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

Disequilibrium between the classic renin-angiotensin system and its opposing arm in SARS-CoV-2-related lung injury

Published Online:https://doi.org/10.1152/ajplung.00189.2020

Abstract

A dysregulation of the renin-angiotensin system (RAS) has been involved in the genesis of lung injury and acute respiratory distress syndrome from different causes, including several viral infections. The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection of pneumocytes, the hallmark of the pandemic coronavirus disease 2019 (COVID-19) involving both alveolar interstitium and capillaries, is linked to angiotensin-converting enzyme 2 (ACE2) binding and its functional downregulation. ACE2 is a key enzyme for the balance between the two main arms of the RAS: the ACE/angiotensin (Ang) II/Ang II type 1 receptor axis (“classic RAS”) and the ACE2/Ang(1–7)/Mas receptor (MasR) axis (“anti-RAS”). The ACE2 downregulation, as a result of SARS-coronaviruses binding, enhances the classic RAS, leading to lung damage and inflammation with leaky pulmonary blood vessels and fibrosis, when the attenuation mediated by the anti-RAS arm is reduced. ACE inhibitors (ACE-I) and Ang II type 1 receptor blockers (ARB), effective in cardiovascular diseases, were found to prevent and counteract acute lung injury in several experimental models by restoring the balance between these two opposing arms. The evidence of RAS arm disequilibrium in COVID-19 and the hypothesis of a beneficial role of RAS modulation supported by preclinical and clinical studies are the focus of the present review. Preclinical and clinical studies on drugs balancing RAS arms might be the right way to counter COVID-19.

INTRODUCTION

The coronavirus disease 2019 (COVID-19) caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has been declared a pandemic by the World Health Organization on March 11, 2020. Children and younger healthy adults usually have mild symptoms or may even be asymptomatic, whereas older adults are at higher risk of severe complications and death (25, 63). Furthermore, increasing data have linked obesity to a more severe course of COVID-19 and death even in younger people (82). Next to aging and obesity, cardiovascular (CV) comorbidities are likely to play a key role in the prognosis of COVID-19. It is not surprising that hypertension and other CV diseases (CVD) are so common among patients who died from COVID-19 (112). Hypertension is highly prevalent in the general population, being the first CV risk factor in older people, while CVD are known to be the leading cause of death worldwide (99). High prevalence of hypertension has also been found in patients who died after Middle East Respiratory Syndrome (MERS)-CoV and influenza viruses infections (2), indicating its nonspecificity for COVID-19, but most likely representing a confounder, linked to age. Heart failure (HF) is also likely to be highly prevalent among patients deceased for COVID-19, even though published data on this association are still scarce (71).

Angiotensin-converting enzyme 2 (ACE2) is the cellular receptor for SARS-CoV-2, as it was for SARS-CoV (33, 104). ACE inhibitors (ACE-I) and angiotensin II type 1 receptor blockers (ARB) are cornerstone drugs for the management of both hypertension and CVD, and their use has been previously found to be associated with increased ACE2 levels in some tissues or fluids. On this basis, some authors hypothesized a greater susceptibility to SARS-CoV-2 infection and a higher risk for severe disease associated with these drugs (18, 110), despite the absence of solid experimental evidence. Promptly, almost all major international scientific societies and drug authorities clarified that no experimental data support these speculations, stressing the risks due to withdrawal, especially in patients at higher CV risk. Furthermore, several clinical observational studies recently published have largely refuted this hypothesis (50). Actually, the exact opposite appears to be more likely from previous experimental studies on virus-mediated lung injury. Indeed, a dysregulation of the renin-angiotensin system (RAS) plays a key role in the genesis of lung damage after SARS-CoV-2 infection, and RAS inhibitors may restore the equilibrium between the “classic RAS” arm and its counterregulatory arm, here defined as “anti-RAS,” of which ACE2 is the cornerstone. These two pathways and the possible role of a “pathophysiological” drug therapy based on RAS inhibitors, aiming at restoring equilibrium and avoiding severe lung damage and death in COVID-19, are described in depth below (Fig. 1).

Fig. 1.

Fig. 1.Graphic synthesis of the proposed disequilibrium caused by severe acute respiratory syndrome coronavirus 2 infection in lung tissue (top) restored by angiotensin II type 1 receptor blocker (ARB) therapy (bottom). Ang II, angiotensin II; ACE, angiotensin-converting enzyme; AT1R, angiotensin II type 1 receptor; ARDS, acute respiratory distress syndrome; MasR, Mas receptor; MrgD, Mas-related G protein-coupled receptor D.


PHYSIOLOGY OF RAS PATHWAYS IN CARDIOVASCULAR AND PULMONARY HOMEOSTASIS

The Classic RAS and the Anti-RAS Arms

The RAS plays a central role in both the homeostatic control of CV and renal systems and the regulation of both fluid volume and blood pressure, but also exerts trophic and pathomorphological effects on several tissues, including lungs, mainly acting on the microvasculature and mesenchymal cells. The RAS blockade, or better, the RAS modulation, represents a therapeutic cornerstone of both arterial hypertension and its related CV sequelae, such as ischemic heart disease and HF (67).

The classic RAS consists in a cascade of both systemic and tissue enzymatic reactions, mainly resulting in generation of angiotensin (Ang) II (Fig. 2A, top). Renin, a protease released by the kidneys in its active form, cleaves angiotensinogen, produced by the liver in large quantities, but also by the adipose tissue (79), to form Ang I, both locally and systemically, which is then hydrolyzed by the widespread endothelial ACE, producing the octapeptide Ang II. The ACE is very abundant in microvasculature-rich tissues, such as lungs, being highly expressed in the endothelium. Therefore, the pulmonary vascular bed is capable to rapidly convert Ang I to Ang II. It is important to note that this ability may play a key role in the overproduction of Ang II in conditions characterized by increased circulating levels of Ang I, such as obesity or overweight with central (visceral) obesity (79). This biologically active peptide binds Ang II type 1 and type 2 receptors (AT1R and AT2R, respectively). The AT1R binding induces vasoconstriction, but also direct vascular damage when excessively activated (66). Furthermore, in the adrenals, Ang II stimulates aldosterone secretion, promoting sodium reabsorption and blood pressure increase.

Fig. 2.

Fig. 2.Schematic of the “classic renin-angiotensin system (RAS)” and “anti-RAS” arms in different conditions as follows: physiology (A, top), severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection (A, bottom), angiotensin II type 1 receptor blocker (ARB) therapy (B, top), and angiotensin-converting enzyme inhibitor (ACE-I) therapy (B, bottom). The schematic includes circulating factors and local synthesis by the main cells involved (pneumocytes type 1 and 2, interstitial mesenchymal cells, and capillary endothelium). The thickness of the connecting lines and arrows or dashed arrows represents proportional increases or reductions of the pathways. SARS-CoV-2 infection might lead to a downregulation of ACE2 with a consequent decrease of the anti-RAS arm, mainly Ang(1–7) and alamandine. Treatment with ARB could counteract this imbalance, through angiotensin II type 1 receptor (AT1R) blockade and the angiotensin II (Ang II) increase, moving the pathway toward anti-RAS prevalence. The reequilibrium of the anti-RAS arm could be done also likely by treatment with ACE-I through a different mechanism [increased availability of Ang(1–9), decreased degradation of Ang(1–7)] that also involves angiotensin II type 2 receptor (AT2R) stimulation. ACE, angiotensin-converting enzyme; ACE2, angiotensin-converting enzyme 2; Ang, angiotensin; AD, aspartate decarboxylase; MasR, Mas receptor; MrgD, Mas-related G protein-coupled receptor D; AT1R, angiotensin II type 1 receptor; AT2R, angiotensin II type 2 receptor.


The dominant physiological and pathophysiological role of the classic RAS pathway was rethought 20 years ago, after the discovery of ACE2 (17). ACE2 is a type I transmembrane metallocarboxypeptidase, with homology to ACE and, just like ACE, abundant in lung tissue, consisting of an extracellular NH2-terminal domain containing a catalytic site and intracellular COOH-terminal tail. ACE2 has many peptides as substrates, but mainly cleaves Ang I into a nonapeptide [Ang(1–9)] that binds AT2R and Ang II into a heptapeptide [Ang(1–7)] that binds an endogenous orphan receptor (Mas receptor, MasR) (Fig. 2A, top) (73). The catalytic efficiency of ACE2 is ~400-fold higher for Ang II than Ang I (95). Ang(1–9) can also be rapidly converted in Ang(1–7) by ACE. Therefore, ACE2 mainly produces Ang(1–7) from Ang II, unless ACE is inhibited, leading to decreased Ang II and increased Ang I levels so that ACE2 can switch to more Ang(1–9) being produced.

Differences in the modulation of ACE2 and RAS by ACE-I and ARB are then expected. ACE-I reduce the Ang II-AT1R pathway activity by reducing Ang II synthesis with increased Ang I substrate for ACE2 and a reduced conversion of Ang(1–9) to Ang(1–7). Therefore, ACE-I can specifically enhance the activity of Ang(1–9) on AT2R, especially when Ang II is reduced (Fig. 2B, bottom). At the same time, given that ACE also cleaves Ang(1–7) to the inactive Ang(1–5), ACE-I may also increase the lifespan of Ang(1–7) (Fig. 2B, bottom) (9). Differently, ARB compete with Ang II at the AT1R level, leading to a secondary increase of Ang II due to the reduced negative feedback on renin release and thus increasing Ang II substrate for ACE2 (Fig. 2B, top).

ACE2 also interacts with other angiotensin peptides (called alatensins) in which the NH2-terminal aspartate is replaced by alanine, leading to Ala-Ang 1–7 (called alamandine) that has been found to bind the Mas-related G protein-coupled receptor D (MrgD) (74), showing similar effects to Ang(1–7) stimulating MasR (Fig. 2A, top). The largest part of ACE2 is membrane bound, whereas its shorter soluble form circulates in blood at very low levels but can be increased through cleavage of full-length ACE2 membrane-anchor by the disintegrin and metalloprotease 17, which is directly upregulated by the Ang II-AT1R axis (102). The “shedding” of ACE2, induced by the classic RAS effector Ang II/AT1R, is likely to result in local downregulation of the tissue enzymatic activity, similarly from what is observed following SARS-CoV infection (24, 39).

Role of ACE2 in Cardiovascular and Pulmonary Homeostasis

Initially thought to be expressed only in limited organs (mainly heart and kidney), ACE2 was later found to be ubiquitous, and highly expressed in the lungs (98), exerting its key roles in maintaining tissues and body homeostasis by counterbalancing classic RAS activity. ACE2 is abundantly expressed on the cell surface of alveolar type II epithelial cells (type II pneumocytes), which are small cylindrical cells very close to pulmonary capillaries and responsible for the synthesis of alveolar surfactant. ACE2 is also widely expressed in other airway cells, from the epithelial cells of oral mucosa (101) to the bronchial transient secretory cells (49). Furthermore, it is also well detectable on the enterocytes of the small intestine, as well as on vascular endothelium of several human organs and systems, including cardiovascular system, kidneys, lungs, and brain (31, 74), thus probably explaining the ability of SARS-CoV-2 to affect so many organs.

The two main catalytic roles of ACE2 in human CV homeostasis, in the absence of RAS-interfering drugs, are the degradation of Ang II, which is the major effector of the classic RAS arm that promotes vasoconstriction, sodium retention, oxidative stress, inflammation, and fibrosis, and the sharp increase of bioactive peptide Ang(1–7), which binds MasR and counterregulates the Ang II-AT1R axis through its vasodilating, anti-inflammatory, and antifibrotic effects (Fig. 2A, top) (66). ACE2 could mediate both blood pressure reduction and protection from CV damage in preclinical studies (14, 47). According to experimental models, the activation of the Ang(1–7)-MasR axis attenuates inflammation and fibrosis in several tissues. The ACE2-Ang(1–7) pathway also plays a key protective role against HF and coronary heart disease (4). Treatment with the Ang(1–7)/MasR antagonist d-Ala7-Ang-(1–7) (“DALA”) worsened Ang II-induced cardiac fibrosis and increased the expression of multiple cytokines with profibrotic actions (58). In ACE2 knockout mice, Ang II infusion induces hypertension, myocardial hypertrophy, fibrosis, and diastolic dysfunction, whereas recombinant ACE2 (rhACE2) attenuates myocardial remodeling, which is the main feature of HF with preserved ejection fraction (HFpEF) (66, 111).

The ACE2-driven anti-RAS arm is also potentiated by alamandine and its specific receptor that, reinforcing the MasR-dependent effects, may protect against inflammation and fibrosis by improving the endothelial dysfunction and mitigating vascular remodeling also in the lung (16, 74). Moreover, alamandine counteracts cardiac hypertrophy induced by Ang II and left ventricular dysfunction (38). Although AT2R is believed to be the anti-AT1R by some authors, the role of the ACE2-Ang(1–9)-AT2R pathway is still less clear, mostly because the postreceptorial cascade after AT2R activation is still not fully understood and because it is likely to play a role in pathological conditions only (74). AT2R activation likely leads to both vasodilation and a general attenuated response to Ang II/AT1R (34). Administration of Ang(1–9) reduced blood pressure in hypertensive rats through AT2R (62).

In synthesis, many well-conducted studies in different animal models and humans lead to the solid knowledge that a decrease of ACE2 activity leaves unopposed the effects of the Ang II-AT1R axis, contributing to progression of microvascular damage and CVD, whereas increased ACE2 activity leads to the activation of both Ang(1–7)-MasR and alamandine-MrgD pathways and maybe also the Ang(1–9)-AT2R axis, contributing to protect against microvascular damage and CVD due to its antihypertrophic, antifibrotic, and anti-inflammatory activities (66).

SPECIFIC ROLE OF RAS PATHWAYS IN LUNG DISEASE

Viral Infection and Lung Damage

The coronaviruses involved in severe acute respiratory syndromes, such as SARS-CoV-2, can frequently cause interstitial pneumonia. Alveolar epithelial injury is the first crucial step for the development of interstitial pneumonia in humans, especially when SARS coronaviruses are involved. Indeed, these features are shared by SARS-CoV, SARS-CoV-2, and MERS-CoV (103). Even if too much attention has been given to ancillary direct viral damage of other tissues, infection of the lung respiratory epithelium and alveolar damage are the hallmark of COVID-19 (65). This damage is rapidly associated with the involvement of both capillaries and lung interstitium.

High-resolution chest computed tomography (HR-CT) well reflects the evolution of pathological lung changes and the course of COVID-19 (72). In the early stage, radiological images are characterized by multilobar ground-glass opacities (GGO) with a peripheral or posterior distribution. GGO are commonly seen also in other lung diseases characterized by microvascular transudate or exudate filling alveolar spaces. Next, maybe depending also on the viral load, there is an increase in both number and size of GGO and progressive transformation into bilateral multifocal consolidative opacities and septal thickening, with development of a “crazy paving” pattern and a possible evolution in fibrosis (72).

In the early phases, typical pathological findings are enlarged microvessels, alveolar exudation of plasma, and hyaline membrane formation, in the absence of significant inflammatory cell infiltration (93, 103). The very few anatomical and histological studies in humans (93, 103) and macaques (70) published to date found pneumocyte necrosis (involving not only type II but also type I pneumocytes) with reparative replication and vascular (capillary) leakage of protein and fibrin-rich edema filling patchy lung areas. Neutrophils and lymphocytes are not prominent (70), and the subsequent cytokine production after lung damage is likely part of the reparative response. The evolution of alveolar exudation from damaged and leaky capillaries can be toward extensive fibrosis due to secondary hyperstimulation of local fibroblasts, which is believed to be driven by Ang II/AT1R. The clinical picture is finally ascribable to the acute respiratory distress syndrome (ARDS), a common arrival point for many acute viral respiratory diseases in which the dysregulation of the local RAS pathways has been found to exert a key role (44).

Disequilibrium Between Classic RAS and Anti-RAS Arms in Lung Diseases

An imbalance between the classic RAS and the ACE2-driven anti-RAS arms is very likely to promote and accelerate lung injury. The ACE-Ang II-AT1R pathway, especially when inappropriately activated, or in the context of ACE2 downregulation, induces pulmonary vasoconstriction and microvascular damage with increased vascular permeability, resulting in enlarged and “leaky” pulmonary microvessels and damaged alveoli filled with plasma proteins. Furthermore, secondary production of inflammatory cytokines, accelerated apoptosis in alveolar epithelial cells, and promotion of both mesenchymal cell growth and extracellular matrix synthesis leading to lung fibrosis have been demonstrated (94). All of these features are well-documented in coronavirus-mediated lung injury, as described above, and suggest an active role of the Ang II-AT1R axis. On the contrary, Ang(1–7) was found to reduce inflammation, attenuate lung fibrosis, and improve oxygenation in acute lung injury, a noteworthy factor in protecting against ARDS-related poor prognosis (108). The use of different ARB or the less studied ACE-I, through reduction of Ang II/AT1R stimulation and likely induction of ACE2 activity, has been effective in decreasing lung injury in animal models of ARDS (84).

A brief history on the topic tells us that ACE2 returned to the limelight in 2003, following the discovery that it serves as a receptor for the binding and entry of SARS-CoV (44). Similarly, SARS-CoV-2 recognizes ACE2 as its host receptor, binding to viral Spike protein, with a 10- to 20-fold higher affinity compared with SARS-CoV (100). After binding to ACE2, SARS-CoV-2 Spike protein conformational change triggers a proteolytic digestion mediated by host cell transmembrane protease serine 2 (TMPRSS2), allowing the virus entry into the cell (33). ACE2, besides being the gateway for the virus, exerts many favorable actions in lung. In fact, previous well-conducted studies found that ACE2 can protect lungs from severe acute injury in mice through different mechanisms, while the Ang II-AT1R pathway promotes lung disease, leading to leaky pulmonary blood vessels, and impairs lung function (35). Ye and Liu showed how ACE2 injection can attenuate inflammatory response, by inhibiting the lipopolysaccharide (LPS)-TLR4 pathway in mice with LPS-induced acute lung injury (107). Furthermore, previous studies on SARS and MERS showed that the binding of the viral surface Spike protein to ACE2 leads to its downregulation, through its internalization and probably its shedding (35). Importantly, ACE2 downregulation itself results in a hypereffective Ang II-AT1R pathway, promoting ARDS development. It has also been hypothesized that ACE2 downregulation could be linked to the systemic endothelial damage and coagulopathy observed in several COVID-19 cases, through an abnormal secretion of sFlt-1, a decoy receptor of placental growth factor (PLGF), mediated by Ang II, such as in preeclampsia (23). A study conducted in Shenzhen on 12 COVID-19 patients found that plasma samples from infected patients had significantly increased levels of Ang II compared with healthy individuals, and these levels were strongly associated with viral load and lung injury severity (48). Also, in H5N1 influenza, another viral infection that can lead to ARDS, a marked elevation of Ang II levels was linked to disease severity and fatal outcomes in humans (113). This suggests that Ang II, although not always easy to assay, is likely a good biomarker with a predictive value for lung injury.

Experimental SARS-CoV infection of wild-type mice in vivo led to reduced ACE2 expression in lungs, suggesting that reduced ACE2 expression might have a role in SARS-CoV-mediated severe acute lung disease (44). In a mouse model of acid aspiration-induced lung injury, the addition of SARS-CoV Spike protein decreased ACE2 and increased Ang II levels. After the AT1R blockade with losartan, ACE2 increased, and the acute severe lung injury in Spike-Fc-treated mice was attenuated. More importantly, even pulmonary edema was attenuated after AT1R blockade (44, 84). Therefore, SARS-CoV infections are likely to induce acute lung injury and failure through an imbalance between the classic RAS and the ACE2-driven anti-RAS arm, a process that could be attenuated by the AT1R blockade (35). Further insights of this mechanism were elucidated in a cellular and rodent model, where ACE2 was found to physically interact with AT1R on the cell membrane, and, through ACE2-AT1R stabilization, AT1R blockade with ARB prevented the internalization and lysosomal degradation of ACE2 mediated by Ang II (15). In addition to SARS coronaviruses, ACE2 also mediated the severe acute lung injury induced by respiratory syncytial virus infection in an experimental mouse model in which ACE2 deficiency worsened the disease pathogenesis mainly by its action on the AT1R (26). The potential benefit of ACE2 in viral-mediated lung injury was also observed in mice infected by H7N9 influenza virus (106) in which worse lung injury and survival were observed in ACE2 knockout mice. Moreover, in three different acute lung injury models, loss of ACE2 expression precipitated severe acute lung failure, whereas treatment with rhACE2 attenuated ARDS and further reduced Ang II levels in lungs (35). The severity of acid-induced lung injury in ACE2 knockout mice was also reduced through pharmacological AT1R blockade, whereas inhibition of AT2R had no apparent effect on the acute lung injury phenotypes, confirming the causative role of Ang II in acute lung failure mediated mainly by AT1R activation, instead of AT2R (35).

All of these findings suggest an important protective role of AT1R blockade in virus-related lung injury, in which ACE2 downregulation promotes a dangerous imbalance between the RAS pathways (Fig. 2A, bottom), subverting the alveolar-capillary homeostasis through what we like to define an “Ang II/AT1R storm.” The acute increase in the local ratio of Ang II/Ang(1–7) following ACE2 internalization is likely to exacerbate the pulmonary tissue damage initially provoked by pneumocyte viral infection in SARS disease, and most likely also in COVID-19. However, while a reduced ACE2 expression after SARS-CoV infection has been demonstrated (64), whether SARS-CoV-2 also induces ACE2 downregulation in humans is yet to be fully determined, although it can be extrapolated.

At the same time, the ACE2 overexpression following ACE-I and ARB administration is not fully consistent among studies and may differ between the two drug classes and the organs taken into account. Among ARB, olmesartan, losartan, valsartan, candesartan, telmisartan, and irbesartan all increased ACE2 levels in a mouse model, mainly referred to heart tissue (96), while there is very limited data for ACE-I showing upregulation of ACE2. ARB are likely to upregulate ACE2 activity because of the AT1R blockade and the increased substrate load of Ang II (Fig. 2B, top). On the other hand, ACE-I enhance the Ang(1–9) stimulation of AT2R (Fig. 2B, bottom) with specific protective consequences, including reduction of inflammatory cytokines in a rat model of induced lung injury and pulmonary hypertension (8). Moreover, a distinction should be made between membrane-bound and soluble ACE2 because an increase in soluble ACE2 might imply a decrease in membrane-bound ACE2 even with opposite consequences. In several animal models, both ACE-I and ARB could upregulate ACE2, especially in heart tissue (19, 37, 61), but it is not known to happen in human lungs. Human studies reporting ACE2 upregulation following ARB treatment took into account only soluble ACE2 levels (1, 22, 68), as long as in vivo membrane-bound ACE2 measuring in human tissues is technically challenging. Urinary ACE2 was found to increase after treatment with ARB in hypertensive and nephropathic patients, but it is not possible to draw conclusions given the scarce evidence and the complexity of the measurement (1, 22). We do not know if ACE2 plasma levels reflect its tissue expression or if upregulation would occur in all tissues in the same way. Further evidence is needed to clarify this topic. In any case, it is necessary to distinguish between the ability of these drugs to increase ACE2 expression, yet to be demonstrated, and their ability to modulate/increase the ACE2 activity, for example, through the regulation of substrates, which instead appears more conceivable by the different experimental models (Fig. 2B).

As mentioned above, the lung disease following SARS-CoV-2 infection can evolve into pulmonary fibrosis. In lung tissues, the activation of the Ang(1–7)-MasR axis attenuates inflammation, endothelial dysfunction, tissue damage, and fibrosis in different pathological models, such as pulmonary hypertension, asthma, pulmonary emphysema, and idiopathic pulmonary fibrosis (6, 51, 52, 8587). The attenuation of key signaling events in the initiation and progression of cellular fibrosis, such as MAP kinases ERK1/2, p38, and JNK, and the reduced Smad phosphorylation, may be involved in the antifibrotic actions of Ang(1–7), counteracting Ang II-induced migration and both transforming growth factor-β1 (TGF-β1) and collagen expression of pulmonary myofibroblasts (59). TGF-β1 is a key factor in vascular-related fibrosis induction (76). Ang(1–7) effectively inhibits epithelial-mesenchymal transition induced by TGF-β1 via disruption of the TGF-β1-Smad signaling pathway in alveolar epithelial cells (83). Nevertheless, Ang(1–9) was found to activate AT2R in monocrotaline-induced pulmonary hypertensive rats, ameliorating both lung injury and pulmonary vascular remodeling together with a reduction of apoptosis genes and inflammatory cytokine levels (8).

The pathophysiological potential benefits of RAS inhibitors in acute lung injury based on experimental studies are described in Table 1.

Table 1. Pathophysiological and clinical potential benefits of RAS inhibitors in COVID-19, based on experimental and clinical studies

Pathophysiological Benefits Clinical Implications
Direct effects on the lung
Less injury to lung alveolar cells and capillary endothelial cells and enhanced repair by MSCs Preservation of functioning alveolo-capillary membrane for proper gases exchanges
Less vascular permeability Reduced alveolar/interstitial transudation of fluids and exudation of proteins and cells
Less proinflammatory cytokines production Reduced inflammation
Less interstitial-mesenchymal cell proliferation and less production of TGF-β Reduced pulmonary fibrosis and preserved lung compliance
Less coagulation cascade activation within the pulmonary vasculature Reduced pulmonary vascular thrombosis
Less lung oxidative stress (less ROS) and vasodilation Reduced pulmonary arterial pressure
Indirect effects on the lung (cardiovascular, renal, and metabolic effects)
Less angiotensin II and aldosterone production with less sodium reabsorption and vasodilation Reduced blood pressure and HMOD
Less VSMC and myofibroblast proliferation and migration Reduced myocardial, glomerular, and systemic vascular fibrosis
Less oxidative stress (less ROS) with improved endothelial function and less inflammation Reduced atherosclerosis and proteinuria
Less coagulation cascade activation within the systemic vasculature Reduced risk of systemic thromboembolism
Less mitochondrial dysfunction Reduced insulin resistance and balanced energy metabolism

TGF-β, transforming growth factor-β; MSCs, mesoderm-derived mesenchymal stem cells; ROS, reactive oxygen species; HMOD, hypertension-mediated organ damage; VSMC, vascular smooth muscle cells.

Evidence of RAS Modulation Efficacy in Lung Injury: From Experimental Animal Models to Clinical Data

Some clinical studies support the concept that ACE-I and ARB may be useful in patients with acute lung injuries. However, evidence in humans regarding RAS modulation and lung disease are limited to small pilot studies or retrospective studies, although all agree on favorable outcome. On the other hand, clinical data on the possible direct benefits of ACE2/Ang(1–7)/MasR arm overstimulation in humans are still lacking to date. A meta-analysis of 37 studies showed reduced risk of pneumonia-related mortality in patients treated with ACE-I or ARB compared with control treatments (7). Patients with ARDS taking ACE-I or ARB showed better survival compared with controls in a retrospective case-control study (42). Another retrospective study on 539 patients with viral pneumonia showed that ACE-I use was associated with lower rates of intubation and death (32). The rhACE2 infusion in a randomized double-blind pilot trial on ARDS patients decreased Ang II and increased Ang(1–7) with a decrease in interleukin-6, although it had no power to detect changes in acute physiology or clinical outcomes (41).

Regarding COVID-19, several observational retrospective studies on different populations have been published recently. All studies found neutral or even beneficial effects of RAS inhibitors in COVID-19, regarding both the risk of getting infected and the risk of greater disease severity or death (50). Patients taking RAS inhibitors certainly do not have a higher risk of SARS-CoV-2 infection (3, 20, 53, 56, 69). Furthermore, treatment with ACE-I/ARB does not affect hospitalization for COVID-19 (13) or disease severity (28, 69). A retrospective analysis on 1,128 Chinese hypertensive patients found a significant lower risk of death in patients taking ACE-I/ARB compared with other antihypertensive drugs after adjustment for confounders (109). In small samples of COVID-19, ACE-I/ARB therapy affected both IL-6 and peripheral T cell levels (57), being associated with lower levels of inflammatory indices (105). Other observational studies found no association between COVID-19 mortality and ACE-I/ARB treatment (20, 40, 45). Nine observational studies on COVID-19 hypertensive patients have been recently included in a meta-analysis showing that patients taking ACE-I/ARB treatment had a 43% lower risk of mortality for COVID-19 compared with patients not taking ACE-I/ARB (28).

The consistent favorable data on both in vitro and in vivo studies on animals regarding the imbalance of RAS arms and virus-mediated lung injury are leading to randomized controlled trials (RCTs) aimed at obtaining consistent clinical data in COVID-19 patients. One of these RCTs regards the use of rhACE2, aiming at evaluating viral load over time (NCT04287686), while two other RCTs with losartan are ongoing to investigate the possible benefits in COVID-19 patients requiring or not requiring hospitalization (NCT04312009; NCT04311177).

Independent from the mechanisms in acute lung injury, RAS inhibitor withdrawal may cause severe clinical decompensation in high-risk CV patients (30) in which lung infection, per se, is associated with increased risk of CV events (12). This should be a fundamental warning for patients with COVID-19, which is likely to result in a RAS overactivation mediated by Ang II activity, thus exacerbating the cardiac damage that may occur in critically ill patients (48), such as myocardial injury (27, 48, 88), myocardial stress, and cardiomyopathy (5). Indeed, a possible direct cardiac involvement of SARS-CoV-2 has been demonstrated (36). Furthermore, prevalence of HF has not been adequately assessed in the first reports from China, but it is likely to be very common, especially in older COVID-19 patients. Up to 32% of deaths for COVID-19 are due to a failing heart after myocardial injury in a Chinese case series (71). Older inpatients with multiple comorbidities often have an underlying HF (81). Previous studies found that ACE-I/ARB therapy was associated with lower risk of death, likely due to a better CV protection and a higher CV resistance to acute illness, in older patients hospitalized for medical conditions, including infections (91). This association could be theoretically true even for COVID-19 patients. Therefore, it is important to stress the concept that patients affected by HF or other CVD taking ACE-I and ARB as per indication still continue to adhere to their prescribed regimens, even in the COVID-19 era (77). The main potential clinical benefits of RAS inhibitors in COVID-19 are described in Table 1.

Possible Role of RAS in the Severity of COVID-19 According to Sex and Obesity

Based on currently available data, especially in U.S. and European cohorts, obesity is one of the main risk factors for severe COVID-19 course and death, particularly among younger patients without comorbidities (82). Obese patients aged <60 yr have a double risk of critical care admission compared with normal-weight patients that become seven-fold higher in patients with a body mass index (BMI) ≥35 kg/m2, and obesity is associated with higher need of invasive mechanical ventilation (46, 89). Obesity, per se, is related to the main CV risk factors and CVD, such as coronary heart disease, chronic kidney disease, and HF, predisposing to adverse outcomes (75, 92). The RAS dysregulation in obese patients is well known, as well as the role of adipose tissue in upregulating RAS activity, both locally and systemically (79). Furthermore, food intake plays a key role (21). Indeed, upon overfeeding stress, almost 20–30% of circulating angiotensinogen is produced by white adipose tissue, favoring obesity-related hypertension (54), with Ang II acting as a “growth factor” for adipocytes. In obese hypertensive patients, “inappropriately normal” levels of plasma renin activity and aldosterone have been documented, even in patients taking ACE-I or ARB, with consequent vascular damage (78, 90). It is reasonable to speculate that the Ang II/AT1R storm promoted at the lung level by the SARS-CoV-2-induced ACE2 downregulation, might be even much worse in overweight/obese patients, who have both Ang I and Ang II increased levels coming from visceral adipose tissue to the systemic venous circulation, and then to the pulmonary microcirculation (82).

Near obesity, male sex has been associated with a worse COVID-19 course and a greater need for intensive care compared with female sex (60). An Italian report found that 82% of COVID-19 patients referred to intensive care units were males (25). Animal models of obesity have shown sex differences in expression, activity, and tissue responsiveness of RAS components. Importantly and differently from males, obese females were likely protected from hypertension and metabolic complications induced by Ang II-AT1R pathway activation, through upregulation of the anti-RAS pathway, such as ACE2/Ang(1–7)/MasR (97). Estrogenic activity decreases the vascular response [vasoconstriction and NAD(P)H oxidase activation] to Ang II and facilitates the action of ACE2/Ang(1–7) in animal models (43). Moreover, obese women have more subcutaneous than proinflammatory visceral adipose tissue, and adipocytes are smaller, more lipogenic, and insulin sensitive than obese males (97). Finally, the ACE2 gene is X linked, and females have higher ACE2/Ang(1–7) levels at least in animal models (29, 43). All of these findings could affect the clinical course of COVID-19.

Potential Role of Angiotensin Receptor-Neprilysin Inhibitor in Lung Disease and COVID-19

All of the pharmacological strategies able to prevent CV complications should be implemented in COVID-19 patients, given their relevance on disease prognosis (11). ACE-I and ARB are cornerstone not only for hypertension but also for HF treatment. An emerging topic of interest is the potential role of angiotensin receptor-neprilysin inhibitor (ARNI) sacubitril/valsartan in patients with pulmonary vascular remodeling, pulmonary hypertension, and right ventricular overload related to lung diseases (10). The synergistic effects of RAS and neprilysin inhibition, versus RAS inhibition alone, may magnify the protective benefits of natriuretic peptides by reducing fibrosis and vascular remodeling and promoting vasodilation and natriuresis (80). Given the prevalence and the negative prognostic role of myocardial injury in COVID-19 patients (11), ARNI may not only be effective in reducing morbidity and mortality in patients with chronic symptomatic HF with reduced ejection fraction (HFrEF) (55) but may also counterbalance the Ang II-AT1R pathway, which has been implicated in maladaptive right ventricular hypertrophy and fibrosis associated with pulmonary hypertension (10). Indeed, neprilysin inhibition is intimately related to RAS activation, but it is unclear whether ARNI has any impact on ACE2 or in COVID-19 patients. Caution is needed because circulating neprilysin can convert Ang I in the lung-protective Ang(1–7) (66). Therefore, sacubitril could partially mitigate this conversion. ACE-I could amplify the attenuation of this pathway by increasing Ang I levels, and the association with neprilysin inhibitors is contraindicated, while the concomitant use of ARB could theoretically counteract this negative effect by increasing Ang II levels and bypassing the direct role of neprilysin in the RAS pathway.

CONCLUSION AND PERSPECTIVES

The balance between RAS pathways in the lung is likely to be of fundamental importance in the genesis and promotion of acute lung injury, but it is complex, and more data on humans are needed. Hasty speculations on a negative relationship between RAS blockers and COVID-19 are not justified by our present knowledge. Hypertensive patients, especially if having CVD such as HF, should continue to take these drugs with high adherence to the prescribed regimens. On the other hand, the administration of ACE-I or ARB aiming at preventing COVID-19 or reducing the disease severity is still not supported by conclusive data, although preclinical evidence and several observational findings are encouraging. Therefore, if on one hand there is solid evidence of the benefits of RAS modulation in CVD, on the other hand caution and clinical equipoise must be maintained regarding RAS modulation in COVID-19. There is a clinical rush toward several COVID-19 “therapies,” such as old antimalarial drugs with possible serious cardiac side effects or low-molecular-weight heparin, assuming that COVID-19 is primarily a coagulation disorder of the pulmonary circulation, or monoclonal antibodies against many different interleukins to counter the cytokine “storm” caused by the direct virus-driven lung damage. In this chaos, research on drugs that interact with RAS pathways is probably the most founded on a solid pathophysiological basis. Further evidence is urgently needed to discover more effective therapies for COVID-19.

DISCLOSURES

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

AUTHOR CONTRIBUTIONS

C.D.P. prepared figures; R.S., F.G., C.D.P., and F.S. drafted manuscript; R.S., F.G., and P.G. edited and revised manuscript; R.S., F.G., C.D.P., P.G., and F.S. approved final version of manuscript.

REFERENCES

  • 1. Abe M, Oikawa O, Okada K, Soma M. Urinary angiotensin-converting enzyme 2 increases in diabetic nephropathy by angiotensin II type 1 receptor blocker olmesartan. J Renin Angiotensin Aldosterone Syst 16: 159–164, 2015. doi:10.1177/1470320314551443.
    Crossref | PubMed | Web of Science | Google Scholar
  • 2. Al-Baadani AM, Elzein FE, Alhemyadi SA, Khan OA, Albenmousa AH, Idrees MM. Characteristics and outcome of viral pneumonia caused by influenza and Middle East respiratory syndrome-coronavirus infections: a 4-year experience from a tertiary care center. Ann Thorac Med 14: 179–185, 2019. doi:10.4103/atm.ATM_179_18.
    Crossref | PubMed | Web of Science | Google Scholar
  • 3. Amat-Santos IJ, Santos-Martinez S, López-Otero D, Nombela-Franco L, Gutiérrez-Ibanes E, Del Valle R, Muñoz-García E, Jiménez-Diaz VA, Regueiro A, González-Ferreiro R, Benito T, Sanmartin-Pena XC, Catalá P, Rodríguez-Gabella T, Delgado-Arana JR, Carrasco-Moraleja M, Ibañez B, San Román JA. Ramipril in high risk patients with COVID-19. J Am Coll Cardiol 76: 268–276, 2020. doi:10.1016/j.jacc.2020.05.040.
    Crossref | PubMed | Web of Science | Google Scholar
  • 4. Arendse LB, Danser AHJ, Poglitsch M, Touyz RM, Burnett JC Jr, Llorens-Cortes C, Ehlers MR, Sturrock ED. Novel therapeutic approaches targeting the renin-angiotensin system and associated peptides in hypertension and heart failure. Pharmacol Rev 71: 539–570, 2019. doi:10.1124/pr.118.017129.
    Crossref | PubMed | Web of Science | Google Scholar
  • 5. Arentz M, Yim E, Klaff L, Lokhandwala S, Riedo FX, Chong M, Lee M. Characteristics and outcomes of 21 critically ill patients with COVID-19 in Washington State. JAMA 323: 1612–1614, 2020. doi:10.1001/jama.2020.4326.
    Crossref | PubMed | Web of Science | Google Scholar
  • 6. Bastos AC, Magalhães GS, Gregório JF, Matos NA, Motta-Santos D, Bezerra FS, Santos RAS, Campagnole Santos MJ, Rodrigues-Machado MG. Oral formulation angiotensin-(1-7) therapy attenuates pulmonary and systemic damage in mice with emphysema induced by elastase. Immunobiology 225: 151893 , 2020. doi:10.1016/j.imbio.2019.12.002.
    Crossref | PubMed | Web of Science | Google Scholar
  • 7. 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 , 2012. doi:10.1136/bmj.e4260.
    Crossref | PubMed | Google Scholar
  • 8. Cha SA, Park BM, Kim SH. Angiotensin-(1-9) ameliorates pulmonary arterial hypertension via angiotensin type II receptor. Korean J Physiol Pharmacol 22: 447–456, 2018. doi:10.4196/kjpp.2018.22.4.447.
    Crossref | PubMed | Web of Science | Google Scholar
  • 9. Chappell MC, Pirro NT, Sykes A, Ferrario CM. Metabolism of angiotensin-(1-7) by angiotensin-converting enzyme. Hypertension 31: 362–367, 1998. doi:10.1161/01.HYP.31.1.362.
    Crossref | PubMed | Web of Science | Google Scholar
  • 10. Clements RT, Vang A, Fernandez-Nicolas A, Kue NR, Mancini TJ, Morrison AR, Mallem K, McCullough DJ, Choudhary G. Treatment of pulmonary hypertension with angiotensin II receptor blocker and neprilysin inhibitor sacubitril/valsartan. Circ Heart Fail 12: e005819 , 2019. doi:10.1161/CIRCHEARTFAILURE.119.005819.
    Crossref | PubMed | Web of Science | Google Scholar
  • 11. Clerkin KJ, Fried JA, Raikhelkar J, Sayer G, Griffin JM, Masoumi A, Jain SS, Burkhoff D, Kumaraiah D, Rabbani L, Schwartz A, Uriel N. Coronavirus disease 2019 (COVID-19) and cardiovascular disease. Circulation 141: 1648–1655, 2020. doi:10.1161/CIRCULATIONAHA.120.046941.
    Crossref | PubMed | Web of Science | Google Scholar
  • 12. Corrales-Medina VF, Musher DM, Wells GA, Chirinos JA, Chen L, Fine MJ. Cardiac complications in patients with community-acquired pneumonia: incidence, timing, risk factors, and association with short-term mortality. Circulation 125: 773–781, 2012. doi:10.1161/CIRCULATIONAHA.111.040766.
    Crossref | PubMed | Web of Science | Google Scholar
  • 13. de Abajo FJ, Rodríguez-Martín S, Lerma V, Mejía-Abril G, Aguilar M, García-Luque A, Laredo L, Laosa O, Centeno-Soto GA, Ángeles Gálvez M, Puerro M, González-Rojano E, Pedraza L, de Pablo I, Abad-Santos F, Rodríguez-Mañas L, Gil M, Tobías A, Rodríguez-Miguel A, Rodríguez-Puyol D, Barreira-Hernandez D, Zubiaur P, Santos-Molina E, Pintos-Sánchez E, Navares-Gómez M, Aparicio RM, García-Rosado V, Gutiérrez-Ortega C, Pérez C, Ascaso A, Elvira C; MED-ACE2-COVID19 study group. 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 | Web of Science | Google Scholar
  • 14. De Maria ML, Araújo LD, Fraga-Silva RA, Pereira LA, Ribeiro HJ, Menezes GB, Shenoy V, Raizada MK, Ferreira AJ. Anti-hypertensive effects of diminazene aceturate: an angiotensin- converting enzyme 2 activator in rats. Protein Pept Lett 23: 9–16, 2016. doi:10.2174/0929866522666151013130550.
    Crossref | PubMed | Web of Science | Google Scholar
  • 15. Deshotels MR, Xia H, Sriramula S, Lazartigues E, Filipeanu CM. Angiotensin II mediates angiotensin converting enzyme type 2 internalization and degradation through an angiotensin II type I receptor-dependent mechanism. Hypertension 64: 1368–1375, 2014 [Erratum in Hypertension 64: e8, 2014]. doi:10.1161/HYPERTENSIONAHA.114.03743.
    Crossref | PubMed | Web of Science | Google Scholar
  • 16. de Souza-Neto FP, Silva MME, Santuchi MC, de Alcântara-Leonídio TC, Motta-Santos D, Oliveira AC, Melo MB, Canta GN, de Souza LE, Irigoyen MCC, Campagnole-Santos MJ, Guatimosim S, Santos RAS, da Silva RF. Alamandine attenuates arterial remodelling induced by transverse aortic constriction in mice. Clin Sci (Lond) 133: 629–643, 2019. doi:10.1042/CS20180547.
    Crossref | PubMed | Web of Science | Google Scholar
  • 17. Donoghue M, Hsieh F, Baronas E, Godbout K, Gosselin M, Stagliano N, Donovan M, Woolf B, Robison K, Jeyaseelan R, Breitbart RE, Acton S. A novel angiotensin-converting enzyme-related carboxypeptidase (ACE2) converts angiotensin I to angiotensin 1-9. Circ Res 87: E1–E9, 2000. doi:10.1161/01.RES.87.5.e1.
    Crossref | PubMed | Web of Science | Google Scholar
  • 18. 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 | Web of Science | Google Scholar
  • 19. 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 | Web of Science | Google Scholar
  • 20. Fosbøl EL, Butt JH, Østergaard L, Andersson C, Selmer C, Kragholm K, Schou M, Phelps M, Gislason GH, Gerds TA, Torp-Pedersen C, Køber L. Association of angiotensin-converting enzyme inhibitor or angiotensin receptor blocker use with COVID-19 diagnosis and mortality. JAMA 324: 168–177, 2020. doi:10.1001/jama.2020.11301.
    Crossref | PubMed | Web of Science | Google Scholar
  • 21. Frederich RC Jr, Kahn BB, Peach MJ, Flier JS. Tissue-specific nutritional regulation of angiotensinogen in adipose tissue. Hypertension 19: 339–344, 1992. doi:10.1161/01.HYP.19.4.339.
    Crossref | PubMed | Web of Science | Google Scholar
  • 22. Furuhashi M, Moniwa N, Mita T, Fuseya T, Ishimura S, Ohno K, Shibata S, Tanaka M, Watanabe Y, Akasaka H, Ohnishi H, Yoshida H, Takizawa H, Saitoh S, Ura N, Shimamoto K, Miura T. Urinary angiotensin-converting enzyme 2 in hypertensive patients may be increased by olmesartan, an angiotensin II receptor blocker. Am J Hypertens 28: 15–21, 2015. doi:10.1093/ajh/hpu086.
    Crossref | PubMed | Web of Science | Google Scholar
  • 23. Giardini V, Carrer A, Casati M, Contro E, Vergani P, Gambacorti-Passerini C. Increased sFLT1/PlGF ratio in COVID-19: a novel link to Angiotensin II-mediated endothelial dysfunction. Am J Hematol. In press. doi:10.1002/ajh.25882.
    Crossref | PubMed | Web of Science | Google Scholar
  • 24. Glowacka I, Bertram S, Herzog P, Pfefferle S, Steffen I, Muench MO, Simmons G, Hofmann H, Kuri T, Weber F, Eichler J, Drosten C, Pöhlmann S. Differential downregulation of ACE2 by the spike proteins of severe acute respiratory syndrome coronavirus and human coronavirus NL63. J Virol 84: 1198–1205, 2010. doi:10.1128/JVI.01248-09.
    Crossref | PubMed | Web of Science | Google Scholar
  • 25. Grasselli G, Zangrillo A, Zanella A, Antonelli M, Cabrini L, Castelli A, Cereda D, Coluccello A, Foti G, Fumagalli R, Iotti G, Latronico N, Lorini L, Merler S, Natalini G, Piatti A, Ranieri MV, Scandroglio AM, Storti E, Cecconi M, Pesenti A; NCOVID-19 Lombardy ICU Network. Baseline characteristics and outcomes of 1591 patients infected with SARS-CoV-2 admitted to ICUs of the Lombardy region, Italy. JAMA 323: 1574–1581, 2020. doi:10.1001/jama.2020.5394.
    Crossref | PubMed | Web of Science | Google Scholar
  • 26. Gu H, Xie Z, Li T, Zhang S, Lai C, Zhu P, Wang K, Han L, Duan Y, Zhao Z, Yang X, Xing L, Zhang P, Wang Z, Li R, Yu JJ, Wang X, Yang P. Angiotensin-converting enzyme 2 inhibits lung injury induced by respiratory syncytial virus. Sci Rep 6: 19840, 2016. doi:10.1038/srep19840.
    Crossref | PubMed | Web of Science | Google Scholar
  • 27. 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: 1–8, 2020. doi:10.1001/jamacardio.2020.1017.
    Crossref | PubMed | Web of Science | Google Scholar
  • 28. Guo X, Zhu Y, Hong Y. Decreased mortality of COVID-19 with renin-angiotensin-aldosterone system inhibitors therapy in patients with hypertension: a meta-analysis. Hypertension 76: e13–e14, 2020. doi:10.1161/HYPERTENSIONAHA.120.15572.
    Crossref | PubMed | Web of Science | Google Scholar
  • 29. 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 | Web of Science | Google Scholar
  • 30. Halliday BP, Wassall R, Lota AS, Khalique Z, Gregson J, Newsome S, Jackson R, Rahneva T, Wage R, Smith G, Venneri L, Tayal U, Auger D, Midwinter W, Whiffin N, Rajani R, Dungu JN, Pantazis A, Cook SA, Ware JS, Baksi AJ, Pennell DJ, Rosen SD, Cowie MR, Cleland JGF, Prasad SK. Withdrawal of pharmacological treatment for heart failure in patients with recovered dilated cardiomyopathy (TRED-HF): an open-label, pilot, randomised trial. Lancet 393: 61–73, 2019. doi:10.1016/S0140-6736(18)32484-X.
    Crossref | PubMed | Web of Science | Google Scholar
  • 31. Hamming I, Timens W, Bulthuis ML, Lely AT, Navis G, 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 | Web of Science | Google Scholar
  • 32. Henry C, Zaizafoun M, Stock E, Ghamande S, Arroliga AC, White HD. Impact of angiotensin-converting enzyme inhibitors and statins on viral pneumonia. Proc Bayl Univ Med Cent 31: 419–423, 2018. doi:10.1080/08998280.2018.1499293.
    Crossref | PubMed | Google Scholar
  • 33. Hoffmann M, Kleine-Weber H, Schroeder S, Kruger N, Herrler T, Erichsen S, Schiergens TS, Herrler G, Wu NH, Nitsche A, Muller MA, Drosten C, Pohlmann S. SARS-CoV-2 cell entry depends on ACE2 and TMPRSS2 and is blocked by a clinically proven protease inhibitor. Cell 181: 271–280.e8, 2020. doi:10.1016/j.cell.2020.02.052.
    Crossref | PubMed | Web of Science | Google Scholar
  • 34. Ichiki T, Labosky PA, Shiota C, Okuyama S, Imagawa Y, Fogo A, Niimura F, Ichikawa I, Hogan BL, Inagami T. Effects on blood pressure and exploratory behaviour of mice lacking angiotensin II type-2 receptor. Nature 377: 748–750, 1995. doi:10.1038/377748a0.
    Crossref | PubMed | Web of Science | Google Scholar
  • 35. 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 | Web of Science | Google Scholar
  • 36. Inciardi RM, Lupi L, Zaccone G, Italia L, Raffo M, Tomasoni D, Cani DS, Cerini M, Farina D, Gavazzi E, Maroldi R, Adamo M, Ammirati E, Sinagra G, Lombardi CM, Metra M. Cardiac involvement in a patient with coronavirus disease 2019 (COVID-19). JAMA Cardiol. In press. doi:10.1001/jamacardio.2020.1096.
    Crossref | PubMed | Web of Science | Google Scholar
  • 37. 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 | Web of Science | Google Scholar
  • 38. Jesus ICG, Scalzo S, Alves F, Marques K, Rocha-Resende C, Bader M, Santos RAS, Guatimosim S. Alamandine acts via MrgD to induce AMPK/NO activation against ANG II hypertrophy in cardiomyocytes. Am J Physiol Cell Physiol 314: C702–C711, 2018. doi:10.1152/ajpcell.00153.2017.
    Link | Web of Science | Google Scholar
  • 39. Jia HP, Look DC, Tan P, Shi L, Hickey M, Gakhar L, Chappell MC, Wohlford-Lenane C, McCray PB Jr. Ectodomain shedding of angiotensin converting enzyme 2 in human airway epithelia. Am J Physiol Lung Cell Mol Physiol 297: L84–L96, 2009. doi:10.1152/ajplung.00071.2009.
    Link | Web of Science | Google Scholar
  • 40. Jung SY, Choi JC, You SH, Kim WY. Association of renin-angiotensin-aldosterone system inhibitors with COVID-19-related outcomes in Korea: a nationwide population-based cohort study. Clin Infect Dis. In press. doi:10.1093/cid/ciaa624.
    Crossref | PubMed | Web of Science | Google Scholar
  • 41. Khan A, Benthin C, Zeno B, Albertson TE, Boyd J, Christie JD, Hall R, Poirier G, Ronco JJ, Tidswell M, Hardes K, Powley WM, Wright TJ, Siederer SK, Fairman DA, Lipson DA, Bayliffe AI, Lazaar AL. A pilot clinical trial of recombinant human angiotensin-converting enzyme 2 in acute respiratory distress syndrome. Crit Care 21: 234, 2017. doi:10.1186/s13054-017-1823-x.
    Crossref | PubMed | Web of Science | Google Scholar
  • 42. Kim J, Choi SM, Lee J, Park YS, Lee CH, Yim JJ, Yoo CG, Kim YW, Han SK, Lee SM. Effect of renin-angiotensin system blockage in patients with acute respiratory distress syndrome: a retrospective case control study. Korean J Crit Care Med 32: 154–163, 2017. doi:10.4266/kjccm.2016.00976.
    Crossref | PubMed | Google Scholar
  • 43. Komukai K, Mochizuki S, Yoshimura M. Gender and the renin-angiotensin-aldosterone system. Fundam Clin Pharmacol 24: 687–698, 2010. doi:10.1111/j.1472-8206.2010.00854.x.
    Crossref | PubMed | Web of Science | Google Scholar
  • 44. 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 | Web of Science | Google Scholar
  • 45. 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: 1–6, 2020. doi:10.1001/jamacardio.2020.1624.
    Crossref | PubMed | Web of Science | Google Scholar
  • 46. 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. In press. doi:10.1093/cid/ciaa415.
    Crossref | PubMed | Web of Science | Google Scholar
  • 47. Liu P, Wysocki J, Souma T, Ye M, Ramirez V, Zhou B, Wilsbacher LD, Quaggin SE, Batlle D, Jin J. Novel ACE2-Fc chimeric fusion provides long-lasting hypertension control and organ protection in mouse models of systemic renin angiotensin system activation. Kidney Int 94: 114–125, 2018. doi:10.1016/j.kint.2018.01.029.
    Crossref | PubMed | Web of Science | Google Scholar
  • 48. 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 | Web of Science | Google Scholar
  • 49. Lukassen S, Chua RL, Trefzer T, Kahn NC, Schneider MA, Muley T, Winter H, Meister M, Veith C, Boots AW, Hennig BP, Kreuter M, Conrad C, Eils R. SARS-CoV-2 receptor ACE2 and TMPRSS2 are primarily expressed in bronchial transient secretory cells. EMBO J 39: e105114 , 2020. doi:10.15252/embj.20105114.
    Crossref | PubMed | Web of Science | Google Scholar
  • 50. Mackey K, King VJ, Gurley S, Kiefer M, Liederbauer E, Vela K, Sonnen P, Kansagara D. Risks and impact of angiotensin-converting enzyme inhibitors or angiotensin-receptor blockers on SARS-CoV-2 infection in adults. Ann Intern Med. In press. doi:10.7326/M20-1515.
    Crossref | PubMed | Web of Science | Google Scholar
  • 51. Magalhaes GS, Barroso LC, Reis AC, Rodrigues-Machado MG, Gregório JF, Motta-Santos D, Oliveira AC, Perez DA, Barcelos LS, Teixeira MM, Santos RAS, Pinho V, Campagnole-Santos MJ. Angiotensin-(1-7) promotes resolution of eosinophilic inflammation in an experimental model of asthma. Front Immunol 9: 58, 2018. doi:10.3389/fimmu.2018.00058.
    Crossref | PubMed | Web of Science | Google Scholar
  • 52. Magalhães GS, Rodrigues-Machado MG, Motta-Santos D, Silva AR, Caliari MV, Prata LO, Abreu SC, Rocco PR, Barcelos LS, Santos RA, Campagnole-Santos MJ. Angiotensin-(1-7) attenuates airway remodelling and hyperresponsiveness in a model of chronic allergic lung inflammation. Br J Pharmacol 172: 2330–2342, 2015. doi:10.1111/bph.13057.
    Crossref | PubMed | Web of Science | Google Scholar
  • 53. 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 | Web of Science | Google Scholar
  • 54. Massiéra F, Bloch-Faure M, Ceiler D, Murakami K, Fukamizu A, Gasc JM, Quignard-Boulange A, Negrel R, Ailhaud G, Seydoux J, Meneton P, Teboul M. Adipose angiotensinogen is involved in adipose tissue growth and blood pressure regulation. FASEB J 15: 2727–2729, 2001. doi:10.1096/fj.01-0457fje.
    Crossref | PubMed | Web of Science | Google Scholar
  • 55. McMurray JJ, Packer M, Desai AS, Gong J, Lefkowitz MP, Rizkala AR, Rouleau JL, Shi VC, Solomon SD, Swedberg K, Zile MR; PARADIGM-HF Investigators and Committees. Angiotensin-neprilysin inhibition versus enalapril in heart failure. N Engl J Med 371: 993–1004, 2014. doi:10.1056/NEJMoa1409077.
    Crossref | PubMed | Web of Science | Google Scholar
  • 56. Mehta N, Kalra A, Nowacki AS, Anjewierden S, Han Z, Bhat P, Carmona-Rubio AE, Jacob M, Procop GW, Harrington S, Milinovich A, Svensson LG, Jehi L, Young JB, Chung MK. Association of use of angiotensin-converting enzyme inhibitors and angiotensin II receptor blockers with testing positive for coronavirus disease 2019 (COVID-19) (Online). JAMA Cardiol. In press. doi:10.1001/jamacardio.2020.1855.
    Crossref | PubMed | Web of Science | Google Scholar
  • 57. Meng J, Xiao G, Zhang J, He X, Ou M, Bi J, Yang R, Di W, Wang Z, Li Z, Gao H, Liu L, Zhang G. Renin-angiotensin system inhibitors improve the clinical outcomes of COVID-19 patients with hypertension. Emerg Microbes Infect 9: 757–760, 2020. doi:10.1080/22221751.2020.1746200.
    Crossref | PubMed | Web of Science | Google Scholar
  • 58. Meng W, Zhao W, Zhao T, Liu C, Chen Y, Liu H, Sun Y. Autocrine and paracrine function of Angiotensin 1-7 in tissue repair during hypertension. Am J Hypertens 27: 775–782, 2014. doi:10.1093/ajh/hpt270.
    Crossref | PubMed | Web of Science | Google Scholar
  • 59. Meng Y, Yu CH, Li W, Li T, Luo W, Huang S, Wu PS, Cai SX, Li X. Angiotensin-converting enzyme 2/angiotensin-(1-7)/Mas axis protects against lung fibrosis by inhibiting the MAPK/NF-κB pathway. Am J Respir Cell Mol Biol 50: 723–736, 2014. doi:10.1165/rcmb.2012-0451OC.
    Crossref | PubMed | Web of Science | Google Scholar
  • 60. Mo P, Xing Y, Xiao Y, Deng L, Zhao Q, Wang H, Xiong Y, Cheng Z, Gao S, Liang K, Luo M, Chen T, Song S, Ma Z, Chen X, Zheng R, Cao Q, Wang F, Zhang Y. Clinical characteristics of refractory COVID-19 pneumonia in Wuhan, China. Clin Infect Dis. In press. doi:10.1093/cid/ciaa270.
    Crossref | PubMed | Web of Science | Google Scholar
  • 61. 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 | Web of Science | Google Scholar
  • 62. Ocaranza MP, Moya J, Barrientos V, Alzamora R, Hevia D, Morales C, Pinto M, Escudero N, García L, Novoa U, Ayala P, Díaz-Araya G, Godoy I, Chiong M, Lavandero S, Jalil JE, Michea L. Angiotensin-(1-9) reverses experimental hypertension and cardiovascular damage by inhibition of the angiotensin converting enzyme/Ang II axis. J Hypertens 32: 771–783, 2014. doi:10.1097/HJH.0000000000000094.
    Crossref | PubMed | Web of Science | Google Scholar
  • 63. Onder G, Rezza G, Brusaferro S. Case-fatality rate and characteristics of patients dying in relation to COVID-19 in Italy. JAMA 323:1775–1776, 2020. doi:10.1001/jama.2020.4683.
    Crossref | PubMed | Web of Science | Google Scholar
  • 64. 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 | Web of Science | Google Scholar
  • 65. Pan F, Ye T, Sun P, Gui S, Liang B, Li L, Zheng D, Wang J, Hesketh RL, Yang L, Zheng C. Time course of lung changes on chest CT during recovery from 2019 novel coronavirus (COVID-19) pneumonia. Radiology 295:715–721, 2020. doi:10.1148/radiol.2020200370.
    Crossref | PubMed | Web of Science | Google Scholar
  • 66. Paz Ocaranza M, Riquelme JA, García L, Jalil JE, Chiong M, Santos RAS, Lavandero S. Counter-regulatory renin-angiotensin system in cardiovascular disease. Nat Rev Cardiol 17: 116–129, 2020. doi:10.1038/s41569-019-0244-8.
    Crossref | PubMed | Web of Science | Google Scholar
  • 67. Ponikowski P, Voors AA, Anker SD, Bueno H, Cleland JGF, Coats AJS, Falk V, González-Juanatey JR, Harjola VP, Jankowska EA, Jessup M, Linde C, Nihoyannopoulos P, Parissis JT, Pieske B, Riley JP, Rosano GMC, Ruilope LM, Ruschitzka F, Rutten FH, van der Meer P; ESC Scientific Document Group. 2016 ESC Guidelines for the diagnosis and treatment of acute and chronic heart failure: The Task Force for the diagnosis and treatment of acute and chronic heart failure of the European Society of Cardiology (ESC)Developed with the special contribution of the Heart Failure Association (HFA) of the ESC. Eur Heart J 37: 2129–2200, 2016. doi:10.1093/eurheartj/ehw128.
    Crossref | PubMed | Web of Science | Google Scholar
  • 68. Ramchand J, Patel SK, Kearney LG, Matalanis G, Farouque O, Srivastava PM, Burrell LM. Plasma ACE2 activity predicts mortality in aortic stenosis and is associated with severe myocardial fibrosis. JACC Cardiovasc Imaging 13: 655–664, 2020. doi:10.1016/j.jcmg.2019.09.005.
    Crossref | PubMed | Web of Science | Google Scholar
  • 69. 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 | Web of Science | Google Scholar
  • 70. Rockx B, Kuiken T, Herfst S, Bestebroer T, Lamers MM, Oude Munnink BB, de Meulder D, van Amerongen G, van den Brand J, Okba NMA, Schipper D, van Run P, Leijten L, Sikkema R, Verschoor E, Verstrepen B, Bogers W, Langermans J, Drosten C, Fentener van Vlissingen M, Fouchier R, de Swart R, Koopmans M, Haagmans BL. Comparative pathogenesis of COVID-19, MERS, and SARS in a nonhuman primate model. Science 368: 1012–1015, 2020. doi:10.1126/science.abb7314.
    Crossref | PubMed | Web of Science | Google Scholar
  • 71. 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. doi:10.1007/s00134-020-05991-x.
    Crossref | PubMed | Web of Science | Google Scholar
  • 72. Salehi S, Abedi A, Balakrishnan S, Gholamrezanezhad A. Coronavirus disease 2019 (COVID-19): a systematic review of imaging findings in 919 patients. AJR Am J Roentgenol 215: 87–93, 2020. doi:10.2214/AJR.20.23034.
    Crossref | PubMed | Web of Science | Google Scholar
  • 73. Santos RA, Simoes e Silva AC, Maric C, Silva DM, Machado RP, de Buhr I, Heringer-Walther S, Pinheiro SV, Lopes MT, Bader M, Mendes EP, Lemos VS, Campagnole-Santos MJ, Schultheiss HP, Speth R, Walther T. Angiotensin-(1-7) is an endogenous ligand for the G protein-coupled receptor Mas. Proc Natl Acad Sci USA 100: 8258–8263, 2003. doi:10.1073/pnas.1432869100.
    Crossref | PubMed | Web of Science | Google Scholar
  • 74. Santos RAS, Oudit GY, Verano-Braga T, Canta G, Steckelings UM, Bader M. The renin-angiotensin system: going beyond the classical paradigms. Am J Physiol Heart Circ Physiol 316: H958–H970, 2019. doi:10.1152/ajpheart.00723.2018.
    Link | Web of Science | Google Scholar
  • 75. Sarzani R, Bordicchia M, Spannella F, Dessì-Fulgheri P, Fedecostante M. Hypertensive heart disease and obesity: a complex interaction between hemodynamic and not hemodynamic factors. High Blood Press Cardiovasc Prev 21: 81–87, 2014. doi:10.1007/s40292-014-0054-3.
    Crossref | PubMed | Google Scholar
  • 76. Sarzani R, Brecher P, Chobanian AV. Growth factor expression in aorta of normotensive and hypertensive rats. J Clin Invest 83: 1404–1408, 1989. doi:10.1172/JCI114029.
    Crossref | PubMed | Web of Science | Google Scholar
  • 77. Sarzani R, Giulietti F, Pentima CD, Giordano P, Spannella F. Severe acute respiratory syndrome coronavirus 2 infection, angiotensin-converting enzyme 2 and treatment with angiotensin-converting enzyme inhibitors or angiotensin II type 1 receptor blockers. Eur J Prev Cardiol. In press. doi:10.1177/2047487320918421.
    Crossref | Web of Science | Google Scholar
  • 78. Sarzani R, Guerra F, Mancinelli L, Buglioni A, Franchi E, Dessì-Fulgheri P. Plasma aldosterone is increased in class 2 and 3 obese essential hypertensive patients despite drug treatment. Am J Hypertens 25: 818–826, 2012. doi:10.1038/ajh.2012.47.
    Crossref | PubMed | Web of Science | Google Scholar
  • 79. Sarzani R, Salvi F, Dessì-Fulgheri P, Rappelli A. Renin-angiotensin system, natriuretic peptides, obesity, metabolic syndrome, and hypertension: an integrated view in humans. J Hypertens 26: 831–843, 2008. doi:10.1097/HJH.0b013e3282f624a0.
    Crossref | PubMed | Web of Science | Google Scholar
  • 80. Sarzani R, Spannella F, Giulietti F, Balietti P, Cocci G, Bordicchia M. Cardiac natriuretic peptides, hypertension and cardiovascular risk. High Blood Press Cardiovasc Prev 24: 115–126, 2017. doi:10.1007/s40292-017-0196-1.
    Crossref | PubMed | Web of Science | Google Scholar
  • 81. Sarzani R, Spannella F, Giulietti F, Fedecostante M, Giordano P, Gattafoni P, Espinosa E, Busco F, Piccinini G, Dessì-Fulgheri P. NT-proBNP and its correlation with in-hospital mortality in the very elderly without an admission diagnosis of heart failure. PLoS One 11: e0153759 , 2016. doi:10.1371/journal.pone.0153759.
    Crossref | PubMed | Web of Science | Google Scholar
  • 82. Sattar N, McInnes IB, McMurray JJV. Obesity is a risk factor for severe COVID-19 infection: multiple potential mechanisms. Circulation 142: 4–6, 2020. doi:10.1161/CIRCULATIONAHA.120.047659.
    Crossref | PubMed | Web of Science | Google Scholar
  • 83. Shao M, Wen ZB, Yang HH, Zhang CY, Xiong JB, Guan XX, Zhong WJ, Jiang HL, Sun CC, Luo XQ, He XF, Zhou Y, Guan CX. Exogenous angiotensin (1–7) directly inhibits epithelial-mesenchymal transformation induced by transforming growth factor-beta1 in alveolar epithelial cells. Biomed Pharmacother 117: 109193 , 2019. doi:10.1016/j.biopha.2019.109193.
    Crossref | PubMed | Web of Science | Google Scholar
  • 84. Shen L, Mo H, Cai L, Kong T, Zheng W, Ye J, Qi J, Xiao Z. Losartan prevents sepsis-induced acute lung injury and decreases activation of nuclear factor kappaB and mitogen-activated protein kinases. Shock 31: 500–506, 2009. doi:10.1097/SHK.0b013e318189017a.
    Crossref | PubMed | Web of Science | Google Scholar
  • 85. Shenoy V, Ferreira AJ, Qi Y, Fraga-Silva RA, Díez-Freire C, Dooies A, Jun JY, Sriramula S, Mariappan N, Pourang D, Venugopal CS, Francis J, Reudelhuber T, Santos RA, Patel JM, Raizada MK, Katovich MJ. The angiotensin-converting enzyme 2/angiogenesis-(1-7)/Mas axis confers cardiopulmonary protection against lung fibrosis and pulmonary hypertension. Am J Respir Crit Care Med 182: 1065–1072, 2010. doi:10.1164/rccm.200912-1840OC.
    Crossref | PubMed | Web of Science | Google Scholar
  • 86. Shenoy V, Gjymishka A, Jarajapu YP, Qi Y, Afzal A, Rigatto K, Ferreira AJ, Fraga-Silva RA, Kearns P, Douglas JY, Agarwal D, Mubarak KK, Bradford C, Kennedy WR, Jun JY, Rathinasabapathy A, Bruce E, Gupta D, Cardounel AJ, Mocco J, Patel JM, Francis J, Grant MB, Katovich MJ, Raizada MK. Diminazene attenuates pulmonary hypertension and improves angiogenic progenitor cell functions in experimental models. Am J Respir Crit Care Med 187: 648–657, 2013. doi:10.1164/rccm.201205-0880OC.
    Crossref | PubMed | Web of Science | Google Scholar
  • 87. Shenoy V, Kwon KC, Rathinasabapathy A, Lin S, Jin G, Song C, Shil P, Nair A, Qi Y, Li Q, Francis J, Katovich MJ, Daniell H, Raizada MK. Oral delivery of angiotensin-converting enzyme 2 and angiotensin-(1-7) bioencapsulated in plant cells attenuates pulmonary hypertension. Hypertension 64: 1248–1259, 2014. doi:10.1161/HYPERTENSIONAHA.114.03871.
    Crossref | PubMed | Web of Science | Google Scholar
  • 88. 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, China. JAMA Cardiol. In press. doi:10.1001/jamacardio.2020.0950.
    Crossref | PubMed | Web of Science | Google Scholar
  • 89. Simonnet A, Chetboun M, Poissy J, Raverdy V, Noulette J, Duhamel A, Labreuche J, Mathieu D, Pattou F, Jourdain M; LICORN and the Lille COVID-19 and Obesity study group. High prevalence of obesity in severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) requiring invasive mechanical ventilation. Obesity 28: 1195–1199, 2020. doi:10.1002/oby.22831.
    Crossref | PubMed | Web of Science | Google Scholar
  • 90. Spannella F, Giulietti F, Balietti P, Borioni E, Lombardi FE, Ricci M, Cocci G, Landi L, Sarzani R. Plasma renin activity to plasma aldosterone concentration ratio correlates with night-time and pulse pressures in essential hypertensive patients treated with angiotensin-converting enzyme inhibitors/AT1 blockers. J Hypertens 35: 2315–2322, 2017. doi:10.1097/HJH.0000000000001438.
    Crossref | PubMed | Web of Science | Google Scholar
  • 91. Spannella F, Giulietti F, Balietti P, Cocci G, Landi L, Lombardi FE, Borioni E, Bernardi B, Rosettani G, Bordoni V, Sarzani R. Renin-angiotensin system blockers and statins are associated with lower in-hospital mortality in very elderly hypertensives. J Am Med Dir Assoc 19: 342–347, 2018. doi:10.1016/j.jamda.2017.09.023.
    Crossref | PubMed | Web of Science | Google Scholar
  • 92. Spannella F, Giulietti F, Di Pentima C, Sarzani R. Prevalence and control of dyslipidemia in patients referred for high blood pressure: the disregarded “double-trouble” lipid profile in overweight/obese. Adv Ther 36: 1426–1437, 2019. doi:10.1007/s12325-019-00941-6.
    Crossref | PubMed | Web of Science | Google Scholar
  • 93. Tian S, Hu W, Niu L, Liu H, Xu H, Xiao SY. Pulmonary pathology of early-phase 2019 novel coronavirus (COVID-19) pneumonia in two patients with lung cancer. J Thorac Oncol 15: 700–704, 2020. doi:10.1016/j.jtho.2020.02.010.
    Crossref | PubMed | Web of Science | Google Scholar
  • 94. Uhal BD, Li X, Piasecki CC, Molina-Molina M. Angiotensin signalling in pulmonary fibrosis. Int J Biochem Cell Biol 44: 465–468, 2012. doi:10.1016/j.biocel.2011.11.019.
    Crossref | PubMed | Web of Science | Google Scholar
  • 95. Vickers C, Hales P, Kaushik V, Dick L, Gavin J, Tang J, Godbout K, Parsons T, Baronas E, Hsieh F, Acton S, Patane M, Nichols A, Tummino P. Hydrolysis of biological peptides by human angiotensin-converting enzyme-related carboxypeptidase. J Biol Chem 277: 14838–14843, 2002. doi:10.1074/jbc.M200581200.
    Crossref | PubMed | Web of Science | Google Scholar
  • 96. Wang X, Ye Y, Gong H, Wu J, Yuan J, Wang S, Yin P, Ding Z, Kang L, Jiang Q, Zhang W, Li Y, Ge J, Zou Y. The effects of different angiotensin II type 1 receptor blockers on the regulation of the ACE-AngII-AT1 and ACE2-Ang(1-7)-Mas axes in pressure overload-induced cardiac remodeling in male mice. J Mol Cell Cardiol 97: 180–190, 2016. doi:10.1016/j.yjmcc.2016.05.012.
    Crossref | PubMed | Web of Science | Google Scholar
  • 97. White MC, Fleeman R, Arnold AC. Sex differences in the metabolic effects of the renin-angiotensin system. Biol Sex Differ 10: 31, 2019. doi:10.1186/s13293-019-0247-5.
    Crossref | PubMed | Web of Science | Google Scholar
  • 98. Wiener RS, Cao YX, Hinds A, Ramirez MI, Williams MC. Angiotensin converting enzyme 2 is primarily epithelial and is developmentally regulated in the mouse lung. J Cell Biochem 101: 1278–1291, 2007. doi:10.1002/jcb.21248.
    Crossref | PubMed | Web of Science | Google Scholar
  • 99. Williams B, Mancia G, Spiering W, Agabiti Rosei E, Azizi M, Burnier M, Clement DL, Coca A, de Simone G, Dominiczak A, Kahan T, Mahfoud F, Redon J, Ruilope L, Zanchetti A, Kerins M, Kjeldsen SE, Kreutz R, Laurent S, Lip GYH, McManus R, Narkiewicz K, Ruschitzka F, Schmieder RE, Shlyakhto E, Tsioufis C, Aboyans V, Desormais I; Authors/Task Force Members. 2018 ESC/ESH Guidelines for the management of arterial hypertension: The Task Force for the management of arterial hypertension of the European Society of Cardiology and the European Society of Hypertension: The Task Force for the management of arterial hypertension of the European Society of Cardiology and the European Society of Hypertension. J Hypertens 36: 1953–2041, 2018. doi:10.1097/HJH.0000000000001940.
    Crossref | PubMed | Web of Science | Google Scholar
  • 100. 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 | Web of Science | Google Scholar
  • 101. 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: 8, 2020. doi:10.1038/s41368-020-0074-x.
    Crossref | PubMed | Web of Science | Google Scholar
  • 102. 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 | Web of Science | Google Scholar
  • 103. 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. doi:10.1016/S2213-2600(20)30076-X.
    Crossref | PubMed | Web of Science | Google Scholar
  • 104. 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 | Web of Science | Google Scholar
  • 105. Yang G, Tan Z, Zhou L, Yang M, Peng L, Liu J, Cai J, Yang R, Han J, Huang Y, He S. Effects of angiotensin II receptor blockers and ACE (angiotensin-converting enzyme) inhibitors on virus infection, inflammatory status, and clinical outcomes in patients with COVID-19 and hypertension: a single-center retrospective study. Hypertension 76: 51–58, 2020. doi:10.1161/HYPERTENSIONAHA.120.15143.
    Crossref | PubMed | Web of Science | Google Scholar
  • 106. Yang P, Gu H, Zhao Z, Wang W, Cao B, Lai C, Yang X, Zhang L, 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, 2014. doi:10.1038/srep07027.
    Crossref | PubMed | Web of Science | Google Scholar
  • 107. 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 | Web of Science | Google Scholar
  • 108. Zambelli V, Bellani G, Borsa R, Pozzi F, Grassi A, Scanziani M, Castiglioni V, Masson S, Decio A, Laffey JG, Latini R, Pesenti A. Angiotensin-(1-7) improves oxygenation, while reducing cellular infiltrate and fibrosis in experimental acute respiratory distress syndrome. Intensive Care Med Exp 3: 8, 2015. doi:10.1186/s40635-015-0044-3.
    Crossref | PubMed | Google Scholar
  • 109. Zhang P, Zhu L, Cai J, Lei F, Qin JJ, Xie J, Liu YM, Zhao YC, Huang X, Lin L, Xia M, Chen MM, Cheng X, Zhang X, Guo D, Peng Y, Ji YX, Chen J, She ZG, Wang Y, Xu Q, Tan R, Wang H, Lin J, Luo P, Fu S, Cai H, Ye P, Xiao B, Mao W, Liu L, Yan Y, Liu M, Chen M, Zhang XJ, Wang X, Touyz RM, Xia J, Zhang BH, Huang X, Yuan Y, Rohit L, Liu PP, Li H. Association of inpatient use of angiotensin converting enzyme inhibitors and angiotensin II receptor blockers with mortality among patients with hypertension hospitalized with COVID-19. Circ Res 126: 1671–1681, 2020. doi:10.1161/CIRCRESAHA.120.317134.
    Crossref | PubMed | Web of Science | Google Scholar
  • 110. 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 | Web of Science | Google Scholar
  • 111. 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 | Web of Science | Google Scholar
  • 112. 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 | Web of Science | Google Scholar
  • 113. 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 | Web of Science | Google Scholar

AUTHOR NOTES