ReviewThe Pathophysiology of COVID-19 and SARS-CoV-2 Infection

Angiotensin-converting enzyme 2 and COVID-19: patients, comorbidities, and therapies

Published Online:https://doi.org/10.1152/ajplung.00259.2020

Abstract

On March 11, 2020, the World Health Organization declared coronavirus disease 2019 (COVID-19) a pandemic, and the reality of the situation has finally caught up to the widespread reach of the disease. The presentation of the disease is highly variable, ranging from asymptomatic carriers to critical COVID-19. The availability of angiotensin-converting enzyme 2 (ACE2) receptors may reportedly increase the susceptibility and/or disease progression of COVID-19. Comorbidities and risk factors have also been noted to increase COVID-19 susceptibility. In this paper, we hereby review the evidence pertaining to ACE2’s relationship to common comorbidities, risk factors, and therapies associated with the susceptibility and severity of COVID-19. We also highlight gaps of knowledge that require further investigation. The primary comorbidities of respiratory disease, cardiovascular disease, renal disease, diabetes, obesity, and hypertension had strong evidence. The secondary risk factors of age, sex, and race/genetics had limited-to-moderate evidence. The tertiary factors of ACE inhibitors and angiotensin II receptor blockers had limited-to-moderate evidence. Ibuprofen and thiazolidinediones had limited evidence.

INTRODUCTION

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) caused over 3 million infections and over 250,000 deaths worldwide 2 months after the World Health Organization (WHO) declared the virus-induced disease a pandemic. Mathematical models have shown drastic exponential growth in these infections. However, the coronavirus disease 2019 (COVID-19) does not indiscriminately affect the population. Certain comorbidities and risk factors have shown increased susceptibility and disease severity (1, 2).

The average observed case-fatality ratio is around 4% but varies widely (3). Earlier reports suggested that elderly patients with comorbidities, particularly diabetes, cardiovascular disease, and hypertension, suffered greater mortality than younger patients (2, 46). Nevertheless, younger patients with no apparent disease also appeared prone to rapid progression, severe/critical disease, and death.

Angiotensin-converting enzyme 2 (ACE2) is an important regulator of the renin-angiotensin system (RAS), and SARS-CoV-2 propagates on to host ACE2 receptors as a vehicle for invasion of human cells (7). ACE2 is found mostly in the endothelium, lungs, heart, kidneys, and intestines, which parallels the detection of SARS-CoV-2 in multiple organs found in silico analysis (710). Moreover, certain comorbidities have been shown to increase ACE2 levels, which may increase COVID-19’s severity and susceptibility potential.

This article reviews the relationship among ACE2 levels, common comorbidities, and risk factors in COVID-19 cases. An electronic search was conducted in LitCovid, PubMed, Google Scholar, WHO, and Centers for Disease Control and Prevention (CDC) databases. Search terms included COVID-19, SARS-CoV-2, 2019-nCOV, angiotensin-converting enzyme 2, and ACE2. Manuscripts published were reviewed; relevant references were checked. The information in this review is current as of this article’s writing (November 3, 2020), since new data will likely emerge during the pandemic.

PHYSIOLOGY OF ACE2

ACE2/ANG-1–7/Mas axis is the prominent counterregulator against activated RAS pathophysiology (Fig. 1). SARS-CoV-2 uses the receptor ACE2 for host cell entry and viral replication.

Figure 1.

Figure 1.Schematic representation of renin-angiotensin system (RAS) and homeostatic features. RAS regulates vascular function, blood pressure, and fluid and electrolyte balance. The liver synthesizes and releases angiotensinogen into the circulatory system. Angiotensinogen is then converted to the decapeptide angiotensin I until it reaches the lungs, where angiotensin-converting enzyme (ACE) converts it to the octapeptide angiotensin II (ANG II) (11, 12). ANG II, a powerful vasoconstrictor, has short-term presence in the blood before it is metabolized (13). The proinflammatory effects of ANG II are further mediated by ANG II type I receptor, which stimulates aldosterone secretion from the adrenal medulla and antidiuretic hormone from the posterior pituitary. A key regulator of RAS is ACE2, a monocarboxypeptidase, that metabolizes and inactivates ANG II to the hepapeptide angiotensin 1–7 (ANG-1–7), which, after binding with the G protein-coupled receptor MAS receptor, decreases the vasoconstrictor stimulus (11, 12). ANG-1–7 can also be produced directly via zinc metallopeptidase neprilysin/prolyl endopeptidases or through conversion of angiotensin 1–9 by ACE but with lower efficiency (11, 12). ANG-1–7’s other protective effects include anti-fibrotic, anti-inflammatory, antioxidant and antihypertrophic qualities (14). Knockout mice have shown that reduced ACE2 increases tissue and circulating levels of angiotensin II (13, 15). Additionally, the therapeutic treatment mechanism is shown here: β-blockers inhibit renin and prevent conversion of angiotensinogen to angiotensin I; ACE inhibitor blocks ACE and prevents conversion of angiotensin I to II; angiotensin receptor blockers (ARBs) prevent angiotensin II from binding to its receptor.


SARS-CoV-2 is transmitted via respiratory droplets (>5 μm) or aerosol transmission (<5 μm) (16, 17). Early in infection, the virus targets the nasal and oral mucosa, bronchial epithelial cells, and pneumocytes as they are enriched with ACE2 receptors, the entry points in facilitating SARS-CoV-2’s access to the airways and the body in general (18, 19). On the virion surface, Spike’s glycoprotein (S protein) mediates receptor recognition and membrane fusion onto ACE2’s peptidase domain, together with the type 2 transmembrane serine protease (TMPRSS2), with a high affinity of Kd ∼15 nM (2022). Viral entry further upregulates ADAM17 protease activity, which downregulates ACE2 by cleaving the receptor from the cell surface (“shedding”), thereby shifting protective ACE2/angiotensin 1–7/Mas axis toward the disease state and accumulation in angiotensin II (ANG II) (12, 14). Recent studies show that SARS-CoV-2 binds with greater affinity than the prior SARS-CoV (2325) and resiliently binds to ACE2 receptors with a low-species barrier (26). Additionally, current studies suggest antigenic drift variations in the harboring S protein to possibly present with increased affinity to ACE2 docking sites. For example, 614 G variants positively correlated with infectivity and fatality rates (2729), despite reduced affinity for ACE2 due to faster dissociation rates (30). The continuing evolution of SARS-CoV-2’s durable and adaptable bind with ACE2 receptors could partly explain the transmissibility affecting most nations.

PHYSIOLOGY OF EXTRAPULMONARY MANIFESTATIONS

COVID-19 infection ranges from asymptomatic to critical. Figure 2 highlights key phases of disease progression from presymptomatic/asymptomatic to late phase/severe disease involving direct organ-toxicity, systemic inflammatory response syndrome (cytokine storm), and endothelial damage and thromboinflammation (12, 32).

Figure 2.

Figure 2.Infected individuals may remain asymptomatic up to a week before encountering mild to moderate symptoms of fever, dry cough, sore throat, loss of smell and taste, or head and body aches. Eventually severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), an enveloped, nonsegmented positive-sense RNA virus, infects angiotensin-converting enzyme 2 (ACE2)-expressing type II alveolar epithelial cells in the lower respiratory tract, where 90% of symptomatic individuals have reported pneumonitis (31). Progression to severe/critical involves viral toxicity, disruption of the epithelial-endothelial barrier, complement depositions, and hyperinflammation, commonly requiring ventilation and/or life support for multiorgan injury (31). Severe COVID-19 is defined as dyspnea, respiratory rate ≥30/min, SpO2  ≤93%, PaO2 / FiO2  <300, and/or lung infiltrates >50% within 24–48 h, compared with critical disease, which further involves respiratory failure, septic shock, and organ dysfunction/failure (5). [Figure reproduced with permission from Matheson and Lehner (31).]


ACE2 and Organ-Toxicity

SARS-CoV-2’s direct tropism onto ACE2 receptors on varying tissue, aside from lung parenchyma, is believed to cause multiorgan injury (710). Histopathological reports confirm SARS-CoV-2’s direct organotropism, thus speculate preexisting conditions to aggravate COVID-19’s course (9, 3338). The mechanism of the virus’s systematic spread, whether lymphatic, hematogenous, or otherwise, remains elusive.

ACE2 and Cytokine Storm

Host cells lyse during rapid viral replication directly producing proinflammatory cytokines such as TNF-α, IL-1β, IL-6, INF-γ, monocyte chemoattractant protein-1, and TGF-β. INF-γ and IL-6 are prominent inflammatory markers linked to the worst outcomes (3945). Specifically, IL-6’s signaling pathway elevates the following serum inflammatory markers: C-reactive protein (CRP), procalcitonin, erythrocyte sedimentation rate, D-dimer, fibrinogen, lactate dehydrogenase, ferritin, and cardiac stress markers (12, 32, 46). Excessive inflammatory responses may also predispose patients to thrombosis due to platelet activation, endothelial dysfunction, and stasis (47). Thrombosis further exacerbates proinflammatory effects of endothelial injury and phagocytic responses.

In addition, the imbalance of the ACE2/ANG-1–7/Mas axis contributes to systemic inflammation. INF-γ and SARS-CoV-2 downregulate ACE2, resulting in elevated ANG II, which causes pulmonary vasoconstriction, additional inflammation, and oxidative and fibrotic organ damage, ultimately advancing toward acute respiratory distress syndrome (4850). Studies have demonstrated that the loss or downregulation of ACE2 perpetuates acute lung injury (ALI); in conjunction, rhACE2 improves it (49, 5153). Conversely, Ziegler et al. (54) found that INF-γ and influenza increased ACE2 in human nasal epithelia and lung tissue, suggesting a finer balance between INF-γ and SARS-CoV-2 regulation of ACE2.

A combination of viral-induced lysis, thrombosis, and dysregulation of ACE2 overwhelms the systemic release of cytokines, resulting in cytokine storm. Emerging studies reveal a positive correlation of type-I INF deficiency to clinical severity (55, 56). Therapeutics in study, e.g., dexamethasone, interferon-α-2b, interferon β-1b, aim to regulate hyperinflammatory responses (5759).

ACE2 and Thrombosis

Risk of arterial/venous thrombosis, acute coronary syndrome, and stroke in severe/critical patients remains high: 29.4%–50% (6063). Endothelial dysfunction in COVID-19 patients results from ACE2-mediated binding of SARS-CoV-2, hyperinflammation, and prothrombin upregulation (32, 33, 64). Direct viral toxicity creates microthrombi and fibrinous exudates that dysregulate coagulant pathways (33, 38, 6567). In conjunction, endothelial injury (i.e., elevated von Willebrand) and endothelialitis facilitate proinflammatory effects by triggering elevated D-dimer, prothrombin time, and activated partial thromboplastin time prolongation, fibrinogen, and complement. Endothelialitis involves neutrophil and macrophage-facilitated cytokine release and neutrophil extracellular trap formation, which further damage the endothelium and activate extrinsic and intrinsic coagulation pathways (32, 63, 68). Moreover, hypoxia-mediated upregulation of hypoxia inducible factor-1 (HIF-1) activates cytokines, tissue factor, and plasminogen activator inhibitor type-1 (63, 67). These manifestations promote a procoagulative state, aggravating Virchow’s triad (32, 63, 68, 69). Heparin and other anticoagulants reduce thrombotic complications, decreasing COVID-19 patient mortality (47, 70, 71).

ACE2 and Susceptibility/Severity of COVID-19

ACE2 may mediate unique susceptibility to SARS-CoV-2, but the enzyme’s dysfunction evidently contributes to the severity of COVID-19.

Prior studies on SARS-CoV illustrate significant correlations to ACE2 expression in vitro (7274). However, evidence between ACE2 expression and SARS-CoV-2 remains new. Limited reports of hACE2 mice models depict increased SARS-CoV-2 viral load, compared with controls (7577). Rapidly emerging human studies mirror similar sentiments. Early COVID-19 histopathological reports noted increased SARS-CoV-2 expression and ACE2-positive receptors in affected lung, cardiac, and renal tissue (9, 3338). For instance, increased ACE2-positive endothelial cells were observed in significant endothelial injury, potentially correlating ACE2 expression to increased disease progression (33). Also, ACE2 RNA expression was associated with increased viral load from 430 nasopharyngeal swabs (78) and high admission viral load predicted in-hospital mortality in n = 2,914. Here, the mortality rate was 38.8% in high viral-load, 24.1% in medium viral load, and 15.3% in low viral-load patients (P < 0.001) (79). In conjunction, viral load was an independent predictor of mortality in a large hospitalized cohort, n = 1145—5.2 versus 6.4 mean log10 copies/mL, respectively, in alive versus deceased patients (80).

Furthermore, limited clinical studies depict elevated plasma ANG II and aldosterone levels correlating to COVID-19 severity (8184). Significant plasma ANG II level elevations were seen in 90.2% of the observed COVID-19 cases, especially in 100% of the critical COVID-19 cases (84); although Henry et al. (85) saw no differences in ANG II regarding disease severity. Liu et al. further delineated markedly increased ANG II levels linearly associated to viral loads and lung injury (82), and multivariate analyses observed aldosterone levels positively associated with severity (83).

Initial data indicate that increased ACE2 receptor availability poses greater severity to COVID-19, including an increased viral load, organ-toxicity, hyperinflammation, and endothelial dysfunction. Later on in the disease progression, ADAM17, together with inflammatory markers, directly downregulate/dysregulate ACE2, which leads to imbalance of ACE2/ANG II toward the disease state of elevated ANG II and aldosterone levels, further worsening conditions (52, 86). Novel therapeutics in study, e.g., camostat mesylate, nanobodies, decoy receptors, aim to disrupt S protein to inhibit viral entry into the host ACE2 (23, 8789). Early phase 2 investigations of human recombinant soluble ACE2 antibodies (NCT04335136) were seen to reduce SARS-CoV-2 viral loads in infected Vero-E6 cells by a factor of 1,000–5,000 and inhibit viral infections of kidney and vascular organoids, potentially decreasing direct organotropism and disease progression (90).

PRIMARY RISK FACTORS

Primary comorbidities increase COVID-19 susceptibility and severity. Reports have indicated that most COVID-19 patients have more than comorbidities; of these, ACE2 activity has been widely studied in experimental and clinical trials (Fig. 3).

Figure 3.

Figure 3.Respiratory disease: angiotensin-converting enzyme 2 (ACE2) receptors in the lower airways, most prominently in alveolar type II and epithelial cells (7). Although ACE2 in the lungs is lower compared with nasopharyngeal mucosa and other organs, ACE2 receptors are not evenly distributed throughout the lungs, which may be perceived as decreased ACE2 expression in immunohistological stains (19). ACE2 prevents prolonged increased ANG II production, which triggers pulmonary edema and acute respiratory distress syndrome (49). Knockout mice models for ACE2 led to severe lung injury when mice contracted H5N1, but treating knockout mice with rhACE2 decreased injury (91). Cardiovascular disease: ACE2 receptors localized in cardiac myocytes and intramyocardial vessels extending into the aortic intima. Elevated ACE2 metabolizes ANG II, a critical inotrope and growth factor for remodeling the cardiac extracellular matrix. Knockout mice illustrate that ACE2 loss results in early hypertrophy, accelerated myocardial infarction, fibrosis, and dilated cardiomyopathy from oxidative stress, pathologic hypertrophy, increased neutrophilic infiltration, and inflammatory cytokines INF-γ, IL-6, and the chemokine monocyte chemoattractant protein-1 (10, 12, 92, 93). Conversely, overexpression of ACE2/ANG-1–7 significantly reduces deleterious myocardial infarction-induced cardiac remodeling (94, 95). Hypertension: Experimental models have solidified ACE2 as a protector against hypertension, while deficiency exacerbates hypertension, defining the enzyme’s essential role for maintaining healthy blood pressure (9698). Models further illustrated that rhACE2 prevents hypertension by reducing plasma ANG II while increasing plasma ANG-1–7 levels (99); rhACE2 also has an established record for treating pulmonary arterial hypertension (NCT01597635 and NCT03177603) (100, 101). Renal disease: expressed predominantly in the proximal tubule, endothelial, podocytes, and smooth muscle cells of renal vessels (102, 103). Experimental animal models propose the importance of ACE2 in regulation of renal diseases to prevent injury and fibrosis, e.g., ACE2-deficient mice have been reported to increase age-related glomerulosclerosis (104). Diabetes mellitus: many organs involved in controlling blood sugar are rich in ACE2 (105). Although ACE2’s function here is unknown, it is implicated to cause β-cell proliferation and insulin secretion by decreasing islet fibrosis, possibly reducing type 2 diabetes (T2D) onset (106). Obesity: ACE2 expression was found to be higher in human subcutaneous adipose tissue and human visceral adipose tissue (107). ACE2 expresses potent anti-inflammatory effects in adipose tissue of obese, as seen in T2D mice (108). Gastrointestinal disease: presence of ACE2 was found in intestinal glandular cells, as well as gastric, duodenal and rectal epithelial cells. ACE2 may regulate homeostasis of intestinal amino acids, expression of antimicrobial peptides, and ecology of gut (109111). Cerebrovascular disease: ACE2 receptors are nonspecifically located in brain tissue but more prominently found in brain vasculature. ACE2 was observed to have beneficial effects on neurogenic blood pressure, stress response, anxiety, cognition, brain injury, and neurogenesis (32, 112). PT, prothrombin time; aPTT, activated partial thromboplastin time. [Figure reproduced with permission from Vabret et al. (46).]


Respiratory Disease

Strong evidence pertains to chronic obstructive pulmonary disease (COPD) and emphysema associated with increased risk of COVID-19 susceptibility and severity (1, 6, 113, 114). Meta-analysis of preexisting respiratory disease between severe and nonsevere COVID-19 patients was odds ratio (OR): 2.46 (1.76–3.44) (6), although mixed evidence is analyzed in asthmatics for risk and severity (1, 115117). School reopenings are shifting the focus toward safety of young asthmatic individuals (115, 118), with reports of 27% of hospitalized COVID-19 cases in young adults (119). In addition, smoking was associated with increased severity, intensive care unit (ICU) admission, and death in hospitalized COVID-19 patients [OR: 2.0(1.3–3.1) – 2.2(1.3–3.7)] (120124). Although likely related to severity, evidence to quantify the risk to smokers is unavailable (122).

RAS activity is intrinsically high in the lungs. Histopathological reports acknowledge direct viral toxicity, resulting in atypia and detachment of type II pneumocytes, hyaline membrane formation, interstitial inflammatory response, and endothelial dysfunction (33, 35, 37, 38, 125). A pathophysiological timeline of 65 cases stressed direct lung damage from viral organotropism during the first week but later transitioned to host inflammatory and hypercoagulable responses 10–28 days into the disease phase (125). Reports show mainly bronchitis and pneumonitis in mild/moderate individuals (31) and pneumonia, acute respiratory distress syndrome, and pulmonary embolism in severe/critical patients (125).

ACE2 elevation exists in acute and chronic lung disease to prevent lung injury. A study on the previous SARS-CoV virus revealed it in autopsy specimens from severe SARS patients with ALI elevated ACE2, SARS‐CoV S protein, RNA, and proinflammatory cytokines (126). Chronic lung disease studies have pursued ACE2’s role in COPD, emphysema, asthma, and other ailments.

Recent literature provides overwhelming evidence that COPD upregulates ACE2 and TMPRSS2 expression in the nasal, bronchial and lower airways (127134). Significant inverse relationships between ACE2 gene expression and predicted forced expiratory volume for 1 s (r = −[0.24 – 0.40]; P < 0.05) were reported (127, 129), and higher ACE2 mRNA and protein levels in lung tissue were seen in moderate to severe COPD (128). Toru et al. (135) discovered significant serum ACE2 level increases in 27 COPD patients. However, reductions in endothelin-1, which downregulate ACE2, were found during exacerbations of COPD, suggesting that ACE2 dysregulation exacerbates disease potentiation (136, 137). Although the mechanisms of ACE2 upregulation in COPD patients is unknown, potential gene regulators related to histone modifications, e.g., HAT, HDAC2, and KDM5B, were correlated to ACE2 expression (131).

Moreover, inflammatory cytokines have proved to be key mediators. Studies have found that TNF‐α concentrations increased in COPD patients’ sputum and plasma (138). TNF‐α, IL‐12, and IL‐17A were found to upregulate ACE2 expression in BEAS‐2B cells derived from lung tissue (134). Interestingly, the preprint of Finney et al. (139) reported that inhaled corticosteroid therapy reduced ACE2 pulmonary expression through suppression of type I interferon in in vitro and in vivo COPD models and thus may reduce COVID-19 susceptibility.

However, mixed and limited evidence supports ACE2’s role in asthma pathophysiology, which parallels the divided consensus on asthma as a significant COVID-19 comorbidity. In fact, patients with COPD had higher frequencies of severe cases than asthmatics (57.1% vs. 4.6%, P < 0.01) (134). Saheb Sharif-Askari et al. (133) found ACE2 and TMPRSS2 lung airway expression to be upregulated slightly, while plasma ACE2 was significantly upregulated in asthmatics. Radzikowska et al. (132) similarly detected elevated TMPRSS2 expression in children and adult asthmatics from available RNA-Seq databases. ACE2, correlated gene signatures were found to be significant in a subset of type 2, low patients with asthma with characteristics resembling known risk factors for severe COVID-19 (140). However, other recent studies concluded no significant differences or reductions in ACE2 expression in the lower airways with allergic sensitization and asthma, suggesting additional factors beyond ACE2 modulation in the response to COVID-19 in individuals with allergies (130, 134, 141). Heterogeneity of asthma endotypes, type 2 (allergic) versus nontype 2 (nonallergic), formulate possible confounders; IgE and blood eosinophils have been shown to lower ACE2 expression, potentially regulating ACE2 in type 2 inflammation (134, 141). Additionally, hypoxia is known to regulate ACE2 expression (142, 143), which could explain significant ACE2 expression in COPD patients and not asthmatics.

Additionally, studies consistently correlate tobacco use to an increase in ACE2 and TMPRSS2 gene expressions in bronchial and alveolar epithelial, bronchial alveolar lavage, and protein in blood and lung tissue, which may intensify COVID-19 severity in smokers (127134, 144). Current smokers had significantly higher gene expression than ex-smokers and nonsmokers (2.77 ± 0.91 vs. 2.00 ± 1.23 vs. 1.78 ± 0.39, respectively; P = 0.024) (128, 129). The additive factor of smoking in COPD patients caused them to have higher ACE2 expression, compared with nonsmoking COPD patients, suggesting that smoking may emulate a drastic modifying factor rather than a pathological disease. Therefore, hypoxia from smoking may contribute to elevated ACE2. These studies suggest that smokers are at an increased risk for COVID-19 susceptibility and severity, although evidence associates smoking only with increased severity.

In sum, ACE2 significantly contributes to lung injury repair in acute and chronic lung disease. This potentially increases COPD patients’ and smokers’ risk and susceptibility to severe/critical COVID-19, although evidence for asthma is less clear.

Cardiovascular Disease

Individuals with serious cardiac conditions, e.g., heart failure, coronary artery disease, cardiomyopathies, have the strongest, most consistent evidence for increased COVID-19 susceptibility and severity (1, 6, 124, 145). Interestingly, a large retrospective study (n = 144,279) from the United Kingdom found no difference between preexisting ischemic heart diseases in COVID-19 (11.4%) and non-COVID-19 (12%) deaths (146).

Mounting evidence from pathological reports observed viral myocarditis from SARS-CoV-2 direct cardiotoxicity as the presumed etiology of primary cardiac injury (9, 147, 148), while hyperinflammation is another mechanism (32, 37, 149). Reports indicate myocardial injury, cardiomyopathy, acute coronary syndrome, cor pulmonale, arrhythmias, and cardiogenic shock (32). In China, acute and chronic cardiovascular damage were seen in nearly 20% of 416 patients, arrhythmias in 44% of ICU patients, and significant higher risk of all-cause mortality in hospitalized patients (150153). Studies in New York City found that 6% of 4,250 patients have prolonged QTc at admission and atrial arrhythmias were in critical patients but not noncritical ones (17.7% vs. 1.9%) (4, 154). Moreover, echocardiography findings from 69 countries revealed heart damage in over half of COVID-19 patients (155). Postcomplications were noted from cardiovascular magnetic resonance imaging of ongoing cardiac involvement and myocardial inflammation in 78% and 60%, respectively, of German patients independent of comorbidities and severity (156).

Although the pathophysiology for cardiovascular manifestation is probably multifactorial, ACE2 abounds in cardiac tissue, suggesting a mechanism of direct SARS-CoV-2 infestation (157, 158). Individuals with underlying cardiovascular disease have upregulated RAS as a compensatory mechanism to maintain cardiac output mainly through the elevated ANG II pathophysiology (Fig. 3). Therefore, ACE2-generating cardiac diseases may sensitize the myocardium to elevated SARS-CoV-2 entry and viral replication, leading to ACE2 shedding. This would cause the myocardium to lose protective effects of ANG-1–7, causing inflammation, reactive oxygen species, and vasoconstriction for cardiac damage (159, 160).

Clinical and experimental studies have demonstrated relations between cardiovascular disease and ACE2. Elevated serum ACE2 was found within animal models of myocardial infarction, atherosclerotic development, reduced left ventricular ejection fraction, cardiomyopathies, and heart failure (12, 92). Burrell et al. (157) found that myocardial infarction significantly increased ACE2 mRNA in day 3 and 28 postmyocardial infarction for rats, which paralleled explanted failing hearts in human subjects. ACE2 was the most upregulated gene, and a fivefold increase in ACE2 protein was found in hypertrophic cardiomyopathy human cardiac tissue, compared with that of controls (159). Plasma ACE2 activity directly related to persistent atrial fibrillation (AF; 22.8 pmol/min/mL) and paroxysmal AF (16.9 pmol/min/mL), compared with control (13.3 pmol/min/mL) (161). Also, a study of 79 obstructive coronary artery disease patients revealed that they had significantly elevated plasma ACE2, which correlated with increased adverse long-term cardiovascular outcomes (162).

Moreover, numerous studies confirmed ACE2’s elevation in heart failure. Recent studies measured elevated plasma ACE2 concentrations in 1,485 men and 537 women with heart failure and a threefold increase in myocardial ACE2 gene expression in heart-failure patients (163, 164). Another study showed near-doubling of ACE2 activity in acute heart-failure patients, compared with healthy controls: 52.5 pmol/h/ml versus 22.5 pmol/h/ml, respectively. Chronic heart-failure patients also had increased ACE2 levels (33.6 pmol/h/ml) (165). Other studies demonstrate that soluble ACE2 activity positively correlates with heart failure severity (166, 167). Novel drugs targeting ACE2 are being studied in heart failure patients and are shown to attenuate effects of systolic and diastolic dysfunction (168). Ongoing clinical trials are investigating modulation of ACE2/ANG-1–7 balance with rhACE2 (NCT00886353) and cardiac progenitor cells (NCT02348515) (11).

These findings suggest that elevated ACE2 in preexisting cardiovascular disease may increase susceptibility and severity to SARS-CoV-2.

Hypertension

This is a notable risk factor for increased morbidity and mortality from COVID-19 (1, 6, 124), but whether hypertensive individuals are more susceptible to infection is unclear (169). Hypertension prevalence in COVID-19 patients seen in a meta-analysis denotes 21.1% (13.0–27.2) (6), in severe COVID-19 [OR: 2.36(1.46–3.83) – 2.72(1.60–4.64)] (6, 124). Furthermore, a hazard ratio (HR): 1.7–3.05 was reported for mortality in COVID-19 patients with hypertension (44, 45). SARS-CoV-2 predisposition may be due to ACE2 polymorphisms in individuals with hypertension (170).

Experimental models have solidified ACE2 as a protector against hypertension (Fig. 3). Hypertension’s integrative role with other organs affects RAS systemically and locally, which is activated by distinct signals from differing physiologic and pathophysiologic conditions (171, 172). Essential hypertension can be delineated into two groups based on plasma renin activity: those with low activity, and those with normal-to-high activity (173). Studies show that hypertension with high plasma renin activity relates to increased cardiovascular complications and vascular damage (173175). The deleterious effects may destroy ACE2 mRNA and protein activity in tissue, as hypertensive clinical and experimental models show, primarily in the kidneys and heart and thus depict hypertensive progression decreasing local ACE2 activity (97, 176180). We believe that organ damage from hypertension causes local destruction of ACE2 within the targeted organ. An autopsy study of 20 patients diagnosed with hypertensive cardiomyopathy or nephropathy revealed decreased local ACE2 expression (177).

However, human and animal models positively correlate plasma ACE2 activity with increased systolic blood pressure (167, 181187). The Leeds Family study detected significantly higher systolic and diastolic blood pressure, compared with those without detectable plasma ACE2 (182). The study also provided evidence of genetic effects on plasma ACE2, estimating that hereditable factors influenced 65% of variability in ACE2 levels. Meta-analysis revealed possible association of ACE2 G8790A and rs2106809 polymorphisms with essential hypertension risk (188). Furthermore, Li et al. (181) determined higher serum ACE2 concentrations in hypertensive patients than in healthy subjects (170.31 [83.50–707.12]p g/ml vs. 59.28 [39.71–81.81] pg/ml, respectively; P < 0.001). Another study revealed that ACE2 activity was 1.5 times greater in 239 hypertensive patients than in healthy volunteers (167).

We believe that shedding from endothelial cells release the catalytically active ectodomain ACE2 as a soluble form in plasma (12, 186) and that compensatory release of ACE2 from other organs provides appropriate systemic response to regulate homeostatic changes similar to the anti-inflammatory and antifibrotic ones observed in rhACE2 that increased systemic and/or local ACE2 levels (189192). Other studies have reported no association between plasma ACE2 and hypertension (193, 194). Whether SARS-CoV-2 utilizes plasma ACE2 as an active domain to replicate is unclear, but ACE2 shedding amplifies endothelial dysfunction and hyperinflammation, thus potentially increasing COVID-19 severity.

However, mixed results were noted in the relationship between secondary hypertension and ACE2 (178). Variability in experimental models may relate to acute and chronic models, mechanism of the type of secondary hypertension, and compensatory effects of other organs. RAS and ACE2 activity is notable in neurogenic hypertension, which is characterized by an increase in sympathetic activity and often resistance to drug treatments. The rostral ventrolateral medulla reportedly stimulates sympathetic preganglionic neurons, which activate ACE2 to regulate blood pressure. ACE2’s role downregulates ANG II-mediated presser and fluid retention effects (195). Loss of compensatory activity during neurogenic hypertension was seen in 27 patients with increased ACE2 activity in the cerebrospinal fluid, correlating with systolic blood pressure. Moreover, increased TNF-α was found, suggesting the upregulation of ADAM17 (186).

Experimental studies and pathological reports clearly find a decrease in local tissue ACE2 activity, which may relate to progressive organ damage from hypertension. However, strong evidence suggests associations between hypertension and elevated plasma ACE2 and upregulation of ADAM17, which may increase COVID-19 severity in hypertensive patients. Large clinical studies are needed to clarify ACE2’s role in essential and secondary hypertension, acute and chronic hypertension, and hypertensive disease progression. Determination of plasma renin activity in both hypertensive and nonhypertensive COVID-19 patients may also indicate prognosis and treatment decisions (173).

Renal Disease

Chronic kidney disease (CKD) has strong, consistent evidence for increased COVID-19 susceptibility and severity (1, 196, 197). Meta-analysis of 25 studies indicated CKD to have the most significant relative risk of mortality for COVID-19, 3.25(1.13–9.28) (197). Genetic risks are amplified for infected individuals with APOL1 gene variant, occurring in 14% of Blacks, causing deadly viral-induced collapsing focal segmental glomerulosclerosis (198).

The kidney is reportedly a target for SARS-CoV-2, which replicates there in almost 30% of infected patients (199). Increased renal disease prevalence before admission and acute kidney injury development was seen during hospitalization (200). Histopathological reports from postmortem COVID-19 patients indicated direct virulence in the kidneys (9, 36). Light microscopy observed proximal tubule injury with the loss of brush border, vasculitis, nonisometric vacuolar degeneration, and necrosis, indicating the combined effects of viral toxicity, thrombosis, and cytokine storm (9, 32, 36, 64).

Abundant ACE2 in the kidneys (Fig. 3) threatens to be a culprit for risks of susceptibility and severe/critical COVID-19. Overwhelming evidence of experimental and human models has been reported in local tissue reduction in ACE2 mRNA and protein activity in glomerular and tubular pathology in primary glomerulopathy, IgA nephropathy, hypertension, nephrosclerosis, and nephrectomy (177, 179, 201206). Conversely, Lely et al. (103) detected ACE2 increases in glomeruli and tubules in primary and secondary renal disease and renal transplant patients. However, only eight patients were observed, and the variation in ACE2 expression markers among tissue samples from diabetic nephropathy patients was not considered.

The pathophysiological process of renal disease damages glomerular and tubulointerstitial tissue, causing ACE2 loss locally. However, diabetic nephropathy provides mixed observation within studies. Most experimental and clinical studies find significant decreases in glomerular ACE2 and increases in tubular ACE2 (207210), although an early animal study showed that ACE2 protein levels decreased ∼30% in glomerular tissue in diabetic kidneys (211). Diabetic nephropathy may progress slower than other renal diseases, partly due to disease process and early aggressive treatments. Thus the damage is seen primarily at the glomerulus. As diabetic nephropathy becomes more severe, however, it may also damage the tubulointerstitium and hence decrease ACE2 in tubular tissue (203, 204).

Although local renal ACE2 production was decreased, plasma ACE2 was significantly elevated, many studies show (15, 92, 183, 184, 187, 212217). Experiments demonstrated a twofold increase in circulating ACE2 in diabetic nephrotic mice (15). Clinical studies also support elevated circulating ACE2 in diabetic and renal disease patients. Roberts et al. (183) found increased plasma ACE2 in CKD patients. Increased levels were further supported by predialysis CKD stages of three to five patients (n = 1,456), compared with patients on dialysis (n = 546) (212, 213). Furthermore, a study of 859 type 1 diabetes patients found serum ACE2 increases in diabetic individuals with micro- and macro-albuminuria, which negatively correlated with glomerular filtration rates (184). As mentioned, shedding and systemic compensatory production may elevate serum ACE2 during development of renal disease.

In sum, major evidence correlates renal disease, reduced local ACE2, and elevated plasma ACE2.

Endocrinological Disorders

Type II diabetes mellitus (T2D) and/or obesity (body mass index ≥ 30) patients are evidently at high risk of COVID-19 susceptibility and severity (1, 4, 218222), while evidence for risk in type I diabetes mellitus (T1D) is limited (1, 4, 218, 220). However, recent data in England’s death registry showed that T1D had 3.5 times the risk of COVID-19 in-hospital death versus two times in T2D (218, 223). Studies had strong evidence for the relationship of T2D and severe COVID-19 [OR: 2.75(2.09–3.62)] and mortality [OR: 1.90(1.37–2.64)] (224). Complications include new-onset diabetes, hyperglycemia, preexisting diabetes, euglycemic ketosis, hyperosmolarity, and classic diabetic ketoacidosis (225). These manifestations pose challenges in clinical management with glucose-lowering medications, due to ineffective results in diabetic COVID-19 patients (219).

Many blood sugar controlling organs are rich in ACE2 (105). Aside from diabetic nephropathy modulating ACE2 expression as mentioned, pancreatic islet cells reportedly have elevated enzyme expression and have been postulated as a target for SARS-CoV-2 infection (226228). Thus pancreatic viral toxicity potentially explains for new-onset diabetes or worsening metabolic control in patients with diabetes (225). Bindom et al. (229) found that islet ACE2 expression increases early in the disease course and decreases with disease progression in T2D mice. A similar trend is noted with β-cells as with renal ACE2 production in renal disease: damage of local tissue from the disease seemingly prevents local ACE2 production, although systemic ACE2 is upregulated.

Based on one of the largest genome-wide studies on T2D to date (n = 898,130), T2D was causally linked to raised ACE2 expression (P = 2.91E-03; Mendelian randomization-inverse‐variance weighted) (230). IL-6 and INS genes were also associated with diabetic patients in 700 lung transcriptome samples (131). INS gene encodes the insulin hormone, and insulin is associated with the NAD-dependent histone deacetylase sirtuin-1, which reportedly regulates ACE2 (142). Animal models in diabetic (STZ induction) mice observed elevated serum ACE2 levels (15, 187). Although evidence for clinical models is limited for circulating ACE2 levels in diabetic patients, Soro-Paavonen et al. (184) noted that ACE2 activity was increased in men with T1D and microalbuminuria, compared with patients without albuminuria or controls: 30.2 ± 1.5 versus 27.0 ± 0.5 versus 25.6 ± 0.8 ngE/ml, respectively; P < 0.05. Furthermore, a preprint of a single center population-based study of 5,457 Icelanders found significant associations of elevated serum ACE2 levels in smokers and obese or diabetic individuals (231). Elevated ACE2 is likely caused from shedding, as seen in increased urinary ACE2 and ADAM17 in 40 T2D patients (232), as well as possible compensatory ACE2 mechanism in vascular and renal function regulation.

One study determined the pathogenesis of increased glucose to directly increase viral load, ACE2, and IL-1β expression in SARS-CoV-2-infected monocytes in a dose-dependent manner. Subsequent treatment with glycolysis inhibitors completely inhibited viral replication in infected monocytes and decreased ACE2 and IL-1B expression (233). In addition, HIF-1 was a strong inducer in glycolysis, also involved in inflammatory response and endothelial dysfunction, suggesting elevated glucose as a catalyst for severe COVID-19 (233).

While pathophysiology in increased risk of worse outcomes in diabetics is most likely multifactorial, obesity has strong associations to T2D and increased risk to COVID-19 susceptibility and severity (1, 42, 221, 222, 234). Of 257 critically ill patients hospitalized in New York City, 36% were diabetic, 46% obese (40). Pooled analysis from 75 studies found that obese individuals were more at risk for COVID-19, 46.0% [OR: 1.46(1.30–1.65)]; for hospitalization, 113% [OR: 2.13(1.74–2.60)]; for ICU admission, 74% [OR: 1.74(1.46–2.08)]; and for mortality, 48% increase in deaths [OR: 1.48(1.22–1.80)] (222).

ACE2 expression was found to be higher in human subcutaneous adipose tissue and human visceral adipose tissue than in human lung tissue, suggesting that adipose tissue may be more vulnerable to COVID-19 (107). Strong evidence in mice models observed increased ACE2 mRNA expression, protein, and circulating ACE2 levels (235239). Moreover, augmented cardiac ACE2 was detected in lean and obese mice (240). No recent published clinical studies were examined for ACE2 and obesity; thus they need to be for future evidence.

Possible physiology includes viral toxicity on adipocytes. The prior SARS‐CoV was found to use cholesterol to facilitate viral budding following S-protein binding of cellular ACE2 receptors. Depletion of cholesterol in ACE2-expressing cells resulted in markedly reduced viral S-protein binding (241). Viral organotropism may explain elevated aspartate aminotransferase concentrations and poorer prognosis in nonalcoholic fatty liver disease and nonalcoholic steatohepatitis in COVID-19 patients (242). After viral binding, deactivation of ACE2 suggests increased macrophage polarization to proinflammatory response such as TNFα, IL-6, IL-8, leptin, and adiponectin, as seen in epicardial adipose tissue, suggesting aggravations to cytokine storm (32, 242244).

In sum, elevated glucose levels and increased adipose tissue evidently correlate to increased COVID-19 susceptibility and severity by contributing to both increased viral replication and cytokine production. Limited studies of elevated ACE2 and adipose tissues demand further investigation of correlation between obesity and ACE2 levels.

Other Manifestations

Other comorbidities, e.g., gastrointestinal diseases, cerebrovascular disease, cancer, may affect COVID-19 infection. Although gastrointestinal diseases, e.g., inflammatory bowel disease, irritable bowel syndrome, peptic ulcer disease, gastroparesis, have not been readily studied as risk factors for COVID-19, more than 20% of COVID-19 patients suffer from gastrointestinal symptoms (111). A U.S. multicenter study found that almost two-thirds of patients hospitalized with COVID-19 presented at least one gastrointestinal symptom (anorexia, 34.8%; diarrhea, 33.7%; nausea, 26.4%; vomiting, 15.4%) (245). Studies suggested fecal-oral transmission, as reported in a meta-analysis detecting viral mRNA in feces in 40.5% (27.4–55.1) of patients (246). Pathophysiology of gastrointestinal infiltration in COVID-19, presumably multifactorial, direct viral mediated ACE2 damage, may contribute to gut dysbiosis. More studies are needed to clarify the relationship of SARS-CoV-2 to gastrointestinal pathophysiology.

Mixed evidence exists for patients with cerebrovascular disease to have an increased risk in severity and mortality, with limited evidence of COVID-19 susceptibility (197, 247, 248). Cerebrovascular disease was associated with increased mortality with borderline significance 2.04(1.43,2.91) – 2.16(0.97,4.80) (197, 248). ACE2 expression has been found in the neurovascular system, suggesting that viral aggravation causes nonspecific neurological symptoms in up to 40% of patients, including severer presentations such as acute stroke and acute necrotizing encephalopathy (32). In combination with direct viral toxicity, ACE2 also contributes to endothelial dysfunction and inflammatory responses, causing meningoencephalitis and acute stroke of varying arterial and venous mechanisms. Limited evidence supports a relevant role for ACE2 in several neuropsychiatry conditions, cerebrovascular ischemic and hemorrhagic lesions, and neurodegenerative diseases (249). A mouse model indicated that cerebral ischemic lesions increased regional cerebral and circulating ANG-1–7 at 12 h, compared with control (7.276 ± 0.320 vs. 2.466 ± 0.410 ng/mg, serum; 1.024 ± 0.056 vs. 0.499 ± 0.032, brain; P < 0.05) (250). In addition, ACE2 expression increased in the cortex penumbra in pathological reports of rats after ischemic injuries and smoking (251).

Further investigations are needed for ACE2’s role in gastrointestinal diseases, cerebrovascular diseases, and other comorbidities, e.g., cancer, sickle-cell disease, solid organ transplantation, that present consistent evidence of increased COVID-19 susceptibility and severity (1).

SECONDARY RISK FACTORS

Data from the WHO, CDC, and other health organizations uncover a bias in infections toward elderly males. Trends also indicate that COVID-19 has affected certain races, ethnicities, and countries more than others, suggesting that genetics may contribute to infection.

Age

Much is debated regarding increased susceptibility and age. In the early phase of the outbreak, COVID-19 appeared to occur in older people in most world regions (242, 252). A meta-analysis of 32 studies suggests that susceptibility in children/adolescents is half that of adults (253). However, other data indicate that young adults and children are as susceptible to the disease as older adults (254, 255), possibly for social reasons, making optimal strategy for reopening schools and universities difficult. In fact, significantly greater amounts of viral nucleic acid were detected in children (<5 yr; n = 46) than in adults (18–65 yr; n = 48) (256). While the infection prevalence’s relation to age is unclear, increased risk for severe illness evidently increases with age, making elders a high-risk population (4, 5, 257259).

The CDC reports that 8 of 10 COVID-19 deaths in the U.S. are over age 65 (260). Case fatality rates by age group were observed in Italy and China: 96.5% of total deaths in Italy and 81% of those in China were past age 60 (258); 0.3% of total deaths in Italy and 2.6% of those in China were below age 39. A study of critically ill patients in China found that, compared with survivors, nonsurvivors were older (64.6 ± 11.2 yr vs. 51.9 ± 12.9 yr) and had comorbidities (259). Moreover, in-patient mortality rates in New York City were higher in those older than 65 versus those aged 18–65 (4). Although data suggest that the elderly develop more complications and young adults will less likely become symptomatic (261), a case series highlights the presence of genetic variants in young men with severe COVID-19. Rare putative loss of function TLR7 variants were associated with impaired IFN responses (262).

In earlier reports, ACE2 receptor availability was thought to decrease in the elderly in most tissues, increasing their risk of severe illness. Since ACE2 is a key regulator for inflammatory and immune responses, ACE2 reduction could result from a weakened immune system, increasing COVID-19 infection vulnerability. Experimental models demonstrate significantly lower ACE2 levels in older 24-mo-old male and female mice and aging endothelial cells, suggesting that ACE2 receptors in older adults could be decreased in the respiratory tract, possibly weakening the immune system (263265), although other animal models depict no significant difference in ACE2 expression in cerebrovascular tissue (266, 267). More recently, RNA-Seq gene profiling in 30 tissues across thousands of individuals found ACE2 expression to significantly decrease with age in Caucasian males, although data remained nonsignificant in other ethnic groups (268). However, other analyses of gene expression profiling repository data found no substantial differences in ACE2 and TMPRSS2 with ages >60 yr versus <60 yr and ≤49 yr versus >49 yr (269, 270). Moreover, a clinical study found no statistical differences in activity of ACE/ACE2 among four age groups ranging from neonates to seniors (271), although the study had limited samples of 17–29 individuals in each group. Recent studies suggest otherwise.

Mouse models revealed that elderly mice had higher ACE2 and TMPRSS2 expression in nasal mucosa than younger ones (272, 273). Similarly, clinical models found significant linear trends in nasal epithelial ACE2 gene expression with advancing age groups and olfactory and gustatory deficits in a cohort of 305 individuals aged 4–60, indicating that children could have milder COVID-19 symptoms (274, 275). Though limited in age, nasopharyngeal, oropharyngeal, and/or blood specimens were collected in 192 children aged 0–22. The study revealed that age did not impact viral load, but older children (>10 yr) had significantly higher ACE2 expression than younger ones. Furthermore, a positively weak correlation (r = 0.20, P = 0.02) between ACE2 expression and age suggests that future studies must clarify a plausible linear relationship (276).

Likewise, Saheb Sharif-Askari et al. (133) observed significantly elevated ACE2 and TMPRSS2 in adults compared with children in nasal and bronchial tissue in multiple transcriptomic datasets. Increased expression of ACE2 and TMPRSS2 in upper and lower airways of adults may contribute to increased severity of infection in adults. Children may hypothetically be protected from severe COVID-19 due to cross-protective antibodies and/or decreased ACE2 receptors to SARS-CoV-2 in the lower airway (277), possibly because their lungs do not fully mature until age 20–25. Moreover, novel findings of ACE2 expression in gastrointestinal tissue moderately correlate with age. Findings have evaluated ACE2 mRNA in duodenal and ileal biopsies, which correlated with age (r = 0.32, P = 0.0099; r = 0.64, P = 0.0099, respectively) (278, 279). The different ACE2-associated amino acid transporter (B0AT1, SIT1) expression at the brush borders in older patients may impact susceptibility to intestinal symptoms and/or increased disease severity (279).

In addition, age differences in serum ACE2 may provide insights for COVID-19. Studies of 118 healthy individuals aged 41–70 and 213 patients with newly diagnosed mild-to-moderate hypertension determined a positive association between age and ACE2 serum activity (280, 281). Moreover, a large cohort of participants (n = 2,051) in a commercial wellness program found significantly higher plasma ACE2 levels in older individuals; age association was more pronounced in women pre- and postmenopausal (282). A recent longitudinal study observed low serum ACE2 in males and females up to age 12 and significantly increased serum in adolescents and young adults, implying that androgen sensitivity in puberty may exacerbate ACE2 production (283).

Although increased circulating ACE2 enzyme offers protection against influenza A (H7N9) virus-induced ALI (284), the apparent paradox for older individuals to have elevated soluble ACE2 levels potentially heightens COVID-19 susceptibility and/or severity in older adults. Although concerns present for direct viral organ-toxicity and ACE2 dysregulation, age-associated frailty involves increased baseline inflammation, called “inflammaging,” which can cause exuberant inflammatory responses in older individuals with high baseline IL-6 and IL-8 (285). For example, the preprint of Baker et al. (286) detected increased ACE2 expression with age in the setting of alveolar damage observed in patients on mechanical ventilation, providing a potential mechanism of exacerbated inflammatory responses for increased COVID-19 mortality in the elderly.

Sex

Despite similar sex distribution of individuals infected with SARS-CoV-2 (male 51%, female 49%), a sex difference is notable in COVID-19 fatality rates: males could be at higher risk than females for contracting severe COVID-19 (6). China’s CDC confirmed increased case fatality rates in males compared with females (2.8% vs. 1.7%, respectively), revealing that 64% of deaths in China have been male (287). Italy had similar trends: male mortality is apparently twice that of females in every age group. Italy’s Public Health Research Agency noted that 59.8% of SARS-CoV-2 cases and 70% of national deaths so far have been male (288). New York City had increased hospitalized males more than females (60.3% vs. 39.7%, respectively; n = 5,700) (4). Moreover, Open SAFELY, an analytics platform covering over 17 million records in England, revealed that males have over one-half the mortality risk of females, HR: 1.59(1.53–1.65) (145). Reduced SARS-CoV-2 infection in females could be attributed to their increased protection from viral infections by an additional X chromosome and varied sex hormones (289). The ACE2 gene, which lies on Xp22.2, is affected by sex hormonal regulations.

Mice models show ACE2 activity to be higher in males than females in specific tissue and serum. Studies mainly revealed elevated local ACE2 activity in male kidneys, compared with female ones (290293). Conversely, Sampson et al. (294) noted increased ACE2 receptor expression within female kidneys. Likewise, serum ACE2 levels were elevated in male normotensive mice (293, 295). However, levels were contrary in hypertensive mice and comparable to those of hypertensive patients (187, 293, 296), suggesting comorbidity-dependent hormonal modulation on ACE2 gene activity.

Although animal models provided increased evidence supporting elevated ACE2 activity in renal tissues, human studies indicated mixed results in large transcriptome public databases. Bulk RNA-seq profiles determined increased ACE2 expression in male lung tissue, largely in type II pneumocytes, and male plasma (297299). Other transcriptome studies revealed comparable ACE2 and TMPRSS2 concentrations in multiple tissue (270, 300), while Chen et al. (268) revealed Asian females to have higher ACE2 expression than males out of three studied ethnic groups, suggesting ACE2 regulation through genetic differences. Limited independent human tissue studies and recent plasma studies are nonetheless evident for males exhibiting significantly higher ACE2 expression levels. Male bronchial biopsy and bronchoalveolar lavage generally led to increased expression of ACE2 and ACE2-related genes in smokers and nonsmokers (132, 278). ACE2 concentrations were reported higher in male smokers than in male nonsmokers, implicating gender-specific behavioral differences (299). Furthermore, ACE2 expression in healthy ileal biopsies (n = 154) were found to be 130% greater in men than in women (P = 0.0256) (278).

Recent serum studies indicate strong evidence of increased circulating ACE2 levels in healthy males and males with comorbidities (132, 164, 217, 280, 282, 283, 301). Studies are revealing comorbidities, e.g., endocrine manifestations, hypertension, heart failure, end-stage renal disease, exacerbating increased serum ACE2 in men (164, 217, 282, 301). Sama et al. (164) observed male sex as the strongest predictor of elevated concentrations of ACE2 in control and heart failure cohorts (coefficient = 0.19 and 0.26, respectively; P < 0.001), and Stienen et al. (301) detected lower ACE2 levels in females with preserved ejection fraction heart failure. These effects suggest that gender-specific factors and behaviors may potentiate ACE2 receptors and thus demand investigation.

In contrast, no sex differences were detected for ACE2 in 118 healthy men and women, possibly because ACE2 activity in females was affected by postmenopausal period (281). Kornilov et al. (282) confirmed higher levels of plasma ACE2 in postmenopausal women than in premenopausal women (P = 0.02). A longitudinal study found similar low-serum ACE2 in both sexes until age 12, where ACE2 increased more in boys than in girls, emphasizing possible sex hormonal regulation (283). In fact, mechanisms for sex hormonal regulation of ACE2 and TMPRSS2 remain controversial with limited data.

ACE2 expression may be upregulated in females by estrogen [i.e., 17β‐estradiol (E2)], X chromosome inactivation, or reduced ACE2 methylation, providing increased ACE2 levels to maintain RAS equilibrium (302). Experimental models have observed estrogen upregulating ACE2, AT2R, and MAS expression levels through effects at estrogen receptor-mediated binding at the ACE2 promoter (303, 304). Yet, low androgen levels and elevated E2 levels in females may suppress ACE2 expression, providing a protective factor against COVID-19 (302). The only known stimuli of TMPRSS2 gene transcription are androgens, and hospitalized COVID-19 patients exhibited androgenic alopecia (305). E2-treated NHBE cells expressed lower levels of ACE2 mRNA (306), while ovariectomy (loss of E2) increased ACE2 activity, and orchiectomy (loss of androgens) decreased enzyme activity (292, 307, 308). Further studies correlating sex hormones to ACE2 are needed.

Other factors, e.g., population composition, sex-related comorbidities, immunological responses, must be considered. More males than females appear affected in China and Italy, as mentioned, though males comprise a larger percentage of the Chinese but not Italian population (105.6 males per 100 females, 93 males per 100 females, respectively) (309). Therefore, population composition cannot solely explain the sex discrepancy. Another confounding variable: males are more likely than females to have comorbidities and gender-specific behavior that increase ACE2 levels, e.g., cardiovascular disease, hypertension, obesity, and smoking. For example, obesity was found to be a major risk factor for COVID-19 mortality in men but not women (310). Smoking habits are also disproportionately increased in males more than females, as seen in China (288 million and 12.6 million, respectively) and globally (40% and 9% tobacco use, respectively) (311, 312). Additionally, males with severe COVID-19 reportedly have higher CRP concentration than females, independent of comorbidities and age (313). Males have elevated proinflammatory cytokine responses, but females apparently have robust T-cell activation (305, 314, 315). Elevated cytokine response and poor T-cell response correlate with worse disease outcomes (305, 315). Therefore, sex differences may be less drastic than initially noted, due to the effects of confounding variables with ACE2 levels.

Although large transcriptome studies are inconsistent across multiple tissues, animal and human models favor elevated ACE2 in males, compared with females. We believe that moderate evidence of increased ACE2 expression relates to sex differences. However, studies directly comparing ACE2 and TMPRSS2 expression by sex and COVID-19 outcomes/severity are needed.

Race, Ethnicity, and Genetics

Current epidemiological data strongly indicate variability of case-fatality rates from as high as 15.1% in U.K., 14.2% in Italy, 3.3% in the U.S., and 2.1% in South Korea (3, 316). Furthermore, increasing evidence suggests that COVID-19 disproportionately affects some racial and ethnic minority groups (317321). The OpenSAFELY extensive analytic platform indicated HR: 1.62–1.88 (adjusted for age and sex) for Black, South Asian, and mixed ethnicity, compared with white patients; and HR: 1.43–1.48 after adjustment of all included factors (145). Moreover, race/ethnicity data from the Morbidity and Mortality Weekly Report revealed that 33% of hospitalizations were for Blacks, two times the U.S. Black population. Conversely, 45% of those hospitalized were white, less than half of the white population (119). Likewise, the Johns Hopkins University and American Community Survey reported that infection and death rates were more than three times and six times higher, respectively, in Black counties than in white ones in the U.S (322).

Unfortunately, scant data examine the biological mechanisms underlying ethnic differences in COVID-19 susceptibility and severity. Preliminary data of nasal epithelial gene expression identified significantly higher nasal gene expression of TMPRSS2 in Black than in other races and ethnicities, suggesting that ACE2 variations among ethnic groups may partially contribute to the higher burden of the disease among Blacks (274). However, the study fails to present demographics, comorbidities, and smoking history of the 305 participants, which may affect ACE2 expression.

We may question whether genetic variations in ACE2 and TMPRSS2 among racial and ethnic groups exist. Fujikura and Vesaka (323) detected 349 and 551 single nucleotide variations in human ACE2 and TMPRSS2, respectively, in 156,513 individuals. The single nucleotide variations were rare but population specific and deleterious, suggesting susceptibility of different populations to SARS-CoV-2. Relevant ACE2 polymorphisms under review include rs2285666, rs1978124, and rs714205 (324). Also, deleterious variants in ACE2 differed among nine populations in gnomAD, specifically Black and non-Finnish European ones: 39% versus 54%, respectively (325).

Moreover, a recent study noted significantly higher ACE2 expression among Asians than Blacks and whites (326). Analysis of GTEx and other public data examined higher allele frequencies in expression quantitative locis (eQTLs) associated with elevated ACE2 expression in Eastern Asian population tissues; eQTLs were calculated close to 100% in Eastern Asians and >30% higher than other ethnic groups (268, 327). Conversely, Hou et al. (325) found no eQTLs for ACE2 across different populations, but polymorphisms (i.e., p.Val160Met [rs12329760]) were found in TMPRSS2. Furthermore, studies found that Asians and other races express similar levels of genetic polymorphisms of the SARS-CoV-2 entry receptor (328, 329), and transcriptomic datasets of lung tissue revealed no significant ACE2 gene expression disparities between Asians and whites (330). Therefore, genetics consortia report no direct association between SNVs and eQTLs in ACE2 and TMPRSS2.

Other haplotypes comprising ACE1, ABO-locus, 9q34.2, and 3p21.31 are of interest (324, 331). The ACE1 II genotype had a strong negative correlation with SARS-CoV-2 cases and deaths (332). The study noted that the European population had lower ACE1 II genotype frequency and higher prevalence of mortality than the Asian population (332). This may factor into Central Europe experiencing far more COVID-19 cases and deaths than East Asia.

ABO blood groups and COVID-19 incidence and mortality are being studied as well (333). In a meta-analysis of 318 studies, blood group A has a significant partial risk factor [OR: 1.16(1.02–1.33)] for COVID-19 infection, whereas AB has an insignificant but considerable one [OR: 1.25(0.84–1.86)] (334). Blood group O was considered as a possible protective factor [OR: 0.73(0.60–0.88)] against infection (334). Additionally, ABO blood groups versus severe COVID-19 extrapolated similar sentiments with blood group A and blood group O: OR: 1.45(1.2–1.75), OR: 0.65(0.53–0.79), respectively. O blood type carriers may have lower ACE levels and higher regulated IL-6 levels, suggesting increased balance in the ACE/ANG II axis (331). Guillon et al. (335) also observed that anti-A antibodies blocked S-protein/ACE2-dependent adhesion, indicating that anti-A antibodies may block the interaction between the virus and its receptor. In addition, locus 9q34.2 coincides with the ABO blood group locus with an increased OR: 1.32(1.20–1.47) with severe COVID-19.

Another genomic region of interest relates to several genes on chromosome 3 (SLC6A20, LZTFL1, CCR9, FYCO1, CXCR6 and XCR1) (333); 3p21.31 presented an increased OR: 1.77(1.48–2.11) with severe COVID-19. One gene in particular, SLC6A20, encodes for an amino acid transporter that interacts with ACE2, which may alter the binding affinity to the virus. Other genes in cluster affect immunological responses related to chemokine receptors, C-X-C motif chemokine receptor 6, and CC-motif chemokine receptor 9, in response to T-cell differentiation and recruitment.

Researchers have also hypothesized possible genetic predisposition to ACE2 polymorphisms linked to diabetes, stroke, and hypertension (170). A meta-analysis (n = 11,051) provided strong evidence that ACE2 gene polymorphism G8790A had an increased risk factor for essential hypertension across different ethnic populations in female subjects and Han-Chinese male subjects (336). Genetic predispositions associated with increased comorbidities may factor into higher prevalence of heart disease, hypertension, diabetes, and obesity in minority groups, leading to increased COVID-19 susceptibility and severity. Although current but limited studies indicate possible genetic variability in COVID-19 contraction and/or severity, much is debated. Further research is needed to understand the molecular and pathophysiological mechanisms underlying the relationship among genetics, race/ethnic disparities, and COVID-19 infection and severity.

While our review emphasizes physiologic factors, we acknowledge that COVID-19 susceptibility and severity largely result from socioeconomic inequities, which need further study. Inequities in social determinants of health affecting minority groups, e.g., racism, barriers to healthcare access and use, higher representation in occupations with increased chance of exposure, education, income, wealth gaps, crowded housing conditions, are interrelated and influence disease outcome and mortality. These are only a few factors associated with increased rates of COVID-19 cases, hospitalizations, and deaths within minority racial and ethnic communities. Behavioral and environmental factors also increase rates of certain medical conditions, such as CKD, COPD, and T2D, which affect COVID-19 incidence and severity. Easier access to information, affordable testing, and medical care are needed to mitigate inequity and empower individuals. Further research is needed to loosen systemic barriers for appropriate and critical care.

TERTIARY RISK FACTORS

The thought of certain medications increasing risk of SARS-CoV-2 infection remains controversial. ACE inhibitors (ACEi) and angiotensin receptor blockers (ARBs) are particularly criticized for possibly aggravating lung injury in infected patients due to increased ACE2 expression in the body (337). Other concerns are directed toward diabetic medications and ibuprofen (170).

Angiotensin-Converting Enzyme Inhibitors/Angiotensin Receptor Blockers

Many COVID-19 patients have cardiovascular and renal comorbidities that usually require these medications. Both medications were believed to increase ACE2 expression, potentially increasing susceptibility/severity to COVID-19.

ACEi inhibit ACE and ARBs block RAS’s effects, causing angiotensin I accumulation; ACE2 then metabolizes it into angiotensin (33, 92, 107, 112, 118, 212,213, 285, 288) (Fig. 1). ACEi do not inhibit ACE2, because ACE2’s S2′ pocket in the active site is smaller than ACE’s corresponding pocket (338). Most studies were reported in animal models (Table 1), while few were conducted in human tissue. One animal study showed that ACEi increased ACE2 mRNA 1.8-fold and that losartan showed a significant ACE2 mRNA increase (338). Other studies reported similar trends in ACE2 mRNA for enalapril, olmesartan, and valsartan (339, 341343). Interestingly, ARBs have shown to improve lung recovery in ACE2-downregulated mice infected by the previous SARS-CoV (52). Others have also suggested that ACE2’s compensatory elevation may cause the benefit of ACEi/ARBs in non-COVID-19 viral pneumonia. However, this elevation may not be beneficial in a COVID-19 context, as SARS-CoV-2 uses ACE2 as a vector for infection (337, 345). Thus patients treated with ACEi/ARBs could be at higher risk of severe COVID-19 due to increased S-protein coronavirus binding sites in their lungs (277).

Table 1. Evidence of the relationship between ACEi/ARBs and ACE2 levels in animal models

Study ACEi/ARB Tissue ACE2 mRNA ACE2 Serum/Protein
Angiotensin-converting enzyme inhibitor
 Ocaranza et al. (339) Enalapril Heart
 Ferrario et al. (338) Lisinopril Heart
 Tikellis et al. (10) Perindopril Kidney
 Lezama-Martinez et al. (340) Captopril Aorta N/A
 Hamming et al. (102) Lisinopril Kidney
 Ferrario et al. (338) Lisinopril Kidney
 Burrell et al. (157) Ramipril Heart
Angiotensin receptor blocker
 Ishiyama et al. (341) Losartan Heart N/A
 Ishiyama et al. (341) Olmesartan Heart N/A
 Whaley-Connell et al. (342) Valsartan Kidney N/A
 Takeda et al. (343) Candesartan Heart
 Ferrario et al. (338) Losartan Heart
 Kuba et al. (52) Losartan Lung N/A
 Ferrario et al. (338) Losartan Kidney
 Lezama-Martinez et al. (340) Losartan Aorta N/A

Summary of the evidence of the relationship between ACEi/ARBs and ACE2 levels. More comprehensive table reviewed in Kreutz et al. (344; Table 1): effect of renin-angiotensin system blockers on ACE2. ACEi: angiotensin-converting enzyme inhibitor; ARB: angiotensin receptor blocker; ACE2: angiotensin-converting enzyme 2.

Although evidence supports increases in ACE2 by ACEi/ARBs, it remains controversial. Perindopril has shown to reduce plasma and cortical ACE2 activity in both healthy and diabetic mice (10). Similarly, captopril, lisinopril, and losartan decrease ACE2 mRNA in mice (102, 340). In addition, Burrell et al. (157) indicated that ramipril does not affect cardiac ACE2 mRNA and proteins.

In sum, a review of the available literature revealed the use of ARBs to potentially elevate ACE2 mRNA and proteins in experimental models, while ACEi has inconsistent changes (344). However, the Human Lung Tissue Expression Quantitative Trait Loci Study, which analyzed the gene expression of ACE2, TMPRSS2, and ADAM17, observed no alterations in gene expression by ARBs but a reduction in ACE2 and TMPRSS2 expressions by ACEi (346). The study suggests that long-term ACEi use may downregulate lung ACE2 expression by reducing substrate (e.g., ANG II) availability, thereby reducing risk of SARS-CoV-2 infection. The significance of these findings is unclear. Clinical studies are needed to elucidate the relationship of ACEi/ARBs to ACE2.

Ending ACEi/ARB use in COVID-19 patients has been debated. However, observational-cohort studies denoted no statistical significance in samples of 173 and 634 patients on ACEi/ARBs, β-blockers or other anti-hypertensives to critical COVID-19’s severity and outcomes (347, 348). Population-based case-control studies in Italy (n = 6,272) and Spain (n = 1,139) reached similar conclusions (349, 350). A systematic review confirmed scant evidence for the use of ACEi/ARBs, prophylactically or therapeutically, against COVID-19 (351). In fact, the Council on Hypertension urges patients to “continue treatment with their usual antihypertensive therapy because there is no clinical or scientific evidence to suggest that treatment with ACEi/ARBs should be discontinued” (352). Also, medication changes would require patients to visit a pharmacy or obtain bloodwork, which can increase COVID-19 exposure. Since ACEi/ARBs are widely prescribed for cardiovascular and renal disease, more attention should be paid to COVID-19 patients and close contacts using ACEi/ARBs (353). β-Blockers also inhibit renin production, but their relation to ACE2 regulation and COVID-19 is unknown. Also, no evidence shows that calcium channel blockers increase ACE2 activity, so these could be alternative treatments for these patients (170). Additional pharmacological studies of the relation of ACE2 levels and COVID-19 are needed.

Thiazolidinediones and Ibuprofen

Other common medications, such as thiazolidinediones and ibuprofen, have been brought into question. Thiazolidinediones are popular diabetes treatments. These medications act on peroxisome proliferator-activated receptor-γ, which subsequently acts on RAS. One study found that pioglitazone significantly increases ACE2 protein expression in the liver, adipose tissue, and skeletal muscle, compared with the high-fat diet group (354). Yet, evidence for this is limited.

Furthermore, the French health minister warned patients to use acetaminophen over nonsteroidal anti-inflammatory drugs based on observing serious side effects from ibuprofen. A study of the link between ibuprofen and cardiac fibrosis in diabetic rats revealed an association between ibuprofen and elevated ACE2 (355). However, no strong, consistent evidence shows that ibuprofen increases ACE2. The WHO and European Medicines Agency do not condemn ibuprofen, because no clinical or population-based data on this topic are available (356).

CONCLUSIONS

ACE2 conducts extensive vascular and organ protection in significantly underlying risk factors for COVID-19. Table 2 summarizes the primary, secondary, and tertiary factors and ACE2, as discussed above. Men aged >60 with underlying comorbidities are reportedly more vulnerable to COVID-19 susceptibility and severity. Whether patients’ demographics, therapies, or other factors influence this is unclear, but COVID-19 appears related to ACE2’s role.

Table 2. Evidence of the relationship between ACE2 and primary, secondary, and tertiary risk factors in COVID-19 patients

Risk Factors Susceptibility Severity ACE2 Relationship Experimental Models Human Models
Primary
 Respiratory disease • Strong evidence: COPD, emphysema, smokers• Mixed evidence: asthma • Strong evidence: COPD and emphysema• Mixed evidence: asthma• Limited evidence: smokers • Strong: COPD, emphysema, smoking• Limited: asthma • Increased ACE2: autopsy specimens from severe SARS patients with ALI had elevated ACE2, SARS‐CoV S protein, RNA, and proinflammatory cytokines (126)• Increased ACE2: COPD upregulates ACE2 and TMPRSS2 expression in the nasal, bronchial and lower airways (127134)• Increased ACE2: significant inverse relationship between ACE2 gene expression and FEV1% (r = −[0.24–0.40]; P < 0.05). (127, 129)• Increased ACE2: in moderate/severe COPD lung tissue (128)• Increased ACE2: significant serum ACE2 level increases in 27 COPD patients (135)• Increased ACE2/TMPRSS2: slightly upregulated ACE2 and TMPRSS2 lung airway expression in asthmatics, while plasma ACE2 was significantly upregulated in asthmatics (133)• Increased TMPRSS2: elevated in children and adult asthmatics from available RNA-Seq databases (132)• Increased ACE2: ACE2-correlated gene signatures were found to be significant in a subset of type 2-low patients with asthma with characteristics resembling known risk factors for severe COVID-19 (140)• Increased ACE2/TMPRSS2: increase in ACE2 and TMPRSS2 gene expressions in bronchial and alveolar epithelial, bronchial alveolar lavage, and protein in lung tissue and blood (127134, 144)• Increased ACE2: current smokers had significantly higher gene expression than ex-smokers and nonsmokers (2.77 ± 0.91 vs. 2.00 ± 1.23 vs. 1.78 ± 0.39, respectively; P = 0.024) (128, 129)• No difference in ACE2: no significant difference in ACE2 expression in the lower airways with allergic sensitization and asthma (130, 134, 141)
 Cardiovascular Disease • Strong evidence: heart failure, coronary artery disease, or cardiomyopathies (6, 124, 145, 337, 357) • Strong evidence: heart failure, coronary artery disease, or cardiomyopathies • Strong • Increased ACE2: elevated serum ACE2 myocardial infarction, atherosclerotic development, reduced left ventricular ejection fraction, cardiomyopathies, and heart failure (12, 92)• Increased ACE2: elevated ACE2 mRNA days 3 and 28 postmyocardial infarction (157) • Increased ACE2: ACE2 gene was most upregulated and a fivefold increase in ACE2 protein in hypertrophic cardiomyopathy human cardiac tissue, compared with that of controls (159)• Increased ACE2: plasma ACE2 activity directly related to persistent AF (22.8 pmol/min/mL) and paroxysmal AF (16.9 pmol/min/mL), compared with control (13.3 pmol/min/mL) (161)• Increased ACE2: elevated plasma ACE2 concentrations in 1,485 men and 537 women with heart failure and a threefold increase in myocardial ACE2 gene expression in patients with heart failure (163, 164)Increased ACE2: elevated ACE2 serum in acute heart failure (52.5 pmol/h/ml) and chronic heart failure (33.6 pmol/h/ml), compared with healthy controls (22.5 pmol/h/ml) (165)• Increased ACE2: elevated ACE2 mRNA in explanted failing hearts (157)• Increased ACE2: in patients with heart failure and correlated with disease severity (166, 167)• Increased ACE2: elevated plasma ACE2 in 79 patients with coronary artery disease and correlated with adverse long-term outcomes (162)
Hypertension • Limited evidence: essential and secondary hypertension • Strong evidence: essential and secondary hypertension • Strong: essential hypertension• Limited: secondary hypertension • Increased ACE2: positive plasma ACE2 correlation with increased systolic blood pressure (187)• Decreased ACE2: decreased local ACE2 mRNA and protein activity in tissue, primarily in the kidneys and heart, thus depict hypertensive progression to decrease local ACE2 activity (97, 176180) • Increased ACE2: positive plasma ACE2 correlation with increased systolic blood pressure (167, 181187)• Increased ACE2: plasma ACE2 1.5 times greater in 239 hypertensive patients (167)• Increased ACE2: patients with detectable plasma ACE2 had significantly higher systolic and diastolic blood pressure, compared with those without (182)• Increased ACE2: elevated serum ACE2 concentrations in hypertensive patients, compared with healthy subjects (170.31 [83.50–707.12] pg/ml vs. 59.28 [39.71–81.81] pg/ml, respectively; P < 0.001) (181)• Mixed ACE2 results: noted in the relationship between secondary hypertension and ACE2 (178)• Decreased ACE2: decreased local ACE2 mRNA and protein activity in tissue, primarily in the kidneys and heart, thus depict hypertensive progression to decrease local ACE2 activity (97, 176180)
 Renal Disease • Strong evidence: chronic kidney disease • Strong evidence: chronic kidney disease • Strong • Increased ACE2: (twofold) in diabetic nephrotic mice (15)• Decreased ACE2: local tissue reduction in ACE2 mRNA and protein activity in glomerular and tubular tissue in primary glomerulopathy, IgA nephropathy, hypertension, nephrosclerosis, and nephrectomy (177, 179, 201, 205) • Increased ACE2: elevated plasma ACE2 in CKD stages 3-5 predialysis patients, n = 1,456 (212, 213)• Increased ACE2: local ACE2 increases in glomeruli and tubules in primary and secondary renal disease, as well as renal transplant patients (103)• Increased ACE2: significantly elevated plasma ACE2 (15, 92, 183, 184, 187, 212217)• Increased ACE2: elevated serum ACE2 in type 1 diabetes, n = 859, with micro- and macro-albuminuria; negatively correlated with glomerular filtration rate (184)• Decreased ACE2: local tissue reduction in ACE2 mRNA and protein activity in glomerular and tubular tissue in primary glomerulopathy, IgA nephropathy, hypertension, nephrosclerosis, and nephrectomy (177, 202204, 206)
 Diabetes Mellitus • Strong evidence: type 2 diabetes• Limited evidence: type 1 diabetes • Strong evidence: type 2 diabetes• Limited evidence: type 1 diabetes • Strong • Increased ACE2: twofold increase serum ACE2 in diabetic nephrotic mice (15)• Increased ACE2: islet expression early in the disease and decreased with disease progression in type 2 diabetic mice (229)• Increased ACE2: elevated serum ACE2 observed in diabetic mice (15, 187)• Increased ACE2: increased glucose to directly increase viral load, ACE2, and IL-1β expression in SARS-CoV-2 infected monocytes in a dose-dependent manner.• Decreased ACE2: decreased local ACE2 by ∼30% in diabetic kidneys (211) • Increased ACE2: type 2 diabetes was causally linked to raised ACE2 expression (P = 2.91E-03; MR-IVW) (230)• Increased ACE2: elevated serum ACE2 in Type 1 diabetes, n = 859, with micro- and macro-albuminuria; negatively correlated with glomerular filtration rate (184)• Increased ACE2: elevated expression in the pancreas of healthy subjects (n = 74); slightly higher than in the lungs (227)• Increased ACE2: 5,457 Icelanders found significant associations of elevated serum ACE2 levels in smokers and in obese or diabetic individuals (231)• Increased ACE2: increased urinary ACE2 and ADAM17 in 40 T2D patients (232)
Obesity • Strong evidence: BMI >30 kg/m2 • Strong evidence: BMI >30 kg/m2 • Strong • Increased ACE2: seen in obese, type 2 diabetic mice (108)• Increased ACE2: elevated ACE2 mRNA expression, protein, and circulating ACE2 levels (235239)• Increased ACE2: elevated local ACE2 in augmented cardiomyopathy in lean/obese mice (240) • Increased ACE2: ACE2 expression was found to be higher in human subcutaneous adipose tissue and human visceral adipose tissue than in human lung tissue (107)
Gastrointestinal Disease • Limited evidence: Inflammatory bowel disease, irritable bowel syndrome, peptic ulcer disease, gastroparesis • Limited evidence: Inflammatory bowel disease, irritable bowel syndrome, peptic ulcer disease, gastroparesis • Limited • Increased TMPRSS2: Crohn’s disease ileum was 70% higher (P < 0.05) than in controls. Ulcerative colitis ileum was 30% higher (P < 0.05) than in controls (278)• Increased ACE2: Crohn’s disease colonic was 30% higher (P < 0.05) than in controls. Ulcerative colitis colonic was 70% higher (P < 0.05) than in controls (278)• No difference ACE2: ulcerative colitis ileum did not differ compared with controls (278)• No difference TMPRSS2: Crohn’s disease colonic did not differ compared with controls (278)• Decreased ACE2: Crohn’s disease ileum was 60% lower (P < 0.05) than in controls (278)
Cerebrovascular Disease • Limited evidence • Mixed evidence • Limited • Increased ACE2: in mice with cerebral ischemic lesions, resulting in a significant increase in regional cerebral and circulating ANG-1-7 at 12 h, compared with control (7.276 ± 0.320 vs. 2.466 ± 0.410 ng/mg, serum; 1.024 ± 0.056 vs. 0.499 ± 0.032, brain; P < 0.05) (250)• Increased ACE2: elevated ACE2 expression in the cortex penumbra in pathological reports of rats after ischemic injuries and smoking (251) • Limited ACE2 evidence: in several neuropsychiatry conditions, cerebrovascular ischemic and hemorrhagic lesions, and neurodegenerative diseases (249)
Secondary
Age • Mixed evidence: Prevalence of infection is related to age • Strong evidence: Increased risk for severe illness increases with age, making elders a high-risk population (4, 5, 257259) • Moderate • Increased ACE2/TMPRSS2: elderly mice had higher expression of ACE2 and TMPRSS2 in nasal mucosa, compared with younger mice (272, 273)• No difference ACE2: no significant difference in ACE2 expression in cerebrovascular tissue and age in older mice (266, 267)• Decreased ACE2: significantly lower ACE2 levels in older 24-mo-old male and female mice and aging endothelial cells (263265) • Positive ACE2 correlation: with age, n = 118, 41-70 yr (281)• Positive ACE2 gene expression correlation: With age groups; n = 305, 4-60 yr (274)• Increased ACE2: Significant linear trends were found in nasal epithelial ACE2 gene expression with advancing age groups in a cohort of 305 individuals aged 4-60 (274, 275)• Increased ACE2: positively weak correlation (r = 0.20, P = 0.02) between ACE2 expression and age (276)• Increased ACE2: significantly elevated ACE2 and TMPRSS2 in adults, compared with children in nasal and bronchial tissue in multiple transcriptomic datasets (133)• Increased ACE2: ACE mRNA in duodenal and ileal biopsies moderately correlated with age (r = 0.32, P = 0.0099; r = 0.64, P = 0.0099, respectively) (278, 279)• Increased ACE2: positive association between age and ACE2 serum activity in 118 healthy and 213 mild-moderate hypertensive patients (280, 281)• Increased ACE2: large cohort of participants (n = 2,051) in a commercial wellness program found significantly higher plasma ACE2 levels in older individuals; age association was more pronounced in women pre- and postmenopausal (282)• Increased ACE2: significantly increased serum in adolescents/young adulthood (283)• No difference in ACE2: between four age groups, neonates to >65 yr (271)• No difference in ACE2/TMPRSS2: no significant differences in ACE2 and TMPRSS2 gene expression with ages >60 yr vs. <60 yr and ages ≤49 yr vs. ages >49 yr (269, 270)• Decreased ACE2: ACE2 expression to significantly decrease with age in Caucasian males, although data remained nonsignificant in other ethnic groups (268)
Sex • Limited evidence • Strong evidence: Males at higher risk than females for contracting severe COVID-19 • Moderate • Increased ACE2: elevated local ACE2 in mice male kidneys (290293)• Increased ACE2: serum ACE2 levels were found elevated in male normotensive mice (293, 295)• Decreased local ACE2: in mice male kidneys (294)• Estrogen upregulates: ACE2, AT2R and MAS expression levels through effects at estrogen receptor-mediated binding at the ACE2 promoter (303, 304)• Estrogen downregulates: E2-treated NHBE cells expressed lower levels of ACE2 mRNA (306), while ovariectomy (loss of E2) increased ACE2 activity, and orchiectomy (loss of androgens) decreased enzyme activity (292, 307, 308) • Increased ACE2: increased ACE2 expression in male lung tissue, largely in type II pneumocytes, and male plasma (297299)• Increased ACE2: ACE2 expression in healthy ileal biopsies (n = 154) were found to be 130% greater in men than in women (P = 0.0256) (278)• Increased ACE2: increased circulating ACE2 levels in both healthy males or males with comorbidities (132, 164, 217, 280, 282, 283, 301)• Increased ACE2: male sex as the strongest predictor of elevated concentrations of ACE2 in control and heart failure cohorts (coefficient = 0.19 and 0.26, respectively; P < 0.001) (164)• Increased ACE2: increased ACE2 levels in males with preserved ejection fraction heart failure (164, 301)• Increased ACE2: ACE2 increased more in boys than in girls, emphasizing possible sex hormonal regulation (283)• No difference in ACE2: no sex differences for ACE2 in 118 healthy men and women (281)• No difference in ACE2: gene expression in gender in transcriptome databases (268, 270, 300, 328, 330)• Decreased ACE2: Asian males to have lower ACE2 expression than females out of three studied ethnic groups (268)
 Race / Ethnicity / Genetics • Limited evidence • Strong evidence: ethnic/racial minorities at higher risk for severe COVID-19. In addition, significant national differences in case-fatality ratios • Limited • Increased TMPRSS2: significantly higher nasal gene expression of TMPRSS2 in Blacks than in other races and ethnicities (274)• Increased ACE2: deleterious variants in ACE2 differ among nine populations in gnomAD, specifically in African Americans and non-Finnish European population: 39% vs. 54%, respectively (325)• Increased ACE2: ACE2 expression is significantly higher among Asians than African Americans and Caucasians (326)• Increased eQTLs: eQTLs associated with elevated ACE2 expression in tissues of Eastern Asian population: close to 100% in Eastern Asians and >30% higher than other ethnic groups (268, 327)• No difference in eQTLs: no difference in eQTLs any eQTLs for ACE2 across different populations, though polymorphisms (i.e., p.Val160Met [rs12329760]) were found in TMPRSS2 (325)• No difference in genetic polymorphisms: Asians and other races express similar levels of genetic polymorphisms of the SARS-CoV-2 entry receptor (328, 329)• No difference in ACE2: expression in Asians compared with other races and no unique genetic polymorphisms (328)• No difference in ACE2: gene expressions in lung tissue between Asians and Caucasians in transcriptome databases (330)• Decreased ACE1 II: European population had lower ACE1 II genotype frequency and a higher prevalence of and mortality than the Asian population (332)• Decreased ACE2: O blood type carriers may have lower ACE levels and higher regulated IL-6 levels, suggesting increased balance in the ACE/ANG II axis (331)
Tertiary
 ACEi/ARBs • Strong evidence: for no significant differences in ACEi/ARBs use in non-COVID-19 and COVID-19 patients • Strong evidence: for no significant differences in ACEi/ARBs use in non-COVID-19 and COVID-19 patients • Moderate/ limited • Increased ACE2 mRNA: in mice treated with losartan, olmesartan, valsartan, candesartan, enalapril, and lisinopril (338, 339, 341343)• Increased ACE2 protein: In mice treated with losartan, candesartan, enalapril, and lisinopril (52, 338, 339, 343)• Decreased/no change in ACE2 mRNA: in mice treated with lisinopril, losartan, perindopril, captopril, losartan, lisinopril, and ramipril (10, 102, 157, 338, 340)• Decreased/no change in ACE2 protein: in mice treated with lisinopril, perindopril, and ramipril (10, 102, 157, 338) • Decreased ACE2 expression: lung eQTL study showed the possibility that long-term ACEi use downregulates lung ACE2 expression by reducing substrate availability (346)• Decreased TMPRSS2 expression: lung eQTL study showed ACEi deceased TMPRSS2 expression (346)• No difference in ACE2 expression: lung eQTL study showed that ARBs did not alter ACE2 gene expression (346)
 Thiazolidinediones • Limited • Increased local ACE2: in the liver, adipose tissue, and skeletal muscle when treated with Pioglitazone (354)
 Ibuprofen • Limited • Increased ACE2: When on ibuprofen in diabetic rats with cardiac fibrosis (355)

Summary of the evidence of the relationship between ACE2 levels and primary, secondary, and tertiary risk factors. ACE2, angiotensin-converting enzyme 2; COVID-19, coronavirus disease; CKD, chronic kidney disease; COPD, chronic obstructive pulmonary disease; ACEi, angiotensin converting enzyme inhibitor; ARBs, angiotensin II receptor blockers; SARS-CoV-2, severe acute respiratory syndrome coronavirus 2; MR-IVW, Mendelian randomization-inverse‐variance weighted is correctly written out as inverse variance weighted; BMI, body mass index; TMPRSS2, type 2 transmembrane serine protease; AF, atrial fibrillation; eQTL, expression quantitative locis. ARBs had moderate evidence for elevated ACE2, while ACEi had weak evidence.

As ACE2 provides a pathway for SARS-CoV-2 invasion, increasing studies are investigating the underlying imbalance in the ACE2/ANG-1-7/Mas axis. Dysregulation of ACE2 during COVID-19 may come at a striking cost of direct organ-toxicity, cytokine storm, and endothelial dysfunction. Previous studies have noted that risk factors elevate plasma ACE2 activity. Whether increased plasma ACE2 activity reflects increased synthesis from tissue, ACE2 mRNA, or increased shedding of tissue ACE2 remains to be determined.

Nevertheless, recent studies are uncovering the correlation of ACE2 activity, viral load, and severity of the disease with the preliminary consensus of 1) increased SARS-CoV-2 expression and ACE2 activity is found in direct viral organ-toxicity in severe/critical COVID-19; and 2) elevated viral load relates to increased severity. However, ACE2 activity and susceptibility of COVID-19 are yet to be determined. Further clinical studies are needed to understand ACE2’s relationship to susceptibility and severity of the disease. Novel therapies against SARS-CoV-2 will undoubtedly explore its association with ACE2.

DISCLOSURES

No conflicts of interest, financial or otherwise, are declared by the authors.

AUTHOR CONTRIBUTIONS

G.P. prepared figures; G.P., P.P.F., and A.R.H. drafted manuscript; G.P., P.P.F., A.R.H. and A.E.A. edited and revised manuscript; G.P., P.P.F., A.R.H. and A.E.A. approved final version of manuscript.

ACKNOWLEDGMENTS

The authors credit Devin A. Chan for assistance with graphic design. We also thank our colleagues, the scientific community, and healthcare workers for doing their part in flattening the curve to fight this pandemic.

REFERENCES

  • 1. Centers for Disease Control and Prevention (CDC). Scientific Evidence for Conditions that Increase Risk of Severe Illness | COVID-19 | CDC (Online). https://www.cdc.gov/coronavirus/2019-ncov/need-extra-precautions/evidence-table.html [24 July 2020].
    Google Scholar
  • 2. Guan W, Ni Z, Hu Y, Liang W, Ou C, He J, China Medical Treatment Expert Group for Covid-19 , , et al.. Clinical characteristics of coronavirus disease 2019 in China. N Engl J Med 382: 1708–1720, 2020. doi:10.1056/NEJMoa2002032.
    Crossref | PubMed | ISI | Google Scholar
  • 3. Johns Hopkins University. 2020 Mortality Analyses–Johns Hopkins Coronavirus Resource Center (Online). https://coronavirus.jhu.edu/data/mortality [5 Aug. 2020].
    Google Scholar
  • 4. 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, the Northwell COVID-19 Research Consortium. Presenting characteristics, comorbidities, and outcomes among 5700 patients hospitalized with COVID-19 in the New York City area. JAMA 323: 2052–2059, 2020. [Erratum in JAMA 323: 2098, 2020]. doi:10.1001/jama.2020.6775.
    Crossref | PubMed | ISI | Google Scholar
  • 5. World Health Organization. Report of the WHO-China Joint Mission on Coronavirus Disease 2019 (Online). https://www.who.int/docs/default-source/coronaviruse/who-china-joint-mission-on-covid-19-final-report.pdf [24 Feb 2020].
    Google Scholar
  • 6. Yang J, Zheng Y, Gou X, Pu K, Chen Z, Guo Q, Ji R, Wang H, Wang Y, Zhou Y. Prevalence of comorbidities and its effects in coronavirus disease 2019 patients: A systematic review and meta-analysis. Int J Infect Dis 94: 91–95, 2020. doi:10.1016/j.ijid.2020.03.017.
    Crossref | PubMed | ISI | Google Scholar
  • 7. Zou X, Chen K, Zou J, Han P, Hao J, Han Z. Single-cell RNA-seq data analysis on the receptor ACE2 expression reveals the potential risk of different human organs vulnerable to 2019-nCoV infection. Front Med 14: 185–192, 2020. doi:10.1007/s11684-020-0754-0.
    Crossref | PubMed | ISI | Google Scholar
  • 8. Hikmet F, Méar L, Edvinsson Å, Micke P, Uhlén M, Lindskog C. The protein expression profile of ACE2 in human tissues. Mol Syst Biol 16: e9610 , 2020. msb.20209610.
    PubMed | ISI | Google Scholar
  • 9. Puelles VG, Lütgehetmann M, Lindenmeyer MT, Sperhake JP, Wong MN, Allweiss L, Chilla S, Heinemann A, Wanner N, Liu S, Braun F, Lu S, Pfefferle S, Schröder 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 383: 590–592, 2020. doi:10.1056/NEJMc2011400.
    Crossref | PubMed | ISI | Google Scholar
  • 10. Tikellis C, Thomas MC. Angiotensin-converting enzyme 2 (ACE2) is a key modulator of the renin angiotensin system in health and disease. Int J Pept 2012: 1–8, 2012. doi:10.1155/2012/256294.
    Crossref | PubMed | Google Scholar
  • 11. Brojakowska A, Narula J, Shimony R, Bander J. Clinical implications of SARS-CoV-2 interaction with renin angiotensin system: JACC review topic of the week. J Am Coll Cardiol 75: 3085–3095, 2020. dois:10.1016/S0735-1097(20)33712-8, 10.1016/j.jacc.2020.04.028.
    Crossref | PubMed | ISI | Google Scholar
  • 12. 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. Circ Res 126: 1456–1474, 2020. doi:10.1161/CIRCRESAHA.120.317015.
    Crossref | PubMed | ISI | Google Scholar
  • 13. Norwood VF, Fernandez LG, Tufro A, Gomez RA. Development of the renin-angiotensin system. Fetal and Neonatal Physiology: Third Edition. Philadelphia, PA: Saunders, 2003, p. 1249–1256.
    Google Scholar
  • 14. Wang K, Gheblawi M, Oudit GY. Angiotensin converting enzyme 2. Circulation 142: 426–428, 2020. doi:10.1161/CIRCULATIONAHA.120.047049.
    Crossref | PubMed | ISI | Google Scholar
  • 15. Tikellis C, Bialkowski K, Pete J, Sheehy K, Su Q, Johnston C, Cooper ME, Thomas MC. ACE2 deficiency modifies renoprotection afforded by ACE inhibition in experimental diabetes. Diabetes 57: 1018–1025, 2008 [Erratum in Diabetes 60: 360, 2011]. doi:10.2337/db07-1212.
    Crossref | PubMed | ISI | Google Scholar
  • 16. Klompas M, Baker MA, Rhee C. Airborne transmission of SARS-CoV-2. JAMA 324: 441 , 2020. doi:10.1001/jama.2020.12458.
    Crossref | PubMed | ISI | Google Scholar
  • 17. Van Doremalen N, Bushmaker T, Morris DH, Holbrook MG, Gamble A, Williamson BN, Tamin A, Harcourt JL, Thornburg NJ, Gerber SI, Lloyd-Smith JO, De Wit E, Munster VJ. Aerosol and surface stability of SARS-CoV-2 as compared to SARS-CoV-1. N Engl J Med 382: 1564–1567, 2020. doi:10.1056/NEJMc2004973.
    Crossref | PubMed | ISI | Google Scholar
  • 18. Wiersinga WJ, Rhodes A, Cheng AC, Peacock SJ, Prescott HC. Pathophysiology, transmission, diagnosis, and treatment of coronavirus disease 2019 (COVID-19): a review. JAMA 324: 782–793, 2020. doi:10.1001/jama.2020.12839.
    Crossref | PubMed | ISI | Google Scholar
  • 19. Xu H, Zhong L, Deng J, Peng J, Dan H, Zeng X, Li T, Chen Q. High expression of ACE2 receptor of 2019-nCoV on the epithelial cells of oral mucosa. Int J Oral Sci 12: 1–5, 2020. doi:10.1038/s41368-019-0067-9.
    Crossref | PubMed | ISI | Google Scholar
  • 20. Atri D, Siddiqi HK, Lang JP, Nauffal V, Morrow DA, Bohula EA. COVID-19 for the cardiologist: basic virology, epidemiology, cardiac manifestations, and potential therapeutic strategies. JACC Basic Transl Sci 5: 518–536, 2020. doi:10.1016/j.jacbts.2020.04.002.
    Crossref | PubMed | Google Scholar
  • 21. Xu X, Chen P, Wang J, Feng J, Zhou H, Li X, Zhong W, Hao P. Evolution of the novel coronavirus from the ongoing Wuhan outbreak and modeling of its spike protein for risk of human transmission. Sci China Life Sci 63: 457–460, 2020. doi:10.1007/s11427-020-1637-5.
    Crossref | PubMed | ISI | Google Scholar
  • 22. Yan R, Zhang Y, Li Y, Xia L, Guo Y, Zhou Q. Structural basis for the recognition of SARS-CoV-2 by full-length human ACE2. Science 367: 1444–1448, 2020. doi:10.1126/science.abb2762.
    Crossref | PubMed | ISI | Google Scholar
  • 23. Lei C, Qian K, Li T, Zhang S, Fu W, Ding M, Hu S. Neutralization of SARS-CoV-2 spike pseudotyped virus by recombinant ACE2-Ig. Nat Commun 11: 1–5, 2020. doi:10.1038/s41467-019-13993-7.
    Crossref | PubMed | ISI | Google Scholar
  • 24. Wang Q, Zhang Y, Wu L, Niu S, Song C, Zhang Z, Lu G, Qiao C, Hu Y, Yuen KY, Wang Q, Zhou H, Yan J, Qi J. Structural and functional basis of SARS-CoV-2 entry by using human ACE2. Cell 181: 894–904.e9, 2020. doi:10.1016/j.cell.2020.03.045.
    Crossref | PubMed | ISI | Google Scholar
  • 25. Wrapp D, Wang N, Corbett KS, Goldsmith JA, Hsieh CL, Abiona O, Graham BS, McLellan JS. Cryo-EM structure of the 2019-nCoV spike in the prefusion conformation. Science 367: 1260–1263, 2020. doi:10.1126/science.abb2507.
    Crossref | PubMed | ISI | Google Scholar
  • 26. Zhai X, Sun J, Yan Z, Zhang J, Zhao J, Zhao Z, Gao Q, He WT, Veit M, Su S. Comparison of SARS-CoV-2 spike protein binding to ACE2 receptors from human, pets, farm animals, and putative intermediate hosts. J Virol 94, 2020. doi:10.1128/JVI.00831-20.
    Crossref | PubMed | ISI | Google Scholar
  • 27. Eaaswarkhanth M, Al Madhoun A, Al-Mulla F. Could the D614G substitution in the SARS-CoV-2 spike (S) protein be associated with higher COVID-19 mortality. Int J Infect Dis 96: 459–460, 2020. doi:10.1016/j.ijid.2020.05.071.
    Crossref | PubMed | ISI | Google Scholar
  • 28. Korber B, Fischer WM, Gnanakaran S, Yoon H, Theiler J, Abfalterer W, Sheffield COVID-19 Genomics Group , , et al.. Tracking changes in SARS-CoV-2 spike: evidence that d614g increases infectivity of the COVID-19 virus. Cell 182: 812–827.e19, 2020. doi:10.1016/j.cell.2020.06.043.
    Crossref | PubMed | ISI | Google Scholar
  • 29. Toyoshima Y, Nemoto K, Matsumoto S, Nakamura Y, Kiyotani K. SARS-CoV-2 genomic variations associated with mortality rate of COVID-19. J Hum Genet 65: 1075–1082, 2020. doi:10.1038/s10038-020-0808-9.
    Crossref | PubMed | ISI | Google Scholar
  • 30. Yurkovetskiy L, Wang X, Pascal KE, Tomkins-Tinch C, Nyalile T, Wang Y, Baum A, Diehl WE, Dauphin A, Carbone C, Veinotte K, Egri SB, Schaffner SF, Lemieux JE, Munro J, Rafique A, Barve A, Sabeti PC, Kyratsous CA, Dudkina N, Shen K, Luban J. Structural and functional analysis of the D614G SARS-CoV-2 spike protein variant. Cell 183: 739–751.e8, 2020. doi:10.1016/j.cell.2020.09.032.
    Crossref | PubMed | ISI | Google Scholar
  • 31. Matheson NJ, Lehner PJ. How does SARS-CoV-2 cause COVID-19? Science 369: 510–511, 2020. doi:10.1126/science.abc6156.
    Crossref | PubMed | ISI | Google Scholar
  • 32. Gupta A, Madhavan MV, Sehgal K, Nair N, Mahajan S, Sehrawat TS, Bikdeli B, Ahluwalia N, Ausiello JC, Wan EY, Freedberg DE, Kirtane AJ, Parikh SA, Maurer MS, Nordvig AS, Accili D, Bathon JM, Mohan S, Bauer KA, Leon MB, Krumholz HM, Uriel N, Mehra MR, Elkind MV, Stone GW, Schwartz A, Ho DD, Bilezikian JP, Landry DW. Extrapulmonary manifestations of COVID-19. Nat Med 26: 1017–1032, 2020. doi:10.1038/s41591-020-0968-3.
    Crossref | PubMed | ISI | Google Scholar
  • 33. Ackermann M, Verleden SE, Kuehnel M, Haverich A, Welte T, Laenger F, Vanstapel A, Werlein CMD, 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 383: 120–128, 2020. doi:10.1056/NEJMoa2015432.
    Crossref | PubMed | ISI | Google Scholar
  • 34. Dolhnikoff M, Ferreira Ferranti J, de Almeida Monteiro RA, Duarte-Neto AN, Soares Gomes-Gouvêa M, Viu Degaspare N, Figueiredo Delgado A, Montanari Fiorita C, Nunes Leal G, Rodrigues RM, Taverna Chaim K, Rebello Pinho JR, Carneiro-Sampaio M, Mauad T, Ferraz da Silva LF, Brunow de Carvalho W, Saldiva PH, Garcia Caldini E. SARS-CoV-2 in cardiac tissue of a child with COVID-19-related multisystem inflammatory syndrome. Lancet Child Adolesc Heal 4: 790–794, 2020. doi:10.1016/S2352-4642(20)30257-1.
    Crossref | PubMed | ISI | Google Scholar
  • 35. Hanley B, Naresh KN, Roufosse C, Nicholson AG, Weir J, Cooke GS, Thursz M, Manousou P, Corbett R, Goldin R, Al-Sarraj S, Abdolrasouli A, Swann OC, Baillon L, Penn R, Barclay WS, Viola P, Osborn M. Histopathological findings and viral tropism in UK patients with severe fatal COVID-19: a post-mortem study. Lancet Microbe 1: e245–e253, 2020. doi:10.1016/S2666-5247(20)30115-4.
    Crossref | PubMed | Google Scholar
  • 36. Su H, Yang M, Wan C, Yi LX, Tang F, Zhu HY, Yi F, Yang HC, Fogo AB, Nie X, Zhang C. Renal histopathological analysis of 26 postmortem findings of patients with COVID-19 in China. Kidney Int 98: 219–227, 2020. doi:10.1016/j.kint.2020.04.003.
    Crossref | PubMed | ISI | Google Scholar
  • 37. 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 Respir Med 8: 420–422, 2020 [Erratum in Lancet Respir Med 8: e26, 2020]. doi:10.1016/S2213-2600(20)30076-X.
    Crossref | PubMed | ISI | Google Scholar
  • 38. Zhang H, Zhou P, Wei Y, Yue H, Wang Y, Hu M, Zhang S, Cao T, Yang C, Li M, Guo G, Chen X, Chen Y, Lei M, Liu H, Zhao J, Peng P, Wang CY, Du R. Histopathologic changes and SARS-CoV-2 immunostaining in the lung of a patient with COVID-19. Ann Intern Med 172: 629–632, 2020. doi:10.7326/M20-0533.
    Crossref | PubMed | ISI | Google Scholar
  • 39. Channappanavar R, Fehr AR, Vijay R, Mack M, Zhao J, Meyerholz DK, Perlman S. Dysregulated type I interferon and inflammatory monocyte-macrophage responses cause lethal pneumonia in SARS-CoV-infected mice. Cell Host Microbe 19: 181–193, 2016. doi:10.1016/j.chom.2016.01.007.
    Crossref | PubMed | ISI | Google Scholar
  • 40. Cummings MJ, Baldwin MR, Abrams D, Jacobson SD, Meyer BJ, Balough EM, Aaron JG, Claassen J, Rabbani LR, Hastie J, Hochman BR, Salazar-Schicchi J, Yip NH, Brodie D, O’Donnell MR. Epidemiology, clinical course, and outcomes of critically ill adults with COVID-19 in New York City: a prospective cohort study. Lancet 395: 1763–1770, 2020. doi:10.1016/S0140-6736(20)31189-2.
    Crossref | PubMed | ISI | Google Scholar
  • 41. 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 395: 497–506, 2020. doi:10.1016/S0140-6736(20)30183-5.
    Crossref | PubMed | ISI | Google Scholar
  • 42. Petrilli CM, Jones SA, Yang J, Rajagopalan H, O’Donnell L, Chernyak Y, Tobin KA, Cerfolio RJ, Francois F, Horwitz LI. Factors associated with hospital admission and critical illness among 5279 people with coronavirus disease 2019 in New York City: prospective cohort study. BMJ 369, m1966 , 2020. doi:10.1136/bmj.m1966.
    Crossref | PubMed | Google Scholar
  • 43. 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 46: 846–848, 2020 [Erratum in Intensive Care Med 46: 1294-1297, 2020]. doi:10.1007/s00134-020-05991-x.
    Crossref | PubMed | ISI | Google Scholar
  • 44. Wu C, Chen X, Cai Y, Xia JJ, Zhou XX, Xu S, Huang H, Zhang L, Zhou XX, Du C, Zhang Y, Song J, Wang S, Chao Y, Yang Z, Xu J, Zhou XX, Chen D, Xiong W, Xu L, Zhou F, Jiang J, Bai C, Zheng J, Song Y. Risk factors associated with acute respiratory distress syndrome and death in patients with coronavirus disease 2019 pneumonia in Wuhan, China. JAMA Intern Med 180: 934–943, 2020. doi:10.1001/jamainternmed.2020.0994.
    Crossref | PubMed | ISI | Google Scholar
  • 45. Zhou F, Yu T, Du R, Fan G, Liu Y, Liu Z, Xiang J, Wang Y, Song B, Gu X, Guan L, Wei Y, Li H, Wu X, Xu J, Tu S, Zhang Y, Chen H, Cao B. Clinical course and risk factors for mortality of adult inpatients with COVID-19 in Wuhan, China: a retrospective cohort study. Lancet 395: 1054–1062, 2020. doi:10.1016/S0140-6736(20)30566-3.
    Crossref | PubMed | ISI | Google Scholar
  • 46. Vabret N, Britton GJ, Gruber C, Hegde S, Kim J, Kuksin M, Sinai Immunology Review Project , , et al.. Immunology of COVID-19: current state of the science. Immunity 52: 910–941, 2020. doi:10.1016/j.immuni.2020.05.002.
    Crossref | PubMed | ISI | Google Scholar
  • 47. Bikdeli B, Madhavan MV, Jimenez D, Chuich T, Dreyfus I, Driggin E, Global COVID-19 Thrombosis Collaborative Group, Endorsed by the ISTH, NATF, ESVM, and the IUA, Supported by the ESC Working Group on Pulmonary Circulation and Right Ventricular Function , , et al.. COVID-19 and thrombotic or thromboembolic disease: implications for prevention, antithrombotic therapy, and follow-up. J Am Coll Cardiol 75: 2950–2973, 2020. doi:10.1016/j.jacc.2020.04.031, 10.1016/S0735-1097(20)33577-4.
    Crossref | PubMed | ISI | Google Scholar
  • 48. de Lang A, Osterhaus AD, Haagmans BL. Interferon-γ and interleukin-4 downregulate expression of the SARS coronavirus receptor ACE2 in Vero E6 cells. Virology 353: 474–481, 2006. doi:10.1016/j.virol.2006.06.011.
    Crossref | PubMed | ISI | Google Scholar
  • 49. Imai Y, Kuba K, Rao S, Huan Y, Guo F, Guan B, Yang P, Sarao R, Wada T, Leong-Poi H, Crackower MA, Fukamizu A, Hui CC, Hein L, Uhlig S, Slutsky AS, Jiang C, Penninger JM. Angiotensin-converting enzyme 2 protects from severe acute lung failure. Nature 436: 112–116, 2005. doi:10.1038/nature03712.
    Crossref | PubMed | ISI | Google Scholar
  • 50. Verdecchia P, Cavallini C, Spanevello A, Angeli F. The pivotal link between ACE2 deficiency and SARS-CoV-2 infection. Eur J Intern Med 76: 14–20, 2020. doi:10.1016/j.ejim.2020.04.037.
    Crossref | PubMed | ISI | Google Scholar
  • 51. Jia H. Pulmonary angiotensin-converting enzyme 2 (ACE2) and inflammatory lung disease. Shock 46: 239–248, 2016. doi:10.1097/SHK.0000000000000633.
    Crossref | PubMed | ISI | Google Scholar
  • 52. Kuba K, Imai Y, Rao S, Gao H, Guo F, Guan B, Huan Y, Yang P, Zhang Y, Deng W, Bao L, Zhang B, Liu G, Wang Z, Chappell M, Liu Y, Zheng D, Leibbrandt A, Wada T, Slutsky AS, Liu D, Qin C, Jiang C, Penninger JM. A crucial role of angiotensin converting enzyme 2 (ACE2) in SARS coronavirus-induced lung injury. Nat Med 11: 875–879, 2005. doi:10.1038/nm1267.
    Crossref | PubMed | ISI | Google Scholar
  • 53. Ye R, Liu Z. ACE2 exhibits protective effects against LPS-induced acute lung injury in mice by inhibiting the LPS-TLR4 pathway. Exp Mol Pathol 113: 104350 , 2020. doi:10.1016/j.yexmp.2019.104350.
    Crossref | PubMed | ISI | Google Scholar
  • 54. Ziegler CG, Allon SJ, Nyquist SK, Mbano IM, Miao VN, Tzouanas CN, , et al.. SARS-CoV-2 receptor ACE2 is an interferon-stimulated gene in human airway epithelial cells and is detected in specific cell subsets across tissues. Cell 181: 1016–1035.e19, 2020. doi:10.1016/j.cell.2020.04.035.
    Crossref | PubMed | ISI | Google Scholar
  • 55. Hadjadj J, Yatim N, Barnabei L, Corneau A, Boussier J, Smith N, Péré H, Charbit B, Bondet V, Chenevier-Gobeaux C, Breillat P, Carlier N, Gauzit R, Morbieu C, Pène F, Marin N, Roche N, Szwebel TA, Merkling SH, Treluyer JM, Veyer D, Mouthon L, Blanc C, Tharaux PL, Rozenberg F, Fischer A, Duffy D, Rieux-Laucat F, Kernéis S, Terrier B. Impaired type I interferon activity and inflammatory responses in severe COVID-19 patients. Science 369: 718–724, 2020. doi:10.1126/science.abc6027.
    Crossref | PubMed | ISI | Google Scholar
  • 56. Plenge RM. Molecular underpinnings of severe coronavirus disease 2019. JAMA 324: 638 , 2020. doi:10.1001/jama.2020.14015.
    Crossref | PubMed | ISI | Google Scholar
  • 57. Hung IF, Lung KC, Tso EY, Liu R, Chung TW, Chu MY, , et al.. Triple combination of interferon beta-1b, lopinavir–ritonavir, and ribavirin in the treatment of patients admitted to hospital with COVID-19: an open-label, randomised, phase 2 trial. Lancet 395: 1695–1704, 2020. doi:10.1016/S0140-6736(20)31042-4.
    Crossref | PubMed | ISI | Google Scholar
  • 58. Horby P, Lim WS, Emberson JR, Mafham M, Bell JL, 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, RECOVERY Collaborative Group. Dexamethasone in hospitalized patients with Covid-19—preliminary report. New Engl J Med. In press. doi:10.1056/nejmoa2021436.
    Crossref | PubMed | ISI | Google Scholar
  • 59. Wang N, Zhan Y, Zhu L, Hou Z, Liu F, Song P, Qiu F, Wang X, Zou X, Wan D, Qian X, Wang S, Guo Y, Yu H, Cui M, Tong G, Xu Y, Zheng Z, Lu Y, Hong P. Retrospective multicenter cohort study shows early interferon therapy is associated with favorable clinical responses in COVID-19 patients. Cell Host Microbe 28: 455–464.e2, 2020. doi:10.1016/j.chom.2020.07.005.
    Crossref | PubMed | ISI | Google Scholar
  • 60. Bilaloglu S, Aphinyanaphongs Y, Jones S, Iturrate E, Hochman J, Berger JS. Thrombosis in hospitalized patients with COVID-19 in a New York City Health System. JAMA 324: 799–801, 2020. doi:10.1001/jama.2020.13372.
    Crossref | PubMed | ISI | Google Scholar
  • 61. Klok FA, Kruip MJ, van der Meer NJ, Arbous MS, Gommers D, Kant KM, Kaptein FH, van Paassen J, Stals MA, Huisman MV, Endeman H. Incidence of thrombotic complications in critically ill ICU patients with COVID-19. Thromb Res 191: 145–147, 2020. doi:10.1016/j.thromres.2020.04.013.
    Crossref | PubMed | ISI | Google Scholar
  • 62. Nahum J, Morichau-Beauchant T, Daviaud F, Echegut P, Fichet J, Maillet JM, Thierry S. Venous thrombosis among critically ill patients with coronavirus disease 2019 (COVID-19). JAMA Netw Open 3: e2010478 , 2020. doi:10.1001/jamanetworkopen.2020.10478.
    Crossref | PubMed | ISI | Google Scholar
  • 63. Price LC, McCabe C, Garfield B, Wort SJ. Thrombosis and COVID-19 pneumonia: the clot thickens. Eur Respir J 56: 2001608 , 2020. doi:10.1183/13993003.01608-2020.
    Crossref | PubMed | ISI | Google Scholar
  • 64. 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 395: 1417–1418, 2020. doi:10.1016/S0140-6736(20)30937-5.
    Crossref | PubMed | ISI | Google Scholar
  • 65. Deshpande C. Thromboembolic findings in COVID-19 autopsies: pulmonary thrombosis or embolism? Ann Intern Med 173: 394–395, 2020. doi:10.7326/M20-3255.
    Crossref | PubMed | ISI | Google Scholar
  • 66. Giannis D, Ziogas IA, Gianni P. Coagulation disorders in coronavirus infected patients: COVID-19, SARS-CoV-1, MERS-CoV and lessons from the past. J Clin Virol 127: 104362 , 2020. doi:10.1016/j.jcv.2020.104362.
    Crossref | PubMed | ISI | Google Scholar
  • 67. Gupta N, Zhao YY, Evans CE. The stimulation of thrombosis by hypoxia. Thromb Res 181: 77–83, 2019. doi:10.1016/j.thromres.2019.07.013.
    Crossref | PubMed | ISI | Google Scholar
  • 68. Teuwen L-A, Geldhof V, Pasut A, Carmeliet P. COVID-19: the vasculature unleashed. Nat Rev Immunol 20: 389–391, 2020 [Erratum in Nat Rev Immunol 20: 448, 2020]. doi:10.1038/s41577-020-0343-0.
    Crossref | PubMed | ISI | Google Scholar
  • 69. Lowenstein CJ, Solomon SD. Severe COVID-19 is a microvascular disease. Circulation 142: 1609–1611, 2020. doi:10.1161/CIRCULATIONAHA.120.050354.
    Crossref | PubMed | ISI | Google Scholar
  • 70. Ayerbe L, Risco C, Ayis S. The association between treatment with heparin and survival in patients with Covid-19. J Thromb Thrombolysis 50: 298–301, 2020. doi:10.1007/s11239-020-02162-z.
    Crossref | PubMed | ISI | Google Scholar
  • 71. Stessel B, Vanvuchelen C, Bruckers L, Geebelen L, Callebaut I, Vandenbrande J, Pellens B, van Tornout M, Ory JP, van Halem K, Messiaen P, Herbots L, Ramaekers D, Dubois J. Impact of implementation of an individualised thromboprophylaxis protocol in critically ill ICU patients with COVID-19: a longitudinal controlled before-after study. Thromb Res 194: 209–215, 2020. doi:10.1016/j.thromres.2020.07.038.
    Crossref | PubMed | ISI | Google Scholar
  • 72. Hofmann H, Geier M, Marzi A, Krumbiegel M, Peipp M, Fey GH, Gramberg T, Pöhlmann S. Susceptibility to SARS coronavirus S protein-driven infection correlates with expression of angiotensin converting enzyme 2 and infection can be blocked by soluble receptor. Biochem Biophys Res Commun 319: 1216–1221, 2004. doi:10.1016/j.bbrc.2004.05.114.
    Crossref | PubMed | ISI | Google Scholar
  • 73. Jia HP, Look DC, Shi L, Hickey M, Pewe L, Netland J, Farzan M, Wohlford-Lenane C, Perlman S, McCray PB. ACE2 Receptor expression and severe acute respiratory syndrome coronavirus infection depend on differentiation of human airway epithelia. J Virol 79: 14614–14621, 2005. doi:10.1128/JVI.79.23.14614-14621.2005.
    Crossref | PubMed | ISI | Google Scholar
  • 74. Nie Y, Wang P, Shi X, Wang G, Chen J, Zheng A, Wang W, Wang Z, Qu X, Luo M, Tan L, Song X, Yin X, Chen J, Ding M, Deng H. Highly infectious SARS-CoV pseudotyped virus reveals the cell tropism and its correlation with receptor expression. Biochem Biophys Res Commun 321: 994–1000, 2004. doi:10.1016/j.bbrc.2004.07.060.
    Crossref | PubMed | ISI | Google Scholar
  • 75. Bao L, Deng W, Huang B, Gao H, Liu J, Ren L, , et al.. The pathogenicity of SARS-CoV-2 in hACE2 transgenic mice. Nature 583: 830–833, 2020. doi:10.1038/s41586-020-2312-y.
    Crossref | PubMed | ISI | Google Scholar
  • 76. Hassan AO, Case JB, Winkler ES, Thackray LB, Kafai NM, Bailey AL, McCune BT, Fox JM, Chen RE, Alsoussi WB, Turner JS, Schmitz AJ, Lei T, Shrihari S, Keeler SP, Fremont DH, Greco S, McCray PB, Perlman S, Holtzman MJ, Ellebedy AH, Diamond MS. A SARS-CoV-2 infection model in mice demonstrates protection by neutralizing antibodies. Cell 182: 744–753.e4, 2020. doi:10.1016/j.cell.2020.06.011.
    Crossref | PubMed | ISI | Google Scholar
  • 77. Moreau GB, Burgess SL, Sturek JM, Donlan AN, Petri WA, Mann BJ. Evaluation of K18-hACE2 mice as a model of SARS-CoV-2 infection. Am J Trop Med Hyg 103: 1215–1219, 2020. doi:10.4269/ajtmh.20-0762.
    Crossref | PubMed | ISI | Google Scholar
  • 78. Lieberman NAP, Peddu V, Xie H, Shrestha L, Huang M-L, Mears MC, Cajimat MN, Bente DA, Shi P-Y, Bovier F, Roychoudhury P, Jerome KR, Moscona A, Porotto M, Greninger AL. In vivo antiviral host transcriptional response to SARS-CoV-2 by viral load, sex, and age. PLOS Biol 18: e3000849 , 2020. doi:10.1371/journal.pbio.3000849.
    Crossref | PubMed | ISI | Google Scholar
  • 79. Westblade LF, Brar G, Pinheiro LC, Paidoussis D, Rajan M, Martin P, Goyal P, Sepulveda JL, Zhang L, George G, Liu D, Whittier S, Plate M, Small CB, Rand JH, Cushing MM, Walsh TJ, Cooke J, Safford MM, Loda M, Satlin MJ. SARS-CoV-2 viral load predicts mortality in patients with and without cancer who are hospitalized with COVID-19. Cancer Cell 38: 661–671.e2, 2020. doi:10.1016/j.ccell.2020.09.007.
    Crossref | PubMed | ISI | Google Scholar
  • 80. Pujadas E, Chaudhry F, McBride R, Richter F, Zhao S, Wajnberg A, Nadkarni G, Glicksberg BS, Houldsworth J, Cordon-Cardo C. SARS-CoV-2 viral load predicts COVID-19 mortality. Lancet Respir Med 8: e70 , 2020. doi:10.1016/S2213-2600(20)30354-4.
    Crossref | PubMed | ISI | Google Scholar
  • 81. Liu N, Hong Y, Chen RG, Zhu HM. High rate of increased level of plasma angiotensin II and its gender difference in COVID-19: an analysis of 55 hospitalized patients with COVID-19 in a single hospital, WuHan, China (Preprint). medRxiv 2020.04.27.20080432 , 2020. doi:10.1101/2020.04.27.20080432.
    Crossref | Google Scholar
  • 82. Liu Y, Yang Y, Zhang C, Huang F, Wang F, Yuan J, Wang Z, Li J, Li J, Feng C, Zhang Z, Wang L, Peng L, Chen L, Qin Y, Zhao D, Tan S, Yin L, Xu J, Zhou C, Jiang C, Liu L. Clinical and biochemical indexes from 2019-nCoV infected patients linked to viral loads and lung injury. Sci China Life Sci 63: 364–374, 2020. doi:10.1007/s11427-020-1643-8.
    Crossref | PubMed | ISI | Google Scholar
  • 83. Villard O, Morquin D, Molinari N, Raingeard I, Nagot N, Cristol JP, Jung B, Roubille C, Foulongne V, Fesler P, Lamure S, Taourel P, Konate A, Maria AT, Makinson A, Bertchansky I, Larcher R, Klouche K, Le Moing V, Renard E, Guilpain P. The plasmatic aldosterone and C-reactive protein levels, and the severity of Covid-19: The Dyhor-19 Study. J Clin Med 9: 2315 , 2020. doi:10.3390/jcm9072315.
    Crossref | PubMed | ISI | Google Scholar
  • 84. Wu Z, Hu R, Zhang C, Ren W, Yu A, Zhou X. Elevation of plasma angiotensin II level is a potential pathogenesis for the critically ill COVID-19 patients. Crit Care 24: 2315 , 2020. doi:10.1186/s13054-020-03015-0.
    Crossref | PubMed | ISI | Google Scholar
  • 85. Henry BM, Benoit S, Lippi G, Benoit J. Letter to the Editor–Circulating plasma levels of angiotensin II and aldosterone in patients with coronavirus disease 2019 (COVID-19): a preliminary report. Prog Cardiovasc Dis 63: 702–703, 2020. doi:10.1016/j.pcad.2020.07.006.
    Crossref | PubMed | ISI | Google Scholar
  • 86. Oudit GY, Kassiri Z, Jiang C, Liu PP, Poutanen SM, Penninger JM, Butany J. SARS-coronavirus modulation of myocardial ACE2 expression and inflammation in patients with SARS. Eur J Clin Invest 39: 618–625, 2009. doi:10.1111/j.1365-2362.2009.02153.x.
    Crossref | PubMed | ISI | Google Scholar
  • 87. Chan KK, Dorosky D, Sharma P, Abbasi SA, Dye JM, Kranz DM, Herbert AS, Procko E. Engineering human ACE2 to optimize binding to the spike protein of SARS coronavirus 2. Science 369: 1261–1265, 2020. doi:10.1126/science.abc0870.
    Crossref | PubMed | ISI | Google Scholar
  • 88. Hoffmann M, Kleine-Weber H, Schroeder S, Krüger N, Herrler T, Erichsen S, Schiergens TS, Herrler G, Wu NH, Nitsche A, Müller MA, Drosten C, Pöhlmann S. SARS-CoV-2 Cell Entry Depends on ACE2 and TMPRSS2 and Is Blocked by a Clinically Proven Protease Inhibitor. Cell 181: 271–280.e8, 2020. doi:10.1016/j.cell.2020.02.052.
    Crossref | PubMed | ISI | Google Scholar
  • 89. Huo J, Le Bas A, Ruza RR, Duyvesteyn HM, Mikolajek H, Malinauskas T, Tan TK, , et al.. Neutralizing nanobodies bind SARS-CoV-2 spike RBD and block interaction with ACE2. Nat Struct Mol Biol 27: 846–854, 2020 [Erratum in Nat Struct Mol Biol 27: 1094, 2020]. doi:10.1038/s41594-020-0469-6.
    Crossref | PubMed | ISI | Google Scholar
  • 90. Monteil V, Kwon H, Prado P, Hagelkrüys 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 181: 905–913.e7, 2020. doi:10.1016/j.cell.2020.04.004.
    Crossref | PubMed | ISI | Google Scholar
  • 91. Zou Z, Yan Y, Shu Y, Gao R, Sun Y, Li X, Ju X, Liang Z, Liu Q, Zhao Y, Guo F, Bai T, Han Z, Zhu J, Zhou H, Huang F, Li C, Lu H, Li N, Li D, Jin N, Penninger JM, Jiang C. Angiotensin-converting enzyme 2 protects from lethal avian influenza A H5N1 infections. Nat Commun 5: 3594 , 2014. doi:10.1038/ncomms4594.
    Crossref | PubMed | ISI | Google Scholar
  • 92. Anguiano L, Riera M, Pascual J, Soler MJ. Circulating ACE2 in cardiovascular and kidney diseases. Curr Med Chem 24: 3231–3241, 2017. doi:10.2174/0929867324666170414162841.
    Crossref | PubMed | ISI | Google Scholar
  • 93. Oudit GY, Kassiri Z, Patel MP, Chappell M, Butany J, Backx PH, Tsushima RG, Scholey JW, Khokha R, Penninger JM. Angiotensin II-mediated oxidative stress and inflammation mediate the age-dependent cardiomyopathy in ACE2 null mice. Cardiovasc Res 75: 29–39, 2007. doi:10.1016/j.cardiores.2007.04.007.
    Crossref | PubMed | ISI | Google Scholar
  • 94. Qi YF, Zhang J, Wang L, Shenoy V, Krause E, Oh SP, Pepine CJ, Katovich MJ, Raizada MK. Angiotensin-converting enzyme 2 inhibits high-mobility group box 1 and attenuates cardiac dysfunction post-myocardial ischemia. J Mol Med 94: 37–49, 2016. doi:10.1007/s00109-015-1356-1.
    Crossref | PubMed | ISI | Google Scholar
  • 95. Wang Y, Qian C, Roks AJ, Westermann D, Schumacher SM, Escher F, Schoemaker RG, Reudelhuber TL, van Gilst WH, Schultheiss HP, Tschöpe C, Walther T. Circulating rather than cardiac angiotensin-(1-7) stimulates cardioprotection after myocardial infarction. Circ Heart Fail 3: 286–293, 2010. doi:10.1161/CIRCHEARTFAILURE.109.905968.
    Crossref | PubMed | ISI | Google Scholar
  • 96. Crackower MA, Sarao R, Oudit GY, Yagil C, Kozieradzki I, Scanga SE, Oliveira-dos-Santos AJ, da Costa J, Zhang L, Pei Y, Scholey J, Ferrario CM, Manoukian AS, Chappell MC, Backx PH, Yagil Y, Penninger JM. Angiotensin-converting enzyme 2 is an essential regulator of heart function. Nature 417: 822–828, 2002. doi:10.1038/nature00786.
    Crossref | PubMed | ISI | Google Scholar
  • 97. Díez-Freire C, Vázquez J, Correa de Adjounian MF, Ferrari MF, Yuan L, Silver X, Torres R, Raizada MK. ACE2 gene transfer attenuates hypertension-linked pathophysiological changes in the SHR. Physiol Genomics 27: 12–19, 2006. doi:10.1152/physiolgenomics.00312.2005.
    Link | ISI | Google Scholar
  • 98. Rentzsch B, Todiras M, Iliescu R, Popova E, Campos LA, Oliveira ML, Baltatu OC, Santos RA, Bader M. Transgenic angiotensin-converting enzyme 2 overexpression in vessels of SHRSP rats reduces blood pressure and improves endothelial function. Hypertension 52: 967–973, 2008. doi:10.1161/HYPERTENSIONAHA.108.114322.
    Crossref | PubMed | ISI | Google Scholar
  • 99. Wysocki J, Ye M, Rodriguez E, González-Pacheco FR, Barrios C, Evora K, Schuster M, Loibner H, Brosnihan KB, Ferrario CM, Penninger JM, Batlle D. Targeting the degradation of angiotensin II with recombinant angiotensin-converting enzyme 2: prevention of angiotensin II-dependent hypertension. Hypertension 55: 90–98, 2010. doi:10.1161/HYPERTENSIONAHA.109.138420.
    Crossref | PubMed | ISI | Google Scholar
  • 100. Cole-Jeffrey CT, Liu M, Katovich MJ, Raizada MK, Shenoy V. ACE2 and microbiota: emerging targets for cardiopulmonary disease therapy. J Cardiovasc Pharmacol 66: 540–550, 2015. doi:10.1097/FJC.0000000000000307.
    Crossref | PubMed | ISI | Google Scholar
  • 101. Hemnes AR, Rathinasabapathy A, Austin EA, Brittain EL, Carrier EJ, Chen X, Fessel JP, Fike CD, Fong P, Fortune N, Gerszten RE, Johnson JA, Kaplowitz M, Newman JH, Piana R, Pugh ME, Rice TW, Robbins IM, Wheeler L, Yu C, Loyd JE, West JA. Potential therapeutic role for angiotensin-converting enzyme 2 in human pulmonary arterial hypertension. Eur Respir J 51: 1702638 , 2018. doi:10.1183/13993003.02638-2017.
    Crossref | PubMed | ISI | Google Scholar
  • 102. Hamming I, Van Goor H, Turner AJ, Rushworth CA, Michaud AA, Corvol P, Navis G. Differential regulation of renal angiotensin-converting enzyme (ACE) and ACE2 during ACE inhibition and dietary sodium restriction in healthy rats. Exp Physiol 93: 631–638, 2008. doi:10.1113/expphysiol.2007.041855.
    Crossref | PubMed | ISI | Google Scholar
  • 103. Lely AT, Hamming I, van Goor H, Navis GJ. Renal ACE2 expression in human kidney disease. J Pathol 204: 587–593, 2004. doi:10.1002/path.1670.
    Crossref | PubMed | ISI | Google Scholar
  • 104. Oudit GY, Herzenberg AM, Kassiri Z, Wong D, Reich H, Khokha R, Crackower MA, Backx PH, Penninger JM, Scholey JW. Loss of angiotensin-converting enzyme-2 leads to the late development of angiotensin II-dependent glomerulosclerosis. Am J Pathol 168: 1808–1820, 2006. doi:10.2353/ajpath.2006.051091.
    Crossref | PubMed | ISI | Google Scholar
  • 105. Hamming I, Timens W, Bulthuis MLC, Lely AT, Navis GJ, van Goor H. Tissue distribution of ACE2 protein, the functional receptor for SARS coronavirus. A first step in understanding SARS pathogenesis. J Pathol 203: 631–637, 2004. doi:10.1002/path.1570.
    Crossref | PubMed | ISI | Google Scholar
  • 106. Batlle D, Soler MJ, Ye M. ACE2 and diabetes: ACE of ACEs? Diabetes 59: 2994–2996, 2010. doi:10.2337/db10-1205.
    Crossref | PubMed | ISI | Google Scholar
  • 107. Al-Benna S. Association of high level gene expression of ACE2 in adipose tissue with mortality of COVID-19 infection in obese patients. Obes Med 19: 100283 , 2020. doi:10.1016/j.obmed.2020.100283.
    Crossref | PubMed | Google Scholar
  • 108. Mori J, Patel VB, Ramprasath T, Alrob OA, DesAulniers J, Scholey JW, Lopaschuk GD, Oudit GY. Angiotensin 1–7 mediates renoprotection against diabetic nephropathy by reducing oxidative stress, inflammation, and lipotoxicity. Am J Physiol Renal Physiol 306: F812–F821, 2014. doi:10.1152/ajprenal.00655.2013.
    Link | ISI | Google Scholar
  • 109. Duan Y, Prasad R, Feng D, Beli E, Li Calzi S, Longhini ALF, Lamendella R, Floyd JL, Dupont M, Noothi SK, Sreejit G, Athmanathan B, Wright J, Jensen AR, Oudit GY, Markel TA, Nagareddy PR, Obukhov AG, Grant MB. Bone marrow-derived cells restore functional integrity of the gut epithelial and vascular barriers in a model of diabetes and ACE2 deficiency. Circ Res 125: 969–988, 2019. doi:10.1161/CIRCRESAHA.119.315743.
    Crossref | PubMed | ISI | Google Scholar
  • 110. Hashimoto T, Perlot T, Rehman A, Trichereau J, Ishiguro H, Paolino M, Sigl V, Hanada T, Hanada R, Lipinski S, Wild B, Camargo SMR, Singer D, Richter A, Kuba K, Fukamizu A, Schreiber S, Clevers H, Verrey F, Rosenstiel P, Penninger JM. ACE2 links amino acid malnutrition to microbial ecology and intestinal inflammation. Nature 487: 477–481, 2012. doi:10.1038/nature11228.
    Crossref | PubMed | ISI | Google Scholar
  • 111. Ye Q, Wang B, Zhang T, Xu J, Shiqiang Shang X. The mechanism and treatment of gastrointestinal symptoms in patients with COVID-19. Am J Physiol Gastrointest Liver Physiol 319: G245–G252, 2020. doi:10.1152/ajpgi.00148.2020.
    Link | ISI | Google Scholar
  • 112. Alenina N, Bader M. ACE2 in brain physiology and pathophysiology: evidence from transgenic animal models. Neurochem Res 44: 1323–1329, 2019. doi:10.1007/s11064-018-2679-4.
    Crossref | PubMed | ISI | Google Scholar
  • 113. Chow N, Fleming-Dutra K, Gierke R, Hall A, Hughes M, Pilishvili T, Ritchey M, Roguski K, Skoff T, Ussery E. Preliminary estimates of the prevalence of selected underlying health conditions among patients with coronavirus disease 2019–United States, February 12–March 28, 2020. MMWR Morb Mortal Wkly Rep 69: 382–386, 2020. doi:10.15585/mmwr.mm6913e2.
    Crossref | PubMed | ISI | Google Scholar
  • 114. Lippi G, Henry BM. Chronic obstructive pulmonary disease is associated with severe coronavirus disease 2019 (COVID-19). Respir Med 167: 105941 , 2020. doi:10.1016/j.rmed.2020.105941.
    Crossref | PubMed | ISI | Google Scholar
  • 115. Chao JY, Derespina KR, Herold BC, Goldman DL, Aldrich M, Weingarten J, Ushay HM, Cabana MD, Medar SS. Clinical characteristics and outcomes of hospitalized and critically ill children and adolescents with coronavirus disease 2019 at a tertiary care medical center in New York City. J Pediatr 223: 14–19.e2, 2020. doi:10.1016/j.jpeds.2020.05.006.
    Crossref | PubMed | ISI | Google Scholar
  • 116. DeBiasi RL, Song X, Delaney M, Bell M, Smith K, Pershad J, Ansusinha E, Hahn A, Hamdy R, Harik N, Hanisch B, Jantausch B, Koay A, Steinhorn R, Newman K, Wessel D. Severe coronavirus disease-2019 in children and young adults in the Washington, DC, metropolitan region. J Pediatr 223: 199–203.e1, 2020 doi:10.1016/j.jpeds.2020.05.007.
    Crossref | PubMed | ISI | Google Scholar
  • 117. Mahdavinia M, Foster KJ, Jauregui E, Moore D, Adnan D, Andy-Nweye AB, Khan S, Bishehsari F. Asthma prolongs intubation in COVID-19. J Allergy Clin Immunol Pract 8: 2388–2391, 2020. doi:10.1016/j.jaip.2020.05.006.
    Crossref | PubMed | ISI | Google Scholar
  • 118. Abrams EM, McGill G, Bhopal SS, Sinha I, Fernandes RM. COVID-19, asthma, and return to school. Lancet Respir Med 8: 847–849, 2020. doi:10.1016/S2213-2600(20)30353-2.
    Crossref | PubMed | ISI | Google Scholar
  • 119. Garg S, Kim L, Whitaker M, O'Halloran A, Cummings C, Holstein R, , et al.. Hospitalization rates and characteristics of patients hospitalized with laboratory-confirmed coronavirus disease 2019–COVID-NET, 14 states, March 1–30, 2020. MMWR Morb Mortal Wkly Rep 69: 458–464, 2020. doi:10.15585/mmwr.mm6915e3.
    Crossref | PubMed | ISI | Google Scholar
  • 120. Guo FR. Active smoking is associated with severity of coronavirus disease 2019 (COVID-19): an update of a meta-analysis. Tob Induc Dis 18: 1653 , 2020. doi:10.18332/tid/121915.
    Crossref | PubMed | ISI | Google Scholar
  • 121. Patanavanich R, Glantz SA. Smoking is associated with COVID-19 progression: a meta-analysis. Nicotine Tob Res 22: 1653–1656, 2020. doi:10.1093/ntr/ntaa082.
    Crossref | PubMed | ISI | Google Scholar
  • 122. World Health Organization. Smoking and COVID-19 (Online). https://www.who.int/news-room/commentaries/detail/smoking-and-covid-19 [9 Aug 2020].
    Google Scholar
  • 123. Zhao Q, Meng M, Kumar R, Wu Y, Huang J, Lian N, Deng Y, Lin S. The impact of COPD and smoking history on the severity of COVID-19: a systemic review and meta-analysis. J Med Virol 92: 1915–1921, 2020. doi:10.1002/jmv.25889.
    Crossref | PubMed | ISI | Google Scholar
  • 124. Zheng Z, Peng F, Xu B, Zhao J, Liu H, Peng J, Li Q, Jiang C, Zhou Y, Liu S, Ye C, Zhang P, Xing Y, Guo H, Tang W. Risk factors of critical & mortal COVID-19 cases: a systematic literature review and meta-analysis. J Infect 81: e16–e25, 2020. doi:10.1016/j.jinf.2020.04.021.
    Crossref | PubMed | ISI | Google Scholar
  • 125. Polak SB, Van Gool IC, Cohen D, von der Thüsen JH, van Paassen J. A systematic review of pathological findings in COVID-19: a pathophysiological timeline and possible mechanisms of disease progression. Mod Pathol 33: 2128–2138, 2020. doi:10.1038/s41379-020-0603-3.
    Crossref | PubMed | ISI | Google Scholar
  • 126. He L, Ding Y, Zhang Q, Che X, He Y, Shen H, Wang H, Li Z, Zhao L, Geng J, Deng Y, Yang L, Li J, Cai J, Qiu L, Wen K, Xu X, Jiang S. Expression of elevated levels of pro-inflammatory cytokines in SARS-CoV-infected ACE2+ cells in SARS patients: relation to the acute lung injury and pathogenesis of SARS. J Pathol 210: 288–297, 2006. doi:10.1002/path.2067.
    Crossref | PubMed | ISI | Google Scholar
  • 127. Higham A, Singh D. Increased ACE2 expression in bronchial epithelium of COPD patients who are overweight. Obesity 28: 1586–1589, 2020. doi:10.1002/oby.22907.
    Crossref | PubMed | ISI | Google Scholar
  • 128. Jacobs M, Van Eeckhoutte HP, Wijnant SR, Janssens W, Joos GF, Brusselle GG, Bracke KR. Increased expression of ACE2, the SARS-CoV-2 entry receptor, in alveolar and bronchial epithelium of smokers and COPD subjects. Eur Respir J 56: 2002378 , 2020. doi:10.1183/13993003.02378-2020.
    Crossref | PubMed | ISI | Google Scholar
  • 129. Leung JM, Yang CX, Tam A, Shaipanich T, Hackett TL, Singhera GK, Dorscheid DR, Sin DD. ACE-2 expression in the small airway epithelia of smokers and COPD patients: implications for COVID-19. Eur Respir J 55: 2000688 , 2020. doi:10.1183/13993003.00688-2020.
    Crossref | PubMed | ISI | Google Scholar
  • 130. Li G, He X, Zhang L, Ran Q, Wang J, Xiong A, Wu D, Chen F, Sun J, Chang C. Assessing ACE2 expression patterns in lung tissues in the pathogenesis of COVID-19. J Autoimmun 112: 102463 , 2020. doi:10.1016/j.jaut.2020.102463.
    Crossref | PubMed | ISI | Google Scholar
  • 131. Pinto BG, Oliveira AE, Singh Y, Jimenez L, Gonçalves AN, Ogava RL, Creighton R, Schatzmann Peron JP, Nakaya HI. ACE2 expression is increased in the lungs of patients with comorbidities associated with severe COVID-19. J Infect Dis 222: 556–563, 2020. doi:10.1093/infdis/jiaa332.
    Crossref | PubMed | ISI | Google Scholar
  • 132. Radzikowska U, Ding M, Tan G, Zhakparov D, Peng Y, Wawrzyniak P, Wang M, Li S, Morita H, Altunbulakli C, Reiger M, Neumann AU, Lunjani N, Traidl‐Hoffmann C, Nadeau KC, O’Mahony L, Akdis C, Sokolowska M. Distribution of ACE2, CD147, CD26, and other SARS‐CoV‐2 associated molecules in tissues and immune cells in health and in asthma, COPD, obesity, hypertension, and COVID‐19 risk factors. Allergy 75: 2829–2845, 2020. doi:10.1111/all.14429.
    Crossref | PubMed | ISI | Google Scholar
  • 133. Saheb Sharif-Askari N, Saheb Sharif-Askari F, Alabed M, Temsah MH, Al Heialy S, Hamid Q, Halwani R. Airways expression of SARS-CoV-2 receptor, ACE2, and TMPRSS2 is lower in children than adults and increases with smoking and COPD. Mol Ther Methods Clin Dev 18: 1–6, 2020. doi:10.1016/j.omtm.2020.05.013.
    Crossref | PubMed | ISI | Google Scholar
  • 134. Song J, Zeng M, Wang H, Qin C, Hou H, Sun Z, Xu S, Wang G, Guo C, Deng Y, Wang Z, Ma J, Pan L, Liao B, Du Z, Feng Q, Liu Y, Xie J, Liu Z. Distinct effects of asthma and COPD comorbidity on disease expression and outcome in patients with COVID‐19. Allergy. In press. doi:10.1111/all.14517.
    Crossref | PubMed | ISI | Google Scholar
  • 135. Toru Ü, Ayada C, Genç O, Sahin S, Arik Ö, Bulut I. Serum levels of RAAS components in COPD. In: European Respiratory Journal. Lausanne, Switzerland: European Respiratory Society, 2020, p. PA3970.
    Google Scholar
  • 136. Roland M, Bhowmik A, Sapsford RJ, Seemungal TA, Jeffries DJ, Warner TD, Wedzicha JA. Sputum and plasma endothelin-1 levels in exacerbations of chronic obstructive pulmonary disease. Thorax 56: 30–35, 2001. doi:10.1136/thorax.56.1.30.
    Crossref | PubMed | ISI | Google Scholar
  • 137. Zhang H, Li Y, Zeng Y, Wu R, Ou J. Endothelin-1 downregulates angiotensin-converting enzyme-2 expression in human bronchial epithelial cells. Pharmacology 91: 297–304, 2013. doi:10.1159/000350395.
    Crossref | PubMed | ISI | Google Scholar
  • 138. Barnes PJ. Unexpected failure of anti-tumor necrosis factor therapy in chronic obstructive pulmonary disease. Am J Respir Crit Care Med 175: 866–867, 2007. doi:10.1164/rccm.200702-253ED.
    Crossref | PubMed | ISI | Google Scholar
  • 139. Finney L, Glanville N, Farne H, Aniscenko J, Fenwick P, Kemp S, Trujillo-Torralbo M-B, Calderazzo MA, Wedzicha J, Mallia P, Bartlett N, Johnston S, Singanayagam A. Inhaled corticosteroids downregulate the SARS-CoV-2 receptor ACE2 in COPD through suppression of type I interferon. J Allergy Clin Immunol. In press. doi:10.1016/j.jaci.2020.09.034.
    Crossref | PubMed | ISI | Google Scholar
  • 140. Camiolo MJ, Gauthier M, Kaminski N, Ray A, Wenzel SE. Expression of SARS-CoV-2 receptor ACE2 and coincident host response signature varies by asthma inflammatory phenotype. J Allergy Clin Immunol 146: 315–324.e7, 2020. doi:10.1016/j.jaci.2020.05.051.
    Crossref | PubMed | ISI | Google Scholar
  • 141. Jackson DJ, Busse WW, Bacharier LB, Kattan M, O’Connor GT, Wood RA, Visness CM, Durham SR, Larson D, Esnault S, Ober C, Gergen PJ, Becker P, Togias A, Gern JE, Altman MC. Association of respiratory allergy, asthma, and expression of the SARS-CoV-2 receptor ACE2. J Allergy Clin Immunol 146: 203–206.e3, 2020. doi:10.1016/j.jaci.2020.04.009.
    Crossref | PubMed | ISI | Google Scholar
  • 142. Clarke NE, Belyaev ND, Lambert DW, Turner AJ. Epigenetic regulation of angiotensin-converting enzyme 2 (ACE2) by SIRT1 under conditions of cell energy stress. Clin Sci 126: 507–516, 2014. doi:10.1042/CS20130291.
    Crossref | PubMed | ISI | Google Scholar
  • 143. Joshi S, Wollenzien H, Leclerc E, Jarajapu YP. Hypoxic regulation of angiotensin‐converting enzyme 2 and Mas receptor in human CD34 + cells. J Cell Physiol 234: 20420–20431, 2019. doi:10.1002/jcp.28643.
    Crossref | PubMed | ISI | Google Scholar
  • 144. Brake SJ, Barnsley K, Lu W, McAlinden KD, Eapen MS, Sohal SS. Smoking upregulates angiotensin-converting enzyme-2 receptor: a potential adhesion site for novel coronavirus SARS-CoV-2 (Covid-19). J Clin Med 9: 841 , 2020. doi:10.3390/jcm9030841.
    Crossref | PubMed | ISI | Google Scholar
  • 145. Williamson EJ, Walker AJ, Bhaskaran K, Bacon S, Bates C, Morton CE, Curtis HJ, Mehrkar A, Evans D, Inglesby P, Cockburn J, McDonald HI, MacKenna B, Tomlinson L, Douglas IJ, Rentsch CT, Mathur R, Wong AY, Grieve R, Harrison D, Forbes H, Schultze A, Croker R, Parry J, Hester F, Harper S, Perera R, Evans SJ, Smeeth L, Goldacre B. Factors associated with COVID-19-related death using OpenSAFELY. Nature 584: 430–436, 2020. doi:10.1038/s41586-020-2521-4.
    Crossref | PubMed | ISI | Google Scholar
  • 146. Mohamed MO, Gale CP, Kontopantelis E, Doran T, de Belder M, Asaria M, Luscher T, Wu J, Rashid M, Stephenson C, Denwood T, Roebuck C, Deanfield J, Mamas MA. Sex-differences in mortality rates and underlying conditions for COVID-19 deaths in England and Wales. Mayo Clin Proc 95: 2110–2124, 2020. doi:10.1016/j.mayocp.2020.07.009.
    Crossref | PubMed | ISI | Google Scholar
  • 147. Tavazzi G, Pellegrini C, Maurelli M, Belliato M, Sciutti F, Bottazzi A, Sepe PA, Resasco T, Camporotondo R, Bruno R, Baldanti F, Paolucci S, Pelenghi S, Iotti GA, Mojoli F, Arbustini E. Myocardial localization of coronavirus in COVID-19 cardiogenic shock. Eur J Heart Fail 22: 911–915, 2020. doi:10.1002/ejhf.1828.
    Crossref | PubMed | ISI | Google Scholar
  • 148. Wichmann D, Sperhake JP, Lütgehetmann M, Steurer S, Edler C, Heinemann A, Heinrich F, Mushumba H, Kniep I, Schröder AS, Burdelski C, de Heer G, Nierhaus A, Frings D, Pfefferle S, Becker H, Bredereke-Wiedling H, de Weerth A, Paschen H-R, Sheikhzadeh-Eggers S, Stang A, Schmiedel S, Bokemeyer C, Addo MM, Aepfelbacher M, Püschel K, Kluge S. Autopsy findings and venous thromboembolism in patients with COVID-19. Ann Intern Med 173: 268–277, 2020. doi:10.7326/M20-2003.
    Crossref | PubMed | ISI | Google Scholar
  • 149. Sala S, Peretto G, Gramegna M, Palmisano A, Villatore A, Vignale D, De Cobelli F, Tresoldi M, Cappelletti AM, Basso C, Godino C, Esposito A. Acute myocarditis presenting as a reverse Tako-Tsubo syndrome in a patient with SARS-CoV-2 respiratory infection. Eur Heart J 41: 1861–1862, 2020. doi:10.1093/eurheartj/ehaa286.
    Crossref | PubMed | ISI | Google Scholar
  • 150. Ni W, Yang X, Liu J, Bao J, Li R, Xu Y, Guo W, Hu Y, Gao Z. Acute myocardial injury at hospital admission is associated with all-cause mortality in COVID-19. J Am Coll Cardiol 76: 124–125, 2020. doi:10.1016/j.jacc.2020.05.007.
    Crossref | PubMed | ISI | Google Scholar
  • 151. Shi S, Qin M, Shen B, Cai Y, Liu T, Yang F, Gong W, Liu X, Liang J, Zhao Q, Huang H, Yang B, Huang C. Association of cardiac injury with mortality in hospitalized patients with COVID-19 in Wuhan. JAMA Cardiol 5: 802 , 2020. doi:10.1001/jamacardio.2020.0950.
    Crossref | PubMed | ISI | Google Scholar
  • 152. 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 323: 1061–1069, 2020. doi:10.1001/jama.2020.1585.
    Crossref | PubMed | ISI | Google Scholar
  • 153. Zheng YY, Ma YT, Zhang JY, Xie X. COVID-19 and the cardiovascular system. Nat Rev Cardiol 17: 259–260, 2020. doi:10.1038/s41569-020-0360-5.
    Crossref | PubMed | ISI | Google Scholar
  • 154. Goyal P, Choi JJ, Pinheiro LC, Schenck EJ, Chen R, Jabri A, Satlin MJ, Campion TR, Nahid M, Ringel JB, Hoffman KL, Alshak MN, Li HA, Wehmeyer GT, Rajan M, Reshetnyak E, Hupert N, Horn EM, Martinez FJ, Gulick RM, Safford MM. Clinical characteristics of COVID-19 in New York City. N Engl J Med 382: 2372–2374, 2020. doi:10.1056/NEJMc2010419.
    Crossref | PubMed | ISI | Google Scholar
  • 155. Dweck MR, Bularga A, Hahn RT, Bing R, Lee KK, Chapman AR, White A, Salvo GD, Sade LE, Pearce K, Newby DE, Popescu BA, Donal E, Cosyns B, Edvardsen T, Mills NL, Haugaa K. Global evaluation of echocardiography in patients with COVID-19. Eur Heart J Cardiovasc Imaging 21: 949–958, 2020. doi:10.1093/ehjci/jeaa178.
    Crossref | PubMed | ISI | Google Scholar
  • 156. Puntmann VO, Carerj ML, Wieters I, Fahim M, Arendt C, Hoffmann J, Shchendrygina A, Escher F, Vasa-Nicotera M, Zeiher AM, Vehreschild M, Nagel E. Outcomes of cardiovascular magnetic resonance imaging in patients recently recovered from coronavirus disease 2019 (COVID-19). JAMA Cardiol 5: 1265 , 2020. doi:10.1001/jamacardio.2020.3557.
    Crossref | PubMed | ISI | Google Scholar
  • 157. Burrell LM, Risvanis J, Kubota E, Dean RG, MacDonald PS, Lu S, Tikellis C, Grant SL, Lew RA, Smith AI, Cooper ME, Johnston CI. Myocardial infarction increases ACE2 expression in rat and humans. Eur Heart J 26: 369–375, 2005. doi:10.1093/eurheartj/ehi114.
    Crossref | PubMed | ISI | Google Scholar
  • 158. Zulli A, Burrell LM, Widdop RE, Black MJ, Buxton BF, Hare DL. Immunolocalization of ACE2 and AT2 receptors in rabbit atherosclerotic plaques. J Histochem Cytochem 54: 147–150, 2006. doi:10.1369/jhc.5C6782.2005.
    Crossref | PubMed | ISI | Google Scholar
  • 159. Bos JM, Hebl VB, Oberg AL, Sun Z, Herman DS, Teekakirikul P, Seidman JG, Seidman CE, dos Remedios CG, Maleszewski JJ, Schaff HV, Dearani JA, Noseworthy PA, Friedman PA, Ommen SR, Brozovich FV, Ackerman MJ. Marked up-regulation of ace2 in hearts of patients with obstructive hypertrophic cardiomyopathy: implications for SARS-CoV-2-mediated COVID-19. Mayo Clin Proc 95: 1354–1368, 2020. doi:10.1016/j.mayocp.2020.04.028.
    Crossref | PubMed | ISI | Google Scholar
  • 160. Liu PP, Blet A, Smyth D, Li H. The science underlying COVID-19: implications for the cardiovascular system. Circulation 142: 68–78, 2020. doi:10.1161/CIRCULATIONAHA.120.047549.
    Crossref | PubMed | ISI | Google Scholar
  • 161. Walters TE, Kalman JM, Patel SK, Mearns M, Velkoska E, Burrell LM. 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 19: 1280–1287, 2016.
    PubMed | ISI | Google Scholar
  • 162. 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 13: e0198144 , 2018. doi:10.1371/journal.pone.0198144.
    Crossref | PubMed | ISI | Google Scholar
  • 163. 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 116: 1097–1100, 2020. [Erratum in Cardiovasc Res 116: 1994]. doi:10.1093/cvr/cvaa078.
    Crossref | PubMed | ISI | Google Scholar
  • 164. Sama IE, Ravera A, Santema BT, van Goor H, ter Maaten JM, Cleland JG, Rienstra M, Friedrich AW, Samani NJ, Ng LL, Dickstein K, Lang CC, Filippatos G, Anker SD, Ponikowski P, Metra M, van Veldhuisen DJ, Voors AA. Circulating plasma concentrations of angiotensin-converting enzyme 2 in men and women with heart failure and effects of renin-angiotensin-aldosterone inhibitors. Eur Heart J 41: 1810–1817, 2020. doi:10.1093/eurheartj/ehaa373.
    Crossref | PubMed | ISI | Google Scholar
  • 165. Basu R, Poglitsch M, Yogasundaram H, Thomas J, Rowe BH, Oudit GY. Roles of angiotensin peptides and recombinant human ACE2 in heart failure. J Am Coll Cardiol 69: 805–819, 2017. doi:10.1016/S0735-1097(17)34194-3, 10.1016/j.jacc.2016.11.064.
    Crossref | PubMed | ISI | Google Scholar
  • 166. Epelman S, Tang WH, Chen SY, Van Lente F, Francis GS, Sen S. Detection of soluble angiotensin-converting enzyme 2 in heart failure. insights into the endogenous counter-regulatory pathway of the renin-angiotensin-aldosterone system. J Am Coll Cardiol 52: 750–754, 2008. doi:10.1016/j.jacc.2008.02.088.
    Crossref | PubMed | ISI | Google Scholar
  • 167. Uri K, Fagyas M, Kertesz A, Borbely A, Jenei C, Bene O, , et al.. Circulating ACE2 activity correlates with cardiovascular disease development. J Renin-Angiotensin-Aldosterone Syst 17: 1470320316668435 , 2016. doi:10.1177/1470320316668435.
    Crossref | PubMed | ISI | Google Scholar
  • 168. Zhong J, Basu R, Guo D, Chow FL, Byrns S, Schuster M, Loibner H, Wang XH, Penninger JM, Kassiri Z, Oudit GY. Angiotensin-converting enzyme 2 suppresses pathological hypertrophy, myocardial fibrosis, and cardiac dysfunction. Circulation 122: 717–728, 2010. doi:10.1161/CIRCULATIONAHA.110.955369.
    Crossref | PubMed | ISI | Google Scholar
  • 169. Pranata R, Lim MA, Huang I, Raharjo SB, Lukito AA. Hypertension is associated with increased mortality and severity of disease in COVID-19 pneumonia: A systematic review, meta-analysis and meta-regression. J Renin-Angiotensin-Aldosterone Syst 21: 1470320320926899 , 2020. doi:10.1177/1470320320926899.
    Crossref | PubMed | ISI | Google Scholar
  • 170. Fang L, Karakiulakis G, Roth M. Are patients with hypertension and diabetes mellitus at increased risk for COVID-19 infection? Lancet Respir Med 8: e21 , 2020. doi:10.1016/S2213-2600(20)30116-8.
    Crossref | PubMed | ISI | Google Scholar
  • 171. Nehme A, Zouein FA, Zayeri ZD, Zibara K. An update on the tissue renin angiotensin system and its role in physiology and pathology. J Cardiovasc Dev Dis 6: 14 , 2019. doi:10.3390/jcdd6020014.
    Crossref | PubMed | ISI | Google Scholar
  • 172. Sevá Pessôa B, van der Lubbe N, Verdonk K, Roks AJ, Hoorn EJ, Danser AH. Key developments in renin-angiotensin-aldosterone system inhibition. Nat Rev Nephrol 9: 26–36, 2013. doi:10.1038/nrneph.2012.249.
    Crossref | PubMed | ISI | Google Scholar
  • 173. Silva-Aguiar RP, Peruchetti DB, Rocco PR, Schmaier AH, Silva PM, Martins MA, Carvalho VF, Pinheiro AA, Caruso-Neves C. Role of the renin-angiotensin system in the development of severe COVID-19 in hypertensive patients. Am J Physiol Lung Cell Mol Physiol 319: L596–L602, 2020. doi:10.1152/ajplung.00286.2020.
    Link | ISI | Google Scholar
  • 174. Genest J, Boucher R, Kuchel O, Nowaczynski W. Renin in hypertension: how important as a risk factor? Can Med Assoc J 109: 475–478, 1973.
    PubMed | ISI | Google Scholar
  • 175. Verma S, Gupta M, Holmes DT, Xu L, Teoh H, Gupta S, Yusuf S, Lonn EM. Plasma renin activity predicts cardiovascular mortality in the Heart Outcomes Prevention Evaluation (HOPE) study. Eur Heart J 32: 2135–2142, 2011. doi:10.1093/eurheartj/ehr066.
    Crossref | PubMed | ISI | Google Scholar
  • 176. Chappell MC. Does ACE2 contribute to the development of hypertension? Hypertens Res 33: 107–109, 2010. doi:10.1038/hr.2009.207.
    Crossref | PubMed | ISI | Google Scholar
  • 177. Koka V, Xiao RH, Chung ACK, Wang W, Truong LD, Lan HY. Angiotensin II up-regulates angiotensin i-converting enzyme (ACE), but down-regulates ACE2 via the AT1-ERK/p38 MAP kinase pathway. Am J Pathol 172: 1174–1183, 2008. doi:10.2353/ajpath.2008.070762.
    Crossref | PubMed | ISI | Google Scholar
  • 178. Patel SK, Velkoska E, Freeman M, Wai B, Lancefield TF, Burrell LM. From gene to protein experimental and clinical studies of ACE2 in blood pressure control and arterial hypertension. Front Physiol 5: 227 , 2014. doi:10.3389/fphys.2014.00227.
    Crossref | PubMed | ISI | Google Scholar
  • 179. Prieto MC, González-Villalobos RA, Botros FT, Martin VL, Pagán J, Satou R, Lara LS, Feng Y, Fernandes FB, Kobori H, Casarini DE, Navar GG. Reciprocal changes in renal ACE/ANG II and ACE2/ANG 1-7 are associated with enhanced collecting duct renin in goldblatt hypertensive rats. Am J Physiol Renal Physiol 300: F749–F755, 2011. doi:10.1152/ajprenal.00383.2009.
    Link | ISI | Google Scholar
  • 180. Tikellis C, Cooper ME, Bialkowski K, Johnston CI, Burns WC, Lew RA, Smith AI, Thomas MC. Developmental expression of ACE2 in the SHR kidney: a role in hypertension? Kidney Int 70: 34–41, 2006. doi:10.1038/sj.ki.5000428.
    Crossref | PubMed | ISI | Google Scholar
  • 181. Li S, Wang Z, Yang X, Hu B, Huang Y, Fan S. Association between circulating angiotensin-converting enzyme 2 and cardiac remodeling in hypertensive patients. Peptides 90: 63–68, 2017. doi:10.1016/j.peptides.2017.02.007.
    Crossref | PubMed | ISI | Google Scholar
  • 182. Rice GI, Jones AL, Grant PJ, Carter AM, Turner AJ, Hooper NM. Circulating activities of angiotensin-converting enzyme, its homolog, angiotensin-converting enzyme 2, and neprilysin in a family study. Hypertension 48: 914–920, 2006. doi:10.1161/01.HYP.0000244543.91937.79.
    Crossref | PubMed | ISI | Google Scholar
  • 183. Roberts MA, Velkoska E, Ierino FL, Burrell LM. Angiotensin-converting enzyme 2 activity in patients with chronic kidney disease. Nephrol Dial Transplant 28: 2287–2294, 2013. doi:10.1093/ndt/gft038.
    Crossref | PubMed | ISI | Google Scholar
  • 184. Soro-Paavonen A, Gordin D, Forsblom C, Rosengard-Barlund M, Waden J, Thorn L, Sandholm N, Thomas MC, Groop PH. Circulating ACE2 activity is increased in patients with type 1 diabetes and vascular complications. J Hypertens 30: 375–383, 2012. doi:10.1097/HJH.0b013e32834f04b6.
    Crossref | PubMed | ISI | Google Scholar
  • 185. Úri K, Fagyas M, Mányiné Siket I, Kertész A, Csanádi Z, Sándorfi G, Clemens M, Fedor R, Papp Z, Édes I, Tóth A, Lizanecz E. New perspectives in the renin-angiotensin-aldosterone system (RAAS) IV: circulating ACE2 as a biomarker of systolic dysfunction in human hypertension and heart failure. PLoS One 9: e87845 , 2014. doi:10.1371/journal.pone.0087845.
    Crossref | PubMed | ISI | Google Scholar
  • 186. Xu J, Sriramula S, Xia H, Moreno-Walton L, Culicchia F, Domenig O, Poglitsch M, Lazartigues E. Clinical relevance and role of neuronal AT1 receptors in ADAM17-mediated ACE2 shedding in neurogenic hypertension. Circ Res 121: 43–55, 2017. doi:10.1161/CIRCRESAHA.116.310509.
    Crossref | PubMed | ISI | Google Scholar
  • 187. Yamaleyeva LM, Gilliam-Davis S, Almeida I, Bridget Brosnihan K, Lindsey SH, Chappell MC. Differential regulation of circulating and renal ACE2 and ACE in hypertensive mRen2.Lewis rats with early-onset diabetes. Am J Physiol Renal Physiol 302: F1374–F1384, 2012. doi:10.1152/ajprenal.00656.2011.
    Link | ISI | Google Scholar
  • 188. Yang M, Zhao J, Xing L, Shi L. The association between angiotensinconverting enzyme 2 polymorphisms and essential hypertension risk: A metaanalysis involving 14,122 patients. J Renin Angiotensin Aldosterone Syst 16: 1240–1244, 2015. doi:10.1177/1470320314549221.
    Crossref | PubMed | ISI | Google Scholar
  • 189. Choi HS, Kim IJ, Kim CS, Ma SK, Scholey JW, Kim SW, Bae EH. Angiotensin-[1–7] attenuates kidney injury in experimental Alport syndrome. Sci Rep 10: 4225 , 2020. doi:10.1038/s41598-020-61250-5.
    Crossref | PubMed | ISI | Google Scholar
  • 190. Goru SK, Kadakol A, Malek V, Pandey A, Sharma N, Gaikwad AB. Diminazene aceturate prevents nephropathy by increasing glomerular ACE2 and AT2 receptor expression in a rat model of type1 diabetes. Br J Pharmacol 174: 3118–3130, 2017. doi:10.1111/bph.13946.
    Crossref | PubMed | ISI | Google Scholar
  • 191. Husková Z, Kopkan L, Červenková L, Doleželová Š, Vaňourková Z, Škaroupková P, Nishiyama A, Kompanowska-Jezierska E, Sadowski J, Kramer HJ, Červenka L. Intrarenal alterations of the angiotensin-converting enzyme type 2/angiotensin 1-7 complex of the renin-angiotensin system do not alter the course of malignant hypertension in Cyp1a1-Ren-2 transgenic rats. Clin Exp Pharmacol Physiol 43: 438–449, 2016. doi:10.1111/1440-1681.12553.
    Crossref | PubMed | ISI | Google Scholar
  • 192. Wysocki J, Ye M, Khattab AM, Fogo A, Martin A, David NV, Kanwar Y, Osborn M, Batlle D. Angiotensin-converting enzyme 2 amplification limited to the circulation does not protect mice from development of diabetic nephropathy. Kidney Int 91: 1336–1346, 2017. doi:10.1016/j.kint.2016.09.032.
    Crossref | PubMed | ISI | Google Scholar
  • 193. Keidar S, Strizevsky A, Raz A, Gamliel-Lazarovich A. ACE2 activity is increased in monocyte-derived macrophages from prehypertensive subjects. Nephrol Dial Transplant 22: 597–601, 2006. doi:10.1093/ndt/gfl632.
    Crossref | PubMed | ISI | Google Scholar
  • 194. Ortiz-Pérez JT, Riera M, Bosch X, De Caralt TM, Perea RJ, Pascual J, Soler MJ. Role of circulating angiotensin converting enzyme 2 in left ventricular remodeling following myocardial infarction: a prospective controlled study. PLoS One 8: e61695 , 2013. doi:10.1371/journal.pone.0061695.
    Crossref | PubMed | ISI | Google Scholar
  • 195. Feng Y, Xia H, Cai Y, Halabi CM, Becker LK, Santos RA, Speth RC, Sigmund CD, Lazartigues E. Brain-selective overexpression of human angiotensin-converting enzyme type 2 attenuates neurogenic hypertension. Circ Res 106: 373–382, 2010. doi:10.1161/CIRCRESAHA.109.208645.
    Crossref | PubMed | ISI | Google Scholar
  • 196. Myers LC, Parodi SM, Escobar GJ, Liu VX. Characteristics of hospitalized adults with COVID-19 in an integrated health care system in California. JAMA 323: 2195–2198, 2020. doi:10.1001/jama.2020.7202.
    Crossref | PubMed | ISI | Google Scholar
  • 197. Ssentongo P, Ssentongo AE, Heilbrunn ES, Ba DM, Chinchilli VM. Association of cardiovascular disease and 10 other preexisting comorbidities with COVID-19 mortality: a systematic review and meta-analysis. PLoS One 15: e0238215 , 2020. doi:10.1371/journal.pone.0238215.
    Crossref | PubMed | ISI | Google Scholar
  • 198. Velez JC, Caza T, Larsen CP. COVAN is the new HIVAN: the re-emergence of collapsing glomerulopathy with COVID-19. Nat Rev Nephrol 16: 565–567, 2020 [Erratum in Nat Rev Nephrol 16: 614, 2020]. dois:10.1038/s41584-020-0480-7, 10.1038/s41581-020-0332-3.
    Crossref | PubMed | ISI | Google Scholar
  • 199. Diao B, Feng Z, Wang C, Wang H, Liu L, Wang C, Wang R, Liu Y, Liu Y, Wang G, Yuan Z, Wu Y, Chen Y. Human kidney is a target for novel severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection (Preprint). medRxiv 2020.03.04.203311202, 2020. doi:10.1101/2020.03.04.20031120.
    Crossref | Google Scholar
  • 200. Cheng Y, Luo R, Wang K, Zhang M, Wang Z, Dong L, Li J, Yao Y, Ge S, Xu G. Kidney disease is associated with in-hospital death of patients with COVID-19. Kidney Int 97: 829–838, 2020. doi:10.1016/j.kint.2020.03.005.
    Crossref | PubMed | ISI | Google Scholar
  • 201. Eräranta A, Riutta A, Fan M, Koskela J, Tikkanen I, Lakkisto P, Niemelä O, Parkkinen J, Mustonen J, Pörsti I. Dietary phosphate binding and loading alter kidney angiotensin-converting enzyme mRNA and protein content in 5/6 nephrectomized rats. Am J Nephrol 35: 401–408, 2012. doi:10.1159/000337942.
    Crossref | PubMed | ISI | Google Scholar
  • 202. Mizuiri S, Hemmi H, Arita M, Aoki T, Ohashi Y, Miyagi M, Sakai K, Shibuya K, Hase H, Aikawa A. Increased ACE and decreased ACE2 expression in kidneys from patients with IgA nephropathy. Nephron Clin Pract 117: c57–66, 2010. doi:10.1159/000319648.
    Crossref | PubMed | Google Scholar
  • 203. Mizuiri S, Hemmi H, Arita M, Ohashi Y, Tanaka Y, Miyagi M, Sakai K, Ishikawa Y, Shibuya K, Hase H, Aikawa A. Expression of ACE and ACE2 in individuals with diabetic kidney disease and healthy controls. Am J Kidney Dis 51: 613–623, 2008. doi:10.1053/j.ajkd.2007.11.022.
    Crossref | PubMed | ISI | Google Scholar
  • 204. Reich HN, Oudit GY, Penninger JM, Scholey JW, Herzenberg AM. Decreased glomerular and tubular expression of ACE2 in patients with type 2 diabetes and kidney disease. Kidney Int 74: 1610–1616, 2008. doi:10.1038/ki.2008.497.
    Crossref | PubMed | ISI | Google Scholar
  • 205. Velskoska E, Dean RG, Burchill L, Levidiotis V, Burrell LM. Reduction in renal ACE2 expression in subtotal nephrectomy in rats is ameliorated with ACE inhibition. Clin Sci 118: 269–279, 2010. doi:10.1042/CS20090318.
    Crossref | ISI | Google Scholar
  • 206. Wang G, Lai FM, Kwan BC, Lai KB, Chow KM, Li PK, Szeto CC. Expression of ACE and ACE2 in patients with hypertensive nephrosclerosis. Kidney Blood Press Res 34: 141–149, 2011. doi:10.1159/000324521.
    Crossref | PubMed | ISI | Google Scholar
  • 207. Chodavarapu H, Grobe N, Somineni HK, Salem ES, Madhu M, Elased KM. Rosiglitazone treatment of type 2 diabetic db/db mice attenuates urinary albumin and angiotensin converting enzyme 2 excretion. PLoS One 8: e62833 , 2013. doi:10.1371/journal.pone.0062833.
    Crossref | PubMed | ISI | Google Scholar
  • 208. Moon JY, Jeong KH, Lee SH, Lee TW, Ihm CG, Lim SJ. Renal ACE and ACE2 expression in early diabetic rats. Nephron Exp Nephrol 110: e8–e16, 2008. doi:10.1159/000149586.
    Crossref | PubMed | Google Scholar
  • 209. Ye M, Wysocki J, Naaz P, Salabat MR, LaPointe MS, Batlle D. Increased ACE 2 and decreased ACE protein in renal tubules from diabetic mice: a renoprotective combination? Hypertension 43: 1120–1125, 2004. doi:10.1161/01.HYP.0000126192.27644.76.
    Crossref | PubMed | ISI | Google Scholar
  • 210. Ye M, Wysocki J, William J, Soler MJ, Cokic I, Batlle D. Glomerular localization and expression of angiotensin-converting enzyme 2 and angiotensin-converting enzyme: Implications for albuminuria in diabetes. J Am Soc Nephrol 17: 3067–3075, 2006. doi:10.1681/ASN.2006050423.
    Crossref | PubMed | ISI | Google Scholar
  • 211. Tikellis C, Johnston CI, Forbes JM, Burns WC, Burrell LM, Risvanis J, Cooper ME. Characterization of renal angiotensin-converting enzyme 2 in diabetic nephropathy. Hypertension 41: 392–397, 2003. doi:10.1161/01.HYP.0000060689.38912.CB.
    Crossref | PubMed | ISI | Google Scholar
  • 212. Anguiano L, Riera M, Pascual J, Valdivielso JM, Barrios C, Betriu A, Clotet S, Mojal S, Fernández E, Soler MJ. Circulating angiotensin converting enzyme 2 activity as a biomarker of silent atherosclerosis in patients with chronic kidney disease. Atherosclerosis 253: 135–143, 2016. doi:10.1016/j.atherosclerosis.2016.08.032.
    Crossref | PubMed | ISI | Google Scholar
  • 213. Anguiano L, Riera M, Pascual J, Valdivielso JM, Barrios C, Betriu A, , et al.. Circulating angiotensin-converting enzyme 2 activity in patients with chronic kidney disease without previous history of cardiovascular disease. Nephrol Dial Transplant 30: 1176–1185, 2015. doi:10.1093/ndt/gfv025.
    Crossref | PubMed | ISI | Google Scholar
  • 214. Burchill L, Velkoska E, Dean RG, Lew RA, Smith AI, Levidiotis V, Burrell LM. Acute kidney injury in the rat causes cardiac remodelling and increases angiotensin-converting enzyme 2 expression. Exp Physiol 93: 622–630, 2008. doi:10.1113/expphysiol.2007.040386.
    Crossref | PubMed | ISI | Google Scholar
  • 215. Riera M, Anguiano L, Clotet S, Roca-Ho H, Rebull M, Pascual J, Soler MJ. Paricalcitol modulates ACE2 shedding and renal ADAM17 in NOD mice beyond proteinuria. Am J Physiol Renal Physiol 310: F534–F546, 2016. doi:10.1152/ajprenal.00082.2015.
    Link | ISI | Google Scholar
  • 216. Riera M, Márquez E, Clotet S, Gimeno J, Roca-Ho H, Lloreta J, Juanpere N, Batlle D, Pascual J, Soler MJ. Effect of insulin on ACE2 activity and kidney function in the non-obese diabetic mouse. PLoS One 9: e84683 , 2014 [Erratum in PLoS One 9: 1, 2014]. doi:10.1371/journal.pone.0084683.
    Crossref | PubMed | ISI | Google Scholar
  • 217. Soler MJ, Riera M, Crespo M, Mir M, Márquez E, Pascual MJ, Puig JM, Pascual J. Circulating angiotensin-converting enzyme 2 activity in kidney transplantation: a longitudinal pilot study. Nephron 121: c144–c150, 2012. doi:10.1159/000345508.
    Crossref | PubMed | ISI | Google Scholar
  • 218. Barron E, Bakhai C, Kar P, Weaver A, Bradley D, Ismail H, Knighton P, Holman N, Khunti K, Sattar N, Wareham NJ, Young B, Valabhji J. Associations of type 1 and type 2 diabetes with COVID-19-related mortality in England: a whole-population study. Lancet Diabetes Endocrinol 8: 813–822, 2020. doi:10.1016/S2213-8587(20)30272-2.
    Crossref | PubMed | ISI | Google Scholar
  • 219. Chen Y, Yang D, Cheng B, Chen J, Peng A, Yang C, Liu C, Xiong M, Deng A, Zhang Y, Zheng L, Huang K. Clinical characteristics and outcomes of patients with diabetes and covid-19 in association with glucose-lowering medication. Diabetes Care 43: 1399–1407, 2020. doi:10.2337/dc20-0660.
    Crossref | PubMed | ISI | Google Scholar
  • 220. Fadini GP, Morieri ML, Longato E, Avogaro A. Prevalence and impact of diabetes among people infected with SARS-CoV-2. J Endocrinol Invest 43: 867–869, 2020. doi:10.1007/s40618-020-01236-2.
    Crossref | PubMed | ISI | Google Scholar
  • 221. Lighter J, Phillips M, Hochman S, Sterling S, Johnson D, Francois F, Stachel A. Obesity in patients younger than 60 years is a risk factor for COVID-19 hospital admission. Clin Infect Dis 71: 896–897, 2020. doi:10.1093/cid/ciaa415.
    Crossref | PubMed | ISI | Google Scholar
  • 222. Palaiodimos L, Kokkinidis DG, Li W, Karamanis D, Ognibene J, Arora S, Southern WN, Mantzoros CS. Severe obesity is associated with higher in-hospital mortality in a cohort of patients with COVID-19 in the Bronx, New York. Metabolism 108: 154262 , 2020. doi:10.1016/j.metabol.2020.154262.
    Crossref | PubMed | ISI | Google Scholar
  • 223. Holman N, Knighton P, Kar P, O’Keefe J, Curley M, Weaver A, Barron E, Bakhai C, Khunti K, Wareham NJ, Sattar N, Young B, Valabhji J. Risk factors for COVID-19-related mortality in people with type 1 and type 2 diabetes in England: a population-based cohort study. Lancet Diabetes Endocrinol 8: 823–833, 2020. doi:10.1016/S2213-8587(20)30271-0.
    Crossref | PubMed | ISI | Google Scholar
  • 224. Kumar A, Arora A, Sharma P, Anikhindi SA, Bansal N, Singla V, Khare S, Srivastava A. Is diabetes mellitus associated with mortality and severity of COVID-19? A meta-analysis. Diabetes Metab Syndr Clin Res Rev 14: 535–545, 2020. doi:10.1016/j.dsx.2020.04.044.
    Crossref | PubMed | ISI | Google Scholar
  • 225. Rubino F, Amiel SA, Zimmet P, Alberti G, Bornstein S, Eckel RH, Mingrone G, Boehm B, Cooper ME, Chai Z, Del Prato S, Ji L, Hopkins D, Herman WH, Khunti K, Mbanya JC, Renard E. New-onset diabetes in Covid-19. N Engl J Med 383: 789–790, 2020. doi:10.1056/NEJMc2018688.
    Crossref | PubMed | ISI | Google Scholar
  • 226. Harmer D, Gilbert M, Borman R, Clark KL. Quantitative mRNA expression profiling of ACE 2, a novel homologue of angiotensin converting enzyme. FEBS Lett 532: 107–110, 2002. doi:10.1016/S0014-5793(02)03640-2.
    Crossref | PubMed | ISI | Google Scholar
  • 227. Liu F, Long X, Zhang B, Zhang W, Chen X, Zhang Z. ACE2 Expression in pancreas may cause pancreatic damage after SARS-CoV-2 infection. Clin Gastroenterol Hepatol 18: 2128–2130.e2, 2020. doi:10.1016/j.cgh.2020.04.040.
    Crossref | PubMed | ISI | Google Scholar
  • 228. Yang JK, Lin SS, Ji XJ, Guo LM. Binding of SARS coronavirus to its receptor damages islets and causes acute diabetes. Acta Diabetol 47: 193–199, 2010. doi:10.1007/s00592-009-0109-4.
    Crossref | PubMed | ISI | Google Scholar
  • 229. Bindom SM, Hans CP, Xia H, Boulares AH, Lazartigues E. Angiotensin I-converting enzyme type 2 (ACE2) gene therapy improves glycemic control in diabetic mice. Diabetes 59: 2540–2548, 2010. doi:10.2337/db09-0782.
    Crossref | PubMed | ISI | Google Scholar
  • 230. Rao S, Lau A, So HC. Exploring diseases/traits and blood proteins causally related to expression of ACE2, the putative receptor of SARS-CoV-2: a Mendelian randomization analysis highlights tentative relevance of diabetes-related traits. Diabetes Care 43: 1416–1426, 2020. doi:10.2337/dc20-0643.
    Crossref | PubMed | ISI | Google Scholar
  • 231. Emilsson V, Gudmundsson EF, Aspelund T, Jonsson BG, Gudjonsson A, Launer LJ, Lamb JR, Gudmundsdottir V, Jennings LL, Gudnason V. ACE2 levels are altered in comorbidities linked to severe outcome in COVID-19. medRxiv 2020.06.04.20122044 , 2020. doi:10.1101/2020.06.04.20122044.
    Crossref | PubMed | Google Scholar
  • 232. Gutta S, Grobe N, Kumbaji M, Osman H, Saklayen M, Li G, Elased KM. Increased urinary angiotensin converting enzyme 2 and neprilysin in patients with type 2 diabetes. Am J Physiol Renal Physiol 315: F263–F274, 2018. doi:10.1152/ajprenal.00565.2017.
    Link | ISI | Google Scholar
  • 233. Codo AC, Davanzo GG, Monteiro L de B, de Souza GF, Muraro SP, Virgilio-da-Silva JV, , et al.. Elevated glucose levels favor SARS-CoV-2 infection and monocyte response through a HIF-1α/glycolysis-dependent axis. Cell Metab 32: 437–446, 2020. doi:10.1016/j.cmet.2020.07.007.
    Crossref | PubMed | ISI | Google Scholar
  • 234. Popkin BM, Du S, Green WD, Beck MA, Algaith T, Herbst CH, Alsukait RF, Alluhidan M, Alazemi N, Shekar M. Individuals with obesity and COVID‐19: A global perspective on the epidemiology and biological relationships. Obes Rev 21: e13128 , 2020. doi:10.1111/obr.13128.
    Crossref | PubMed | ISI | Google Scholar
  • 235. Frantz ED, Crespo-Mascarenhas C, Barreto-Vianna AR, Aguila MB, Mandarim-de-Lacerda CA. Renin-angiotensin system blockers protect pancreatic islets against diet-induced obesity and insulin resistance in mice. PLoS One 8: e67192 , 2013. doi:10.1371/journal.pone.0067192.
    Crossref | PubMed | ISI | Google Scholar
  • 236. Gupte M, Boustany-Kari CM, Bharadwaj K, Police S, Thatcher S, Gong MC, English VL, Cassis LA. ACE2 is expressed in mouse adipocytes and regulated by a high-fat diet. Am J Physiol Regul Integr Comp Physiol 295: R781–R788, 2008. doi:10.1152/ajpregu.00183.2008.
    Link | ISI | Google Scholar
  • 237. Gupte M, Thatcher SE, Boustany-Kari CM, Shoemaker R, Yiannikouris F, Zhang X, Karounos M, Cassis LA. Angiotensin converting enzyme 2 contributes to sex differences in the development of obesity hypertension in C57BL/6 mice. Arterioscler Thromb Vasc Biol 32: 1392–1399, 2012. doi:10.1161/ATVBAHA.112.248559.
    Crossref | PubMed | ISI | Google Scholar
  • 238. Ridwan R, Natzir R, Rasyid H, Patellongi I, Hatta M, Linggi EB, Bukhari A, Bahrun U. Decreased renal function induced by high-fat diet in Wistar rat: the role of plasma angiotensin converting enzyme 2 (ACE2). Biomed Pharmacol J 12: 1279–1287, 2019. doi:10.13005/bpj/1756.
    Crossref | Google Scholar
  • 239. Shoemaker R, Tannock LR, Su W, Gong M, Gurley SB, Thatcher SE, Yiannikouris F, Ensor CM, Cassis LA. Adipocyte deficiency of ACE2 increases systolic blood pressures of obese female C57BL/6 mice. Biol Sex Differ 10: 45 , 2019. doi:10.1186/s13293-019-0260-8.
    Crossref | PubMed | ISI | Google Scholar
  • 240. Barretti DLM, Magalhães F de C, Fernandes T, do Carmo EC, Rosa KT, Irigoyen MC, Negrão CE, Oliveira EM. Effects of aerobic exercise training on cardiac renin-angiotensin system in an obese zucker rat strain. PLoS One 7: e46114 , 2012. doi:10.1371/journal.pone.0046114.
    Crossref | PubMed | ISI | Google Scholar
  • 241. Glende J, Schwegmann-Wessels C, Al-Falah M, Pfefferle S, Qu X, Deng H, Drosten C, Naim HY, Herrler G. Importance of cholesterol-rich membrane microdomains in the interaction of the S protein of SARS-coronavirus with the cellular receptor angiotensin-converting enzyme 2. Virology 381: 215–221, 2008. doi:10.1016/j.virol.2008.08.026.
    Crossref | PubMed | ISI | Google Scholar
  • 242. Apicella M, Campopiano MC, Mantuano M, Mazoni L, Coppelli A, Del Prato S. COVID-19 in people with diabetes: understanding the reasons for worse outcomes. Lancet Diabetes Endocrinol 8: 782–792, 2020. doi:10.1016/S2213-8587(20)30238-2.
    Crossref | PubMed | ISI | Google Scholar
  • 243. Patel VB, Basu R, Oudit GY. ACE2/Ang 1-7 axis: a critical regulator of epicardial adipose tissue inflammation and cardiac dysfunction in obesity. Adipocyte 5: 306–311, 2016. doi:10.1080/21623945.2015.1131881.
    Crossref | PubMed | ISI | Google Scholar
  • 244. Sanchis-Gomar F, Lavie CJ, Mehra MR, Henry BM, Lippi G. Obesity and outcomes in COVID-19: when an epidemic and pandemic collide. Mayo Clin. Proc. 95: 1445–1453, 2020. doi:10.1016/j.mayocp.2020.05.006.
    Crossref | PubMed | ISI | Google Scholar
  • 245. Redd WD, Zhou JC, Hathorn KE, McCarty TR, Bazarbashi AN, Thompson CC, Shen L, Chan WW. Prevalence and characteristics of gastrointestinal symptoms in patients with severe acute respiratory syndrome coronavirus 2 infection in the United States: a multicenter cohort study. Gastroenterology 159: 765–767.e2, 2020. doi:10.1053/j.gastro.2020.04.045.
    Crossref | PubMed | ISI | Google Scholar
  • 246. Parasa S, Desai M, Thoguluva Chandrasekar V, Patel HK, Kennedy KF, Roesch T, Spadaccini M, Colombo M, Gabbiadini R, Artifon EL, Repici A, Sharma P. Prevalence of gastrointestinal symptoms and fecal viral shedding in patients with coronavirus disease 2019: a systematic review and meta-analysis. JAMA Netw Open 3: e2011335 , 2020. doi:10.1001/jamanetworkopen.2020.11335.
    Crossref | PubMed | ISI | Google Scholar
  • 247. Matsushita K, Ding N, Kou M, Hu X, Chen M, Gao Y, Honda Y, Dowdy D, Mok Y, Ishigami J, Appel LJ. The relationship of COVID-19 severity with cardiovascular disease and its traditional risk factors: a systematic review and meta-analysis. Glob Heart 15: 64 , 2020. doi:10.5334/gh.814.
    Crossref | PubMed | ISI | Google Scholar
  • 248. Pranata R, Huang I, Lim MA, Wahjoepramono EJ, July J. Impact of cerebrovascular and cardiovascular diseases on mortality and severity of COVID-19–systematic review, meta-analysis, and meta-regression. J Stroke Cerebrovasc Dis 29: 104949 , 2020. doi:10.1016/j.jstrokecerebrovasdis.2020.104949.
    Crossref | ISI | Google Scholar
  • 249. Rocha NP, Simoes e Silva AC, Prestes TR, Feracin V, Machado CA, Ferreira RN, Teixeira AL, de Miranda AS. RAS in the central nervous system: potential role in neuropsychiatric disorders. Curr Med Chem 25: 3333–3352, 2018. doi:10.2174/0929867325666180226102358.
    Crossref | PubMed | ISI | Google Scholar
  • 250. Lu J, Jiang T, Wu L, Gao L, Wang Y, Zhou F, Zhang S, Zhang Y. The expression of angiotensin-converting enzyme 2-angiotensin-(1-7)-Mas receptor axis are upregulated after acute cerebral ischemic stroke in rats. Neuropeptides 47: 289–295, 2013. doi:10.1016/j.npep.2013.09.002.
    Crossref | PubMed | ISI | Google Scholar
  • 251. Choi JY, Lee HK, Park JH, Cho SJ, Kwon M, Jo C, Koh YH. Altered COVID-19 receptor ACE2 expression in a higher risk group for cerebrovascular disease and ischemic stroke. Biochem Biophys Res Commun 528: 413–419, 2020. doi:10.1016/j.bbrc.2020.05.203.
    Crossref | PubMed | ISI | Google Scholar
  • 252. Davies NG, Klepac P, Liu Y, Prem K, Jit M, Pearson CAB, Quilty BJ, Kucharski AJ, Gibbs H, Clifford S, Gimma A, van Zandvoort K, Munday JD, Diamond C, Edmunds WJ, Houben RM, Hellewell J, Russell TW, Abbott S, Funk S, Bosse NI, Sun YF, Flasche S, Rosello A, Jarvis CI, Eggo RM, CMMID COVID-19 working group. Age-dependent effects in the transmission and control of COVID-19 epidemics. Nat Med 26: 1205–1211, 2020. doi:10.1038/s41591-020-0962-9.
    Crossref | PubMed | ISI | Google Scholar
  • 253. Viner RM, Mytton OT, Bonell C, Melendez-Torres GJ, Ward J, Hudson L, Waddington C, Thomas J, Russell S, van der Klis F, Koirala A, Ladhani S, Panovska-Griffiths J, Davies NG, Booy R, Eggo RM. Susceptibility to SARS-CoV-2 infection among children and adolescents compared with adults. JAMA Pediatrics 175: 143–156, 2021. doi:10.1001/jamapediatrics.2020.4573.
    Crossref | PubMed | ISI | Google Scholar
  • 254. Lopez AS, Hill M, Antezano J, Vilven D, Rutner T, Bogdanow L, Claflin C, Kracalik IT, Fields VL, Dunn A, Tate JE, Kirking HL, Kiphibane T, Risk I, Tran CH. Transmission dynamics of COVID-19 outbreaks associated with child care facilities–Salt Lake City, Utah, April–July 2020. MMWR Morb Mortal Wkly Rep 69: 1319–1323, 2020. doi:10.15585/mmwr.mm6937e3.
    Crossref | PubMed | ISI | Google Scholar
  • 255. Szablewski CM, Chang KT, Brown MM, Chu VT, Yousaf AR, Anyalechi N, Aryee PA, Kirking HL, Lumsden M, Mayweather E, McDaniel CJ, Montierth R, Mohammed A, Schwartz NG, Shah JA, Tate JE, Dirlikov E, Drenzek C, Lanzieri TM, Stewart RJ. SARS-CoV-2 transmission and infection among attendees of an overnight camp–Georgia, June 2020. MMWR Morb Mortal Wkly Rep 69: 1023–1025, 2020. doi:10.15585/mmwr.mm6931e1.
    Crossref | PubMed | ISI | Google Scholar
  • 256. Heald-Sargent T, Muller WJ, Zheng X, Rippe J, Patel AB, Kociolek LK. Age-related differences in nasopharyngeal severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) levels in patients with mild to moderate coronavirus disease 2019 (COVID-19). JAMA Pediatr 174: 902 , 2020. doi:10.1001/jamapediatrics.2020.3651.
    Crossref | PubMed | ISI | Google Scholar
  • 257. Karagiannidis C, Mostert C, Hentschker C, Voshaar T, Malzahn J, Schillinger G, Klauber J, Janssens U, Marx G, Weber-Carstens S, Kluge S, Pfeifer M, Grabenhenrich L, Welte T, Busse R. Case characteristics, resource use, and outcomes of 10 021 patients with COVID-19 admitted to 920 German hospitals: an observational study. Lancet Respir Med 8: 853–862, 2020. doi:10.1016/S2213-2600(20)30316-7.
    Crossref | PubMed | ISI | Google Scholar
  • 258. Onder G, Rezza G, Brusaferro S. Case-fatality rate and characteristics of patients dying in relation to COVID-19 in Italy. JAMA J Am Med Assoc 323: 1775–1776, 2020. doi:10.1001/jama.2020.4683.
    Crossref | PubMed | ISI | Google Scholar
  • 259. Yang X, Yu Y, Xu J, Shu H, Xia J, Liu H, Wu Y, Zhang L, Yu Z, Fang M, Yu T, Wang Y, Pan S, Zou X, Yuan S, Shang Y. Clinical course and outcomes of critically ill patients with SARS-CoV-2 pneumonia in Wuhan, China: a single-centered, retrospective, observational study. Lancet Respir Med 8: 475–481, 2020. [Erratum in Lancet Respir Med 8: e26, 2020]. doi:10.1016/S2213-2600(20)30079-5.
    Crossref | PubMed | ISI | Google Scholar
  • 260. Bialek S, Boundy E, Bowen V, Chow N, Cohn A, Dowling N, Ellington S, Gierke R, Hall A, MacNeil J, Patel P, Peacock G, Pilishvili T, Razzaghi H, Reed N, Ritchey M, Sauber-Schatz E. Severe outcomes among patients with coronavirus disease 2019 (COVID-19)–United States, February 12–March 16, 2020. MMWR Morb Mortal Wkly Rep 69: 343–346, 2020. doi:10.15585/mmwr.mm6912e2.
    Crossref | PubMed | ISI | Google Scholar
  • 261. Zimmermann P, Curtis N. Coronavirus infections in children including COVID-19: an overview of the epidemiology, clinical features, diagnosis, treatment and prevention options in children. Pediatr Infect Dis J 39: 355–368, 2020. doi:10.1097/INF.0000000000002660.
    Crossref | PubMed | ISI | Google Scholar
  • 262. Van Der Made CI, Simons A, Schuurs-Hoeijmakers J, Van Den Heuvel G, Mantere T, Kersten S, Van Deuren RC, Steehouwer M, Van Reijmersdal SV, Jaeger M, Hofste T, Astuti G, Corominas Galbany J, Van Der Schoot V, Van Der Hoeven H, Hagmolen Of Ten Have W, Klijn E, Van Den Meer C, Fiddelaers J, De Mast Q, Bleeker-Rovers CP, Joosten LA, Yntema HG, Gilissen C, Nelen M, Van Der Meer JW, Brunner HG, Netea MG, Van De Veerdonk FL, Hoischen A. Presence of genetic variants among young men with severe COVID-19. JAMA 324: 663–673, 2020. doi:10.1001/jama.2020.13719.
    Crossref | PubMed | ISI | Google Scholar
  • 263. Xudong X, Junzhu C, Xingxiang W, Furong Z, Yanrong L. Age- and gender-related difference of ACE2 expression in rat lung. Life Sci 78: 2166–2171, 2006. doi:10.1016/j.lfs.2005.09.038.
    Crossref | PubMed | ISI | Google Scholar
  • 264. Yoon HE, Kim EN, Kim MY, Lim JH, Jang IA, Ban TH, Shin SJ, Park CW, Chang YS, Choi BS. Age-associated changes in the vascular renin-angiotensin system in mice. Oxid Med Cell Longev 2016: 1–14, 2016. doi:10.1155/2016/6731093.
    Crossref | PubMed | ISI | Google Scholar
  • 265. Zhang C, Wang J, Ma X, Wang W, Zhao B, Chen Y, Chen C, Bihl JC. ACE2-EPC-EXs protect ageing ECs against hypoxia/reoxygenation-induced injury through the miR-18a/Nox2/ROS pathway. J Cell Mol Med 22: 1873–1882, 2018. doi:10.1111/jcmm.13471.
    Crossref | PubMed | ISI | Google Scholar
  • 266. Gilliam-Davis S, Gallagher PE, Payne VS, Kasper SO, Tommasi EN, Westwood BM, Robbins ME, Chappell MC, Diz DI. Long-term systemic angiotensin II type 1 receptor blockade regulates mRNA expression of dorsomedial medulla renin-angiotensin system components. Physiol Genomics 43: 829–835, 2011. doi:10.1152/physiolgenomics.00167.2010.
    Link | ISI | Google Scholar
  • 267. Peña-Silva RA, Chu Y, Miller JD, Mitchell IJ, Penninger JM, Faraci FM, Heistad DD. Impact of ACE2 deficiency and oxidative stress on cerebrovascular function with aging. Stroke 43: 3358–3363, 2012. doi:10.1161/STROKEAHA.112.667063.
    Crossref | PubMed | ISI | Google Scholar
  • 268. Chen J, Jiang Q, Xia X, Liu K, Yu Z, Tao W, Gong W, Han JD. Individual variation of the SARS-CoV-2 receptor ACE2 gene expression and regulation. Aging Cell 19: 2–5, 2020. doi:10.1111/acel.13168.
    Crossref | PubMed | ISI | Google Scholar
  • 269. Asselta R, Paraboschi EM, Mantovani A, Duga S. ACE2 and TMPRSS2 variants and expression as candidates to sex and country differences in COVID-19 severity in Italy. Aging (Albany NY) 12: 10087–10098, 2020. doi:10.18632/aging.103415.
    Crossref | PubMed | Google Scholar
  • 270. Li MY, Li L, Zhang Y, Wang XS. Expression of the SARS-CoV-2 cell receptor gene ACE2 in a wide variety of human tissues. Infect Dis Poverty 9: 45 , 2020. doi:10.1186/s40249-020-00662-x.
    Crossref | PubMed | ISI | Google Scholar
  • 271. Schouten LR, van Kaam AH, Kohse F, Veltkamp F, Bos LD, de Beer FM, , et al.. Age-dependent differences in pulmonary host responses in ARDS: a prospective observational cohort study. Ann Intensive Care 9: 55 , 2019. doi:10.1186/s13613-019-0529-4.
    Crossref | PubMed | ISI | Google Scholar
  • 272. Bilinska K, Jakubowska P, Von Bartheld CS, Butowt R. Expression of the SARS-CoV-2 entry proteins, ACE2 and TMPRSS2, in cells of the olfactory epithelium: identification of cell types and trends with age. ACS Chem Neurosci 11: 1555–1562, 2020. doi:10.1021/acschemneuro.0c00210.
    Crossref | PubMed | ISI | Google Scholar
  • 273. Brann DH, Tsukahara T, Weinreb C, Lipovsek M, Van den Berge K, Gong B, Chance R, Macaulay IC, Chou H, Fletcher RB, Das D, Street K, de Bezieux HR, Choi Y-G, Risso D, Dudoit S, Purdom E, Mill JS, Hachem RA, Matsunami H, Logan DW, Goldstein BJ, Grubb MS, Ngai J, Datta SR. Non-neuronal expression of SARS-CoV-2 entry genes in the olfactory system suggests mechanisms underlying COVID-19-associated anosmia. Sci Adv 6: eabc5801 , 2020. doi:10.1126/sciadv.abc5801.
    Crossref | PubMed | ISI | Google Scholar
  • 274. Bunyavanich S, Do A, Vicencio A. Nasal gene expression of angiotensin-converting enzyme 2 in children and adults. JAMA 323: 2427–2429, 2020. doi:10.1001/jama.2020.8707.
    Crossref | PubMed | ISI | Google Scholar
  • 275. Somekh I, Yakub Hanna H, Heller E, Bibi H, Somekh E. Age-dependent sensory impairment in COVID-19 infection and its correlation with ACE2 Eexpression. Pediatr Infect Dis J 39: e270–e272, 2020. doi:10.1097/inf.0000000000002817.
    Crossref | PubMed | ISI | Google Scholar
  • 276. Yonker LM, Neilan AM, Bartsch Y, Patel AB, Regan J, Arya P, Gootkind E, Park G, Hardcastle M, St. John A, Appleman L, Chiu ML, Fialkowski A, De la Flor D, Lima R, Bordt EA, Yockey LJ, D'Avino P, Fischinger S, Shui JE, Lerou PH, Bonventre JV, Yu XG, Ryan ET, Bassett IV, Irimia D, Edlow AG, Alter G, Li JZ, Fasano A. Pediatric severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2): clinical presentation, infectivity, and immune responses. J Pediatr 227: 45–52.e5, 2020. doi:10.1016/j.jpeds.2020.08.037.
    Crossref | PubMed | ISI | Google Scholar
  • 277. Diaz JH. Hypothesis: angiotensin-converting enzyme inhibitors and angiotensin receptor blockers may increase the risk of severe COVID-19. J Travel Med 27: taaa041 , 2020. doi:10.1093/jtm/taaa041.
    Crossref | PubMed | ISI | Google Scholar
  • 278. Nowak JK, Lindstrøm JC, Kalla R, Ricanek P, Halfvarson J, Satsangi J. Age, inflammation, and disease location are critical determinants of intestinal expression of SARS-CoV-2 receptor ACE2 and TMPRSS2 in inflammatory bowel disease. Gastroenterology 159: 1151–1154.e2, 2020. doi:10.1053/j.gastro.2020.05.030.
    Crossref | PubMed | ISI | Google Scholar
  • 279. Vuille-dit-Bille RN, Liechty KW, Verrey F, Guglielmetti LC. SARS-CoV-2 receptor ACE2 gene expression in small intestine correlates with age. Amino Acids 52: 1063–1065, 2020. doi:10.1007/s00726-020-02870-z.
    Crossref | PubMed | ISI | Google Scholar
  • 280. Chen YY, Zhang P, Zhou XM, Liu D, Zhong JC, Zhang CJ, Jin LJ, Yu HM. Relationship between genetic variants of ACE2 gene and circulating levels of ACE2 and its metabolites. J Clin Pharm Ther 43: 189–195, 2018. doi:10.1111/jcpt.12625.
    Crossref | PubMed | ISI | Google Scholar
  • 281. Fernández-Atucha A, Izagirre A, Fraile-Bermúdez AB, Kortajarena M, Larrinaga G, Martinez-Lage P, Echevarría E, Gil J. Sex differences in the aging pattern of renin-angiotensin system serum peptidases. Biol Sex Differ 8: 189 , 2017. doi:10.1186/s13293-017-0128-8.
    Crossref | PubMed | ISI | Google Scholar
  • 282. Kornilov SA, Lucas I, Jade K, Dai CL, Lovejoy JC, Magis AT. Plasma levels of soluble ACE2are associated with sex, metabolic syndrome, and its biomarkers in a large cohort, pointing to a possible mechanism for increased severity in COVID-19. Crit Care 24: 452 , 2020. doi:10.1186/s13054-020-03141-9.
    Crossref | PubMed | ISI | Google Scholar
  • 283. Swärd P, Edsfeldt A, Reepalu A, Jehpsson L, Rosengren BE, Karlsson MK. Age and sex differences in soluble ACE2 may give insights for COVID-19. Crit Care 24: 221 , 2020. doi:10.1186/s13054-020-02942-2.
    Crossref | PubMed | ISI | Google Scholar
  • 284. Yang P, Gu H, Zhao Z, Wang W, Cao B, Lai C, Yang X, Zhang LY, Duan Y, Zhang S, Chen W, Zhen W, Cai M, Penninger JM, Jiang C, Wang X. Angiotensin-converting enzyme 2 (ACE2) mediates influenza H7N9 virus-induced acute lung injury. Sci Rep 4: 7027 , 2015. doi:10.1038/srep07027.
    Crossref | PubMed | ISI | Google Scholar
  • 285. Akbar AN, Gilroy DW. Aging immunity may exacerbate COVID-19. Science 369: 256–257, 2020. doi:10.1038/srep07027.
    Crossref | PubMed | ISI | Google Scholar
  • 286. Baker S, Kowk S, Berry G, Montine T. Angiotensin-converting enzyme 2 (ACE2) expression increases with age in patients requiring mechanical ventilation (Preprint). medRxiv 2020.07.05.20140467 , 2020. doi:10.1101/2020.07.05.20140467.
    Crossref | Google Scholar
  • 287. Hua W, Xiaofeng L, Zhenqiang B, Jun R, Ban W, Liming L. Consideration on the strategies during epidemic stage changing from emergency response to continuous prevention and control. Chinese J Endem 41: 297–300, 2020. doi:10.3760/cma.j.issn.0254-6450.2020.02.003.
    Crossref | Google Scholar
  • 288. Andrianou X, Bella A, Manso MD, Urdiales AM, Fabiani M, Bellino S, Boros S, Rota MC, Filia A, Punzo O, Siddu A, Benedetto CD, Tallon M, Ciervo A, Castrucci MR, Pezzotti P, Stefanelli P, Rezza G, Scondotto S; Task force COVID-19 del Dipartimento Malattie Infettive e Servizio di Informatica, Istituto Superiore di Sanità. Epidemia COVID-19, Aggiornamento nazionale (Online). April 10, 2020; https://www.epicentro.iss.it/coronavirus/bollettino/Bollettino-sorveglianza-integrata-COVID-19_9-aprile-2020.pdf.
    Google Scholar
  • 289. Jaillon S, Berthenet K, Garlanda C. Sexual dimorphism in innate immunity. Clin Rev Allergy Immunol 56: 308–321, 2019. doi:10.1007/s12016-017-8648-x.
    Crossref | PubMed | ISI | Google Scholar
  • 290. Chen KAI, Bi J, Su Y, Chappell MC, Rose JC. Sex-specific changes in renal angiotensin-converting enzyme and angiotensin-converting enzyme 2 gene expression and enzyme activity at birth and over the first year of life. Reprod Sci 23: 200–210, 2016. doi:10.1177/1933719115597760.
    Crossref | PubMed | ISI | Google Scholar
  • 291. Ji H, De Souza AM, Bajaj B, Zheng W, Wu X, Speth RC, Sandberg K. Sex-specific modulation of blood pressure and the renin-angiotensin system by ace (angiotensin-converting enzyme) 2. Hypertension 76: 478–487, 2020. doi:10.1161/HYPERTENSIONAHA.120.15276.
    Crossref | PubMed | ISI | Google Scholar
  • 292. Liu J, Ji H, Zheng W, Wu X, Zhu JJ, Arnold AP, Sandberg K. Sex differences in renal angiotensin converting enzyme 2 (ACE2) activity are 17β-oestradiol-dependent and sex chromosome-independent. Biol Sex Differ 1: 6 , 2010. doi:10.1186/2042-6410-1-6.
    Crossref | PubMed | Google Scholar
  • 293. Pendergrass KD, Pirro NT, Westwood BM, Ferrario CM, Brosnihan KB, Chappell MC. Sex differences in circulating and renal angiotensins of hypertensive mRen(2).Lewis but not normotensive Lewis rats. Am J Physiol Hear Circ Physiol 295: H10–H20, 2008. doi:10.1152/ajpheart.01277.2007.
    Link | ISI | Google Scholar
  • 294. Sampson AK, Moritz KM, Denton KM. Postnatal ontogeny of angiotensin receptors and ACE2 in male and female rats. Gend Med 9: 21–32, 2012. doi:10.1016/j.genm.2011.12.003.
    Crossref | PubMed | Google Scholar
  • 295. Cuffe JSM, Burgess DJ, O'Sullivan L, Singh RR, Moritz KM. Maternal corticosterone exposure in the mouse programs sex-specific renal adaptations in the renin-angiotensin-aldosterone system in 6-month offspring. Physiol Rep 4: e12754 , 2016. doi:10.14814/phy2.12754.
    Crossref | PubMed | Google Scholar
  • 296. Zhang Q, Cong M, Wang N, Li X, Zhang H, Zhang K, Jin M, Wu N, Qiu C, Li J. Association of angiotensin-converting enzyme 2 gene polymorphism and enzymatic activity with essential hypertension in different gender: a case–control study. Medicine (Baltimore) 97: e1917 , 2018. doi:10.1097/md.0000000000012917.
    Crossref | PubMed | ISI | Google Scholar
  • 297. Song H, Seddighzadeh B, Cooperberg MR, Huang FW. Expression of ACE2, the SARS-CoV-2 receptor, and TMPRSS2 in prostate epithelial cells (Preprint). bioRxiv 2020.04.24.056259 , 2020.
    Google Scholar
  • 298. Wang Y, Wang Y, Luo W, Huang L, Xiao J, Li F, Qin S, Song X, Wu Y, Zeng Q, Jin F, Wang Y. A comprehensive investigation of the mrna and protein level of ace2, the putative receptor of sars-cov-2, in human tissues and blood cells. Int J Med Sci 17: 1522–1531, 2020. doi:10.7150/ijms.46695.
    Crossref | PubMed | ISI | Google Scholar
  • 299. Zhang H, Rostami MR, Leopold PL, Mezey JG, O'Beirne SL, Strulovici-Barel Y, Crystal RG. Expression of the SARS-CoV-2 ACE2 receptor in the human airway epithelium. Am J Respir Crit Care Med 202: 219–229, 2020. doi:10.1164/rccm.202003-0541OC.
    Crossref | PubMed | ISI | Google Scholar
  • 300. Baughn LB, Sharma N, Elhaik E, Sekulic A, Bryce AH, Fonseca R. Targeting TMPRSS2 in SARS-CoV-2 Infection. Mayo Clin Proc 95: 1989–1999, 2020. doi:10.1016/j.mayocp.2020.06.018.
    Crossref | PubMed | ISI | Google Scholar
  • 301. Stienen S, Ferreira JP, Kobayashi M, Preud’Homme G, Dobre D, Machu JL, Duarte K, Bresso E, Devignes MD, Andrés NL, Girerd N, Aakhus S, Ambrosio G, Rocca HL, Fontes-Carvalho R, Fraser AG, Van Heerebeek L, De Keulenaer G, Marino P, McDonald K, Mebazaa A, Papp Z, Raddino R, Tschöpe C, Paulus WJ, Zannad F, Rossignol P. Sex differences in circulating proteins in heart failure with preserved ejection fraction. Biol Sex Differ 11: 47 , 2020. doi:10.1186/s13293-020-00322-7.
    Crossref | PubMed | ISI | Google Scholar
  • 302. Foresta C, Rocca MS, Di Nisio A. Gender susceptibility to COVID-19: a review of the putative role of sex hormones and X chromosome. J Endocrinol Invest Sex Differ. In press. doi:10.1007/s40618-020-01383-6.
    Crossref | PubMed | Google Scholar
  • 303. Bukowska A, Spiller L, Wolke C, Lendeckel U, Weinert S, Hoffmann J, Bornfleth P, Kutschka I, Gardemann A, Isermann B, Goette A. Protective regulation of the ACE2/ACE gene expression by estrogen in human atrial tissue from elderly men. Exp Biol Med (Maywood) 242: 1412–1423, 2017. doi:10.1177/1535370217718808.
    Crossref | PubMed | ISI | Google Scholar
  • 304. Wang Y, Shoemaker R, Thatcher SE, Batifoulier-Yiannikouris F, English VL, Cassis LA. Administration of 17β-estradiol to ovariectomized obese female mice reverses obesity-hypertension through an ACE2-dependent mechanism. Am J Physiol Endocrinol Metab 308: E1066–E1075, 2015. doi:10.1152/ajpendo.00030.2015.
    Link | ISI | Google Scholar
  • 305. Haitao T, Vermunt J, Abeykoon J, Ghamrawi R, Gunaratne M, Jayachandran M, Narang K, Parashuram S, Suvakov S, Garovic V. COVID-19 and sex differences: mechanisms and biomarkers. Mayo Clin Proc 95: 2189–2203, 2020. doi:10.1016/j.mayocp.2020.07.024.
    Crossref | PubMed | ISI | Google Scholar
  • 306. Stelzig KE, Canepa-Escaro F, Schiliro M, Berdnikovs S, Prakash YS, Chiarella SE. Estrogen regulates the expression of SARS-CoV-2 receptor ACE2 in differentiated airway epithelial cells. Am J Physiol Lung Cell Mol Physiol 318: L1280–L1281, 2020. doi:10.1152/ajplung.00153.2020.
    Link | ISI | Google Scholar
  • 307. Clotet S, Soler MJ, Rebull M, Gimeno J, Gurley SB, Pascual J, Riera M. Gonadectomy prevents the increase in blood pressure and glomerular injury in angiotensin-converting enzyme 2 knockout diabetic male mice. Effects on renin-angiotensin system. J Hypertens 34: 1752–1765, 2016. doi:10.1097/HJH.0000000000001015.
    Crossref | PubMed | ISI | Google Scholar
  • 308. Dalpiaz PLM, Lamas AZ, Caliman IF, Ribeiro RF, Abreu GR, Moyses MR, Andrade TU, Gouvea SA, Alves MF, Carmona AK, Bissoli NS. Sex hormones promote opposite effects on ACE and ACE2 activity, hypertrophy and cardiac contractility in spontaneously hypertensive rats. PLoS One 10: e0133225 , 2015. doi:10.1371/journal.pone.0133225.
    Crossref | PubMed | ISI | Google Scholar
  • 309. Central Intelligence Office. Field Listing: Religions–The World Factbook–Central Intelligence Agency (Online). https://www.cia.gov/library/publications/the-world-factbook/fields/401.html#AF [28 Mar. 2020].
    Google Scholar
  • 310. Tartof SY, Qian L, Hong V, Wei R, Nadjafi RF, Fischer H, Li Z, Shaw SF, Caparosa SL, Nau CL, Saxena T, Rieg GK, Ackerson BK, Sharp AL, Skarbinski J, Naik TK, Murali SB. Obesity and mortality among patients diagnosed with COVID-19: results from an integrated health care organization. Ann Intern Med 173: 773–781, 2020. doi:10.7326/M20-3742.
    Crossref | PubMed | ISI | Google Scholar
  • 311. Cai H. Sex difference and smoking predisposition in patients with COVID-19. Lancet Respir Med 8: e20 , 2020. doi:10.1016/S2213-2600(20)30117-X.
    Crossref | PubMed | ISI | Google Scholar
  • 312. World Health Organization. Gender, Equity, Human Rights (Online). https://www.who.int/gender-equity-rights/en/ [23 Sep. 2020].
    Google Scholar
  • 313. Qin L, Li X, Shi J, Yu M, Wang K, Tao Y, Zhou Y, Zhou M, Xu S, Wu B, Yang Z, Zhang C, Yue J, Cheng C, Liu X, Xie M. Gendered effects on inflammation reaction and outcome of COVID‐19 patients in Wuhan. J Med Virol 92: 2684–2692, 2020. doi:10.1002/jmv.26137.
    Crossref | PubMed | ISI | Google Scholar
  • 314. Bunders MJ, Altfeld M. Implications of sex differences in immunity for SARS-CoV-2 pathogenesis and design of therapeutic interventions. Immunity 53: 487–495, 2020. doi:10.1016/j.immuni.2020.08.003.
    Crossref | PubMed | ISI | Google Scholar
  • 315. Takahashi T, Ellingson MK, Wong P, Israelow B, Lucas C, Klein J, , et al.. Sex differences in immune responses that underlie COVID-19 disease outcomes. Nature 588: 315–320, 2020. doi:10.1038/s41586-020-2700-3.
    Crossref | PubMed | ISI | Google Scholar
  • 316. World Health Organization. Coronavirus Disease 2019 (110) (Online). https://www.who.int/emergencies/diseases/novel-coronavirus-2019. [2020].
    Google Scholar
  • 317. Gold JA, Wong KK, Szablewski CM, Patel PR, Rossow J, da Silva J, , et al.. Characteristics and clinical outcomes of adult patients hospitalized with COVID-19–Georgia, March 2020. MMWR Morb Mortal Wkly Rep 69: 545–550, 2020. doi:10.15585/mmwr.mm6918e1.
    Crossref | PubMed | ISI | Google Scholar
  • 318. Killerby ME, Link-Gelles R, Haight SC, Schrodt CA, England L, Gomes DJ, CDC COVID-19 Response Clinical Team , , et al.. Characteristics associated with hospitalization among patients with COVID-19–Metropolitan Atlanta, Georgia, March–April 2020. MMWR Morb Mortal Wkly Rep 69: 790–794, 2020. doi:10.15585/mmwr.mm6925e1.
    Crossref | PubMed | ISI | Google Scholar
  • 319. Millett GA, Jones AT, Benkeser D, Baral S, Mercer L, Beyrer C, Honermann B, Lankiewicz E, Mena L, Crowley JS, Sherwood J, Sullivan PS. Assessing differential impacts of COVID-19 on black communities. Ann Epidemiol 47: 37–44, 2020. doi:10.1016/j.annepidem.2020.05.003.
    Crossref | PubMed | ISI | Google Scholar
  • 320. Price-Haywood EG, Burton J, Fort D, Seoane L. Hospitalization and mortality among black patients and white patients with Covid-19. N Engl J Med 382: 2534–2543, 2020. doi:10.1056/NEJMsa2011686.
    Crossref | PubMed | ISI | Google Scholar
  • 321. Stokes EK, Zambrano LD, Anderson KN, Marder EP, Raz KM, El Burai Felix S, Tie Y, Fullerton KE. Coronavirus disease 2019 case surveillance–United States, January 22–May 30, 2020. MMWR Morb Mortal Wkly Rep 69: 759–765, 2020. doi:10.15585/mmwr.mm6924e2.
    Crossref | PubMed | ISI | Google Scholar
  • 322. The Washington Post. African Americans are at Higher Risk of Death from Coronavirus (Online). https://www.washingtonpost.com/nation/2020/04/07/coronavirus-is-infecting-killing-black-americans-an-alarmingly-high-rate-post-analysis-shows/?arc404=true [30 Sep. 2020].
    Google Scholar
  • 323. Fujikura K, Uesaka K. Genetic variations in the human severe acute respiratory syndrome coronavirus receptor ACE2 and serine protease TMPRSS2. J Clin Pathol. In press. doi:10.1136/jclinpath-2020-206867.
    Crossref | PubMed | ISI | Google Scholar
  • 324. Gemmati D, Tisato V. Genetic hypothesis and pharmacogenetics side of renin-angiotensin-system in COVID-19. Genes (Basel) 11: 1044 , 2020. doi:10.3390/genes11091044.
    Crossref | PubMed | ISI | Google Scholar
  • 325. Hou Y, Zhao J, Martin W, Kallianpur A, Chung MK, Jehi L, Sharifi N, Erzurum S, Eng C, Cheng F. New insights into genetic susceptibility of COVID-19: an ACE2 and TMPRSS2 polymorphism analysis. BMC Med 18: 216 , 2020. doi:10.1186/s12916-020-01673-z.
    Crossref | PubMed | ISI | Google Scholar
  • 326. Zhao Y, Zhao Z, Wang Y, Zhou Y, Ma Y, Zuo W. Single-Cell RNA expression profiling of ACE2, the receptor of SARS-CoV-2. Am J Respir Crit Care Med 202: 756–759, 2020. doi:10.1164/rccm.202001-0179LE.
    Crossref | PubMed | ISI | Google Scholar
  • 327. Cao Y, Li L, Feng Z, Wan S, Huang P, Sun X, Wen F, Huang X, Ning G, Wang W. Comparative genetic analysis of the novel coronavirus (2019-nCoV/SARS-CoV-2) receptor ACE2 in different populations. Cell Discov 6: 11 , 2020. doi:10.1038/s41420-020-0245-8, 10.1038/s41421-020-0147-1.
    Crossref | PubMed | ISI | Google Scholar
  • 328. Chen Y, Shan K, Qian W. Asians do not exhibit elevated expression or unique genetic polymorphisms for ACE2, the cell-entry receptor of SARS-CoV-2 (Preprint). Preprints 2020020258 , 2020.
    Google Scholar
  • 329. Li Q, Cao Z, Rahman P. Genetic variability of human angiotensin‐converting enzyme 2 (hACE2) among various ethnic populations. Mol Genet Genomic Med 8: e1344 , 2020. doi:10.1002/mgg3.1344.
    Crossref | PubMed | ISI | Google Scholar
  • 330. Cai G. Bulk and single-cell transcriptomics identify tobacco-use disparity in lung gene expression of ACE2, the receptor of 2019-nCov (Preprint). medRxiv 2020.02.05.20020107 , 2020. doi:10.1101/2020.02.05.20020107v3.
    Crossref | Google Scholar
  • 331. Dai X. ABO blood group predisposes to COVID-19 severity and cardiovascular diseases. Eur J Prev Cardiol 27: 1436–1437, 2020. doi:10.1177/2047487320922370.
    Crossref | PubMed | ISI | Google Scholar
  • 332. Yamamoto N, Ariumi Y, Nishida N, Yamamoto R, Bauer G, Gojobori T, Shimotohno K, Mizokami M. SARS-CoV-2 infections and COVID-19 mortalities strongly correlate with ACE1 I/D genotype. Gene 758: 144944 , 2020. doi:10.1016/j.gene.2020.144944.
    Crossref | PubMed | ISI | Google Scholar
  • 333. Ellinghaus D, Degenhardt F, Bujanda L, Buti M, Albillos A, Invernizzi P, , et al.. genomewide association study of severe covid-19 with respiratory failure. N Engl J Med 383: 1522–1534, 2020. doi:10.1056/NEJMoa2020283.
    Crossref | PubMed | ISI | Google Scholar
  • 334. Pourali F, Afshari M, Alizadeh-Navaei R, Javidnia J, Moosazadeh M, Hessami A. Relationship between blood group and risk of infection and death in COVID-19: a live meta-analysis. New Microbes New Infect 37: 100743 , 2020. doi:10.1016/j.nmni.2020.100743.
    Crossref | PubMed | ISI | Google Scholar
  • 335. Guillon P, Clément M, Sébille V, Rivain JG, Chou CF, Ruvoën-Clouet N, Le Pendu J. Inhibition of the interaction between the SARS-CoV Spike protein and its cellular receptor by anti-histo-blood group antibodies. Glycobiology 18: 1085–1093, 2008. doi:10.1093/glycob/cwn093.
    Crossref | PubMed | ISI | Google Scholar
  • 336. Lu N, Yang Y, Wang Y, Liu Y, Fu G, Chen D, Dai H, Fan X, Hui R, Zheng Y. ACE2 gene polymorphism and essential hypertension: an updated meta-analysis involving 11,051 subjects. Mol Biol Rep 39: 6581–6589, 2012. doi:10.1007/s11033-012-1487-1.
    Crossref | PubMed | ISI | Google Scholar
  • 337. 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 5: 811 , 2020. doi:10.1001/jamacardio.2020.1017.
    Crossref | PubMed | ISI | Google Scholar
  • 338. Ferrario CM, Jessup J, Chappell MC, Averill DB, Brosnihan KB, Tallant EA, Diz DI, Gallagher PE. Effect of angiotensin-converting enzyme inhibition and angiotensin II receptor blockers on cardiac angiotensin-converting enzyme 2. Circulation 111: 2605–2610, 2005. doi:10.1161/CIRCULATIONAHA.104.510461.
    Crossref | PubMed | ISI | Google Scholar
  • 339. Ocaranza MP, Godoy I, Jalil JE, Varas M, Collantes P, Pinto M, Roman M, Ramirez C, Copaja M, Diaz-Araya G, Castro P, Lavandero S. Enalapril attenuates downregulation of angiotensin-converting enzyme 2 in the late phase of ventricular dysfunction in myocardial infarcted rat. Hypertension 48: 572–578, 2006. doi:10.1161/01.HYP.0000237862.94083.45.
    Crossref | PubMed | ISI | Google Scholar
  • 340. Lezama-Martinez D, Flores-Monroy J, Fonseca-Coronado S, Hernandez-Campos ME, Valencia-Hernandez I, Martinez-Aguilar L. Combined antihypertensive therapies that increase expression of cardioprotective biomarkers associated with the renin-angiotensin and kallikrein-kinin system. J Cardiovasc Pharmacol 72: 291–295, 2018. doi:10.1097/FJC.0000000000000629.
    Crossref | PubMed | ISI | Google Scholar
  • 341. Ishiyama Y, Gallagher PE, Averill DB, Tallant EA, Brosnihan KB, Ferrario CM. Upregulation of angiotensin-converting enzyme 2 after myocardial infarction by blockade of angiotensin II receptors. Hypertension 43: 970–976, 2004. doi:10.1161/01.HYP.0000124667.34652.1a.
    Crossref | PubMed | ISI | Google Scholar
  • 342. Whaley-Connell AT, Chowdhury NA, Hayden MR, Stump CS, Habibi J, Wiedmeyer CE, Gallagher PE, Tallant EA, Cooper SA, Link CD, Ferrario C, Sowers JR. Oxidative stress and glomerular filtration barrier injury: role of the renin-angiotensin system in the Ren2 transgenic rat. Am J Physiol Renal Physiol 291: F1308–F1314, 2006. [ doi:10.1152/ajprenal.00167.2006.
    Link | ISI | Google Scholar
  • 343. Takeda Y, Zhu A, Yoneda T, Usukura M, Takata H, Yamagishi M. Effects of aldosterone and angiotensin II receptor blockade on cardiac angiotensinogen and angiotensin-converting enzyme 2 expression in Dahl salt-sensitive hypertensive rats. Am J Hypertens 20: 1119–1124, 2007. doi:10.1016/j.amjhyper.2007.05.008.
    Crossref | PubMed | ISI | Google Scholar
  • 344. Kreutz R, Algharably EA, Azizi M, Dobrowolski P, Guzik T, Januszewicz A, Persu A, Prejbisz A, Riemer TG, Wang JG, Burnier M. Hypertension, the renin-angiotensin system, and the risk of lower respiratory tract infections and lung injury: implications for COVID-19. Cardiovasc Res 116: 1688–1699, 2020. doi:10.1093/cvr/cvaa097.
    Crossref | PubMed | ISI | Google Scholar
  • 345. Caldeira D, Alarcão J, Vaz-Carneiro A, Costa J. Risk of pneumonia associated with use of angiotensin converting enzyme inhibitors and angiotensin receptor blockers: systematic review and meta-analysis. BMJ 345: e4260–e4260, 2012. doi:10.1136/bmj.e4260.
    Crossref | PubMed | Google Scholar
  • 346. Milne S, Yang CX, Timens W, Bossé Y, Sin DD. SARS-CoV-2 receptor ACE2 gene expression and RAAS inhibitors. Lancet Respir. Med. 8: e50–e51, 2020. doi:10.1016/S2213-2600(20)30224-1.
    Crossref | PubMed | ISI | Google Scholar
  • 347. Li J, Wang X, Chen J, Zhang H, Deng A. Association of renin-angiotensin system inhibitors with severity or risk of death in patients with hypertension hospitalized for coronavirus disease 2019 (COVID-19) infection in Wuhan, China. JAMA Cardiol 5: 825–830, 2020 [Erratum in JAMA Cardiol 5: 968, 2020]. doi:10.1001/jamacardio.2020.1624.
    Crossref | PubMed | ISI | Google Scholar
  • 348. 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 382: 2441–2448, 2020. doi:10.1056/NEJMoa2008975.
    Crossref | PubMed | ISI | Google Scholar
  • 349. de Abajo FJ, Rodríguez-Martín S, Lerma V, Mejía-Abril G, Aguilar M, García-Luque A, MED-ACE2-COVID19 study group , , et al.. Use of renin–angiotensin–aldosterone system inhibitors and risk of COVID-19 requiring admission to hospital: a case-population study. Lancet 395: 1705–1714, 2020. doi:10.1016/S0140-6736(20)31030-8.
    Crossref | PubMed | ISI | Google Scholar
  • 350. 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 382: 2431–2440, 2020. doi:10.1056/NEJMoa2006923.
    Crossref | PubMed | ISI | Google Scholar
  • 351. Messerli FH, Siontis GC, Rexhaj E. COVID-19 and renin angiotensin blockers: current evidence and recommendations. Circulation 141: 2042–2044, 2020. doi:10.1161/CIRCULATIONAHA.120.047022.
    Crossref | PubMed | ISI | Google Scholar
  • 352. Patel AB, Verma A. COVID-19 and angiotensin-converting enzyme inhibitors and angiotensin receptor blockers: what is the evidence? JAMA 323: 1769–1770, 2020.
    PubMed | ISI | Google Scholar
  • 353. Yang C, Jin Z. An acute respiratory infection runs into the most common noncommunicable epidemic—COVID-19 and cardiovascular diseases. JAMA Cardiol 5: 743–744, 2020. doi:10.1001/jamacardio.2020.0934.
    Crossref | PubMed | ISI | Google Scholar
  • 354. Zhang W, Xu YZ, Liu B, Wu R, Yang YY, Xiao XQ, Zhang X. Pioglitazone upregulates angiotensin converting enzyme 2 expression in insulin-sensitive tissues in rats with high-fat diet-induced nonalcoholic steatohepatitis. Sci World J 2014: 603409 , 2014. doi:10.1155/2014/603409.
    Crossref | ISI | Google Scholar
  • 355. Qiao W, Wang C, Chen B, Zhang F, Liu Y, Lu Q, Guo H, Yan C, Sun H, Hu G, Yin X. Ibuprofen attenuates cardiac fibrosis in streptozotocin-induced diabetic rats. Cardiology 131: 97–106, 2015. doi:10.1159/000375362.
    Crossref | PubMed | ISI | Google Scholar
  • 356. Day M. Covid-19: European drugs agency to review safety of ibuprofen. BMJ 368: m1168 , 2020. doi:10.1136/bmj.m1168.
    Crossref | PubMed | Google Scholar
  • 357. Chen R, Liang W, Jiang M, Guan W, Zhan C, Wang T, Medical Treatment Expert Group for COVID-19 , , et al.. Risk factors of fatal outcome in hospitalized subjects with coronavirus disease 2019 from a nationwide analysis in China. Chest 158: 97–105, 2020. doi:10.1016/j.chest.2020.04.010.
    Crossref | PubMed | ISI | Google Scholar

AUTHOR NOTES