Abstract
Purpose of review
Hospitalizations for COVID-19 dramatically increase with age. This is likely due to increases in fragility across biological repair systems and a weakened immune system, including loss of the cardiorenal protective arm of the renin angiotensin system (RAS), composed of angiotensin converting enzyme-2 (ACE2)/angiotensin-(1–7) [Ang-(1–7)] and its actions through the Mas receptor. The purpose of this review is to explore how cardiac ACE2 changes with age, cardiac pathologies, comorbid conditions and pharmaceutical regimens in order to shed light on a potential hormonal unbalance facilitating SARs-CoV-2 vulnerabilities in older adults.
Recent findings
Increased ACE2 gene expression has been reported in human hearts with myocardial infarction, cardiac remodeling and heart failure. We also found ACE2 mRNA in atrial appendage tissue from cardiac surgical patients to be positively associated with age, elevated by certain comorbid conditions (e.g., COPD and previous stroke) and increased in conjunction with patients’ chronic use of anti-thrombotic agents and thiazides diuretics, but not drugs that block the renin angiotensin system.
Summary
Cardiac ACE2 may have bifunctional roles in COVID-19 since ACE2 mediates cellular susceptibility to SARS-CoV-2 infection, but also protects the heart via the ACE2/Ang-(1–7) pathway. Linking tissue ACE2 from cardiac surgery patients to their comorbid conditions and medical regimens provides a unique platform to address the influence that altered expression of the ACE2/Ang-(1–7)/Mas receptor axis might impact SARs-CoV-2 vulnerability in older adults.
Keywords: Angiotensin converting enzyme 2, aging, comorbidities, coronavirus disease 2019
Introduction
Hospitalizations for coronavirus disease 2019 (COVID-19) increase dramatically with age (1) and the oldest of the old (those 85 years and older) account for nearly one-third of COVID-19 deaths in the United States (https://www.cdc.gov/nchs/nvss/vsrr/covid_weekly/index.htm#AgeAndSex accessed October 30). Fragility and a weakened immune system are likely contributors to the increase in aging-related adverse sequela of Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) infection. Aggravating factors might also include non-O (2–4), and particularly A blood types (5), and altered expression of renin angiotensin system factors, specifically reduced regulatory capacity of ACE2/Ang-(1–7)/Mas-receptor axis. Indeed, the role of ACE2 in the pathogenesis of heart disease extends beyond that of hydrolyzing angiotensin II into angiotensin-(1–7), since its functional activities may not always associate with favorable outcomes (6, 7), and because it is the receptor of SARS-CoV-2 (8). Thus, the purpose of this review is to briefly discuss why COVID-19 targets older adults, and to explore how cardiac ACE2 might change with age, cardiac pathologies, comorbid conditions and pharmaceutical regimens, in order to shed light on a potential biochemical link to SARs-CoV-2 infection and disease severity with advancing age.
Aging and Covid-19
It is well accepted that hospitalization for COVID-19 increases with age. Persons 85 years and older are 130 times more likely to be hospitalized for Covid-19 than those between ages 18 and 29 years (https://www.cdc.gov/coronavirus/2019-ncov/need-extra-precautions/older-adults.html). To better understand the impact of age on Covid-19, a look at how age impacts the risk of death is important. 16.5 % of the US population is 65 years and older, and this age group accounts for 85% of all-cause deaths, while 1 in 20 adults are in the 85 years and up category, accounting for an astonishing one-third of all Covid-19 related-deaths. Besides advanced age as one of the highest risk factors for COVID-19 related deaths, having comorbid diseases, being male, living in a nursing home, being Black, Hispanic or Latinx, sustaining a low socioeconomic status (e.g. annual income < $25,000 [U.S.]), having blood type A, and having two copies of the APOE e4 gene associate with severe SAR-Cov2 infection, including death (9–12).
Two key aspects related to the increase in aging-related SARs-CoV-2 susceptibility is that the aging body is more fragile, and the aging immune system is markedly different from that of a youthful immune system. With respect to the former, aging leads to a slight imbalance between the wear and tear or damage that normally occurs in the body and the ability of the body to repair itself (13). Both nuclear and mitochondrial DNA repair mechanisms, for example, are essential to maintain cell and whole-body health (14). Defects in repair processes of DNA, such as base excision, nucleotide excision, recombination, and mismatch repair, decline with age, leading to DNA damage accumulation, which play a role in many aging-associated diseases. Interestingly, long-term treatment with ACE-inhibitors or angiotensin receptor blockers increase the lifespan in rodents (15–17). Irrespective of its role in decreasing vasoconstriction and limiting cardiac hypertrophy and associated adverse consequences of systemic hyperfunction (e.g., stroke, myocardial infarction and renal insufficiency), RAS blockade’s benefit in aging involves downregulation of mammalian or mechanistic target of rapamycin (mTOR) (18), prevention of mitochondrial decrease and/or damage and reduced formation of reactive oxygen species (19–22).
Proteins, which are a product of DNA, are also key to the maintenance of cell structure, repair of cellular damage, and catalytic reactions. Unfolded and misfolded proteins need to be degraded and eliminated. With aging there is a loss of protein homeostasis, or impaired proteostasis, which leads to “sticky” aggregates and contributes to age-related diseases such Alzheimer’s and Parkinson’s (23). Drugs targeted at augmenting DNA and protein repair processes, such as Metformin are currently being studied by translational geroscience investigators to offset age-related disease vulnerabilities (24), including that of Covid-19 (ClinicalTrials.gov. NCT04625985; Metformin glycinate, treatment of patients with COVID-19 and severe respiratory syndrome secondary to SAR-CoV-2). Interestingly, in a large retrospective study of adults with type 2 diabetes or obesity, metformin was associated with significantly decreased mortality in women admitted to the hospital with COVID-19, with no significant mortality reduction in men (25). As reviewed by Brandi and Giustina (26), and Bienvenu et al. (27) distinct features of older men as compared to women of same age, including more cardiovascular comorbidities, heightened effects of hypogonadism (i.e., increased inflammatory response, reduced muscular strength) as well hyperandrogenism from exogenous testosterone replacement (i.e., activation of virus entrance in the cells, increased venous thromboembolic events), and absence of biallelic expression of immunologic proteins on the X chromosome, which might impede humoral and cell-mediated immune responses to antigenic stimulation, may account for more severe complications and lethality in males versus females infected with SARS-CoV-2. Whether therapeutic dimorphism with respect to treatment contributes to differences in COVID-19 disease severity between sexes, is not yet known. To briefly review, the drugs approved (https://www.covid19treatmentguidelines.nih.gov/therapeutic-management/) or considered investigational (https://www.fda.gov/vaccines-blood-biologics/investigational-new-drug-ind-or-device-exemption-ide-process-cber/recommendations-investigational-covid-19-convalescent-plasma) in the management of SARS-CoV-2 positive patients outside of the hospital, at high risk for disease progression, include monoclonal antibodies (e.g., bamlanivimab and casirivimab plus imdevimab) while those hospitalized with COVID-19 might receive the antiviral redesivir, antibiotics (e.g., azithromycin), steroid treatment with dexamethasone, and/or convalescent plasma.
What we do know is that immune response mechanisms decline with normal aging. When SARS Cov-2 infects cells and replicates, a robust immune system is more likely to fend off the infection than an unhealthy immune system. Older people have weakened immune systems (Table 1), which often become overwhelmed by infectious insults such as influenza or SARs-CoV-2, resulting in prolonged convalescence, long-standing physical disabilities and premature death (28). The only way to limit the adverse effects of COVID-19 disease sequelae is prevention. That is, prevention in terms of maintaining healthy aging, following primary care provider recommendations, regular exercise, a healthy diet, and by practicing good hygiene, including physical distancing, wearing masks, and frequent hand washing. At the time of this review, the European Medicines Agency and the U.S. Food and Drug Administration commenced the distribution of the Pfizer/BioNTech and Moderna RNA-based coronavirus vaccine for emergency use to help protect against SAR2-CoV-2 infection and prevent the spread of Covid-19 pandemic, in conjunction with all above-mentioned preventive measures.
Table 1.
Effects of aging on innate and adaptive immunity and clinical consequences
| Innate Immunity | |||||
| Components | Skin and Mucous | Dendritic Cells | Natural Killer | Neutrophils | Macrophages |
| ↓ Replacement | ↓ Ability to stimulate lymphocytes | ↓ Cytotoxic capacity | ↓ Chemotaxis and phagocytosis | ↓ Expression and function of TLR | |
| ↓ Barrier function | ↓ Chemotaxis | ↓ Secretion of INF-𝛄 | ↓ Superoxide anion production | ↓ Production of MHC | |
| ↓ IgA after age 60 | ↓ Presentation of antigens | ↓ Response to IL-2 | ↓ Activation signal transduction | ↓ Production of TNF-𝛼 and IL-6 | |
| ↓ Production of INF | |||||
| Clinical consequence | ↓ mucociliary function → reduced cough, less viral clearance and mucous clearance | Loss of self-tolerance and increased reactivity to self; increase in autoantibodies | Slower resolution of inflammatory responses; increased incidence of bacterial and fungal infection | Delayed pathogen clearance; increase tissue damage through excess protease activity. Consistent with this, older adults experience greater end-organ damage after severe infections | Reduced fever; less malaise; less efficient viral clearance; reduced wound healing |
| Adaptive immunity | |||||
| Components | B lymphocytes | T lymphocytes | |||
| ↓ Antibody production | ↓ CD4+/CD8+ lymphocytes | ||||
| ↓ Lymphocyte number | ↓ Membrane receptors important in lymphocyte activation | ||||
| ↓ Production of IL-2 | |||||
| Clinical consequence | Reduced ability to effectively respond against viruses and bacteria; decreased antibody response to vaccination. | Reduced protection from vaccine; reduced longevity of protection from vaccine |
Table created using data from Fuentes E, Fuentes M, Alarcon M, Palomo I. Immune system dysfunction in the elderly. An Acad Bras Cienc 2017;89(1):285–299.
NOTE: IgA, Immunoglobulin A; INF- 𝛄, interferon gamma; IL-2, interleukin-2; INF, interferon; TLR, toll-like receptor; MHC, major histocompatibility complex; TNF-𝛼, tumor necrosis factor alpha; IL-6, interleukin-6
ACE2/RAS, aging and COVID-19
As ACE2 is the primary vector for cellular entry of SARS-CoV-2 infection and key component of the counterregulatory renin angiotensin system axis, it is valuable to review ACE2’s biochemical functions and potential role as a “player” in aging-related COVID-19 disease vulnerability and severity. The characterization of an angiotensin converting enzyme (ACE) homologue, named ACE2, functioning as a mono-carboxy peptidase and insensitive to pharmacological inhibition, expanded our knowledge of the biotransformation mechanisms of generating the biological active peptide hormones from the substrate angiotensinogen (29) (Figure 1). ACE2 is now recognized to modulate the net tissue activity of angiotensin II (Ang II) by virtue of its high hydrolytic activity in converting the peptide into angiotensin-(1–7) [Ang-(1–7)]. In turn, Ang-(1–7) by coupling into the Mas receptor initiates a signaling cascade which opposes profibrotic and oxidative stress actions of transforming growth factor beta/TGF-β1/Smad pathway and hypertrophic and proliferative changes in cardiac and arterial vasculature via inhibition of mitogen-activated protein kinase stimulation (16, 30). The counterbalancing axis of the renin angiotensin system (RAS), as originally proposed by Ferrario’s laboratory (31), constitutes a servomechanism limiting Ang II circulatory and tissue mechanisms from triggering imbalance. Acting as a mono-carboxypeptidase, ACE2 also forms Ang-1–9 through the hydrolysis of Ang I (7). This latter reaction is several hundred times slower than Ang II hydrolysis by ACE2 to form Ang(1–7) (32), yet may also have a role in ACE2 -mediated cardioprotection (33). ACE2 also cleaves several other non-RAS peptides including bradykinin into (des-Arg9)-bradykinin, a member of the kininogen-kinin system (34) which has potent inflammatory actions via activation of types 1 and 2 bradykinin receptors (35), apelin-13, a peptide proposed to cause vasoconstriction and known to regulate fluid homeostasis (36), and dynorphin A, one of several potential mediators of cardiovascular-related inflammation (37).
Figure 1.

Schematic of biochemical pathways to biologically active angiotensins. In the classical or canonical RAS pathway, the protease renin cleaves the substrate angiotensinogen to form angiotensin I (Ang I), and then, ACE removes two amino acids from Ang I to yield Ang II. Ang II can bind to the angiotensin type 1 receptor (AT1R) to exert actions, such as cell growth and proliferation, oxidative stress, inflammation, fibrosis and vasoconstriction. To offset these adverse effects, Ang II can be further degraded to Ang -(1–7), and Ang I can be metabolized by neprilysn to form Ang-(1–7). Ang-(1–7) binds to its receptor, Mas, to exert actions that counteract AT1R activation. In addition, Ang I can be hydrolyzed by ACE2 to form Ang-(1–9), and Ang-(1–9) is then metabolized by ACE to form Ang-(1–7). Recently, a noncanonical pathway leading to Ang II formation, via the conversion of Ang I or angiotensin-(1–12) [Ang-(1–12)] by chymase, was deemed to be the primary contributor in humans to tissue Ang II-induced CVD sequela (93).
Evidence for a role of the ACE2/Ang-(1–7)/Mas-receptor in cardiovascular disease pathogenesis is documented by the presence of low Ang-(1–7) expression in untreated essential hypertensive subjects (38, 39), association of blood pressure control with elevated Ang-(1–7) values in patients treated with ACE inhibitors (ACE-I) (40) or Ang II receptor blockers (ARBs) (41, 42), and increased expression of ACE2 in diseases of the heart and the blood vessels (34, 43–48).
Indeed, complexities in RAS biochemistry resurfaced when the harmful effects of ACE2 were linked to COVID-19 virus-mediated death. Individuals with severe SARs-CoV-2 infection have been shown to have high ACE2 and Ang II levels, suggesting a RAS-related role in COVID-19 pathogenesis. As ACE2 is the central molecular target that interacts with spike-like glycoproteins that are present on SARs-CoV-2 viral surface, high viral loads may lead to ACE2 tissue depletion. Whether this, in turn, triggers an increase in ACE2 transcription, with subsequent increases in nitric oxide and radical oxygen species from an exuberant formation of Ang-(1–7), bradykinin and des-Arg9-bradykinin, is not clear. However, critically ill COVID-19 patients in the intensive care unit are often in a state of refractory shock. Whether the cytokine storm underlying this shock state might be triggered by a feed-forward activation of the RAS and kallikrein system, remains speculative. However, given that kallikrein converts prorenin into renin (49), the interaction between the RAS and the kallikrein system becomes highly suspected to explain the severe vasodilatory, pro-inflammatory and procoagulant syndrome associated with COVID-19 disease severity (35).
How ACE2 might change during aging is likely dependent on multiple factors including, sex, tissue and cell type, and pathological conditions. In healthy humans, ACE2 expression decreases with age in colon, blood and adrenal gland, brain, nerve, adipose, and salivary gland in males, but only does so in the first three tissues in female (50). In human dermal fibroblasts, ACE2 expression is increased with age (51). In pathological conditions, no differences in activity of ACE2 in bronchoalveolar lavage fluid were noted between young and old patients with acute respiratory distress syndrome (ARDS) (52), while in ventilated patients, ACE2 strongly upregulated with increasing age (53). Increased ACE2 expression and activity has also been reported in diseases of the human heart and blood vessels (34, 43–48). Taken together with the fact that the heart is among the top ten organs that express ACE2 (54) (55), and that COVID-19 infection can result in myocardial complications (56–58), we retrospectively analyzed right atrial appendage tissue samples for ACE2 gene expression levels from 34 consented cardiac surgical patients [IRB #22619] and compared levels with respect to comorbid conditions and chronic medication usage (59). Our goal was to establish more definitive knowledge of how this tissue enzyme is altered in various pathological conditions and under different chronic medical regimens, particularly since high atrial expression of Ang II-forming substrates and chymase gene expression were found to be augmented in the left atria of patients with resistant atrial fibrillation (60). We hypothesized that an increase in ACE2 gene expression in the atria of patients with pre-existing chronic obstructive lung disease (COPD), atrial fibrillation (AF), hypertension and treated with ACEIs or ARBs could explain, in part, why the myocardium might be particularly vulnerable to the virus.
We summarize our findings herein to enlighten anesthesiologists and perioperative care physicians on possible biochemical reasons for increases in COVID-19 disease vulnerability among older adults. Indeed, age related cardiac syndromes such arrhythmias, myocarditis, and acute coronary syndrome are among the manifestations of the SARs-CoV-2 infection (61–64). Moreover, patients with preexisting cardiovascular disease, a common medical problem among older adults, have heightened vulnerabilities to SARs-CoV-2 and more dire consequences to COVID-19 (65–69).
In brief, our study cohort consisted of patients undergoing heart surgery for CABG (n=23), aortic valve replacement [AVR) (n=6)], CABG + AVR (n=4), and CABG + mitral valve replacement [MVR (n=1)]. The majority of patients had a history of hypertension (82%), and over a third had diabetes (38%). Other comorbidities included past or current history of AF (18%), COPD (15%) and stroke (15%). With regard to baseline medical therapy, 88% were on anti-thrombolytic agents, 76% were on statins, 68% were on beta-blockers, 49% were on ACE inhibitors (ACEIs) or angiotensin receptor blockers (ARBs), and 21% were on thiazide diuretics. Preoperative echocardiographic indices reveal that the baseline ejection fraction of the patients was within normal limits (median LVEF = 55 (IQR, 45– 55%)), while the right atrial diameter was on the upper end of normal (4.2 (IQR, 3.7 – 4.6 cm)). Atrial appendage ACE2 mRNA normalized to respective glyceraldehyde-3-phosphate dehydrogenase (GAPDH) values ranged from 0.49 – 37.01, and levels were positively related to age (Figure 2).
Figure 2.

Relationship between age and atrial appendage ACE2 gene transcripts obtained from 34 cardiac surgical patients.
Cardiac ACE2 and comorbid conditions
To determine if having a pre-existing condition increased atrial appendage ACE2 expression, analysis of variance was performed. Our results showed no effect of ACE2 for hypertension, diabetes, and atrial fibrillation. Patients with COPD and a stroke history had significantly higher cardiac ACE2 gene expression levels than those without COPD (P<0.003) or a previous stroke (P = 0.014). Figure 3 illustrates expression levels of ACE2 mRNA (normalized to respective GAPDH values) across comorbid conditions. As gene transcripts for renin, ACE, and chymase in the right atrial appendage tissue were also higher in COPD patients versus non-COPD patients (data not shown), the increase ACE2 might represent a compensatory mechanism. COPD and related pulmonary hypertension can increase right ventricular afterload and cause stretching of the right atria, leading to an activation of the local RAS (52). Also, since most COPD patients are/were smokers, it is worth noting that smoking can increase ACE2 expression in the human lung (56, 57).
Figure 3.

Histogram of atrial tissue ACE2 mRNA for patients with or without the following comorbid conditions: chronic obstructive pulmonary disease (COPD), stroke history, hypertension, diabetes, and kidney disease. Values represent mean and standard error of the mean. Astereisk (*) denotes a significant main effect for COPD vs. non-COPD and for previous stroke vs. no stroke history with respect to ACE2 gene levels.
Clinical data show the outcomes of SARS-CoV-2 infection are more severe in COPD patients (28, 58). A recent meta-analysis with a total of 2,002 cases reported that COPD presence associated with nearly a 4-fold higher risk of developing severe COVID-19, while death was reported in 60% of COPD patients compared to 34.3% of non-COPD patients (59). Whether COVID-19 severity in COPD patients is due to an indirect effect of elevated cardiac ACE2 on lung function is not known. It also remains to be determined whether there is a similar elevation in lung ACE2 expression in COPD vs. non-COPD individuals.
Having a previous stroke was also linked to higher ACE2 gene expression levels in our study. Atrial appendage tissue renin and ACE mRNA levels were also increased in stroke vs. non-stroke patients. Ang II is a key mediator by which hypertension exerts its deleterious vascular effects and hypertension is a major risk factor for stroke, with detrimental actions on the cerebral circulation which underlies most cerebrovascular diseases (60). In our small cohort, all patients with a history of stroke were also hypertensive, which might indicate an activated RAS leading to increased cardiac renin and ACE gene expression. In this case, the increase in ACE2 mRNA expression might signify a compensatory mechanism in response to an activated RAS. Although it is not clear from our findings what role an ACE2-to-stroke relationship might have in disease sequelae after SARS-CoV-2 infection, a pooled analysis of four clinical studies found that those patients with a stroke history had a 2.5-fold increase risk of having more severe-COVID-19 symptoms (61) than those without a stroke history. Understanding the full effect of the cardiac RAS, and specifically elevated ACE2, in predicting extracardiac sequelae from SARS-CoV-2 infection will require postmortem molecular and biochemical analyses of infected tissues from those who succumb to COVID-19.
Cardiac ACE2 and chronic RAS blockade
Because cardiac ACE2 levels have been shown to be influenced by chronic RAS blockade in animal studies (70, 71), we also sought to determine the impact of various drug regimens on ACE2 mRNA expression. Interestingly the chronic use of thrombolytic agents and thiazide diuretics led to significant elevations in ACE2 gene expression, whereas neither angiotensin receptor blockers (ARBs) nor angiotensin converting enzyme inhibitors (ACEIs) affected mRNA levels of this enzyme. This held true even after ACE2 mRNA levels vs. RAS inhibitors were compared in a stepwise manner to use of beta blockers, calcium channel blockers, nitrates, and thiazide diuretics. ACE2 gene expression levels also failed to associate with statin use.
It is interesting that ACEi and ARBs were not among the medications linked to elevated atrial tissue ACE2 gene expression. In contrast, Hu et al (72) found that ACE2 mRNA in atrial tissue AF not on ACEi. From a pharmacologic point of view, Ferrario et al (70, 71, 73) and others (74) have shown in preclinical models that blocking the ACE/Ang II/AT1R axis, through limiting the formation and actions of Ang II, potentiates the expression and, in some instances, the activity of ACE2 in the heart and other cardiovascular tissues. Whether differences in disease traits could account for discrepancies between these clinical studies remains uncertain. Even so, the finding that cardiac ACE2 gene expression failed to relate to chronic use of RAS blockade provides insight into the ongoing discussion regarding RAS inhibitors and COVID-19 vulnerabilities. That is, if RAS inhibition leads to upregulation of ACE2, acute respiratory syndrome and myocarditis in SARS-CoV-2‒infected patients might be attenuated by reducing Ang II formation and blocking its actions at the level of the angiotensin type-1 receptor, and conceivably through the local formation of Ang-(1–7). Paradoxically, an increase in ACE2 expression may facilitate virus access into host tissues, thus aggravating the clinical picture. To date, a small observational study (42 patients) and one comprehensive monitoring study involving 576 COVID-19 patients report favorable effects with respect to COVID-19 outcomes in patients on RAS blockers (75, 76), while findings from several large retrospective studies and two meta-analyses of the literature show ACEI/ARB use is not associated with an increase or decrease in adverse events from COVID-19 (77–81). Moreover, in the BRACE-CORONA trial, the first randomized data assessing the role of continuing versus stopping ACE inhibitors and ARBs in patients hospitalized with COVID-19, suspending ACEIs and ARBs for 30 days did not impact the number of days alive and out of hospital (82).
These findings strengthen the recommendations of the American Heart Association and international regulatory agencies about not withdrawing or switching ACEI/ARB treatments to modify COVID-19 prognosis. Until prospective, randomized-controlled studies are completed that investigate the extent of risk reduction for in-hospital death from COVID-19, a de novo trial of RAS inhibitor initiation in patients hospitalized with COVID-19 is not advocated.
Cardiac ACE2 and non-RAS related medication
Anti-thrombotic agents are another class of medications commonly used by older patients with ischemic and valvular heart disease; use of these medications were associated with elevated ACE2 gene expression in our study. To our knowledge, no clinical or basic science reports have described the potential modulation of tissue ACE2 expression or activity by antithrombotic therapy. Whether our finding could be confounded by underlying pathological conditions common to cardiovascular disease patients, such as atherosclerosis, should be considered. Previous studies have reported that ACE2 is capable of modulating thrombus formation. Fraga-Silva et al (83) demonstrated in spontaneously hypertensive rats that a decrease in ACE2 activity in thrombi was associated with an increase in thrombus formation, as well as pharmacologic activation of ACE2-attenuated platelet attachment of vessels and thrombus formation. ACE2 overexpression has also been shown in vitro and in vivo to inhibit the development of early atherosclerotic lesions in rabbit aortas (84, 85), while ACE2 deletion, either whole-body deletion or in bone marrow-derived cells, increased the development of atherosclerosis in LDL receptor-null mice (86). Increases in ACE2 expression or activity within a thrombus might also represent a counterregulatory response within atherosclerotic plaques to macrophage-related increases in proinflammatory cytokines (87). In the context of COVID-19, coagulopathic complications are common (88), and anti-coagulant and anti-thrombotic therapies have proven beneficial, as evidenced by the Antithrombotic Therapy to Ameliorate Complications of COVID-19 trial (NCT04372589). Given this, and the positive link between cardiac ACE2 gene expression and anti-thrombotic use observed in our cardiac surgical patient cohort, postmortem studies focused on COVID-19 pathogenicity are needed to determine whether genes encoding proteins involved in viral entry (e.g., ACE2, transmembrane protease serine 2 (TMPRSS2), and ADAM metallopeptidase domain 17 (ADAM17)), and/or the ACE/Ang II/AT1R and/or the interferon/interleukin-related pathways (e.g., interleukin-6) are up or down-regulated in tissues post-thrombotic syndrome and if the expression of these genes correlate with anti-thrombotic treatment. This information could tell us whether direct viral entry is involved in the thrombus formation or if the procoagulant state of the tissue is a reflection of RAS activation and cytokine-mediated inflammation. Some drugs are already available, or are being repurposed, to dissect these pathways that complicate COVID-19 infection (89), with the aim of preventing viral entry, halting viral replication, and limiting tissue damage.
Thiazide diuretics were also positively associated with atrial appendage ACE2 gene expression. Although only 7 of 34 patients were taking thiazides in our small cohort, including hydrochlorothiazide (HCTZ) and chlorthalidone, association of thiazide use with local ACE2 mRNA expression levels is consistent with previous work. In studies by the Ferrario laboratory (90), chronic low-dose HCTZ treatment increased cardiac ACE2 mRNA and activity in normotensive rats, an effect that was not related to its anti-hypertensive actions. The inhibitory effect of HCTZ on carbonic anhydrase activity (91) may account for its interaction with tissue ACE2. Using a systems biology approach, Emameh and colleagues (92) recently reported co-expression of ACE2 with carbonic anhydrase and neprilysin, another RAS-related enzyme that can produce Ang-(1–7), in various human tissues and its potential pathogenesis of SARS-CoV-2. Although we do not know if an ACE2-neprilysin-carbonic anhydrase protein network is affected by thiazide use in our cardiac surgical patients, this possibility is important to consider in the quest to improve our understanding of the relationship between ACE2 and COVID-19 susceptibility, disease progression, and treatment.
Conclusions
While it is not yet established if older individuals are more vulnerable to SARs-CoV-2 infection, they do have worse outcomes once infected when compared to younger adults and children. Normal declines in the biological aging process, such as increases in fragility in biological repair systems and a weakened immune system are deemed to be partially responsible for symptomatic COVID-19 spikes among older adults. Increased SARs-CoV-2 infection prevalence has also been reported to occur in non-O blood types, and disease severity (e.g., increased risk of mechanical ventilation, continuous renal replacement, and prolonged intensive care stay (56) has been linked to blood-type A. Our finding that ACE2 atrial tissue gene transcript increases with age, COPD, stroke history, and chronic use of anti-thrombotic agents and thiazide diuretics, but not ACEI or ARBs, of older cardiac surgery patients becomes provocative. Although there is no direct evidence of the connection of atrial tissue ACE2 mRNA and cardiac complications from SARS-CoV-2 infection, we believe our findings offer some insight into what conditions and drugs may lead to increased ACE2 expression, which in turn might influence the degree of SARS-CoV-2 infection, disease progression and possibly associated cardiac complications of older adults (Figure 4). The information we provide is not meant to change clinical practice of physician anesthesiologists and intensivists who are on the front lines of the COVID-19 pandemic, but rather to enhance the clinician’s understanding of how the functional existence of a disturbance in neurohormonal regulation of tissue RAS might influence the link of an augmented risk for SARs-CoV-2 infection in older adults with pre-existing cardiovascular conditions.
Figure 4.

Visual summary of clinical data showing patient factors that may modulate cardiac ACE2 gene expression levels. Higher ACE2 expression occurred with preexisting COPD (P<0.003) and stroke history (P=0.014), but not atrial fibrillation, hypertension, or diabetes. ACE2 expression also increased by chronic intake of thrombolytic agents (P=0.005) and thiazide diuretics (P=0.03), but not RAS inhibitors or statins. As a reminder, this was a small observational study involving patients who underwent cardiac surgery and, thus, was not designed to establish a causal relationship between ACE2 expression and COVID-19 disease severity. Even so, our findings offer insight into conditions and drugs that may lead to ↑ACE2 expression, which in turn might influence susceptibility to SARS-CoV-2 infection, COVID-19 progression, and associated cardiac complications in older adults.
Key points.
The presence of age-related comorbidities, such as heart disease, COPD, and stroke, associate with COVID-19 disease severity.
Biological changes that occur with advancing age, including impaired DNA repair systems, loss of protein homeostasis and a weakened immune system, may underlie the increases in aging-related vulnerability to SARS-CoV-2 infection.
SARs-CoV-2 infection severity has been linked to blood-type A.
Cardiac ACE2 may have bifunctional roles in COVID-19 since ACE2 mediates cellular susceptibility to SARS-CoV-2 infection, but also protects the heart via the ACE2/Ang-(1–7) pathway.
ACE2 atrial tissue gene transcripts from older cardiac surgery patients increase with age, COPD, stroke history, and chronic use of anti-thrombotic agents and thiazide diuretics, but not ACEI or ARBs.
Acknowledgements
The authors would like to thank Jessica Von Cannon and Kendra N. Wright for their technical support in various aspects of the clinical study, including patient recruitment, informed-consent, tissue processing, and data management. We also acknowledge Dr. Sayaka Nagata’s effort for commencing RAS biochemistry studies involving atrial appendage tissue from consented cardiac surgical patients.
Financial support and sponsorship
This work was supported in part by the National Institutes of Health [HL-051952 to CMF and LG; AG042758 and AG033727 to LG] and the Department of Anesthesiology at Wake Forest School of Medicine, Winston-Salem, North Carolina.
Abbreviations
- ACE2
Angiotensin converting enzyme-2
- ACE
angiotensin converting enzyme
- RAS
renin angiotensin system
- COPD
chronic obstructive pulmonary disease
- COVID-19
coronavirus disease 2019
- SARS-CoV-2
Severe Acute Respiratory Syndrome Coronavirus 2
- Ang II
angiotensin II
- ACE-I
ACE inhibitors
- ARBs
Ang II receptor blockers
Footnotes
Conflicts of Interest
None
References
- 1.Verity R, Okell LC, Dorigatti I, Winskill P, Whittaker C, Imai N, et al. Estimates of the severity of coronavirus disease 2019: a model-based analysis. Lancet Infect Dis. 2020;20(6):669–77. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Valenti L, Villa S, Baselli G, Temporiti R, Bandera A, Scudeller L, et al. Association of ABO blood group and secretor phenotype with severe COVID-19. Transfusion. 2020;60(12):3067–70.* This study shows that carriage of non-O blood groups predisposes to severe COVID-19 with respiratory failure in European individuals and that the secretor phenotype may moderate disease progression.
- 3.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. 2020;383(16):1522–34.* This study identified a 3p21.31 gene cluster as a genetic susceptibility locus in patients with Covid-19 with respiratory failure and confirmed a potential involvement of the ABO blood-group system.
- 4.Zietz M, Zucker J, Tatonetti NP. Associations between blood type and COVID-19 infection, intubation, and death. Nat Commun. 2020;11(1):5761.** Observational healthcare data on 14,112 individuals tested for SARS-CoV-2 with known blood type in the New York Presbyterian (NYP) hospital system assessed the association between ABO and Rh blood types and infection, intubation, and death.
- 5.Golinelli D, Boetto E, Maietti E, Fantini MP. The association between ABO blood group and SARS-CoV-2 infection: A meta-analysis. PLoS One. 2020;15(9):e0239508.** This meta-analysis (7 studies) involving 7503 SARS-CoV-2 positive cases and 2962160 controls shows that SARS-CoV-2 positive individuals are more likely to have blood group A and less likely to have blood group O.
- 6.Donoghue M, Wakimoto H, Maguire CT, Acton S, Hales P, Stagliano N, et al. Heart block, ventricular tachycardia, and sudden death in ACE2 transgenic mice with downregulated connexins. J Mol Cell Cardiol. 2003;35(9):1043–53. [DOI] [PubMed] [Google Scholar]
- 7.Donoghue M, Hsieh F, Baronas E, Godbout K, Gosselin M, Stagliano N, et al. A novel angiotensin-converting enzyme-related carboxypeptidase (ACE2) converts angiotensin I to angiotensin 1–9. Circ Res. 2000;87(5):E1–E9. [DOI] [PubMed] [Google Scholar]
- 8.Zhang H, Penninger JM, Li Y, Zhong N, Slutsky AS. Angiotensin-converting enzyme 2 (ACE2) as a SARS-CoV-2 receptor: molecular mechanisms and potential therapeutic target. Intensive Care Med. 2020;46(4):586–90.* This review article summarizes the evidence that ACE2 is a receptor for SARs-CoV-2, which might be a potential target for COVID-19 treatment.
- 9.Tenforde MW, Billig Rose E, Lindsell CJ, Shapiro NI, Files DC, Gibbs KW, et al. Characteristics of Adult Outpatients and Inpatients with COVID-19 – 11 Academic Medical Centers, United States, March-May 2020. MMWR Morb Mortal Wkly Rep. 2020;69(26):841–6.* The study involving 11 academic medical centers found that inpatients were older than outpatients, and more likely to be Hispanic.
- 10.Kuo CL, Pilling LC, Atkins JC, Masoli J, Delgado J, Tignanelli C, et al. COVID-19 severity is predicted by earlier evidence of accelerated aging. medRxiv. 2020.* Biological aging as captured by PhenoAge, is a better predictor of COVID-19 severity than biological age.
- 11.Kuo CL, Pilling LC, Atkins JL, Masoli JAH, Delgado J, Kuchel GA, et al. ApoE e4e4 Genotype and Mortality With COVID-19 in UK Biobank. J Gerontol A Biol Sci Med Sci. 2020;75(9):1801–3.* ApoE-e4e4 genotype is associated with COVID-19 test positivity at genome wide significance, and associated with a 4-fold increase in mortality after testing positive for COVID-19, in UK Bank.
- 12.Hultström M, Persson B, Eriksson O, Lipcsey M, Frithiof R, Nilsson B. Blood type A associates with critical COVID-19 and death in a Swedish cohort. Crit Care. 2020;24(1):496.** In a Swedish cohort, blood type A or AB associated with increased risks of requiring critical care or death from COVID-19.
- 13.Harman D The aging process. Proc Natl Acad Sci U S A. 1981;78(11):7124–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Chen Y, Geng A, Zhang W, Qian Z, Wan X, Jiang Y, et al. Fight to the bitter end: DNA repair and aging. Ageing Res Rev. 2020;64:101154.* This review discusses the relationships among DNA damage, DNA repair, aging and aging associated diseases, and the possible role of DNA repair in several potential rejuvenation strategies.
- 15.Inserra F, Romano L, Ercole L, de Cavanagh EM, Ferder L. Cardiovascular changes by long-term inhibition of the renin-angiotensin system in aging. Hypertension. 1995;25(3):437–42. [DOI] [PubMed] [Google Scholar]
- 16.Santos EL, de Picoli Souza K, da Silva ED, Batista EC, Martins PJ, D’Almeida V, Pesquero JB. Long term treatment with ACE inhibitor enalapril decreases body weight gain and increases life span in rats. Biochem Pharmacol 2009;78(8):951–58. [DOI] [PubMed] [Google Scholar]
- 17.Basso N, Cini R, Pietrelli A, Ferder L, Terragno NA, Inserra F. Protective effect of long-term angiotensin II inhibition. Am J Physiol Heart Circ Physiol. 2007;293(3):H1351–8. [DOI] [PubMed] [Google Scholar]
- 18.de Cavanagh EM, Inserra F, Ferder L. Angiotensin II blockade: how its molecular targets may signal to mitochondria and slow aging. Coincidences with calorie restriction and mTOR inhibition. Am J Physiol Heart Circ Physiol. 2015;309(1):H15–44. [DOI] [PubMed] [Google Scholar]
- 19.Ferder L, Romano LA, Ercole LB, Stella I, Inserra F. Biomolecular changes in the aging myocardium: the effect of enalapril. Am J Hypertens. 1998;11(11 Pt 1):1297–304. [DOI] [PubMed] [Google Scholar]
- 20.de Cavanagh EM, Piotrkowski B, Basso N, Stella I, Inserra F, Ferder L, et al. Enalapril and losartan attenuate mitochondrial dysfunction in aged rats. Faseb j. 2003;17(9):1096–8. [DOI] [PubMed] [Google Scholar]
- 21.de Cavanagh EM, Flores I, Ferder M, Inserra F, Ferder L. Renin-angiotensin system inhibitors protect against age-related changes in rat liver mitochondrial DNA content and gene expression. Exp Gerontol. 2008;43(10):919–28. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.de Cavanagh EM, Piotrkowski B, Fraga CG. Concerted action of the renin-angiotensin system, mitochondria, and antioxidant defenses in aging. Mol Aspects Med. 2004;25(1–2):27–36. [DOI] [PubMed] [Google Scholar]
- 23.Koga H, Kaushik S, Cuervo AM. Protein homeostasis and aging: The importance of exquisite quality control. Ageing Res Rev. 2011;10(2):205–15. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Justice JN, Gubbi S, Kulkarni AS, Bartley JM, Kuchel GA, Barzilai N. A geroscience perspective on immune resilience and infectious diseases: a potential case for metformin. Geroscience. 2020:1–20.* This review discusses the potential benefits of metformin in limiting various hallmarks of aging.
- 25.Bramante C, Ingraham N, Murray T, Marmor S, Hoversten S, Gronski J, et al. Observational Study of Metformin and Risk of Mortality in Patients Hospitalized with Covid-19. medRxiv. 2020.* Metformin is significantly associated with reduced mortality in women with obesity or T2DM in observational analyses of claims data from individuals hospitalized with Covid-19.
- 26.Brandi ML, Giustina A. Sexual Dimorphism of Coronavirus 19 Morbidity and Lethality. Trends Endocrinol Metab. 2020;31(12):918–27.** This review article discusses the differences in aging males versus aging females and the role of sex hormones in key phenotypes of COVID-19 infection.
- 27.Bienvenu LA, Noonan J, Wang X, Peter K. Higher mortality of COVID-19 in males: sex differences in immune response and cardiovascular comorbidities. Cardiovasc Res. 2020;116(14):2197–206.** This review article discusses biological sex as a fundamental variable of critical relevance to our mechanistic understanding of SARS-CoV-2 infection and the pursuit of effective COVID-19 preventative and therapeutic strategies.
- 28.Gravenstein S, Pop-Vicas A, Ambrozaitis A. The 2009 A/H1N1 pandemic influenza and the nursing home. Med Health R I. 2010;93(12):382–4. [PubMed] [Google Scholar]
- 29.Tipnis SR, Hooper NM, Hyde R, Karran E, Christie G, Turner AJ. A human homolog of angiotensin-converting enzyme. Cloning and functional expression as a captopril-insensitive carboxypeptidase. J Biol Chem. 2000;275(43):33238–43. [DOI] [PubMed] [Google Scholar]
- 30.Ferrario CM, Cheng CP, Varajic J. Angiotensin-(1–7) and the Heart. In: Santos RAS, editor. Angiotensin-(1–7): A Comprehensive Review : Springer International Publishing; 2019. p. 83–100. [Google Scholar]
- 31.Ferrario CM, Chappell MC, Tallant EA, Brosnihan KB, Diz DI. Counterregulatory actions of angiotensin-(1–7). Hypertension. 1997;30(3 Pt 2):535–41. [DOI] [PubMed] [Google Scholar]
- 32.Vickers C, Hales P, Kaushik V, Dick L, Gavin J, Tang J, et al. Hydrolysis of biological peptides by human angiotensin-converting enzyme-related carboxypeptidase. J Biol Chem. 2002;277(17):14838–43. [DOI] [PubMed] [Google Scholar]
- 33.Ocaranza MP, Jalil JE. Protective role of the ACE2/Ang-(1–9) axis in cardiovascular remodeling. Int J Hypertens. 2012;2012:594361. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Turner AJ, Tipnis SR, Guy JL, Rice G, Hooper NM. ACEH/ACE2 is a novel mammalian metallocarboxypeptidase and a homologue of angiotensin-converting enzyme insensitive to ACE inhibitors. Can J Physiol Pharmacol. 2002;80(4):346–53. [DOI] [PubMed] [Google Scholar]
- 35.Sodhi CP, Wohlford-Lenane C, Yamaguchi Y, Prindle T, Fulton WB, Wang S, et al. Attenuation of pulmonary ACE2 activity impairs inactivation of des-Arg(9) bradykinin/BKB1R axis and facilitates LPS-induced neutrophil infiltration. Am J Physiol Lung Cell Mol Physiol. 2018;314(1):L17–L31. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Kazemi-Bajestani SM, Patel VB, Wang W, Oudit GY. Targeting the ACE2 and Apelin Pathways Are Novel Therapies for Heart Failure: Opportunities and Challenges. Cardiol Res Pract. 2012;2012:823193. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Cissom C, J JP, Shariat-Madar Z. Dynorphins in Development and Disease: Implications for Cardiovascular Disease. Curr Mol Med. 2020;20(4):259–74. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Ferrario CM, Martell N, Yunis C, Flack JM, Chappell MC, Brosnihan KB, et al. Characterization of angiotensin-(1–7) in the urine of normal and essential hypertensive subjects. Am J Hypertens. 1998;11(2):137–46. [DOI] [PubMed] [Google Scholar]
- 39.Ferrario CM, Smith RD, Brosnihan B, Chappell MC, Campese VM, Vesterqvist O, et al. Effects of omapatrilat on the renin-angiotensin system in salt-sensitive hypertension. Am J Hypertens. 2002;15(6):557–64. [DOI] [PubMed] [Google Scholar]
- 40.Luque M, Martin P, Martell N, Fernandez C, Brosnihan KB, Ferrario CM. Effects of captopril related to increased levels of prostacyclin and angiotensin-(1–7) in essential hypertension. J Hypertens. 1996;14(6):799–805. [DOI] [PubMed] [Google Scholar]
- 41.Schindler C, Brosnihan KB, Ferrario CM, Bramlage P, Maywald U, Koch R, et al. Comparison of inhibitory effects of irbesartan and atorvastatin treatment on the renin angiotensin system (RAS) in veins: a randomized double-blind crossover trial in healthy subjects. J Clin Pharmacol. 2007;47(1):112–20. [DOI] [PubMed] [Google Scholar]
- 42.Schindler C, Ferrario CM. Olmesartan for the treatment of arterial hypertension. Future Cardiol. 2008;4(4):357–72. [DOI] [PubMed] [Google Scholar]
- 43.Burrell LM, Johnston CI, Tikellis C, Cooper ME. ACE2, a new regulator of the renin-angiotensin system. Trends Endocrinol Metab. 2004;15(4):166–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Chappel MC, Ferrario CM. ACE and ACE2: their role to balance the expression of angiotensin II and angiotensin-(1–7). Kidney Int. 2006;70(1):8–10. [DOI] [PubMed] [Google Scholar]
- 45.Crackower MA, Sarao R, Oudit GY, Yagil C, Kozieradzki I, Scanga SE, et al. Angiotensin-converting enzyme 2 is an essential regulator of heart function. Nature. 2002;417(6891):822–8. [DOI] [PubMed] [Google Scholar]
- 46.Ferrario CM. Angiotensin-converting enzyme 2 and angiotensin-(1–7): an evolving story in cardiovascular regulation. Hypertension. 2006;47(3):515–21. [DOI] [PubMed] [Google Scholar]
- 47.Penninger J, Imai Y, Kuba K. The discovery of ACE2 and its role in acute lung injury. Exp Physiol. 2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Goulter AB, Goddard MJ, Allen JC, Clark KL. ACE2 gene expression is up-regulated in the human failing heart. BMC Med. 2004;2:19. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Schmaier AH. The plasma kallikrein-kinin system counterbalances the renin-angiotensin system. J Clin Invest. 2002;109(8):1007–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Chen J, Jiang Q, Xia X, Liu K, Yu Z, Tao W, et al. Individual variation of the SARS-CoV-2 receptor ACE2 gene expression and regulation. Aging Cell. 2020;19(7).* This study analyzed ACE2 gene expression levels across thousands of individuals and tissues, and found significantly higher levels in Asian females, an age-dependent decrease in all ethnic groups, and a highly significant decrease in type-II diabetic patients.
- 51.Bickler SW, Cauvi DM, Fisch KM, Prieto JM, Gaidry AD, Thangarajah H, et al. Age is associated with increaesed expression of pattern recognition receptor genes and ACE2, the receptor for SARS-COV-2: Implications for the epidemiology of COVID-19 disease bioRxiv. 2020:2020.06.15.134403.* Using a large dataset of genome-wide RNA-seq profiles derived from human dermal fibroblasts (GSE113957), this study revealed that older age was associated with increased expression of pattern recognition receptor (PRR) genes, ACE2 and four genes that encode proteins that have been shown to interact with SARs-CoV-2 proteins.
- 52.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. 2019;9(1):55. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Baker SA, Kowk S, Berry GJ, Montine TJ. Angiotensin-converting enzyme 2 (ACE2) expression increases with age in patients requiring mechanical ventilation. medRxiv. 2020:2020.07.05.20140467.* ACE2 expression levels were measured by immunohistochemistry in the lung tissues of patients with acute hypoxic respiratory failure. In patients receiving mechanical ventilation, ACE2 was strongly upregulated with age, while in non-ventilated patients, ACE2 did not change with age.
- 54.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. 2020;9(1):45.* This study analyzed the GTEx RNA-Seq gene expression profiling datasets (RSEM normalized) for normal adult human tissues from the UCSC Xena project and found that ACE2 expression levels were highest in the small intestine, testis, kidney, heart, thyroid, and adipose tissue, and lowest in the blood, spleen, bone marrow, brain, blood vessels, and muscle. Medium expression levels of ACE2 were noted in the lungs, colon, liver, bladder, and adrenal gland.
- 55.Dai YJ, Hu F, Li H, Huang HY, Wang DW, Liang Y. A profiling analysis on the receptor ACE2 expression reveals the potential risk of different type of cancers vulnerable to SARS-CoV-2 infection. Ann Transl Med. 2020;8(7):481.* This study analyzed the Tissue Atlas database for numerous expression profiles of RNA and protein expressed in human tissues. Of all organs, the lung, heart, kidney, bladder and esophagus have the highest levels of ACE2 expression among healthy individuals.
- 56.Lala A, Johnson KW, Januzzi JL, Russak AJ, Paranjpe I, Richter F, et al. Prevalence and Impact of Myocardial Injury in Patients Hospitalized With COVID-19 Infection. J Am Coll Cardiol. 2020;76(5):533–46.** Covid-19 Patients with cardiovascular disease (CVD) are more likely to have myocardial injury than patients without CVD. Troponin elevation among patients hospitalized with COVID-19 is associated with higher risk of mortality.
- 57.Boukhris M, Hillani A, Moroni F, Annabi MS, Addad F, Ribeiro MH, et al. Cardiovascular Implications of the COVID-19 Pandemic: A Global Perspective. Can J Cardiol. 2020;36(7):1068–80.* This article provides a comprehensive overview of the pathophysiology and cardiovascular implications of COVID-19, its impact on existing pathways of care, the role of modern technologies to tackle the pandemic, and a proposal of novel management algorithms for the most common acute cardiac conditions.
- 58.Hachim MY, Al Heialy S, Senok A, Hamid Q, Alsheikh-Ali A. Molecular Basis of Cardiac and Vascular Injuries Associated With COVID-19. Front Cardiovasc Med. 2020;7:582399.* Findings from the analysis of publicly available transcriptomic datasets identified shared core genes pertinent to cardiac and vascular-related injuries and their probable role in genetic susceptibility to cardiovascular injury in patients with COVID-19.
- 59.Groban L WH, Saha AK, Sun X, Segal S, Kon N, Ferrario CM. Cardiac ACE2: Not just a marker of cardiovascular disease. American Society of Anesthesiologists, 2020, #BOC08 Available at: http;//wwwasaabstractscom/strands/assaabstracts/abstractArchivehtm Accessed October 26, 2020. 2020(BOC08).* This study found that ACE2 mRNA in atrial appendage tissue from 34 cardiac surgical patients was associated with age, elevated by COPD and previous stroke, and increased in conjunction with patients’ chronic use of anti-thrombotic agents and thiazide diuretics, but not drugs that block the renin angiotensin system.
- 60.Ahmad S, Simmons T, Varagic J, Moniwa N, Chappell MC, Ferrario CM. Chymase-dependent generation of angiotensin II from angiotensin-(1–12) in human atrial tissue. PLoS One. 2011;6(12):e28501. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Adão R, Guzik TJ. Inside the heart of COVID-19. Cardiovasc Res. 2020;116(6):e59–e61.** This review article discusses the link between old age and age-related cardiovascular disease, including hypertension, to severity of COVID-19.
- 62.Guzik TJ, Mohiddin SA, Dimarco A, Patel V, Savvatis K, Marelli-Berg FM, et al. COVID-19 and the cardiovascular system: implications for risk assessment, diagnosis, and treatment options. Cardiovasc Res. 2020;116(10):1666–1687.** The mortality of COVID-19 is increased by comorbidities including cardiovascular disease, hypertension, diabetes, chronic pulmonary disease, and cancer. The most common complications from COVID-19 infection include arrhythmias (atrial fibrillatiion, ventricular tachyarrhythmias, and ventricular fibrillation), cardiac injury (elevations in highly sensitive troponin I and creatine kinase), fulminant myocarditis, heart failure, pulmonary embolism, and disseminated intravascular coagulation.
- 63.Long B, Brady WJ, Koyfman A, Gottlieb M. Cardiovascular complications in COVID-19. Am J Emerg Med. 2020;38(7):1504–1507.* COVID-19 can result in systemic inflammation, multiorgan dysfunction, and critical illness. As discussed in this review, the cardiovascular system is also affected, with complications including myocardial injury, myocarditis, acute myocardial infarction, heart failure, dysrhythmias, and venous thromboembolic events.
- 64.Mehra MR, Ruschitzka F. COVID-19 Illness and Heart Failure: A Missing Link? JACC Heart Fail. 2020;8(6):512–4.** This article discusses how age and age-related cardiovascular diseases such as diabetes, hypertension, coronary heart disease, and associated treatments affect the severity and prognosis of COVID-19.
- 65.Wu C, Chen X, Cai Y, Xia J, Zhou X, Xu S, et al. Risk Factors Associated With Acute Respiratory Distress Syndrome and Death in Patients With Coronavirus Disease 2019 Pneumonia in Wuhan, China. JAMA Intern Med. 2020180(7):934–43.** Analysis from 201 COVID-19 patients show that older age and age-related cardiovascular diseases associated with a greater risk of development of acute respiratory distress syndrome and death, likely due to a less rigorous immune response.
- 66.Huang C, Wang Y, Li X, Ren L, Zhao J, Hu Y, et al. Clinical features of patients infected with 2019 novel coronavirus in Wuhan, China. Lancet. 2020;395(10223):497–506.* This is an early publication from the COVID-19 pandemic which analyzed data from 41 hospitalized COVID-19-positive patients. Their findings were one of the first to reveal that patients with pre-existing cardiovascular disease might have heightened vulnerabilities and more dire consequences to SARs-CoV-2 infection.
- 67.Wang D, Hu B, Hu C, Zhu F, Liu X, Zhang J, et al. Clinical characteristics of 138 hospitalized patients with 2019 novel coronavirus-infected pneumonia in Wuhan, China. JAMA. 2020;323(11);1061–69.* This is an early report from China, which provided evidence from 138 hospitalized patients with COVID-19 that hypertension, diabetes, cardiac disease, and malignancy were among the most common coexisting conditions in their SARs-CoV-2 infected patients.
- 68.Guan WJ, Ni ZY, Hu Y, Liang WH, Ou CQ, He JX, et al. Clinical Characteristics of Coronavirus Disease 2019 in China. N Engl J Med. 2020;382(18):1708–20.* This study analyzed 1099 patients with laboratory-confirmed COVID-19. Hypertension and coronary heart disease were among the most common coexisting conditions.
- 69.Tadic M, Cuspidi C, Mancia G, Dell’Oro R, Grassi G. COVID-19, hypertension and cardiovascular diseases: Should we change the therapy? Pharmacol Res. 2020;158:104906.* This review summarizes the prevalence of hypertension and cardiovascular disease in patients with COVID-19, and their influeence on the progression and the prognosis, and the effect of treatment of hypertension and CVD, especially using the inhibitors of the renin-angiotensin-aldosterone system in COVID-19 patients.
- 70.Ferrario CM, Jessup J, Chappell MC, Averill DB, Brosnihan KB, Tallant EA, et al. Effect of angiotensin-converting enzyme inhibition and angiotensin II receptor blockers on cardiac angiotensin-converting enzyme 2. Circulation. 2005;111(20):2605–10. [DOI] [PubMed] [Google Scholar]
- 71.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. 2004;43(5):970–6. [DOI] [PubMed] [Google Scholar]
- 72.Hu XS, Xie XD, Wang XX, Zeng CL, Ni YM, Yu GW, et al. [Effects of angiotensin converting enzyme inhibitor on the expression of angiotensin converting enzyme 2 in atrium of patients with atrial fibrillation]. Zhonghua Xin Xue Guan Bing Za Zhi. 2007;35(7):625–8. [PubMed] [Google Scholar]
- 73.Igase M, Strawn WB, Gallagher PE, Geary RL, Ferrario CM. Angiotensin II AT1 receptors regulate ACE2 and angiotensin-(1–7) expression in the aorta of spontaneously hypertensive rats. Am J Physiol Heart Circ Physiol. 2005;289(3):H1013–H9. [DOI] [PubMed] [Google Scholar]
- 74.Ocaranza MP, Godoy I, Jalil JE, Varas M, Collantes P, Pinto M, et al. Enalapril attenuates downregulation of angiotensin-converting enzyme 2 in the late phase of ventricular dysfunction in myocardial infarcted rat. Hypertension. 2006;48(4):572–8. [DOI] [PubMed] [Google Scholar]
- 75.Meng J, Xiao G, Zhang J, He X, Ou M, Bi J, et al. Renin-angiotensin system inhibitors improve the clinical outcomes of COVID-19 patients with hypertension. Emerg Microbes Infect. 2020;9(1):757–60.* COVID-19 patients receiving ACE-inhibitors or ARBs had a lower rate of severe disease and a trend toward a lower level of IL-6 in peripheral blood.
- 76.Cippà PE, Cugnata F, Ferrari P, Brombin C, Ruinelli L, Bianchi G, et al. A data-driven approach to identify risk profiles and protective drugs in COVID-19. Proc Natl Acad Sci U S A. 2021;118(1).** Using a survival tree, data driven approach based on comprehensive monitoring of 576 COVID-19 patients, the protective role of renin angiotensin inhibitor usage is endorsed.
- 77.Mancia G, Rea F, Ludergnani M, Apolone G, Corrao G. Renin-Angiotensin-Aldosterone System Blockers and the Risk of Covid-19. N Engl J Med. 2020;382(25):2431–40.* In this large, population-based study, the use of ACE inhibitors and ARBs was more frequent among patients with Covid-19 than among controls because of their higher prevalence of cardiovascular disease. However, there was no evidence that ACE inhibitors or ARBs affected the risk of COVID-19.
- 78.Reynolds HR, Adhikari S, Pulgarin C, Troxel AB, Iturrate E, Johnson SB, et al. Renin-Angiotensin-Aldosterone System Inhibitors and Risk of Covid-19. N Engl J Med. 2020;382(25):2441–8.** This study found no substantial increase in the likelihood of a positive test for Covid-19 or in the risk of severe Covid-19 among patients who tested positive in association with five common classes of antihypertensive medications.
- 79.COVID-19 RISk and Treatments (CORIST) Collaboration. RAAS inhibitors are not associated with mortality in COVID-19 patients: Findings from an observational multicenter study in Italy and a meta-analysis of 19 studies. Vascul Pharmacol. 2020;135:106805.** In this observational study and meta-analysis of the literature, ACE-I or ARB use failed to associate with severity or in-hospital mortality in COVID-19 patients.
- 80.Zhang G, Wu Y, Xu R, Du X. Effects of renin-angiotensin-aldosterone system inhibitors on disease severity and mortality in patients with COVID-19: A meta-analysis. J Med Virol. 2020. November 24. doi: 10.1002/jmv.26695. Online ahead of print.** This study shows that prior use of RAAS inhibitor does not associate with increased mortality or disease severity in COVID-19 patients.
- 81.Trifirò G, Massari M, Da Cas R, Menniti Ippolito F, Sultana J, Crisafulli S, et al. Renin-Angiotensin-Aldosterone System Inhibitors and Risk of Death in Patients Hospitalised with COVID-19: A Retrospective Italian Cohort Study of 43,000 Patients. Drug Saf. 2020;43(12):1297–308.** In the first largest cohort of hospitalized COVID-19 patients exposed to RAS inhibitors, ACEI/ARBs failed to associate with either an increased or decreased risk of all-cause mortality, compared with calcium channel blocker usage.
- 82.Mancia G COVID-19, hypertension, and RAAS blockers: the BRACE-CORONA trial. Cardiovasc Res. 2020;116(14):e198–e9.** This trial, which recruited 659 hypertensive patients (from 34 Brazilian medical sites), shows that suspending ACEIs and ARBs for 30 days did not impact the number of days alive and out of hospital.
- 83.Fraga-Silva RA, Sorg BS, Wankhede M, Dedeugd C, Jun JY, Baker MB, et al. ACE2 Activation Promotes Anti-Thrombotic Activity. Mol Med. 2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Zhang C, Zhao YX, Zhang YH, Zhu L, Deng BP, Zhou ZL, et al. Angiotensin-converting enzyme 2 attenuates atherosclerotic lesions by targeting vascular cells. Proc Natl Acad Sci U S A. 2010;107(36):15886–91. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Dong B, Zhang C, Feng JB, Zhao YX, Li SY, Yang YP, et al. Overexpression of ACE2 enhances plaque stability in a rabbit model of atherosclerosis. Arterioscler Thromb Vasc Biol. 2008;28(7):1270–6. [DOI] [PubMed] [Google Scholar]
- 86.Thatcher SE, Zhang X, Howatt DA, Lu H, Gurley SB, Daugherty A, et al. Angiotensin-converting enzyme 2 deficiency in whole body or bone marrow-derived cells increases atherosclerosis in low-density lipoprotein receptor−/− mice. Arterioscler Thromb Vasc Biol. 2011;31(4):758–65. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Simões e Silva AC, Silveira KD, Ferreira AJ, Teixeira MM. ACE2, angiotensin-(1–7) and Mas receptor axis in inflammation and fibrosis. Br J Pharmacol. 2013;169(3):477–92. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Connors JM, Levy JH. COVID-19 and its implications for thrombosis and anticoagulation. Blood. 2020;135(23):2033–40.** This review summarizes the evidence showing that COVID-19 is associated with increased coagulopathy, findings consistent with infection-induced pro-inflammatory changes observed in patients.
- 89.Alexander SPH, Armstrong JF, Davenport AP, Davies JA, Faccenda E, Harding SD, et al. A rational roadmap for SARS-CoV-2/COVID-19 pharmacotherapeutic research and development: IUPHAR Review 29. Br J Pharmacol. 2020;177(21):4942–66.** This article assesses the scope for targeting key host and viral targets, by first screening these targets against drugs already licensed, an agenda for drug repurposing, which should allow rapid translation to clinical trials.
- 90.Jessup JA, Brosnihan KB, Gallagher PE, Chappell MC, Ferrario CM. Differential Effect of Low Dose Thiazides on the Renin Angiotensin System in Genetically Hypertensive and Normotensive Rats. J Am Soc Hypertens. 2008;2(2):106–15. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Supuran CT. Diuretics: from classical carbonic anhydrase inhibitors to novel applications of the sulfonamides. Curr Pharm Des. 2008;14(7):641–8. [DOI] [PubMed] [Google Scholar]
- 92.Zolfaghari Emameh R, Falak R, Bahreini E. Application of System Biology to Explore the Association of Neprilysin, Angiotensin-Converting Enzyme 2 (ACE2), and Carbonic Anhydrase (CA) in Pathogenesis of SARS-CoV-2. Biol Proced Online. 2020;22:11.Using combined system biology and bioinformatic approaches, this study defined the role of coexpression of ACE2, neprilysin or membrane metallo-endopeptidase, and carbonic anhydrases and their association to the pathogenesis of SARs-CoV-2.
- 93.Reyes S, Varagic J, Ahmad S, VonCannon J, Kon ND, Wang H, et al. Novel cardiac intracrine mechanisms based on Ang-(1–12)/chymase axis require a revision of therapeutic approaches in human heart disease. Curr Hypertens Rep. 2017;19(2):16. [DOI] [PMC free article] [PubMed] [Google Scholar]
