COVID-19 and cardiovascular disease: from basic mechanisms to clinical perspectives

Masataka Nishiga, Dao Wen Wang, Yaling Han, David B Lewis, Joseph C Wu, Masataka Nishiga, Dao Wen Wang, Yaling Han, David B Lewis, Joseph C Wu

Abstract

Coronavirus disease 2019 (COVID-19), caused by a strain of coronavirus known as severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), has become a global pandemic that has affected the lives of billions of individuals. Extensive studies have revealed that SARS-CoV-2 shares many biological features with SARS-CoV, the zoonotic virus that caused the 2002 outbreak of severe acute respiratory syndrome, including the system of cell entry, which is triggered by binding of the viral spike protein to angiotensin-converting enzyme 2. Clinical studies have also reported an association between COVID-19 and cardiovascular disease. Pre-existing cardiovascular disease seems to be linked with worse outcomes and increased risk of death in patients with COVID-19, whereas COVID-19 itself can also induce myocardial injury, arrhythmia, acute coronary syndrome and venous thromboembolism. Potential drug-disease interactions affecting patients with COVID-19 and comorbid cardiovascular diseases are also becoming a serious concern. In this Review, we summarize the current understanding of COVID-19 from basic mechanisms to clinical perspectives, focusing on the interaction between COVID-19 and the cardiovascular system. By combining our knowledge of the biological features of the virus with clinical findings, we can improve our understanding of the potential mechanisms underlying COVID-19, paving the way towards the development of preventative and therapeutic solutions.

Figures

Fig. 1. Structure, genome and life cycle…
Fig. 1. Structure, genome and life cycle of SARS-CoV-2.
a | Coronaviruses form an enveloped spherical particle that consists of four structural proteins (spike (S), envelope (E), membrane (M) and nucleocapsid (N)) and a positive-sense, single-stranded RNA (ssRNA) genome that is 30 kb in length. b | The 5′-terminal two-thirds of the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) genome encodes polyproteins pp1a and pp1ab, which are cleaved into 16 different non-structural proteins. Structural proteins are encoded in the 3′-terminal one-third of the genome. The S protein consists of two subunits; the S1 subunit contains a receptor-binding domain (RBD) that binds to angiotensin-converting enzyme 2 (ACE2) on the surface of host cells, whereas the S2 subunit mediates fusion between the membranes of the virus and the host cell. Compared with the S protein of SARS-CoV, the S protein of SARS-CoV-2 has two notable features. First, within the RBD of the S1 subunit, five of the six residues that are crucial for binding to human ACE2 are mutated. Second, an insertion of four amino acid residues at the boundary between the S1 and S2 subunits is present in SARS-CoV-2 but not in SARS-CoV, which introduces a novel furin cleavage site. c | SARS-CoV-2 infection is triggered by the binding of the S protein to ACE2 on the surface of host cells, and the viral complex is incorporated into the cytoplasm either by direct fusion with the cell membrane or via endocytosis with later release into the cytoplasm from the endocytic vesicle. The S protein is cleaved at the S1/S2 boundary and the S2 subunit facilitates membrane fusion. The viral genome RNA is released into the cytoplasm, and the first open reading frame (ORF) is translated into polyproteins pp1a and pp1ab, which are then cleaved by viral proteases into small, non-structural proteins such as RNA-dependent RNA polymerase (RdRP). The viral genomic RNA is replicated by RdRP. Viral nucleocapsids are assembled from genomic RNA and N proteins in the cytoplasm, whereas budding of new particles occurs at the membrane of the endoplasmic reticulum (ER)–Golgi intermediate compartment. Finally, the genomic RNA and structural proteins are assembled into new viral particles, leading to their release via exocytosis. 3CL, 3-chymotrypsin-like protease.
Fig. 2. Bidirectional interaction between cardiovascular diseases…
Fig. 2. Bidirectional interaction between cardiovascular diseases and COVID-19.
Cardiovascular comorbidities such as hypertension and coronary artery disease are associated with high mortality in patients with coronavirus disease 2019 (COVID-19). Drugs used to reduce cardiovascular risk such as angiotensin-converting enzyme (ACE) inhibitors and angiotensin II receptor blockers (ARBs) have numerous effects that might influence susceptibility to or the severity of COVID-19. Furthermore, although the main presentation of COVID-19 is viral pneumonia, COVID-19 can also induce cardiovascular manifestations including myocardial injury, myocarditis, arrhythmias, acute coronary syndrome and thromboembolism. Among these cardiovascular manifestations, myocardial injury has been independently associated with high mortality among patients with COVID-19 (ref.). Finally, medications that have been proposed as treatments for COVID-19 such as hydroxychloroquine and azithromycin have pro-arrhythmic effects. AF, atrial fibrillation; VF, ventricular fibrillation; VT, ventricular tachycardia.
Fig. 3. ACE2 as a part of…
Fig. 3. ACE2 as a part of the RAAS.
Angiotensin II, the main effector molecule in the renin–angiotensin–aldosterone system (RAAS), is upregulated in many pathological conditions, for which inhibition of angiotensin II by RAAS inhibitors is a common therapeutic strategy.Angiotensin-converting enzyme (ACE) produces angiotensin II from angiotensin I, whereas ACE2 inactivates angiotensin II by converting it to angiotensin (1–7). Therefore, ACE2 has a protective effect against cardiovascular disease and lung injury. In the setting of coronavirus disease 2019, downregulation of ACE2 by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection might be involved in mediating cardiovascular damage. ARB, angiotensin II receptor blocker.

References

    1. Zhou P, et al. A pneumonia outbreak associated with a new coronavirus of probable bat origin. Nature. 2020;579:270–273.
    1. Wu F, et al. A new coronavirus associated with human respiratory disease in China. Nature. 2020;579:265–269.
    1. Lu R, et al. Genomic characterisation and epidemiology of 2019 novel coronavirus: implications for virus origins and receptor binding. Lancet. 2020;395:565–574.
    1. Hoffmann M, et al. SARS-CoV-2 cell entry depends on ACE2 and TMPRSS2 and is blocked by a clinically proven protease inhibitor. Cell. 2020;181:271–280.
    1. Tay MZ, Poh CM, Renia L, MacAry PA, Ng LFP. The trinity of COVID-19: immunity, inflammation and intervention. Nat. Rev. Immunol. 2020;20:363–374.
    1. Bikdeli B, et al. COVID-19 and thrombotic or thromboembolic disease: implications for prevention, antithrombotic therapy, and follow-up: JACC state-of-the-art review. J. Am. Coll. Cardiol. 2020;75:2950–2973.
    1. Connors JM, Levy JH. Thromboinflammation and the hypercoagulability of COVID-19. J. Thromb. Haemost. 2020;18:1559–1561.
    1. Andersen KG, Rambaut A, Lipkin WI, Holmes EC, Garry RF. The proximal origin of SARS-CoV-2. Nat. Med. 2020;26:450–452.
    1. Wrapp D, et al. Cryo-EM structure of the 2019-nCoV spike in the prefusion conformation. Science. 2020;367:1260–1263.
    1. Walls AC, et al. Structure, function, and antigenicity of the SARS-CoV-2 spike glycoprotei. Cell. 2020;181:281–292.
    1. Wan Y, Shang J, Graham R, Baric RS, Li F. Receptor recognition by the novel coronavirus from Wuhan: an analysis based on decade-long structural studies of SARS coronavirus. J. Virol. 2020;94:e00127–20.
    1. Madjid M, Safavi-Naeini P, Solomon SD, Vardeny O. Potential effects of coronaviruses on the cardiovascular system: a review. JAMA Cardiol. 2020 doi: 10.1001/jamacardio.2020.1286.
    1. Clerkin KJ, et al. COVID-19 and cardiovascular disease. Circulation. 2020;141:1648–1655.
    1. Driggin E, et al. Cardiovascular considerations for patients, health care workers, and health systems during the COVID-19 pandemic. J. Am. Coll. Cardiol. 2020;75:2352–2371.
    1. Zheng YY, Ma YT, Zhang JY, Xie X. COVID-19 and the cardiovascular system. Nat. Rev. Cardiol. 2020;17:259–260.
    1. Han Y, et al. CSC expert consensus on principles of clinical management of patients with severe emergent cardiovascular diseases during the COVID-19 epidemic. Circulation. 2020;141:e810–e816.
    1. Huang C, et al. Clinical features of patients infected with 2019 novel coronavirus in Wuhan, China. Lancet. 2020;395:497–506. doi: 10.1016/S0140-6736(20)30183-5.
    1. Wang D, et al. Clinical characteristics of 138 hospitalized patients with 2019 novel coronavirus-infected pneumonia in Wuhan, China. JAMA. 2020 doi: 10.1001/jama.2020.1585.
    1. Zhou F, et al. Clinical course and risk factors for mortality of adult inpatients with COVID-19 in Wuhan, China: a retrospective cohort study. Lancet. 2020;395:1054–1062.
    1. Guan WJ, et al. Clinical characteristics of coronavirus disease 2019 in China. N. Engl. J. Med. 2020;382:1708–1720.
    1. Wu Z, McGoogan JM. Characteristics of and important lessons from the coronavirus disease 2019 (COVID-19) outbreak in China: summary of a report of 72314 cases from the Chinese Center for Disease Control and Prevention. JAMA. 2020 doi: 10.1001/jama.2020.2648.
    1. Ruan Q, Yang K, Wang W, Jiang L, Song J. Clinical predictors of mortality due to COVID-19 based on an analysis of data of 150 patients from Wuhan, China. Intensive Care Med. 2020;46:846–848.
    1. Shi S, et al. Association of cardiac injury with mortality in hospitalized patients with COVID-19 in Wuhan, China. JAMA Cardiol. 2020 doi: 10.1001/jamacardio.2020.0950.
    1. Guo T, et al. Cardiovascular implications of fatal outcomes of patients with coronavirus disease 2019 (COVID-19) JAMA Cardiol. 2020 doi: 10.1001/jamacardio.2020.1017.
    1. Shi S, et al. Characteristics and clinical significance of myocardial injury in patients with severe coronavirus disease 2019. Eur. Heart J. 2020;41:2070–2079.
    1. Riphagen S, Gomez X, Gonzalez-Martinez C, Wilkinson N, Theocharis P. Hyperinflammatory shock in children during COVID-19 pandemic. Lancet. 2020;395:1607–1608.
    1. Yin W, et al. Structural basis for inhibition of the RNA-dependent RNA polymerase from SARS-CoV-2 by remdesivir. Science. 2020;368:1499–1504.
    1. Dai W, et al. Structure-based design of antiviral drug candidates targeting the SARS-CoV-2 main protease. Science. 2020;368:1331–1335.
    1. Cui J, Li F, Shi ZL. Origin and evolution of pathogenic coronaviruses. Nat. Rev. Microbiol. 2019;17:181–192.
    1. Du L, et al. The spike protein of SARS-CoV — a target for vaccine and therapeutic development. Nat. Rev. Microbiol. 2009;7:226–236.
    1. Ge XY, et al. Isolation and characterization of a bat SARS-like coronavirus that uses the ACE2 receptor. Nature. 2013;503:535–538.
    1. Bar-On YM, Flamholz A, Phillips R, Milo R. SARS-CoV-2 (COVID-19) by the numbers. Elife. 2020 doi: 10.7554/eLife.57309.
    1. Masson P, et al. ViralZone: recent updates to the virus knowledge resource. Nucleic Acids Res. 2013;41:D579–D583.
    1. Lurie N, Saville M, Hatchett R, Halton J. Developing COVID-19 vaccines at pandemic speed. N. Engl. J. Med. 2020;382:1969–1973.
    1. Callaway E. The race for coronavirus vaccines: a graphical guide. Nature. 2020;580:576–577.
    1. Suthar MS, et al. Rapid generation of neutralizing antibody responses in COVID-19 patients. Cell Rep. Med. 2020;1:100040.
    1. Wang C, et al. A human monoclonal antibody blocking SARS-CoV-2 infection. Nat. Commun. 2020;11:2251.
    1. Ho TY, Wu SL, Chen JC, Li CC, Hsiang CY. Emodin blocks the SARS coronavirus spike protein and angiotensin-converting enzyme 2 interaction. Antivir. Res. 2007;74:92–101.
    1. Robson B. COVID-19 coronavirus spike protein analysis for synthetic vaccines, a peptidomimetic antagonist, and therapeutic drugs, and analysis of a proposed Achilles’ heel conserved region to minimize probability of escape mutations and drug resistance. Comput. Biol. Med. 2020;121:103749–103749.
    1. Perlman, S. & Masters, P. S. in Fields Virology: Emerging Viruses (eds Howley, P. M & knipe, D. M.) 410–448 (Lippincott Williams & Wilkins, 2020).
    1. Sanders JM, Monogue ML, Jodlowski TZ, Cutrell JB. Pharmacologic treatments for coronavirus disease 2019 (COVID-19): a review. JAMA. 2020 doi: 10.1001/jama.2020.6019.
    1. Li G, De Clercq E. Therapeutic options for the 2019 novel coronavirus (2019-nCoV) Nat. Rev. Drug Discov. 2020;19:149–150.
    1. Li SS, et al. Left ventricular performance in patients with severe acute respiratory syndrome: a 30-day echocardiographic follow-up study. Circulation. 2003;108:1798–1803.
    1. Peiris JS, et al. Clinical progression and viral load in a community outbreak of coronavirus-associated SARS pneumonia: a prospective study. Lancet. 2003;361:1767–1772.
    1. The Novel Coronavirus Pneumonia Emergency Response Epidemiology Team Vital surveillances: the epidemiological characteristics of an outbreak of 2019 novel coronavirus diseases (COVID-19) — China, 2020. China CDC Wkly. 2020;2:113–122.
    1. Grasselli G, et al. Baseline characteristics and outcomes of 1591 patients infected with SARS-CoV-2 admitted to ICUs of the Lombardy region, Italy. JAMA. 2020 doi: 10.1001/jama.2020.5394.
    1. Goyal P, et al. Clinical characteristics of COVID-19 in New York City. N. Engl. J. Med. 2020;382:2372–2374.
    1. Arentz M, et al. Characteristics and outcomes of 21 critically ill patients with COVID-19 in Washington state. JAMA. 2020 doi: 10.1001/jama.2020.4326.
    1. Bhatraju PK, et al. COVID-19 in critically ill patients in the Seattle region — case series. N. Engl. J. Med. 2020;382:2012–2022.
    1. Onder G, Rezza G, Brusaferro S. Case-fatality rate and characteristics of patients dying in relation to COVID-19 in Italy. JAMA. 2020 doi: 10.1001/jama.2020.4683.
    1. Richardson S, et al. Presenting characteristics, comorbidities, and outcomes among 5700 patients hospitalized with COVID-19 in the New York City area. JAMA. 2020 doi: 10.1001/jama.2020.6775.
    1. Stefanini GG, et al. ST-elevation myocardial infarction in patients with COVID-19: clinical and angiographic outcomes. Circulation. 2020;141:2113–2116.
    1. Deng Q, et al. Suspected myocardial injury in patients with COVID-19: evidence from front-line clinical observation in Wuhan, China. Int. J. Cardiol. 2020;311:116–121.
    1. Wang D, et al. Chinese Society of Cardiology expert consensus statement on the diagnosis and treatment of adult fulminant myocarditis. Sci. China Life Sci. 2019;62:187–202.
    1. Inciardi RM, et al. Cardiac involvement in a patient with coronavirus disease 2019 (COVID-19) JAMA Cardiol. 2020 doi: 10.1001/jamacardio.2020.1096.
    1. Hu H, Ma F, Wei X, Fang Y. Coronavirus fulminant myocarditis saved with glucocorticoid and human immunoglobulin. Eur. Heart J. 2020 doi: 10.1093/eurheartj/ehaa190.
    1. Xu Z, et al. Pathological findings of COVID-19 associated with acute respiratory distress syndrome. Lancet Respir. Med. 2020;8:420–422.
    1. Tavazzi G, et al. Myocardial localization of coronavirus in COVID-19 cardiogenic shock. Eur. J. Heart Fail. 2020;22:911–915.
    1. Wichmann D, et al. Autopsy findings and venous thromboembolism in patients with COVID-19. Ann. Intern. Med. 2020 doi: 10.7326/M20-2003.
    1. Schaller T, et al. Postmortem examination of patients with COVID-19. JAMA. 2020 doi: 10.1001/jama.2020.8907.
    1. Jiang F, et al. Angiotensin-converting enzyme 2 and angiotensin 1–7: novel therapeutic targets. Nat. Rev. Cardiol. 2014;11:413–426.
    1. Oudit GY, et al. SARS-coronavirus modulation of myocardial ACE2 expression and inflammation in patients with SARS. Eur. J. Clin. Invest. 2009;39:618–625.
    1. Kwong JC, et al. Acute myocardial infarction after laboratory-confirmed influenza infection. N. Engl. J. Med. 2018;378:345–353.
    1. Madjid M, et al. Influenza epidemics and acute respiratory disease activity are associated with a surge in autopsy-confirmed coronary heart disease death: results from 8 years of autopsies in 34,892 subjects. Eur. Heart J. 2007;28:1205–1210.
    1. Chong PY, et al. Analysis of deaths during the severe acute respiratory syndrome (SARS) epidemic in Singapore: challenges in determining a SARS diagnosis. Arch. Pathol. Lab. Med. 2004;128:195–204.
    1. Bangalore S, et al. ST-segment elevation in patients with COVID-19 – a case series. N. Engl. J. Med. 2020;382:2478–2480.
    1. Libby P, Tabas I, Fredman G, Fisher EA. Inflammation and its resolution as determinants of acute coronary syndromes. Circ. Res. 2014;114:1867–1879.
    1. Bentzon JF, Otsuka F, Virmani R, Falk E. Mechanisms of plaque formation and rupture. Circ. Res. 2014;114:1852–1866.
    1. Varga Z, et al. Endothelial cell infection and endotheliitis in COVID-19. Lancet. 2020;395:1417–1418.
    1. Garcia S, et al. Reduction in ST-segment elevation cardiac catheterization laboratory activations in the United States during COVID-19 pandemic. J. Am. Coll. Cardiol. 2020;75:2871–2872.
    1. De Filippo O, et al. Reduced rate of hospital admissions for ACS during COVID-19 outbreak in Northern Italy. N. Engl. J. Med. 2020;383:88–89.
    1. Rodríguez-Leor O, et al. Impact of the COVID-19 pandemic on interventional cardiology activity in Spain. Rec. Interventional Cardiol. Engl. Ed. 2020 doi: 10.24875/recice.M20000123.
    1. De Rosa S, et al. Reduction of hospitalizations for myocardial infarction in Italy in the COVID-19 era. Eur. Heart J. 2020;41:2083–2088.
    1. Baldi E, et al. Out-of-hospital cardiac arrest during the COVID-19 outbreak in Italy. N. Engl. J. Med. 2020 doi: 10.1056/NEJMc2010418.
    1. Pessoa-Amorim G, et al. Admission of patients with STEMI since the outbreak of the COVID-19 pandemic. A survey by the European Society of Cardiology. Eur. Heart J. Qual. Care Clin. Outcomes. 2020 doi: 10.1093/ehjqcco/qcaa046.
    1. Chen T, et al. Clinical characteristics of 113 deceased patients with coronavirus disease 2019: retrospective study. BMJ. 2020;368:m1091.
    1. Mehra MR, Ruschitzka F. COVID-19 illness and heart failure: a missing link? JACC Heart Fail. 2020;8:512–514.
    1. Dewey M, et al. Clinical quantitative cardiac imaging for the assessment of myocardial ischaemia. Nat. Rev. Cardiol. 2020;17:427–450.
    1. Manka R, et al. Myocardial edema in COVID-19 on cardiac MRI. J. Heart Lung Transplant. 2020;39:730–732.
    1. Fried JA, et al. The variety of cardiovascular presentations of COVID-19. Circulation. 2020;141:1930–1936.
    1. Prabhu SD. Cytokine-induced modulation of cardiac function. Circ. Res. 2004;95:1140–1153.
    1. Liu K, et al. Clinical characteristics of novel coronavirus cases in tertiary hospitals in Hubei province. Chin. Med. J. 2020;133:1025–1031.
    1. Lakkireddy DR, et al. Guidance for cardiac electrophysiology during the COVID-19 pandemic from the Heart Rhythm Society COVID-19 task force; electrophysiology section of the American College of Cardiology; and the electrocardiography and arrhythmias committee of the council on clinical cardiology, American Heart Association. Circulation. 2020;141:e823–e831.
    1. Panigada M, et al. Hypercoagulability of COVID-19 patients in intensive care unit. A report of thromboelastography findings and other parameters of hemostasis. J. Thromb. Haemost. 2020;18:1738–1742.
    1. Ranucci M, et al. The procoagulant pattern of patients with COVID-19 acute respiratory distress syndrome. J. Thromb. Haemost. 2020;18:1747–1751.
    1. Taylor FB, Jr., et al. Towards definition, clinical and laboratory criteria, and a scoring system for disseminated intravascular coagulation. Thromb. Haemost. 2001;86:1327–1330.
    1. Oxley TJ, et al. Large-vessel stroke as a presenting feature of COVID-19 in the young. N. Engl. J. Med. 2020;382:e60.
    1. Bellosta R, et al. Acute limb ischemia in patients with COVID-19 pneumonia. J. Vasc. Surg. 2020 doi: 10.1016/j.jvs.2020.04.483.
    1. Paranjpe I, et al. Association of treatment dose anticoagulation with in-hospital survival among hospitalized patients with COVID-19. J. Am. Coll. Cardiol. 2020;76:122–124.
    1. Tang N, et al. Anticoagulant treatment is associated with decreased mortality in severe coronavirus disease 2019 patients with coagulopathy. J. Thromb. Haemost. 2020;18:1094–1099.
    1. Qiu H, et al. Clinical and epidemiological features of 36 children with coronavirus disease 2019 (COVID-19) in Zhejiang, China: an observational cohort study. Lancet Infect. Dis. 2020;20:689–696.
    1. Verdoni L, et al. 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.
    1. Aslam S, Mehra MR. COVID-19: yet another coronavirus challenge in transplantation. J. Heart Lung Transplant. 2020;39:408–409.
    1. Li F, Cai J, Dong N. First cases of COVID-19 in heart transplantation from China. J. Heart Lung Transplant. 2020;39:496–497.
    1. Ketcham SW, et al. Coronavirus disease-2019 in heart transplant recipients in southeastern Michigan: a case series. J. Card. Fail. 2020;26:457–461.
    1. Latif F, et al. Characteristics and outcomes of recipients of heart transplant with coronavirus disease 2019. JAMA Cardiol. 2020 doi: 10.1001/jamacardio.2020.2159.
    1. Gosain R, et al. COVID-19 and cancer: a comprehensive review. Curr. Oncol. Rep. 2020;22:53.
    1. Ganatra S, Hammond SP, Nohria A. The novel coronavirus disease (COVID-19) threat for patients with cardiovascular disease and cancer. JACC CardioOncol. 2020 doi: 10.1016/j.jaccao.2020.03.001.
    1. Liang W, et al. Cancer patients in SARS-CoV-2 infection: a nationwide analysis in China. Lancet Oncol. 2020;21:335–337.
    1. Turner AJ, Hiscox JA, Hooper NM. ACE2: from vasopeptidase to SARS virus receptor. Trends Pharmacol. Sci. 2004;25:291–294.
    1. GTEx Portal (ACE2). Gene expression for ACE2 (2020).
    1. 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.
    1. Litvinukova, M. et al. Cells and gene expression programs in the adult human heart. Preprint at bioRxiv10.1101/2020.04.03.024075 (2020).
    1. Kaiser, J. How sick will the coronavirus make you? The answer may be in your genes. Science10.1126/science.abb9192 (2020).
    1. The COVID-19 Host Genetics Initiative, a global initiative to elucidate the role of host genetic factors in susceptibility and severity of the SARS-CoV-2 virus pandemic. Eur. J. Hum. Genet.28, 715–718 (2020).
    1. Imai Y, et al. Angiotensin-converting enzyme 2 protects from severe acute lung failure. Nature. 2005;436:112–116.
    1. Kuba K, et al. A crucial role of angiotensin converting enzyme 2 (ACE2) in SARS coronavirus-induced lung injury. Nat. Med. 2005;11:875–879.
    1. Crackower MA, et al. Angiotensin-converting enzyme 2 is an essential regulator of heart function. Nature. 2002;417:822–828.
    1. Thomas MC, et al. Genetic Ace2 deficiency accentuates vascular inflammation and atherosclerosis in the ApoE knockout mouse. Circ. Res. 2010;107:888–897.
    1. Zhao, Y. et al. Single-cell RNA expression profiling of ACE2, the receptor of SARS-CoV-2. Preprint at bioRxiv10.1101/2020.01.26.919985 (2020).
    1. Qi F, Qian S, Zhang S, Zhang Z. Single cell RNA sequencing of 13 human tissues identify cell types and receptors of human coronaviruses. Biochem. Biophys. Res. Commun. 2020;526:135–140.
    1. Sungnak W, et al. SARS-CoV-2 entry factors are highly expressed in nasal epithelial cells together with innate immune genes. Nat. Med. 2020;26:681–687.
    1. Vaduganathan M, et al. Renin–angiotensin–aldosterone system inhibitors in patients with COVID-19. N. Engl. J. Med. 2020;382:1653–1659.
    1. Monteil V, et al. Inhibition of SARS-CoV-2 infections in engineered human tissues using clinical-grade soluble human ACE2. Cell. 2020;181:905–913.
    1. Haschke M, et al. Pharmacokinetics and pharmacodynamics of recombinant human angiotensin-converting enzyme 2 in healthy human subjects. Clin. Pharmacokinet. 2013;52:783–792.
    1. Khan A, et al. A pilot clinical trial of recombinant human angiotensin-converting enzyme 2 in acute respiratory distress syndrome. Crit. Care. 2017;21:234.
    1. Wadman M, Couzin-Frankel J, Kaiser J, Matacic C. A rampage through the body. Science. 2020;368:356–360.
    1. Puelles VG, et al. Multiorgan and renal tropism of SARS-CoV-2. N. Engl. J. Med. 2020 doi: 10.1056/NEJMc2011400.
    1. Kuster GM, et al. SARS-CoV-2: should inhibitors of the renin–angiotensin system be withdrawn in patients with COVID-19? Eur. Heart J. 2020;41:1801–1803.
    1. Sommerstein R, Kochen MM, Messerli FH, Grani C. Coronavirus disease 2019 (COVID-19): do angiotensin-converting enzyme inhibitors/angiotensin receptor blockers have a biphasic effect? J. Am. Heart Assoc. 2020;9:e016509.
    1. Ferrario CM, et al. Effect of angiotensin-converting enzyme inhibition and angiotensin II receptor blockers on cardiac angiotensin-converting enzyme 2. Circulation. 2005;111:2605–2610.
    1. Bozkurt, B., Kovacs, R. & Harrinton, B. HFSA/ACC/AHA statement addresses concerns re: using RAAS antagonists in COVID-19. AHA Professional Heart Daily (2020).
    1. de Simone, G. Position statement of the ESC council on hypertension on ACE-inhibitors and angiotensin receptor blockers. ESC escardio (2020).
    1. Chinese Society of Cardiology. Scientific statement on using renin–angiotensin system blockers in patients with cardiovascular disease and COVID-19. Chin. J. Cardiol. 2020;48:E014.
    1. de Abajo FJ, et al. Use of renin–angiotensin–aldosterone system inhibitors and risk of COVID-19 requiring admission to hospital: a case-population study. Lancet. 2020;395:1705–1714.
    1. 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;382:2431–244.
    1. Reynolds HR, et al. Renin–angiotensin–aldosterone system inhibitors and risk of COVID-19. N. Engl. J. Med. 2020;382:2441–2448.
    1. Mehta N, et al. Association of use of angiotensin-converting enzyme inhibitors and angiotensin II receptor blockers with testing positive for coronavirus disease 2019 (COVID-19) JAMA Cardiol. 2020 doi: 10.1001/jamacardio.2020.1855.
    1. Ishiyama Y, et al. Upregulation of angiotensin-converting enzyme 2 after myocardial infarction by blockade of angiotensin II receptors. Hypertension. 2004;43:970–976.
    1. Soler MJ, et al. Localization of ACE2 in the renal vasculature: amplification by angiotensin II type 1 receptor blockade using telmisartan. Am. J. Physiol. Ren. Physiol. 2009;296:F398–F405.
    1. Burrell LM, et al. Myocardial infarction increases ACE2 expression in rat and humans. Eur. Heart J. 2005;26:369–375.
    1. Ocaranza MP, et al. Enalapril attenuates downregulation of angiotensin-converting enzyme 2 in the late phase of ventricular dysfunction in myocardial infarcted rat. Hypertension. 2006;48:572–578.
    1. Luque M, et al. Effects of captopril related to increased levels of prostacyclin and angiotensin-(1–7) in essential hypertension. J. Hypertens. 1996;14:799–805.
    1. Furuhashi M, et al. Urinary angiotensin-converting enzyme 2 in hypertensive patients may be increased by olmesartan, an angiotensin II receptor blocker. Am. J. Hypertens. 2015;28:15–21.
    1. Epelman S, et al. Soluble angiotensin-converting enzyme 2 in human heart failure: relation with myocardial function and clinical outcomes. J. Card. Fail. 2009;15:565–571.
    1. Ramchand J, et al. Plasma ACE2 activity predicts mortality in aortic stenosis and is associated with severe myocardial fibrosis. JACC Cardiovasc. Imaging. 2020;13:655–664.
    1. Walters TE, et al. Angiotensin converting enzyme 2 activity and human atrial fibrillation: increased plasma angiotensin converting enzyme 2 activity is associated with atrial fibrillation and more advanced left atrial structural remodelling. Europace. 2017;19:1280–1287.
    1. Ramchand J, Patel SK, Srivastava PM, Farouque O, Burrell LM. Elevated plasma angiotensin converting enzyme 2 activity is an independent predictor of major adverse cardiac events in patients with obstructive coronary artery disease. PLoS One. 2018;13:e0198144.
    1. Pushpakom S, et al. Drug repurposing: progress, challenges and recommendations. Nat. Rev. Drug Discov. 2019;18:41–58.
    1. Andersen PI, et al. Discovery and development of safe-in-man broad-spectrum antiviral agents. Int. J. Infect. Dis. 2020;93:268–276.
    1. Guy RK, DiPaola RS, Romanelli F, Dutch RE. Rapid repurposing of drugs for COVID-19. Science. 2020;368:829–830.
    1. Roden DM, Harrington RA, Poppas A, Russo AM. Considerations for drug interactions on QTc in exploratory COVID-19 treatment. Circulation. 2020;141:e906–e907.
    1. Chen, Z. et al. Efficacy of hydroxychloroquine in patients with COVID-19: results of a randomized clinical trial. Preprint at medRxiv10.1101/2020.03.22.20040758 (2020).
    1. Gautret, P. et al. Hydroxychloroquine and azithromycin as a treatment of COVID-19: results of an open-label non-randomized clinical trial. Int. J. Antimicrob. Agents, 105949, 10.1016/j.ijantimicag.2020.105949 (2020).
    1. Wang M, et al. Remdesivir and chloroquine effectively inhibit the recently emerged novel coronavirus (2019-nCoV) in vitro. Cell Res. 2020;30:269–271.
    1. Savarino A, Boelaert JR, Cassone A, Majori G, Cauda R. Effects of chloroquine on viral infections: an old drug against today’s diseases? Lancet Infect. Dis. 2003;3:722–727.
    1. ISAC/Elsevier statement. Joint ISAC and Elsevier statement on Gautret et al. paper. International Society of Antimicrobial Chemotherapy (2020).
    1. Yu B, et al. Low dose of hydroxychloroquine reduces fatality of critically ill patients with COVID-19. Sci. China Life. Sci. 2020 doi: 10.1007/s11427-020-1732-2.
    1. Geleris J, et al. Observational study of hydroxychloroquine in hospitalized patients with COVID-19. N. Engl. J. Med. 2020;382:2411–2418.
    1. Fihn SD, Perencevich E, Bradley SM. Caution needed on the use of chloroquine and hydroxychloroquine for coronavirus disease 2019. JAMA Netw. Open. 2020;3:e209035.
    1. Mercuro NJ, et al. Risk of QT interval prolongation associated with use of hydroxychloroquine with or without concomitant azithromycin among hospitalized patients testing positive for coronavirus disease 2019 (COVID-19) JAMA Cardiol. 2020 doi: 10.1001/jamacardio.2020.1834.
    1. Hancox JC, Hasnain M, Vieweg WV, Crouse EL, Baranchuk A. Azithromycin, cardiovascular risks, QTc interval prolongation, Torsade de Pointes, and regulatory issues: a narrative review based on the study of case reports. Ther. Adv. Infect. Dis. 2013;1:155–165.
    1. Rosenberg ES, et al. Association of treatment with hydroxychloroquine or azithromycin with in-hospital mortality in patients with COVID-19 in New York state. JAMA. 2020 doi: 10.1001/jama.2020.8630.
    1. Grein J, et al. Compassionate use of remdesivir for patients with severe COVID-19. N. Engl. J. Med. 2020;382:2327–2336.
    1. Wang Y, et al. Remdesivir in adults with severe COVID-19: a randomised, double-blind, placebo-controlled, multicentre trial. Lancet. 2020;395:1569–1578.
    1. Sheahan TP, et al. Comparative therapeutic efficacy of remdesivir and combination lopinavir, ritonavir, and interferon beta against MERS-CoV. Nat. Commun. 2020;11:222.
    1. Beigel JH, et al. Remdesivir for the treatment of COVID-19 – preliminary report. N. Engl. J. Med. 2020 doi: 10.1056/NEJMoa2007764.
    1. US FDA. Fact sheet for health care providers: emergency use authorization (EUA) of remdesivir (GS-5734™). (2020).
    1. Cao B, et al. A trial of lopinavir–ritonavir in adults hospitalized with severe COVID-19. N. Engl. J. Med. 2020;382:1787–1799.
    1. Stockman LJ, Bellamy R, Garner P. SARS: systematic review of treatment effects. PLoS Med. 2006;3:e343.
    1. Centers for Disease Control and Prevention. Interim laboratory biosafety guidelines for handling and processing specimens associated with coronavirus disease 2019 (COVID-19). (CDC, 2020).
    1. World Health Organization. Laboratory biosafety guidance related to coronavirus disease 2019 (COVID-19). (2020).
    1. Bao L, et al. The pathogenicity of SARS-CoV-2 in hACE2 transgenic mice. Nature. 2020 doi: 10.1038/s41586-020-2312-y.
    1. Rockx B, et al. Comparative pathogenesis of COVID-19, MERS, and SARS in a nonhuman primate model. Science. 2020;368:1012–1015.
    1. Shi J, et al. Susceptibility of ferrets, cats, dogs, and other domesticated animals to SARS-coronavirus 2. Science. 2020;368:1016–1020.
    1. McCray PB, Jr., et al. Lethal infection of K18-hACE2 mice infected with severe acute respiratory syndrome coronavirus. J. Virol. 2007;81:813–821.
    1. Kim YI, et al. Infection and rapid transmission of SARS-CoV-2 in ferrets. Cell Host Microbe. 2020;27:704–709.
    1. Park SJ, et al. Ferret animal model of severe fever with thrombocytopenia syndrome phlebovirus for human lethal infection and pathogenesis. Nat. Microbiol. 2019;4:438–446.
    1. Chan JF, et al. Simulation of the clinical and pathological manifestations of coronavirus disease 2019 (COVID-19) in golden Syrian hamster model: implications for disease pathogenesis and transmissibility. Clin. Infect. Dis. 2020 doi: 10.1093/cid/ciaa325.
    1. Bao, L. et al. Lack of reinfection in rhesus macaques infected with SARS-CoV-2. Preprint at bioRxiv10.1101/2020.03.13.990226 (2020).
    1. Chen IY, Matsa E, Wu JC. Induced pluripotent stem cells: at the heart of cardiovascular precision medicine. Nat. Rev. Cardiol. 2016;13:333–349.
    1. Shi Y, Inoue H, Wu JC, Yamanaka S. Induced pluripotent stem cell technology: a decade of progress. Nat. Rev. Drug Discov. 2017;16:115–130.
    1. Sharma, A. et al. Human iPSC-derived cardiomyocytes are susceptible to SARS-CoV-2 infection. Cell Rep. Med.10.1016/j.xcrm.2020.100052 (2020).
    1. McCauley KB, Hawkins F, Kotton DN. Derivation of epithelial-only airway organoids from human pluripotent stem cells. Curr. Protoc. Stem Cell Biol. 2018;45:e51.
    1. McCauley KB, et al. Efficient derivation of functional human airway epithelium from pluripotent stem cells via temporal regulation of Wnt signaling. Cell Stem Cell. 2017;20:844–857.

Source: PubMed

3
Subskrybuj