author and made available to other researchers on reasonable request. Analysis of RNA sequencing was carried out on ENCODE data from ECs from arterial, venous, and microvascular beds, in comparison with epithelial cells from respiratory, gastrointestinal, and skin sources. Very low or no basal ACE2 expression was found in ECs compared with epithelial cells (Figure A and B). Moreover, in vitro exposure of ECs to inflammatory cytokines reported as elevated in the plasma of patients with severe COVID-19 failed to upregulate ACE2 expression (Figure C). Figure. Analysis of ACE2 expression in human ECs and of coronavirus replication in primary human ECs. A and B, Comparison of ACE2 expression in human primary epithelial cells and ECs using total RNA sequencing data from the ENCODE database shows low or absent expression in ECs. A, The difference of ACE2 expression in epithelial cells and ECs is shown in boxplots with individual, as well as grouped, samples (inner boxplot). Each dot represents a single sample (n=2 per cell type). B, Transcriptome profiles of epithelial cells and ECs are shown in a density plot, using the median of all samples per group (n=19 360 genes). ACE2 expression in each group is marked with a dotted line: ACE2 expression in ECs (red) overlaps with the peak for nonexpressed transcripts (highlighted in grey), while ACE2 expression in epithelial cells (blue) is to the right, indicating detectable expression. Median ACE2 expression in endothelial cells equals -5.6 in log2 CPM, which corresponds with 0 raw read counts, signifying undetectable ACE2 expression in the majority of ECs. Expression values in all plots are represented as log2-transformed CPM, normalized by trimmed mean of M-value (blue: epithelial, red: endothelial). C, ACE2 expression is not regulated by inflammatory cytokines in HUVECs. qPCR analysis of ACE2 mRNA expression in HUVECs treated with a mix of 4 cytokines/chemokines (TNF-α, IL1-β, IL8, and IL6/IL6R chimeric protein) for 4 hours or 24 hours at 0, 0.01, 0.1, or 1.0 ng/mL. Data are normalized to GAPDH and presented as mean±SEM of 3 independent experiments. D and E, Very low-level, rare, and likely contaminating ACE2 transcripts are seen in ECs. D, ACE2 transcript reads are detected preferentially in PCs. UMAP landscapes of publicly available human heart data sets 2 include 100 579 ECs, 77 856 PCs, 16 242 SMCs, and 718 MCs (https://www.heartcellatlas.org/). ACE2 transcript reads are detected preferentially in the PC cluster (enriching for ABCC9) and are rare in the EC cluster (enriching for PECAM1). E, PC transcripts are enriched together with ACE2 in 0.47% of ECs. Dot plot displaying the abundance of top-50 transcripts enriched ACE2+ versus ACE2- ECs, across cell types indicated in D. (The Wilcoxon rank-sum tests with Bonferroni-corrected P values are < 1E-60 for each). F, Epigenetic profiling indicates that the ACE2 gene is inactive in ECs. ChIP-seq binding profiles in HUVECs for histone modifications, RNA Pol2 enrichment, and deoxyribonuclease I hypersensitivity. The x axis represents the genomic position, the transcription start sites are indicated by closed arrow, and the direction of transcription is indicated by open arrows; the y axis shows ChIP-seq signal in reads per million per base pair (rpm/bp). The bottom row represents the chromatin state segmentation. Color key: active promoter, red; enhancers, yellow; transcriptional elongation, green; repressed, grey. G and H, Coronavirus replication in primary human cardiac and pulmonary ECs shows limited replication of SARS-CoV-2. G, Viral replication curves in human pulmonary (HPAEC) and cardiac (HCAEC) endothelial cells following infection with control HCoV-229E GFP (green fluorescent protein) reporter virus (MOI=0.6). Virus replication was measured via GFP fluorescence every 2 hours from 20 to 58 hours postinoculation. Mean±SEM of 3 technical replicates are shown at each time point for each biological replicate. H, Viral growth curves in HPAEC (n=3), HCAEC (n=3), and nonendothelial Vero cells (n=1) after infection with SARS-CoV-2 at MOI=10 or 100. Supernatant were collected at 1, 24, and 48 hours postinfection and virus copy number quantified by RT-qPCR detection of the SARS-CoV-2 N3 gene. ChIP-seq indicates chromatin immunoprecipitation sequencing; CPM, counts per million; EC, endothelial cell; HUVEC, human umbilical vein endothelial cell; IL, interleukin; MC, mesothelial cell; MOI, multiplicity of infection; PC, pericyte; qPCR, quantitative polymerase chain reaction; RT, real time; SARS-CoV-2, severe acute respiratory syndrome coronavirus 2; SMC, smooth muscle cell; TCID, tissue culture infectivity dose; TNF, tumor necrosis factor; and UMAP, Uniform Manifold Approximation and Projection. Publicly available single-cell RNA sequencing of human organ donor hearts 2 showed that, while ACE2 sequence reads are abundant in pericytes, they are rare in ECs (Figure D). Of 100,579 ECs, only 468 (0.47%) were ACE2 +, and in the majority (424), only a single ACE2 transcript was detected. This could reflect true low and rare endothelial ACE2 expression but also contamination from adherent pericyte fragments, a common confounder in vascular single-cell RNA sequencing data. 3 If such fragments contributed to the ACE2 transcripts observed in certain ECs, we would expect to detect other pericyte transcripts in the same cells. Among the top-50 gene transcripts enriched in ACE2 + versus ACE2 − ECs, we noticed several known pericyte markers, including PDGFRB, ABCC9, KCNJ8, and RGS5 (Figure E). Comparison of transcript abundance across the 3 major vascular and mesothelial cells showed that the top-50 gene transcripts were expressed at the highest levels in pericytes (Figure E). This suggests that the rare occurrence of ACE2 transcripts in human heart ECs is likely caused by pericyte contamination. Similar conclusions have been reached previously in mouse tissues. 3 Analysis of the chromatin landscape at the ACE2 gene locus in human umbilical vein ECs using data from ENCODE further supports this concept. The histone modification mark H3K27me3, which indicates repressed chromatin, was enriched at the ACE2 transcription start site; conversely, promoter, enhancer, and gene body activation marks (H3K27ac, H3K4me1, H3K4me2, H3K4me3, and H3K36me3), RNA polymerase-II and deoxyribonuclease I hypersensitivity were absent or low, suggesting that ACE2 is inactive in ECs. In marked contrast, the adjacent gene BMX, an endothelial-restricted nonreceptor tyrosine kinase, displays an epigenetic profile consistent with active endothelial expression (Figure F). Thus, transcriptomic and epigenomic data indicate that ACE2 is not expressed in human ECs. Other cell surface molecules have been suggested as possible receptors for the virus, but their role in supporting SARS-CoV-2 cell infection remains to be demonstrated. We therefore tested directly whether ECs could be capable of supporting coronavirus replication in vitro. Productive levels of replication in primary human cardiac and pulmonary ECs were observed for the human coronavirus 229E GFP (green fluorescent protein) reporter virus, 4 which uses CD13 as its receptor, demonstrating directly that human ECs can support coronavirus replication in principle (Figure G). However, when cells were exposed to SARS-CoV-2, replication levels were extremely low for ECs, even after exposure to very high concentrations of virus compared with more permissive VeroE6 cells (Figure H). The observed low levels of SARS-CoV-2 replication in ECs are likely explained by viral entry via a non-ACE2–dependent route, attributable to exposure to extremely high concentrations of virus in these experiments (multiplicity of infection, 10 and 100). These data indicate that direct endothelial infection by SARS-CoV-2 is not likely to occur. The endothelial damage reported in severely ill patients with COVID-19 is more likely secondary to infection of neighboring cells and/or other mechanisms, including immune cells, platelets and complement activation, and circulating proinflammatory cytokines. Our hypothesis is corroborated by recent evidence that plasma from critically ill and convalescent patients with COVID-19 causes EC cytotoxicity. 5 These findings have implications for therapeutic approaches to tackle vascular damage in severe COVID-19 disease. Acknowledgments For the ENCODE database, please see www.encodeproject.org. Sources of Funding This research was supported by grants from the Imperial College COVID response fund (to Dr Randi), the Imperial College British Heart Foundation (BHF) Research Excellence Award (RE/18/4/34215) (to Drs Randi, Birdsey, and Noseda); BHF Program grant funding (RG/17/4/32662) (to Drs Randi and Birdsey); the National Institute for Health Research Imperial Biomedical Research Center (to Drs Randi and Birdsey); BHF/German Center for Cardiovascular Research grant (SP/19/1/34461) and Chan Zuckerberg Initiative (grant 2019–202666) (to Dr Noseda); the Swedish Science Council, The Swedish Cancer Society, the Knut and Alice Wallenberg, and Erling-Persson Family Foundation (to Drs Betsholtz and Lendahl); the University of Edinburgh BHF Research Excellence Award; BHF Chair of Translational Cardiovascular Sciences and H2020 European Union grant COVIRNA (agreement DLV-101016072); IMI-2 CARE (to Drs Tait-Burkard and Haas); Biotechnology and Biological Sciences Research Council Institute Strategic Program grant funding to The Roslin Institute (BBS/E/D/20241866, BBS/E/D/20002172, and BBS/E/D/20002174) (to Dr Tait-Burkard); and BHF Center for Vascular Regeneration (RM/17/3/33381) (to Drs Baker and Randi). Disclosures None.

          Related collections

          Most cited references5

          • Record: found
          • Abstract: found
          • Article: found
          Is Open Access

          Cells of the adult human heart

          Cardiovascular disease is the leading cause of death worldwide. Advanced insights into disease mechanisms and therapeutic strategies require a deeper understanding of the molecular processes involved in the healthy heart. Knowledge of the full repertoire of cardiac cells and their gene expression profiles is a fundamental first step in this endeavour. Here, using state-of-the-art analyses of large-scale single-cell and single-nucleus transcriptomes, we characterize six anatomical adult heart regions. Our results highlight the cellular heterogeneity of cardiomyocytes, pericytes and fibroblasts, and reveal distinct atrial and ventricular subsets of cells with diverse developmental origins and specialized properties. We define the complexity of the cardiac vasculature and its changes along the arterio-venous axis. In the immune compartment, we identify cardiac-resident macrophages with inflammatory and protective transcriptional signatures. Furthermore, analyses of cell-to-cell interactions highlight different networks of macrophages, fibroblasts and cardiomyocytes between atria and ventricles that are distinct from those of skeletal muscle. Our human cardiac cell atlas improves our understanding of the human heart and provides a valuable reference for future studies.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            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

            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.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Endotheliopathy Is Induced by Plasma From Critically Ill Patients and Associated With Organ Failure in Severe COVID-19

              Lung histological analyses revealed the presence of vascular inflammation and severe endothelial injury as a direct consequence of intracellular severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection and ensuing host inflammatory response in coronavirus disease 2019 (COVID-19). 1 Endothelial cells promote coagulation following injury, leading to widespread formation of microthrombi, provoking microcirculatory failure or large-vessel thrombosis. 2 Growing evidence suggests that microvascular thrombosis is a major pathophysiological event in COVID-19 pathogenesis. Damaged endothelial cells could be closely implicated in the prothrombotic state commonly reported in severe patients in the intensive care unit (ICU). How SARS-CoV-2 exerts its cytopathic effects is still a matter of debate, and ultrastructural evidence of direct viral replication in endothelial cells remains to be demonstrated. Although direct viral tissue damage is a plausible mechanism of injury, 3 endothelial damage and thromboinflammation associated with dysregulated immune responses, inducing microvascular thrombosis, represent an attractive alternative hypothesis. 2 Using cultured human pulmonary microvascular endothelial cells (HPMVEC), we assessed whether plasma collected from patients with COVID-19 at different disease stages could trigger endothelial damage in vitro. The cytotoxicity of plasma samples on HPMVEC was evaluated by assessing mitochondrial activity (WST-1 test) 1 hour after incubation of cells with plasma as previously described. 4 We further investigated the association of plasma-induced cytotoxicity with levels of circulating biomarkers related to organ dysfunction (Pao2 [partial pressure of oxygen in arterial blood]/Fio2 [fraction of inspired oxygen], widely used as an indicator of oxygenation requirements, lactate dehydrogenase, creatinine, and aspartate transaminase), endothelial damage (von Willebrand factor antigen; ADAMTS13; plasminogen activator inhibitor-1; syndecan-1), tissue injury (cell-free DNA, a damage-associated molecular patterns marker), and levels of circulating cytokines related to the activation of innate (interleukin [IL]–6 and tumor necrosis factor–α) and adaptative immune cell responses (soluble IL-2 receptor). Inclusion criteria were individuals aged 18 years or older with a positive SARS-CoV-2 real-time reverse-transcriptase polymerase chain reaction on nasal or tracheal samples admitted to the Lille University Hospital. Patients on treatment with direct oral anticoagulant or vitamin K antagonists were switched to therapeutic heparin therapy on admission. Patients not in the ICU received once daily thromboprophylaxis with enoxaparin according to their body weight. Patients in the ICU received enoxaparin or unfractionated heparin according to their renal status, their body weight, and the need for invasive procedures. This study was approved by the French institutional authority for personal data protection (Commission Nationale de l’Informatique et des Libertés No. DEC20-086) and the ethics committee (IRB 2020-A00763-36), and informed consent was obtained from all participants. HPMVEC viability was assessed after coincubation with plasma sampled on admission from 28 consecutive patients (non-ICU, n=16; ICU, n=12) hospitalized for COVID-19 at the Lille University Hospital between March 30, 2020, and April 8, 2020, in convalescent patients with COVID-19 (n=6 from the 12 patients in the ICU) sampled after ICU discharge (mean±SD, 21±7 days) and in control healthy donors (n=8). Compared with healthy donor plasma, plasma from patients with COVID-19 significantly decreased HPMVEC viability, with plasma from patients in the ICU inducing the greatest cytotoxicity (Figure [A]). It is interesting that HPMVEC viability was partially restored to control when plasma from convalescent patients after ICU discharge was tested and compared with plasma of the same patients at the time of ICU admission. Moreover, markers of organ dysfunction were correlated with plasma-induced cytotoxicity (Figure [B]). HPMVEC viability also correlated with most plasma markers related to endothelial damage or tissue injury (Figure [C]). Soluble IL-2 receptor and tumor necrosis factor–α levels negatively correlated with HPMVEC viability (Figure [D]). Overall, the degree of vascular endothelial cell injury induced by plasma sampled from patients with COVID-19 correlated to both clinical illness severity at admission and the levels of biomarkers related to endothelial injury, tissue injury, and proinflammatory cytokines. Figure. Endothelial cell cytotoxicity induced by plasma sampled from critically ill and convalescent patients with coronavirus disease 2019 (COVID-19). The cytotoxicity of platelet-poor plasma samples (obtained after a double centrifugation of citrate tubes at 2500 g for 15 minutes at room temperature) from patients with COVID-19 and controls on HPMVEC was evaluated with a colorimetric assay using 4-[3-(4-iodophenyl)-2-(4-nitrophenyl)-2H-5-tetrazolio]-1.3-benzene disulfonate (WST-1), which in viable cells is cleaved by mitochondrial dehydrogenases. After incubation, the cells were washed with phosphate buffered saline and incubated with WST-1 (Roche, Basel, Switzerland) at a dilution of 1:10 (10 µL) for 2 h at 37°C. Absorbance was measured using a multiwell plate reader (Synergy HTX multi-mode plate reader, BioTek Instruments, Highland Park, VT) at 450 nm with a reference wavelength of 620 nm. As a positive control for endothelial cell injury, Shigatoxin 145 (Sigma-Aldrich, Saint Quentin Fallavier, France) was spiked in plasma from healthy adults (10 µg/mL final concentration) and incubated at 37°C for 15 minutes, before addition to HPMVECs. Experiments were performed in triplicate for each patient sample. A, HPMVEC viability after exposure to plasma sampled in healthy subjects (n=8), in non-ICU (n=16), and in ICU (n=12) on admission and in 6 convalescent patients with COVID-19 sampled after ICU discharge (mean±SD, 21±7 days). Data points represent individual sample measurements, whereas horizontal bars show the mean (±SD). Comparisons between groups were done using the Mann-Whitney U test, except for comparison between ICU and convalescent patients, where we used Wilcoxon signed-rank test on matched pairs (n=6). Correlations between HPMVEC viability and (B) markers of organ dysfunction: the Pao 2/FiO2 ratio, widely used as an indicator of oxygenation requirements, LDH, creatinine and AST; (C) parameters related to endothelial dysfunction and tissue injury: VWF:Ag, ADAMTS13, ADAMTS13:VWF ratio, PAI-1, syndecan-1, and cell-free DNA; and (D) plasma cytokine concentrations: IL-2R, IL-6. and TNF-α. Correlations were evaluated with the Spearman rank-correlation statistical test. No adjustment for multiple comparisons was done, and the result should be interpreted as hypothesis-generating. AST indicates aspartate transaminase; HPMVEC, human pulmonary microvascular endothelial cells; ICU, intensive care unit; IL, interleukin; IL-2R, soluble IL-2 receptor; LDH, lactate dehydrogenase; PAI-1, plasminogen activator inhibitor-1; TNF–α, tumor necrosis factor–α; and VWF:Ag, von Willebrand factor antigen. Our data shed new light on the pathophysiology of COVID-19 by demonstrating the direct and rapid cytotoxic effect of plasma collected from critically ill patients on vascular endothelial cells. This rapid effect (1 hour after plasma exposure) excludes a direct cytopathic effect of SARS-CoV-2 infection, as the progression of viral infection and visible cytopathogenic effects are in general only apparent 12 to 24 hours after infection. 5 A higher cytotoxic effect of plasma on endothelial cells was associated with a more pronounced hypoxemia and organ dysfunction as reflected by the correlation with Pao 2/FiO2, lactate dehydrogenase, creatinine, and aspartate transaminase. This cytotoxic effect also correlated with circulating markers of endothelial damage, indicating that this in vitro functional assay reflects microvascular endothelial damage in vivo. Different pathways could be involved in endothelial cell injury during the course of COVID-19, ie, complement activation, cellular hypoxia, platelets, and direct cytotoxicity of cytokines such as IL-6, IL-1β, and tumor necrosis factor–α. We observed a relationship between this cytotoxic effect and the level of proinflammatory cytokines, suggesting that cytotoxicity could be related to overproduction of proinflammatory cytokines. However, this article does not provide the supportive evidence of convalescent plasma for treating severe patients with COVID-19. In conclusion, we provide for the first time the results of a functional assay demonstrating a direct effect of dysregulation of immune response on endothelial damage in COVID-19. Endotheliopathy is an essential part of the pathological response on severe COVID-19, leading to respiratory failure, multiorgan dysfunction, and thrombosis. Endothelial and microvascular damage are associated with immunopathology and may occur in parallel with intracellular SARS-CoV-2 infection. Acknowledgments The authors thank all physicians and medical staff involved in patient care. Special thanks are addressed to Eric Boulleaux, Laureline Bourgeois, Aurélie Jospin, Catherine Marichez, Vincent Dalibard, Bénédicte Pradines, Sandrine Vanderziepe, and all the biologists and technicians of the Hemostasis Department for their support during the COVID-19 pandemic. A.R. and A. Dupont collected clinical data, analyzed the data, and wrote the article. J.G., M.C., S. Staessens, M.D.M., E.J., D.C., G.L., F.L., K.F., M.L., D.G., S.D.M., and J.P. collected data. J.G., M.C., S. Staessens, E.J., and S.D.M. analyzed the data. J.L. and A. Duhamel performed the statistical analysis. B.S., E.V., F.V., J.P., E.K., and P.L. provided critical input in the interpretation of data and critically reviewed the article. S. Susen designed the study, analyzed the data, wrote, and critically reviewed the article. All authors provided editorial review and assisted in writing the article. Sources of Funding This study was supported by the French government through the Program Investissement d’Avenir (I-SITE ULNE/ANR-16-IDEX-0004 ULNE). Disclosures None.
                Bookmark

                Author and article information

                Contributors
                Journal
                Circulation
                Circulation
                CIR
                Circulation
                Lippincott Williams & Wilkins (Hagerstown, MD )
                0009-7322
                1524-4539
                06 January 2021
                23 February 2021
                : 143
                : 8
                : 865-868
                Affiliations
                [1 ]Centre for Cardiovascular Science (I.R.M., A.H.B.), University of Edinburgh, Little France Crescent, United Kingdom.
                [2 ]Infection Medicine (A.D., S.J.G., J.G.H.), University of Edinburgh, Little France Crescent, United Kingdom.
                [3 ]National Heart and Lung Institute, National Institute for Health Research Imperial Biomedical Research Centre, Imperial College London, United Kingdom (G.S., A.H., C.P., V.K., G.M.B., M.N., A.M.R.).
                [4 ]Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Sweden (L.H., M.A.-M., C.B.).
                [5 ]The Roslin Institute, Royal (Dick) School of Veterinary Studies, University of Edinburgh, Easter Bush, Midlothian, United Kingdom (S.F., N.M.C., C.T.-B.).
                [6 ]Integrated Cardio Metabolic Centre (L.M., U.L., C.B.), Karolinska Institute, Sweden.
                [7 ]Department of Medicine Huddinge (C.B.), Karolinska Institute, Sweden.
                [8 ]Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin Neurological Institute, Key Laboratory of Post-Neuroinjury Neuro-Repair and Regeneration in Central Nervous System, Ministry of Education and Tianjin City, Tianjin, China (L.H.).
                [9 ]Department of Cell and Molecular Biology, Karolinska Institute, Stockholm, Sweden (U.L.).
                Author notes
                Anna M. Randi, MD, PhD, National Heart and Lung Institute, Vascular Sciences, Imperial College London, Hammersmith Hospital, Du Cane Road, London, W12 0NN, United Kingdom, Email a.randi@ 123456imperial.ac.uk
                Andrew H. Baker, PhD, Centre for Cardiovascular Science, University of Edinburgh, 47 Little France Crescent, Edinburgh, EH16 4TJ, United Kingdom. Email andy.baker@ 123456ed.ac.uk
                Article
                00012
                10.1161/CIRCULATIONAHA.120.052824
                7899720
                33405941
                ba247346-6e5a-4cb9-a3e7-6d0f23bdc27f
                © 2021 American Heart Association, Inc.

                This article is made available via the PMC Open Access Subset for unrestricted re-use and analyses in any form or by any means with acknowledgement of the original source. These permissions are granted for the duration of the COVID-19 pandemic or until permissions are revoked in writing. Upon expiration of these permissions, PMC is granted a perpetual license to make this article available via PMC and Europe PMC, consistent with existing copyright protections.

                History
                Categories
                10009
                10010
                10014
                10024
                10054
                Correspondence
                Research Letter

                angiotensin-converting enzyme 2,covid-19,endothelial cells,endothelial injury,sars-cov-2

                Comments

                Comment on this article