Next Article in Journal
Macrophage Activation in the Dorsal Root Ganglion in Rats Developing Autotomy after Peripheral Nerve Injury
Next Article in Special Issue
Angiotensin Type-2 Receptors: Transducers of Natriuresis in the Renal Proximal Tubule
Previous Article in Journal
Erythrocytes Prevent Degradation of Carnosine by Human Serum Carnosinase
Previous Article in Special Issue
Angiotensin II and the Cardiac Parasympathetic Nervous System in Hypertension
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Alternative RAS in Various Hypoxic Conditions: From Myocardial Infarction to COVID-19

1
Department of Pharmacology and Toxicology, Faculty of Pharmacy, Comenius University, 832 32 Bratislava, Slovakia
2
Institute of Pathophysiology, Faculty of Medicine, Comenius University, 811 08 Bratislava, Slovakia
3
Department of Heart Failure, Clinic of Cardiology, National Institute of Cardiovascular Diseases, 831 01 Bratislava, Slovakia
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2021, 22(23), 12800; https://doi.org/10.3390/ijms222312800
Submission received: 4 November 2021 / Revised: 20 November 2021 / Accepted: 24 November 2021 / Published: 26 November 2021
(This article belongs to the Special Issue Renin-Angiotensin-Aldosterone System in Pathologies)

Abstract

:
Alternative branches of the classical renin–angiotensin–aldosterone system (RAS) represent an important cascade in which angiotensin 2 (AngII) undergoes cleavage via the action of the angiotensin-converting enzyme 2 (ACE2) with subsequent production of Ang(1-7) and other related metabolites eliciting its effects via Mas receptor activation. Generally, this branch of the RAS system is described as its non-canonical alternative arm with counterbalancing actions to the classical RAS, conveying vasodilation, anti-inflammatory, anti-remodeling and anti-proliferative effects. The implication of this branch was proposed for many different diseases, ranging from acute cardiovascular conditions, through chronic respiratory diseases to cancer, nonetheless, hypoxia is one of the most prominent common factors discussed in conjugation with the changes in the activity of alternative RAS branches. The aim of this review is to bring complex insights into the mechanisms behind the various forms of hypoxic insults on the activity of alternative RAS branches based on the different duration of stimuli and causes (acute vs. intermittent vs. chronic), localization and tissue (heart vs. vessels vs. lungs) and clinical relevance of studied phenomenon (experimental vs. clinical condition). Moreover, we provide novel insights into the future strategies utilizing the alternative RAS as a diagnostic tool as well as a promising pharmacological target in serious hypoxia-associated cardiovascular and cardiopulmonary diseases.

1. Introduction

Since its discovery, the alternative RAS axis made quite the entrance into the established molecular cascades and regulatory processes on various tissue and molecular levels. The range of its impact not only resides in physiological regulation and protective counter-regulation of RAS but seems to exert promising results in tissue pathologies as well, hypoxia being one of them. This review is focused on the regulatory impact that ACE2/Ang(1-7)/Mas axis exerts over cardiovascular and pulmonary systems affected by hypoxic changes. In particular, these organ systems are the relevant site of RAS activity, as they are frequently associated with the presence of local RAS. These alone are a topic of discussion as it remains elusive in which way the local systems are connected and operate together [1]. Pathological conditions that we discussed in this review are either acute, chronic or intermittent hypoxic phenotypes with limited efficiency of therapeutical strategies and serious adverse consequences on a patient’s health outcome. We also aspired to show the purpose and clinical relevance of the ACE2/Ang(1-7)/Mas axis for future pharmacological interventions. Targeting the ACE2/Ang(1-7)/Mas axis, either through the enhancement of their activity, prevention of the metabolism or administration of selective activators could reveal the great potential for novel treatment regimens that reside in the alternative arm of RAS.

2. The Other Side of RAS—Protective ACE2/Ang(1-7)/Mas Axis

In contrast to the classical RAS pathway which as thoroughly researched for many years [2], another branch of RAS, specifically ACE2/Ang(1-7)/Mas axis was described as an alternative non-canonical pathway. The protective arm of RAS produces effects that could be referred to as counterbalancing actions that oppose those elicited by traditional RAS AngII/AT1R signaling cascade. Some of these effects intervene with the maintenance of vascular tone [3,4], anti-inflammatory, anti-proliferatory [5] or anti-remodeling properties [6] and create a link to the protective signalization pathways.

2.1. Activity Mode of ACE2-Dependent Regulation

ACE2 is broadly expressed in a variety of organs and almost all kinds of tissues [7]. As a type I transmembrane protein with monocarboxypeptidase activity and 42% amino acid sequence homology in the catalytic domains with ACE [8,9,10], ACE2 manages the part of a key negative regulator of the ACE/AngII axis. The importance of ACE2 is also implemented in its function as a receptor and contact point for severe acute respiratory syndrome coronavirus (SARS-CoV) [11] and SARS-CoV-2 [12], a virus responsible for the ongoing global COVID-19 pandemic. ACE2 exerts counterregulatory actions over ACE/AngII/AT1R axis on two distinct levels: firstly, by cleavage of its main effector AngII or by limiting the availability of ACE substrate through hydrolysis of AngI and secondly, by generating the active peptide Ang(1-7). In brief, ACE2 via carboxypeptidase activity, hydrolyses octapeptide AngII, by cleavage of its C-terminal residue, resulting in the generation of heptapeptide Ang(1-7). Ang(1-7) can be also formed less efficiently via ACE-mediated conversion of Angiotensin 1-9 (Ang1-9) which was previously generated by ACE2-catalyzed hydrolysis of Angiotensin I (AngI) [8,9,13]. The key active product of the non-classical reaction cascade is Ang(1-7) whose physiological role is primarily mediated via receptor Mas (Figure 1) [14]. It is worth mentioning that another peptide product of ACE2, Ang1-9, exhibits its own physiological effects mediated through Mas receptors and AT2Rs as well. Therefore, the physiological activity of Ang1-9 does not have to end with the fact that it is an Ang(1-7) precursor but could be considered an accessory pathway of the protective RAS arm [15].
ACE2 activity is not restricted by substrate specificity for AngII but is able to cleave single-peptide residues from several bioactive peptides such as AngI, kinins (des-Arg9 bradykinin, but not bradykinin), apelins, dynorphins or neurotensins [8,16]. On the other hand, ADAM17, a disintegrin and metalloproteinase also referred to as tumor necrosis factor α (TNFα) converting enzyme, was found to mediate the proteolysis and ectodomain shedding of ACE2, thereby decreasing its activity in tissues and at the same time increasing activity of soluble ACE2 which lacks the membrane anchors as it circulates the blood [17].

2.2. Angiotensin (1-7) and Mas Receptor Interactions

Ang(1-7), formed directly or indirectly by ACE2 or different enzymes from the precursor peptides AngI or AngII [13,16], is an endogenous agonist for G protein-coupled receptor Mas [14]. Mas was initially identified as proto-oncogen [18] but gained a status of orphan GPCR [19] mediating Ang(1-7) signaling later on [14]. A more recent paper however documented contrasting results and suggested that Ang(1-7) does not bind to Mas directly [20], however, authors also stressed that used tissue preparations or utilization of recombinant ligand/receptor could differ from the real in vivo situation. Needless to say, there is conflicting evidence regarding the direct binding of Ang(1-7) to Mas as discussed below (for more detailed information see the review of Karnik et al., 2017 [21]). Interestingly, Mas was implicated in heteromeric interactions with other RAS receptors: forming functional interaction with AT2R [22], while inhibiting AT1R receptor response [23], which is an important fact to take into consideration whilst looking at its downstream molecular pathways. In experimental settings, the use of selective Mas antagonist A779 ((D-Ala7)-angiotensin-(1-7)) [24] as well as Mas deficiency blunted a significant amount of protective effects of the axis and promoted adverse signaling [14,25]. While these types of results do endorse the prominence of Mas in the alternative RAS axis, extended research is still indispensable. Speaking of RAS receptors, besides Mas, Ang(1-7) is known to bind AT2R [2] or the Mas-related G protein-coupled receptor member D (MRGD) [26].

2.3. ACE2/Ang(1-7)/Mas Axis in Cardiovascular and Pulmonary Regulation

Selected molecular pathways affected by ACE2/Ang(1-7)/Mas axis in cardiovascular and pulmonary systems are graphically summarized in Figure 2.

2.3.1. Heart

Local cardiac RAS activity and its interconnection with relevant molecular pathways have been a subject of interest in numerous studies that have gradually improved our understanding of their local range of actions. Specifically, on the cardiomyocyte level in which proliferative/hypertrophic stimuli are relevant factors for future cardiac remodeling, a study by Tallant et al. demonstrated Ang(1-7)-dependent inhibition of cardiomyocyte growth via mitogen-activated protein kinase (MAPK) extracellular signal-regulated kinase (ERK) 1/2 activity inhibition [27]. Ang(1-7) is generally engaged in antihypertrophic signalization, regarding the effect on the hypertrophic nuclear factor of activated T cells (NFAT) or glycogen synthase kinase 3β (GSK3β) signaling cascade [6], and involved in direct cardiac antifibrotic effects [28]. Ang(1-7) also exhibited Mas-dependent activation of endothelial NO synthase (eNOS) and Akt [29], or increased cardiac NOS expression and activity via AT2R- or bradykinin-dependent pathway [30]. Concurrently, cardioprotective actions of Ang(1-7) could be also associated with its antioxidative [31] or anti-inflammatory properties, in particular with modulation of TNF-α, interleukin-6 or interleukin-10 [32].
Notwithstanding the apparent protective effects of the alternative axis against pathological remodeling, the effects on cardiac function under physiological conditions in the settings of ACE2 gene knockout are ambiguous. In the first report by Crackower et al., ACE2-deficient hearts displayed reduced cardiac contractility, aortic and ventricular pressures, and structural changes with no signs of hypertrophy or fibrotic changes which lead them to consider the ACE2 as a potential regulator of cardiac function [33]. Conversely, Gurley et al. found no evidence of ACE2 being involved in the regulation of cardiac structure or function [34]. Other research groups were similarly not able to confirm the cardiac phenotype observed by Crackower and associates [35,36]. In line with all these findings, physiological heart function might not be ACE2-dependent, but ACE2-deficiency could rather cause higher susceptibility to cardiac injury [35,36] and favor the deleterious effects of AngII [37].

2.3.2. Endothelium, Blood Vessels and Thrombosis

As the counterregulatory RAS, ACE2/Ang(1-7)/Mas axis induces vascular relaxation and improves endothelial function through the activation of eNOS and concomitantly stimulates NO/cGMP/PKG signaling pathway possibly via Akt-dependent mechanism [3,4]. As regards the coronary circulation, the data are similar in the matter that Ang(1-7) induced vasodilatation that could be blocked by Mas antagonist A-779 [38]. Anti-proliferative effect on vascular growth was proposed to be mediated by prostacyclin release, subsequent stimulation of cAMP production and suppression of AngII stimulated ERK1/2 MAPK activities as well as NAD(P)H oxidase-derived superoxide anion-mediated PI3K/Akt pathway [5,39]. Moreover, Sousa-Lopes and associates showed that Ang(1-7) negatively modulates ERK1/2 activation via stimulation of mitogen-activated protein phosphatase (MKP) 1 phosphorylation [40].
In addition to the established role in vascular tone modulation, both arms of RAS contribute to the regulation of hemostasis. While the activity of AngII on AT1R was implicated in the prothrombotic mechanisms and potentiation of platelet aggregation [41], the effects mediated through AT2R lead to thromboprotection via NO and prostacyclin production [42]. On the other hand, mounting evidence on Ang(1-7) is suggesting its potent antithrombotic effects that are mediated via the Mas [43] in both endothelial cells [3,44] and platelets [25,45]. In endothelial cells, Ang(1-7) gives rise to NO release and prostacyclin production [3,44], similarly to platelets, where Fraga-Silva et al. documented Ang(1-7)/Mas activation-stimulated NO production that was inhibited by both Mas antagonist A-779 or Mas knockout [25]. On a related note, elevated plasma levels of NO and prostacyclin were found in mice overexpressing AT2R and Mas receptors [45].

2.3.3. Lungs

On the subject of local RAS activity, lungs are known to express all the major components of RAS [46] including pulmonary vasculature endothelial and smooth muscle cells, type I and II alveolar epithelial cells, bronchial epithelial cells [47,48,49,50,51,52] or bronchial smooth muscle [53]. In fact, the existence of the intrapulmonary renin–angiotensin system as a locally active system is suggested although not confirmed and therefore the possibility of lungs being a self-sufficient system in terms of expressing its own mediators without depending on their circulating precursors should be taken into consideration [46].
Although the excessive activation of classical ACE/AngII/AT1R RAS branch is responsible for many detrimental effects on the pulmonary vasculature and lung parenchyma such as vasoconstriction [4], pro-inflammatory [5,54], pro-apoptotic [55] or pro-fibrotic effects [56] as well as the acceleration of thrombosis [57], induction of oxidative stress [58] or growth-promoting actions involved in pulmonary remodeling [59], Angiotensin II-type 2 receptor (AT2R) cascade [60] likewise the alternative arm of RAS exert predominantly opposite effects. ACE2/Ang(1-7) pathway is reportedly involved in the mediation of anti-apoptotic, anti-inflammatory and anti-proliferative response via the attenuation of MAPKs activities and nuclear factor–ĸB (NF-ĸB) pathway [53,55,61,62,63]. Concurrently, the axis is suggested to regulate the activation of MAPK phosphatase-2 [64]. In particular, under ovalbumin-induced settings, Ang(1-7) exhibited anti-inflammatory properties, reduced inflammatory cell infiltration in peribronchial, perivascular or alveolar regions, inhibited phosphorylation of ERK1/2 or IĸB-α, IgE and decreased multiple pro-inflammatory cytokines and chemokines [53,63]. ACE2/Ang(1-7)/Mas was further implicated in the management of cell survival mechanisms and anti-apoptotic regulation given that ACE2/Ang(1-7) prevented alveolar epithelial cell JNK phosphorylation, caspase activation and nuclear fragmentation [61,64,65,66] or lipopolysaccharide-induced apoptosis of pulmonary microvascular endothelial cells (PMVECs) [55,62]. Furthermore, the axis exerts protective effects against lung remodeling by decreasing the collagen deposition and expression [53] and against lung fibrosis in which oxidative stress represents a relevant role along with NOX4-derived ROS-mediated RhoA/Rho kinase pathway [67,68,69]. On top of that, Ang(1-7) axis does seem to inhibit the activity of ADAM17, a shedding enzyme involved in ACE2 proteolysis and pulmonary inflammation [66].

3. ACE2 Deficiency-Related Pathologies Underlying Hypoxia

Hypoxia is a condition in which the body or a region of the body is deprived of an adequate oxygen supply at the tissue level [70]. During acute hypoxia, hypoxic cells temporarily shift from oxidative to anaerobic (lactate) metabolism, providing a small amount of energy [71]. Severe or prolonged hypoxia can lead to cell death [72]. In most tissues of the body, the response to hypoxia is vasodilation, thus allowing greater perfusion. By contrast, in the lungs, the response to hypoxia is vasoconstriction [73]. Ischemic hypoxia (e.g., ischemia-reperfusion injury [74]) and hypoxemic hypoxia (e.g., ARDS [75]) are well-known examples of acute hypoxic conditions. On the other hand, chronic hypoxia is caused by long-lasting reduced oxygen content in the blood [76]. Chronic hypoxia is typically observed in chronic obstructive pulmonary disease (COPD) with the development of emphysema and subsequent decreased air-exchanging capacity of lungs [77], in pulmonary hypertension (PH) accompanied by pulmonary edema [78], or heart failure (HF) characterized by the insufficient tissue perfusion and blood congestion in the pulmonary blood stream (congestive heart failure) [79].

3.1. Activity of ACE2-Ang(1-7)-Mas Axis in Settings of Acute Hypoxia

3.1.1. Ischemia-Reperfusion Injury and Myocardial Infarction

Ischemia-reperfusion injury (I/R injury or IRI) is the tissue damage caused by the paradoxical exacerbation of cellular dysfunction and death when blood supply returns (reperfusion) to previously ischemic tissue (anoxia or hypoxia). The absence of oxygen and nutrients during the ischemic period creates a condition in which the restoration of circulation results in inflammation and oxidative damage. IRI occurs in a wide range of organs including the heart, lungs, kidney, gut, skeletal muscle and brain and may involve not only the ischemic organ itself but may also induce systemic damage to distant organs [80]. An example can be renal IRI leading to renal failure and subsequent cardiovascular pathologies (hypertension, heart failure, etc.) [81]. The renin–angiotensin system is known to be stimulated after myocardial infarction (MI) [82]. Notwithstanding the presence of Ang II is significant in MI, the role of the cardioprotective counterpart, ACE2-Ang(1-7)-Mas axis, as an early form of compensatory mechanism is unknown.
The situation surrounding ACE2/Ang(1-7) axis in the heart is slightly trickier. Renal IRI can cause degenerative changes in heart histology with the onset of oxidative stress and inflammation accompanied by increased ACE/ACE2 and Ang II/Ang(1-7) levels in heart tissue [83]. If we take a closer look at myocardial IR injury, or more precisely animal (rat and murine) models of myocardial infarction (mainly occlusions of the left anterior descending coronary artery), the results almost consistently showcase surprising findings. Three different papers [24,84,85] showed that 4 weeks after MI the protein levels of ACE2 were increased in the infarcted zone, whereas the non-infarcted zone was not affected, although neglecting Ang(1-7) levels. A study by Kassiri et al. [85], however, observed decreased mRNA ACE2 levels in the infarcted zone, which could suggest that the heart itself is in fact ACE2-deficient but the excess of ACE2 might come from the circulating blood into the damaged tissue. On the other hand, the work of Qi et al. [86], made an effort to contradict these observations, but in fact, they collected tissues from the peri-infarct region, not directly from the infarcted zone. Moreover, one experiment showed increased mRNA of ACE and ACE2 in both infarct and non-infarct regions after the same period of reperfusion [87]. Overall, all these findings are still inconclusive and need further investigation.
Despite the discrepancies in these findings, gene-modifying and treatment-utilizing animal models can shed more light on this problem. Similarly, to other I/R models, ACE2-transgenic rats showed reduced left ventricular (LV) volume, the extent of myocardial fibrosis, increased tissue levels of ACE, AngII, and collagen type I in the myocardium and increased LV ejection fraction [88]. Loss of ACE2 enhanced the susceptibility to MI, with increased mortality, infarct expansion, oxidative stress, remodeling, inflammation and adverse ventricular remodeling characterized by ventricular dilation and systolic dysfunction [85]. Treatment of ex vivo hearts by administration of 10−8 M Ang(1-7) into the bath solution using Langendorff technique re-established the impulse conduction during ischemia-reperfusion and reduced incidence of arrhythmias during IR, possibly due to cardiomyocyte hyperpolarization via the activation of sodium pump [89]. Activation of ACE2 by diminazene aceturate (DIZE) treatment [86,90,91,92] led to an improvement of almost all evaluated parameters (however, lacking evidence against amelioration of hypertrophy). These effects were abolished by ACE2 inhibition (“compound 16”) [84,86] or by Mas-receptor antagonist (A779) [24,88]. Nevertheless, there is still insufficient evidence regarding human studies. Just one study by Wang et al. [93] reported that patients with low serum ACE2 levels (≤1.06 ng/mL), 1 h after coronary artery bypass grafting, were associated with an increased risk of postoperative MI. To further investigate the role of ACE2-Ang(1-7)-Mas axis in clinical settings of MI, we need more human trials, especially with randomized treatments. Nonetheless, mentioned studies suggest that despite the inconsistency of observed ACE/ACE2 levels between various IR models and tissues, administration of ACE2 or any modulation of ACE2/Ang(1-7)/Mas axis has a cardioprotective potential in all of these conditions and thus such treatment could be a candidate for future human trials.
The experimental models of the lung (hind-limb constriction [94,95]) and kidney (hind-limb constriction [96] or renal pedicles [83]) IRI conducted on rats and mice lead to a rise of ACE and AngII tissue and plasma levels with a simultaneous decrease in ACE2 and Ang(1-7) levels. Only one study [97] found increased both ACE2, Mas and decreased ACE and AT1R mRNA expression in kidney tissue after 4 h of reperfusion while plasma levels of both Ang(1-7) and AngII were unchanged compared to control group. Nonetheless, the data may differ on tissue or plasma levels, experimental model (duration of reperfusion from 4 to 24 h after ischemia) or expression evaluation method (WB/ELISA/fluorescence versus qRT-PCR). Indeed, the paper by Chen et al. [94] focused on this issue and observed an immediate increase in AngII in lung tissue along with an increase in Ang(1-7) levels after 4 h of reperfusion, however, they subsequently decreased. Yet, in the plasma, AngII was increased from the beginning and Ang(1-7) was decreasing over time. Another model [83] with a longer reperfusion period (24 h) observed increased ACE/ACE2 and also Ang II/Ang(1-7) tissue ratio but unexpectedly the levels of plasma Ang(1-7) were decreased whereas the levels of plasma ACE2 did not change. To further elucidate the role of ACE2/Ang(1-7) in I/R injury in lungs and kidneys, we can make a look at rodent models with modified ACE2 expression. Both lung and kidney post-ischemic tissues exhibited inflammation (increased expression of inflammatory cytokines and enhanced leukocyte infiltration), oxidative stress and activation of apoptosis. ACE2-deficient animals showed more severe symptoms [96,98] compared to the animals with ACE2-induced overexpression eliciting only milder symptoms [95,99] in contrast to the wild-type. Similar results were obtained after the treatment with ACE2 activator DIZE in the lung [95] and renal [83] IRI or by using the AT2R agonist (“compound 21”). These results suggest that renal and lung IRI may ultimately lead to increased ACE/ACE2 tissue ratio and plasma levels, with ACE2 being an important factor in alleviating IRI symptoms.

3.1.2. The Role of ACE2-Ang(1-7)-Mas in Acute Respiratory Distress Syndrome

Acute respiratory distress syndrome (ARDS) is a serious lung condition characterized by rapid onset of widespread inflammation with diffuse alveolar damage and fluid build-up in the lung alveoli [100]. The fluid keeps the lungs from filling with enough air so that a person cannot breathe, the lungs cannot move enough oxygen into the bloodstream and body tissues are restricted from sufficient perfusion [75]. ARDS is also one of the endpoints of COVID-19 disease [101]. In fact, the postulated pathomechanism of COVID-19 mentions a modulation of the ACE2-Ang(1-7) axis, however, other diseases causing ARDS can affect this axis as well (though there is no direct evidence, this idea was based on observation from the SARS epidemic in 2002 [102,103]). For instance, lethal strains of the Influenza A virus (H7N9) were observed to downregulate ACE2 and upregulate AngII levels in mice [104]. Additionally, ACE2 deficiency was found to aggravate the symptoms in this model. These findings are also supported by a human study [105] of various causes of ARDS which observed an increased Ang(1-10)/Ang(1-9) ratio, thus suggesting a decrease in ACE2 activity. On the contrary, one rat study of cigarette smoke-induced ARDS [106] detected raised levels of both ACE and ACE2, although cigarette smoke alone is able to increase ACE2 expression [107,108]. To support the idea of decreased activity of ACE2 in ARDS, the recombinant ACE2 shows activity on decreasing the occurrence of ARDS symptoms in murine [109] and rat [110] models induced by acid aspiration or lipopolysaccharide (LPS). Regarding these findings, ACE2 is expected to have beneficial effects on the prevention of ARDS, mainly based on its anti-inflammatory, as well as anti-remodeling properties.

3.1.3. ACE2-Ang(1-7)-Mas Branch and COVID-19

By the fall of 2021, the COVID-19 pandemic officially reached over 220 million infected and over 4.5 million deaths worldwide. The studies showed that patients suffering from COPD [111], cardiovascular diseases [112] and diabetes [113] are more vulnerable to severe courses of COVID-19. As we previously mentioned, these pathologies are known to increase the ACE/ACE2 ratio in such patients. SARS-CoV-2 utilizes ACE2 to enter the human cells [12], however, we currently have no direct data on how the virus affects this ratio. Nevertheless, patients infected by a closely related SARS-CoV virus in 2002 showed a decrease in ACE2 expression [114,115,116], therefore, it is likely to expect that SARS-CoV-2 infection might increase the ACE/ACE2 ratio as well. It is also important to mention, that many medications might affect the expression level or activity of ACE2, even though, the gathered up-to-date data regarding the deleterious (increasing the risk of infection) or beneficial (protective effects of ACE2 on pulmonary functions) effects these might exert are conflicting. One Korean retrospective study [117] claims that the renin–angiotensin system blockers might increase the risk of infection by SARS-CoV-2, but on the other hand, the manifestation of the disease was milder and did not result in higher mortality. Nevertheless, a recently published cohort study on 8.3 million people refutes this thesis, in fact, ACEi or ARBs seems to decrease the risks of COVID-19 and overall mortality rather than the opposite [118,119]. Additionally, a difference between the ACEi and ARBs towards the ACE2/Ang(1-7) axis should be taken into consideration since ACE inhibitors deplete also ACE2 substrate resulting in a reduction of anti-inflammatory metabolites production [120].
The other important fact is that patients frequently die from suffocation by alveolar damage caused by ARDS [121,122], multi-organ failure caused by cytokine storm [122] or thromboembolism [122]. In respect of the anti-inflammatory [123] and anti-aggregant [124] properties of ACE2, current phase 2 clinical trials might bring promising results in regard to recombinant human ACE2 as a potential treatment of ARDS [125] and severe cases of COVID-19 [126]. ACE2 was found to be protective in murine [109,127,128] and rat [129] models of ARDS. Decreased ACE2 levels in lungs suffering from ARDS caused by RSV (Respiratory Syncytial Virus) were also observed in neonate children [130]. Despite insufficient data about the role of ACE2 in COVID-19 associated complications, soluble ACE2 is thought to have protective effects on a pulmonary form of the disease not only by its anti-inflammatory and anti-remodeling properties but importantly by exhibiting an anti-aggregant activity, which is important for the survival of hospitalized COVID-19 patients.

3.2. ACE2-Ang(1-7)-Mas Arm in Conditions of Intermittent Hypoxia

Intermittent hypoxia (also known as episodic hypoxia) is a condition in which a person or animal undergoes alternating periods of normoxia and hypoxia. Intermittent hypoxia may exert various effects on blood pressure, glucose tolerance, sympathetic activation, cognition, and inflammation depending on the dosage or experimental protocol [131,132]. Underlying mechanisms can be complicated or even unknown. One of the clinically relevant models of intermittent hypoxia is a condition known as obstructive sleep apnea (OSA). OSA is the most common sleep-related breathing disorder characterized by recurrent episodes of complete or partial obstruction of the upper airway leading to reduced or absent breathing during sleep. Relevant outcomes may include a fall in blood oxygen saturation (hypoxia), a disruption in sleep, or both [133]. It is also considered a risk factor for refractory asthma [134].

3.2.1. Cardiovascular System

Chronic intermittent hypoxia (CIH) is the main pathomechanism underlying OSA which is known to affect RAS activity [134]. The utilization of rodent animal models of CIH (7 days of hypoxia followed by normoxia) to mimic the clinical onset of OSA causes many pathological changes in various organs. CIH caused an increase in ACE/ACE2 ratio in blood plasma, renal arterioles and other renal tissues, which led to an elevation of blood pressure, arterial wall thickening (arterial fibrosis), renal fibrosis and generation of reactive oxygen species (ROS) connected to oxidative stress and inflammation [135,136]. However, in a study by Lu et al., most of these effects were attenuated after administration of exogenous Ang(1-7) [137], which proposes a cardioprotective effect in OSA. Elevated levels of ACE and AngII with decreased levels of ACE2 were also observed in the hearts of mice undergoing CIH [138]. This imbalance was reversed after treatment with angiotensin receptor blocker (ARB) telmisartan. Cardiovascular benefits of ACE inhibitors and ARBs are well known in clinical practice, however, implications for the CIH are unclear. Effects of administered ACE2 or Ang(1-7) have not yet been satisfyingly examined and require further examination.

3.2.2. Lungs and Pulmonary Vasculature

In the model of subchronic intermittent hypoxia/hyperoxia (after 72 h) on primary murine lung endothelial cells increased levels of ACE and reduced levels of ACE2 were observed [139]. Another in vitro model observed TGF-β-mediated epithelial-to-mesenchymal transition (EMT) in LPS-treated human bronchial epithelial cells. Angiotensin(1-7) treatment abolished the aggravation of TGF-β-mediated EMT and epithelial fibrosis upon chronic IH exposure [134]. CIH in vivo also showed a high degree of interstitial edema, alveolar atelectasis, oxidative stress, and inflammatory cell infiltration in alveolar epithelial cells [140,141] with ACE and ACE2 expression impairment. Treatment with angiotensin(1-7) was able to reverse a lung injury through attenuation of lung fibrosis [134], inflammation and oxidative stress [141].
It is generally accepted that hypoxia also exacerbates pulmonary fibrosis. Hypoxia in vitro stimulates TGF-β-mediated signaling resulting in higher collagen content, glycosaminoglycan production and accumulation of extracellular matrix in human lung fibroblasts [142,143]. This effect was also observed in chronic intermittent hypoxia in bleomycin-induced lung fibrosis in rat and murine models [144,145,146] associated with the activation of NF-κB pathway, increased levels of growth factors (TGF-β, PDGF), inflammatory cytokines (TNF, IL-1β) and overall mortality.
The effects of ACE2-Ang(1-7)-Mas axis in acute high altitude hypoxia might also be interesting, however, they have not been studied in this context. There is only a little data available and targeted exclusively on RAAS. It seems that RAAS activity is unchanged in healthy participants [147,148,149], while they are maintained in high (3000 m) altitudes. The situation changes after the further ascend to very high (above 5000 m) altitudes [150,151], where the data seem to be contradictory. It makes high altitude hypoxia a good candidate for future studies regarding both RAAS and ACE2-Ang(1-7)-Mas axis. The possible effects of the angiotensin system on high altitude pulmonary edema are also needed to be evaluated.

3.3. ACE2-Ang(1-7)-Mas Branch in Chronic Hypoxic Conditions

3.3.1. ACE2-Ang(1-7) System Association with Heart Failure

Cardiac muscle cannot maintain essential cellular processes under hypoxemic or ischemic conditions and thus a sufficient supply of oxygen is indispensable to sustain cardiac function and viability. However, the role of oxygen in the heart is complex and goes well beyond its role in energy metabolism [152]. ACE2-Ang(1-7) system is known to be imbalanced in many cardiovascular diseases and is also affected by the hypoxic condition, as we display in this review, thus changes in the ACE/ACE2 ratio in plasma or tissue in HF patients can be expected.
The early-stage (up to 4 weeks after surgery) of rat and murine HF seems to be associated with the upregulation of both ACE/Ang II and ACE2/Ang(1-7) pathways [153,154,155,156], whereas the advanced or end-stage (more than 4 weeks after surgery) of HF tend to be associated with downregulation [153,157] of ACE2/angiotensin(1-7) and upregulation of the ACE/AngII pathway. Although these data are consistent in this time-related manner in both plasma and heart tissues, two studies, a canine congestive heart failure model [158] and a rodent HF model with ascending aortic constriction [159], do not support this theory. Our earlier study observed a similar trend as a low dose of daunorubicin, mimicking early-stage of heart failure, produced an increase in cardiac ACE2 mRNA levels whilst the higher dose markedly decreased ACE2, suggesting a direct effect of chronic daunorubicin cardiomyopathy (end-stage heart failure) on ACE2 expression [160]. ACE/ACE2 balance may determine the decompensation of HF in early stages with progressive downregulation as the disease progress.
To elucidate cardioprotective effects of Ang(1-7) in failing hearts, several rodent models of HF showed significant improvements in inflammation, oxidative stress, fibrosis, ejection fraction and overall decreased mortality in ACE2-overexpressing animals [161,162] or after treatment with rhACE2 [163] or Ang(1-7) [164]. These effects were antagonized by the blockade of the Mas receptor. ACE inhibitors are clinically acknowledged to possess such effects, however, ACE2 overexpression seems to have at least comparable cardioprotective effects similar to AT1R antagonists (irbesartan) [37] or it can be even more beneficial compared to ACE inhibitors (cilazapril) in the treatment of doxorubicin-induced cardiomyopathy [161]. The other important fact is that some AT1R antagonists (olmesartan, candesartan, losartan) also show some effects on ACE2/Ang(1-7) axis independently on AngII targeting, and some (irbesartan, valsartan) do not [165]. This idea can be supported by another finding that olmesartan seems to be superior to telmisartan in reduction of myocardial collagen deposition and adverse remodeling [156], explained by the additional effect on the ACE2/Ang(1-7) axis.
According to the clinical data, patients diagnosed with HF show an increasing trend in plasma levels of circulating soluble ACE2 in more severe NYHA groups (from NYHA-II to NYHA-IV) [159,166,167,168]. Surprisingly, LV tissue levels of ACE2 seem to decrease as the disease progresses [169] despite increased ACE2-mRNA expressions [169,170,171]; or even unchanged in late-stage patients [172]. Such phenomenon can be explained by the fact that circulating ACE2 is capable of systemic AngII cleavage, however, it lacks local (myocardial) activity. A study by Messmann et al. [170] even found decreased cardiac Mas mRNA expression. It is also important to mention that actual guidelines indicate the ACE inhibitors or ARB (among others) as first-line therapy in congestive HF patients. As we previously mentioned, these drugs are known to modulate the ACE2/ACE ratio. Unfortunately, most of the studies are completely neglecting this fact, except for one study by Epelman et al. [166] that admits this limitation and also discusses the problem of circulating versus tissue ACE2. There is also a paper by Walters et al. stating that increased ACE2 plasma activity is present in patients with atrial fibrillation since they propose that elevated plasma ACE2 activity levels are connected with increased shedding of ACE2 from the tissue into the circulation and thus increase in tissue AngII is leading to structural left atrial remodeling [173]. Therefore, plasma ACE2 was proposed as a marker of disease severity and structural remodeling in human atrial fibrillation.
The effects of failing hearts on the ACE/ACE2 ratio are as complex as in the case of COPD. Many limitations are required to be eliminated in future studies in order to evaluate the true nature of this relationship. Based on long clinical practice in the use of ACEi and ARBs, their effects on the ACE/ACE2 ratio are essential in the prognosis of HF patients. On the other hand, it makes them one of the biggest limitations of potential human trials elucidating the role of ACE2 in HF. Despite the unknown role of ACE2 in HF pathomechanism, evidence supporting its cardioprotective potential is getting stronger.

3.3.2. Obstructive Coronary Artery Disease and the Role of ACE2-Ang(1-7)-Mas Axis

Coronary artery disease (CAD) or ischemic heart disease (IHD), is one of the most common cardiovascular diseases characterized by the obstruction of heart arteries with consequent reduction of blood flow into the myocardium [174]. CAD is also one of the known complications in the development of heart failure [175].
From a clinical perspective, drugs affecting RAS are commonly prescribed in CAD; however, their efficacy is in question. They are effective in the reduction of cardiovascular events compared to placebo, but not when compared with other therapeutically used drugs in CAD (calcium channel blockers, thiazide diuretics, etc.). Moreover, RAS blockers are recommended only in CAD patients with hypertension or heart failure, not in CAD patients with a lower cardiovascular risk [176,177]. Therefore, the role of RAS in CAD is not very clear, despite its impact on vascular remodeling and inflammation.
Although there is a lack of evidence demonstrating the high importance of RAS in CAD development, increased levels of circulating ACE2 were observed in patients with CAD [178,179]. On the other hand, Ang(1-7) levels remained unchanged, when compared to controls. Unfortunately, there are no interventional studies available for any impact of ACE2-Ang(1-7)-Mas axis on CAD. This link also seems to come apart after the investigation of Campbell et al. [180], where no direct connection was found between angiotensin II and angiotensin (1-7) coronary plasma levels after the treatment with various ACE inhibitors. Authors suggest that Ang(1-7) levels were increased as a result of increased metabolism of AngI, rather than by ACE2-mediated cleavage of angiotensin II.
CAD is often accompanied with the formation of atherosclerotic plaques. In experimental models of atherosclerosis (apolipoprotein E-deficient animals fed with a high-fat diet), coronary and aortic expressions of ACE2 were found to be reduced, while the levels of ACE2 mRNA were not affected [181,182]. Such conditions might be partially explained by excessive ACE2 release from vessels. In humans, ACE2 activity tends to decline with the progression of atherosclerosis [183], which is in accordance with the situation in experimental models. Knockout of ACE2 gene results in increased atherosclerotic plaque area, enhanced macrophage accumulation into the lesions, the proliferation of vascular smooth muscle cells (VSMCs), accompanied with an increase in AngII levels and increased expression of adhesive proteins, as well as increased matrix metallopeptidase 9 (MMP-9) activity [184,185]. Additionally, Mas-receptor deficiency augmented AngII-induced atherosclerosis and plaque rupture through mechanisms involving increased oxidative stress, inflammation, and apoptosis [184]. All of these complications were abolished by overexpression of ACE2 [54,186,187,188,189] and treatment with ACE2 activator (DIZE) [190,191] or subcutaneous Ang(1-7) [192]. Taken together, there is evidence regarding components of ACE2-Ang(1-7)-Mas axis involvement in CAD devolvement and progression; however, to further elucidate the therapeutic potential of modulation of this axis will require utilization of more selective tools to separate the upstream RAS effects provided by ACE inhibitors or AT1 blockers in general CAD therapy.

3.3.3. Involvement of ACE2-Ang(1-7)-Mas Axis in Chronic Obstructive Pulmonary Disease (COPD)

Patients with stable COPD have an impaired ability to excrete sodium [193,194] accompanied by renal fluid retention and edema [195] enhanced by hypercapnia-induced renal vasoconstriction and antidiuresis [195]. Acute hypoxia in COPD patients does not alter or decrease renin, ACE, angiotensin or aldosterone [196,197] plasma levels. Although, renin and angiotensin II levels can be significantly elevated in the presence of an underlying condition, such as chronic hypoxia and hypercapnia [194]. Renin, angiotensin II and aldosterone levels are much higher in COPD patients with edema than in patients without edema, while sodium and water excretion is decreased significantly in edematous COPD patients. The elevation of renin, angiotensin II and aldosterone correlate with the inability to excrete sodium and water. These data suggest, that in the conjunction with hypercapnia/hypoxia-mediated disturbances in renal function, stimulation of RAAS, especially with the increase in aldosterone, may contribute to edema formation in COPD patients [198]. However, impaired ability to excrete sodium might not be simply improved by the administration of an ACE inhibitor, since ACE inhibition lowers plasma levels of aldosterone without improving sodium excretion. This suggests that the inability of patients with hypoxemic COPD to excrete sodium is not caused by their increased plasma levels of aldosterone [193].
As we mentioned in the preceding paragraphs, the ACE2-Ang(1-7)-Mas axis plays generally a protective role against the effects of angiotensin II. On the other hand, downregulation of this axis can potentiate its deleterious counterpart, the ACE-AngII-AT1 axis. Indeed, an increased ACE/ACE2 ratio is suggested to play an important role in the development of various chronic cardiovascular and pulmonary diseases. ACE2 downregulation can lead to vasoconstriction, increased platelet aggregation, inflammation, proliferation, and fibrosis [31,54,124,199]. Moreover, ACE2 downregulation was observed in arterial hypertension [31,164,200,201,202], atherosclerosis [181], coronary heart disease [203], diabetes mellitus [202,204], cardiac fibrosis/remodeling [201,205] and heart failure [31,164]. Such imbalance can also be involved in pulmonary hypertension [199,206] and chronic obstructive pulmonary disease.
Nonetheless, data on COPD and their relation to ACE2 expression are scarce and controversial. Two animal studies by Xue et al. [207] and Zhang et al. [208] suggest that ACE2 is downregulated in cigarette smoke-induced COPD, whereas characteristic inflammation and fibrosis are possible to be blocked by overexpression of ACE2 [207] or by Ang(1-7) subcutaneous infusion [208]. However, human cohort studies [209,210,211] are not so straightforward. Their findings are in contrast with animal studies, where they observed higher levels of ACE2 in patients with COPD associated with cigarette smoking compared to controls or even ex-smokers. It remains unclear whether ACE2 overexpression is an initial protective response or is associated with COPD pathophysiology, although animal studies with induced ACE2-Ang(1-7)-Mas axis activation might suggest the former. On the other hand, we cannot exclude the effects of associated comorbidities and different medications in those patients. ACE inhibitors and AT1R blockers (sartans) are known to upregulate ACE2 [212,213,214] whilst the role of inhaled corticosteroids (ICS), which are used as first-line therapy of COPD in the combination with bronchodilators, is not very clear. A study by Sinha et al. suggests that ICS upregulates ACE2 in vitro [213], while another study by Finney et al. [215] states the opposite. Human studies of Peters et al. [216] and Aliee et al. [217] also observed the decrease in ACE2 after ICS administration, although the effect of other medications cannot be refuted. There are numerous animal studies [181,218,219] suggesting statins, a group of drugs used in the treatment of dyslipidemia and atherosclerosis, might upregulate ACE2 expression [181] as well.
Moreover, it is also important to distinguish between tissue and plasma forms of ACE2. As we mentioned earlier, cleavage of membrane ACE2 into the soluble form is in part dependent on the tumor necrosis factor-α convertase ADAM17 [17], which acts as a metalloproteinase and is upregulated in various diseases accompanied with inflammation and fibrosis [220], such as HF [17,221] or interstitial lung disease [222]. ADAM17 also plays its role in SARS-CoV-2 entry into human cells [12]. Therefore, cleavage of membrane form of ACE2 may be due to the pathological upregulation of this protease, resulting in a relative decrease in local membrane ACE2 levels. Alternatively, cleavage and release of membrane ACE2 as a soluble form may provide a compensatory mechanism resulting from disease. Additionally, Ang(1–7) has a very short half-life (<9 s) and the release of soluble ACE2 from the vascular endothelium may alter systemic Ang(1–7) concentrations and the relative peripheral balance of ACE2/ACE activity [166].
Besides the studies on clinically relevant COPD models, there are also studies describing lung fibrosis and hypoxia, which are closely related to the pathophysiology of COPD [220,223,224,225]. Unfortunately, in the meantime, we can only provide data from murine [208] and rat [226] animal studies, two studies on human fetal lung fibroblasts [227,228] and one paper studying human lung fibrosis [226]. These studies suggest that lung fibrosis is accompanied by decreased ACE2 levels and subsequent treatment by exogenous ACE2 or Ang(1-7) might be beneficial. To our knowledge, no human trials connecting lung fibrosis with ACE2 have been conducted yet. The situation regarding hypoxia is similar, without any human trials. In the animal models of hypoxia-induced pulmonary arterial hypertension [229,230,231,232], treatment with recombinant ACE2 or Ang(1-7) improved symptoms of pulmonary arterial hypertension (PAH) (lowered pulmonary arterial pressure, lung fibrosis, inflammation), however, there is a lack of evidence about the involvement of ACE2 in the models including both hypoxia and COPD. It is also hard to evaluate how hypoxia alone regulates ACE2/Ang(1-7) expression. Studies on hypoxic human pulmonary artery smooth muscle cells (PASMCs) [233] and CD34+ cells [234] show that HIF-1-α mediates an increase in ACE2 and Mas expression, however, in the studies from Wang et al. [235] and Zhang et al. [236] on PASMCs under chronic hypoxia, the expression of HIF-1-α was increased, while ACE2 was decreased, therefore, the link between HIF-1-α and ACE2 remains elusive. Surprisingly, ACE2 activity and Ang(1-7) levels were not affected by chronic hypoxia in rats at all [237].
These results are insufficient and inconclusive in the way how COPD or lung fibrosis affects the ACE/ACE2 ratio, mainly due to different experimental methods, environmental factors or the effects of various drugs. However, targeting of ACE2/Ang(1-7)/Mas axis for pharmacological modulation as a treatment of COPD seems promising for further human trials.

3.3.4. COVID-19 and the Relation with COPD

Even after an eventual decline, COVID-19 may leave some marks over the population for years to come. One of the clinical outcomes of the infection appears to be lung fibrosis [121,238,239,240,241,242,243,244,245]. Lung fibrosis is generally an irreversible condition, which can lead to PAH or COPD [224]. Indeed, fibrosis was more likely to develop in patients with a severe clinical condition, especially with high inflammatory indicators [246], however, a larger cohort needs to be engaged in clinical studies to reliably validate such results. Unfortunately, we lack accurate data on how frequently fibrosis occurs or how does it correlate with the disease severity. Since hundreds of millions of people around the globe have been infected, the possibility of increasing the rates of PAH or COPD incidence could test our medical capacities in the future.

3.3.5. Role of ACE2-Ang(1-7)-Mas Arm in Pulmonary Hypertension

Pulmonary hypertension (PH) is a life-threatening condition characterized by a progressive increase in vascular resistance in the pulmonary vasculature. PH is a serious complication of several cardiovascular and pulmonary diseases, which ultimately leads to right ventricular failure and death [247,248]. The exact cause is unknown, although generally accepted pathomechanism includes a disbalance between endothelin-1 and nitrous oxide (NO) in endothelium [249] with subsequent vasoconstriction, inflammation and remodeling of lung vasculature [250]. These substances are currently the main targets of specific PH therapy [248] along with prostaglandin analogs. Current therapy is, however, only symptomatic with low or no impact on overall mortality [248]. Thus, the urge to identify novel targets for the therapy is still high.
Recently, it was shown that animal models of PAH [251,252], hypoxic pulmonary hypertension (HPH) [253] and human patients with PAH [199,206], including idiopathic pulmonary arterial hypertension (IPAH) [253], might have an increased ratio of ACE/ACE2. With the increase in pulmonary arterial pressure, ACE2 expression [254] and Ang(1-7) plasma levels [255] tend to decrease. In contrast, our research group reported an increase in lung ACE2/ACE mRNA ratio of monocrotaline-treated rats [256]. While the therapy based on blocking of the RAAS have not been proven to be significantly efficient [248] (we were able to find only one clinical study based solely on effects of ACE inhibitors on pulmonary arterial pressure [257]), aiming at the ACE2-Ang(1-7)-Mas axis seems more promising.
Several rat and murine models suggest its beneficial effects in amelioration of monocrotaline-induced PAH symptoms. Animals overexpressing ACE2 [230,251,258,259] and Ang(1-7) [229] had lower right ventricular systolic pressure and pulmonary arterial pressure, less pronounced right ventricular hypertrophy and pulmonary artery remodeling and better hemodynamics in PAH. In a swine model of HPH, similar effects were observed after treatment with recombinant ACE2 [231]. Knock-out of the ACE2 gene resulted in aggravation of the symptoms [258]. Similar results were observed in models utilizing a pharmacological modulation of ACE2 activity [260,261,262,263,264] or by injecting the exogenous Ang(1-7) [265,266]. All those beneficial effects were abolished by a blockade of Mas-receptor [251,260].
Unfortunately, there is still a lack of evidence for the clinical PH in humans. There was conducted only one pilot study using single-dose intravenous (0.2 or 0.4 mg/kg) recombinant human ACE2 as a treatment of PAH [206], although with a good clinical perspective. Nevertheless, data on clinical outcomes in PAH patients are missing.
In the following Table 1, we summarized the above-mentioned studies based on the hypoxic condition and observed changes in ACE2/Ang(1-7)/Mas axis parameters.

4. Clinical Perspectives and Future Directions

To this day, there is no clinically approved ACE2/Ang(1-7)-aimed pharmacotherapy, even though several experimental studies based on gene modulation and activation of the alternative RAS components or recombinant therapy demonstrated the beneficial effect of stimulating the counter-regulatory RAS, which we listed in Table 2. Although, there are some exceptions, such as ARBs or ACE inhibitors, as they often possess indirect effects on this axis (olmesartan is thought to have some direct effects) [156].
Nevertheless, there are still several ongoing and closed clinical trials evaluating the clinical benefits of intravenously administered human recombinant ACE2 (rhACE2). RhACE2 (GSK2586881) was well tolerated in IIa phase of a clinical trial in patients with ARDS [125] and PAH [267], although in the PAH study there are no published results yet. Another rhACE2 (APN01) was studied in 2nd phase trial for the treatment of COVID-19 to block viral entry and to decrease viral replication. This trial was completed in January 2021 and the results are expected soon to be published [268]. However, one trial on COVID-19 was prematurely terminated last year [126]. The idea behind the study was to bind soluble ACE2 instead of tissue ACE2 to the SARS-CoV-2 virus particle and thus decrease its virulence. Such a mechanism was seen as a promising lung- and cardioprotective approach for the administration of rhACE2.
Despite rhACE2 being possibly beneficial in the treatment of COVID-19, it could also have some drawbacks. Firstly, high costs and secondly, it has the potential of immunogenicity or interaction with S-protein-based vaccines. Two parallel clinical trials are trying to overcome some of these problems using recombinant bacterial ACE2-like enzyme (B38-CAP) [269,270]. The studies were announced in May 2020 with the possible launch of phase 1 in late 2021 or in 2022. Another option is the manufacture of the rhACE2-Fc fusion protein. The advantage of this approach in comparison to previous ones is that the fusion of sACE2 to the Fc region of human immunoglobulin can increase its avidity to viral particles, recruit immune effector functions and increase serum stability, which is a desirable quality if intended for prophylaxis [271]. One such application, SI-F019 composed of the rhACE2-Fc fusion protein, was recently assigned to phase 1 clinical trial to evaluate its safety, tolerability, and pharmacokinetic properties [272].
Immunogenicity can also be overcome by the substitution of ACE2 with Ang(1-7). Several clinical studies are preparing or recruiting participants for the 2nd phase trial for the treatment of COVID-19 [273,274,275,276]. Besides COVID-19, three other clinical trials for peripheral arterial disease [277], essential hypertension [278] and obesity-associated hypertension [279] are about to start.
However, we need to mention that all trials described in this chapter were focused on hospital treatment. For general use, especially in chronically ill patients, it is necessary to prepare orally active pharmaceuticals. Intravenous administration is easily provided in hospitals by professionals, but not in the home environment by the patients themselves. Pharmacokinetic parameters (destruction by stomach acid and intestinal enzymes, lack of absorption, short half-life) of soluble ACE2 [280] (and Ang(1-7) as well [166]) prevent its use from oral administration. There was an attempt to improve pharmacokinetic parameters of Ang(1-7) by glycosylation of this peptide [281]. Half-life and blood–brain barrier penetration was improved, although subcutaneous formulation was still required. There were also attempts at an orally active Ang(1-7). The idea was to incorporate Ang(1-7) into specialized delivery systems, including cyclodextrin-based nanoparticles in animal models of diabetes mellitus type 2 [282], hypertensive model of thrombosis [43] and erectile dysfunction in hypercholesterolemia [283]. ACE2 protein was successfully fused with non-toxic cholera toxin subunit B expressed in plant chloroplasts, which allowed ACE2 oral administration in animal models of diabetic retinopathy [284], pulmonary hypertension [285] and uveitis [286].
However, the development of small molecular drugs is only the beginning. The best pharmacological targets in ACE2-Ang(1-7)-Mas axis could be activators of ACE2 or Mas agonists. The other possible approach is an inhibition of Ang(1-7) degradation since Ang(1-7) is primarily degraded by neprilysin (or neutral endopeptidase—NEP) and aminopeptidase A (APA), secondary to ACE. Ang(1-7) is cleaved by either NEP into inactive Ang(1-4) or by ACE into another inactive molecule Ang(1-5) [287]. ACE inhibitors and NEP inhibitors are already in clinical practice, but they lack Ang(1-7) specificity, especially NEP inhibitors, such as sacubitril, target many other peptides as well. APA is a member of the hydrolase enzyme family of aminopeptidases and its main substrates are AngII and Ang(1-7). AngII is cleaved to an active peptide AngIII with similar effects to AngII. Ang(1-7) is cleaved to inactive Ang(1-4) and Ang(2-7) [287,288]. APA was found to be overactivated in MI, thus it might be a good candidate for further studies as a new pharmacological target in several cardiovascular diseases [288]. Additionally, some other compounds were studied for their inhibitory effects on APA, such as 4-amino-4-phosphonobutyric acid (4-APBA) in a mouse model of MI [288], EC33 and its dimer RB150 (firibastat) in the treatment of high blood pressure in rats [289]. There is also an effort for the treatment of lung [95] and renal [83] IRI with ACE2 activator (DIZE) and the treatment of myocardial IRI-induced necrosis by Mas agonist AVE 0991 [290]. AVE 0991 further exhibited anti-inflammatory potential in an ovalbumin-induced acute asthmatic murine model in which Mas agonist significantly reduced macrophage infiltration [291]. However, all these experiments were conducted in animal models only. To our best knowledge, no human trials using small molecular pharmacotherapy are planned yet.

5. Conclusions

The main findings regarding the alternative RAS in hypoxic conditions are shown in Figure 3. In short, in both acute and chronic hypoxic conditions in CVS and the lungs, there is time-dependent regulation of the ACE2/Ang(1-7) axis, increased levels of ACE2/Ang(1-7) in the early stages in contrast to markedly reduced levels with disease progression at the end-stage. An important element of such behavior throughout the studies is the increased circulation of soluble ACE2 connected to the activity of proteolytic enzymes, particularly sheddases, such as ADAM17. However, generally, there are no major differences between most acute and chronic hypoxic conditions since the pathologies are predominantly characterized by enhanced activity of classical ACE/AngII axis and ACE2/Ang(1-7) suppression. The biggest scientific challenge lies in the fact that most of the translational and clinical studies in humans are affected by the effects of drugs that are included in the patient´s treatment regimen on the ACE2/Ang(1-7) axis or by the interaction/alteration of normal RAS activity and its components. Nonetheless, treatment potential resides in enhancing the activity of the alternative RAS arm (activators/agonists, recombinant components), which has accounted for numerous protective effects in different experimental models, contrastingly to the shutdown of this axis which promotes adverse signaling. Fluctuating plasma levels might also be an accountable prognostic/disease marker for future detection and disease outcome/treatment, particularly, increased plasma ACE2 can serve as a novel and reliable marker of acute cardiac damage. Moreover, during the time of writing this article, several clinical studies are being conducted, namely for the treatment of ARDS, COVID-19 and PAH, pointing to the fact, that the alternative RAS branch is one of the most promising neuroendocrine cascades for the future diagnosis and treatment of many hypoxia-related CVS and pulmonary diseases, including diseases such as myocardial infarction, heart failure, PAH or even COVID-19.

Author Contributions

Conceptualization, T.R., L.P. and J.K.; writing—original draft preparation, T.R., P.G. and L.B.; writing—review and editing, T.R., L.P., E.G. and J.K.; visualization, T.R., L.B. and P.G.; supervision, T.R., L.P., E.G. and J.K.; project administration, T.R. and J.K.; funding acquisition, T.R., L.P., E.G. and J.K. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by the Scientific Grant Agency Of The Ministry Of Education Of Slovak Republic, VEGA 1/0775/21 and the Slovak Research And Development Agency, APVV-15-0685.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Simko, F.; Hrenak, J.; Adamcova, M.; Paulis, L. Renin-Angiotensin-Aldosterone System: Friend or Foe-The Matter of Balance. Insight on History, Therapeutic Implications and COVID-19 Interactions. Int. J. Mol. Sci. 2021, 22, 3217. [Google Scholar] [CrossRef]
  2. Forrester, S.J.; Booz, G.W.; Sigmund, C.D.; Coffman, T.M.; Kawai, T.; Rizzo, V.; Scalia, R.; Eguchi, S. Angiotensin II Signal Transduction: An Update on Mechanisms of Physiology and Pathophysiology. Physiol. Rev. 2018, 98, 1627–1738. [Google Scholar] [CrossRef] [PubMed]
  3. Sampaio, W.O.; Souza dos Santos, R.A.; Faria-Silva, R.; da Mata Machado, L.T.; Schiffrin, E.L.; Touyz, R.M. Angiotensin-(1-7) through receptor Mas mediates endothelial nitric oxide synthase activation via Akt-dependent pathways. Hypertension 2007, 49, 185–192. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Zhang, F.; Xu, Y.; Pan, Y.; Sun, S.; Chen, A.; Li, P.; Bao, C.; Wang, J.; Tang, H.; Han, Y. Effects of Angiotensin-(1-7) and Angiotensin II on Acetylcholine-Induced Vascular Relaxation in Spontaneously Hypertensive Rats. Oxid. Med. Cell Longev. 2019, 2019, 6512485. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Zhang, F.; Ren, X.; Zhao, M.; Zhou, B.; Han, Y. Angiotensin-(1-7) abrogates angiotensin II-induced proliferation, migration and inflammation in VSMCs through inactivation of ROS-mediated PI3K/Akt and MAPK/ERK signaling pathways. Sci. Rep. 2016, 6, 34621. [Google Scholar] [CrossRef]
  6. Gomes, E.R.; Lara, A.A.; Almeida, P.W.; Guimarães, D.; Resende, R.R.; Campagnole-Santos, M.J.; Bader, M.; Santos, R.A.; Guatimosim, S. Angiotensin-(1-7) prevents cardiomyocyte pathological remodeling through a nitric oxide/guanosine 3′,5′-cyclic monophosphate-dependent pathway. Hypertension 2010, 55, 153–160. [Google Scholar] [CrossRef] [Green Version]
  7. Li, M.Y.; Li, L.; Zhang, Y.; Wang, X.S. Expression of the SARS-CoV-2 cell receptor gene ACE2 in a wide variety of human tissues. Infect. Dis. Poverty 2020, 9, 45. [Google Scholar] [CrossRef]
  8. Donoghue, M.; Hsieh, F.; Baronas, E.; Godbout, K.; Gosselin, M.; Stagliano, N.; Donovan, M.; Woolf, B.; Robison, K.; Jeyaseelan, R.; et al. A novel angiotensin-converting enzyme-related carboxypeptidase (ACE2) converts angiotensin I to angiotensin 1-9. Circ. Res. 2000, 87, e1–e9. [Google Scholar] [CrossRef]
  9. Tipnis, S.R.; Hooper, N.M.; Hyde, R.; Karran, E.; Christie, G.; Turner, A.J. A human homolog of angiotensin-converting enzyme. Cloning and functional expression as a captopril-insensitive carboxypeptidase. J. Biol. Chem. 2000, 275, 33238–33243. [Google Scholar] [CrossRef] [Green Version]
  10. Towler, P.; Staker, B.; Prasad, S.G.; Menon, S.; Tang, J.; Parsons, T.; Ryan, D.; Fisher, M.; Williams, D.; Dales, N.A.; et al. ACE2 X-ray structures reveal a large hinge-bending motion important for inhibitor binding and catalysis. J. Biol. Chem. 2004, 279, 17996–18007. [Google Scholar] [CrossRef] [Green Version]
  11. Li, W.; Moore, M.J.; Vasilieva, N.; Sui, J.; Wong, S.K.; Berne, M.A.; Somasundaran, M.; Sullivan, J.L.; Luzuriaga, K.; Greenough, T.C.; et al. Angiotensin-converting enzyme 2 is a functional receptor for the SARS coronavirus. Nature 2003, 426, 450–454. [Google Scholar] [CrossRef] [Green Version]
  12. Ou, X.; Liu, Y.; Lei, X.; Li, P.; Mi, D.; Ren, L.; Guo, L.; Guo, R.; Chen, T.; Hu, J.; et al. Characterization of spike glycoprotein of SARS-CoV-2 on virus entry and its immune cross-reactivity with SARS-CoV. Nat. Commun. 2020, 11, 1620. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Rice, G.I.; Thomas, D.A.; Grant, P.J.; Turner, A.J.; Hooper, N.M. Evaluation of angiotensin-converting enzyme (ACE), its homologue ACE2 and neprilysin in angiotensin peptide metabolism. Biochem. J. 2004, 383, 45–51. [Google Scholar] [CrossRef]
  14. Santos, R.A.; e Silva, A.C.S.; Maric, C.; Silva, D.M.; Machado, R.P.; de Buhr, I.; Heringer-Walther, S.; Pinheiro, S.V.; Lopes, M.T.; Bader, M.; et al. Angiotensin-(1-7) is an endogenous ligand for the G protein-coupled receptor Mas. Proc. Natl. Acad. Sci. USA 2003, 100, 8258–8263. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  15. Mendoza-Torres, E.; Oyarzún, A.; Mondaca-Ruff, D.; Azocar, A.; Castro, P.F.; Jalil, J.E.; Chiong, M.; Lavandero, S.; Ocaranza, M.P. ACE2 and vasoactive peptides: Novel players in cardiovascular/renal remodeling and hypertension. Ther. Adv. Cardiovasc. Dis. 2015, 9, 217–237. [Google Scholar] [CrossRef] [PubMed]
  16. Vickers, C.; Hales, P.; Kaushik, V.; Dick, L.; Gavin, J.; Tang, J.; Godbout, K.; Parsons, T.; Baronas, E.; Hsieh, F.; et al. Hydrolysis of biological peptides by human angiotensin-converting enzyme-related carboxypeptidase. J. Biol. Chem. 2002, 277, 14838–14843. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  17. Lambert, D.W.; Yarski, M.; Warner, F.J.; Thornhill, P.; Parkin, E.T.; Smith, A.I.; Hooper, N.M.; Turner, A.J. Tumor necrosis factor-alpha convertase (ADAM17) mediates regulated ectodomain shedding of the severe-acute respiratory syndrome-coronavirus (SARS-CoV) receptor, angiotensin-converting enzyme-2 (ACE2). J. Biol. Chem. 2005, 280, 30113–30119. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  18. Young, D.; Waitches, G.; Birchmeier, C.; Fasano, O.; Wigler, M. Isolation and characterization of a new cellular oncogene encoding a protein with multiple potential transmembrane domains. Cell 1986, 45, 711–719. [Google Scholar] [CrossRef]
  19. Davenport, A.P.; Alexander, S.P.; Sharman, J.L.; Pawson, A.J.; Benson, H.E.; Monaghan, A.E.; Liew, W.C.; Mpamhanga, C.P.; Bonner, T.I.; Neubig, R.R.; et al. International Union of Basic and Clinical Pharmacology. LXXXVIII. G protein-coupled receptor list: Recommendations for new pairings with cognate ligands. Pharmacol. Rev. 2013, 65, 967–986. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  20. Gaidarov, I.; Adams, J.; Frazer, J.; Anthony, T.; Chen, X.; Gatlin, J.; Semple, G.; Unett, D.J. Angiotensin (1-7) does not interact directly with MAS1, but can potently antagonize signaling from the AT1 receptor. Cell Signal. 2018, 50, 9–24. [Google Scholar] [CrossRef] [PubMed]
  21. Karnik, S.S.; Singh, K.D.; Tirupula, K.; Unal, H. Significance of angiotensin 1-7 coupling with MAS1 receptor and other GPCRs to the renin-angiotensin system: IUPHAR Review 22. Br. J. Pharmacol. 2017, 174, 737–753. [Google Scholar] [CrossRef]
  22. Leonhardt, J.; Villela, D.C.; Teichmann, A.; Münter, L.M.; Mayer, M.C.; Mardahl, M.; Kirsch, S.; Namsolleck, P.; Lucht, K.; Benz, V.; et al. Evidence for Heterodimerization and Functional Interaction of the Angiotensin Type 2 Receptor and the Receptor MAS. Hypertension 2017, 69, 1128–1135. [Google Scholar] [CrossRef] [PubMed]
  23. Kostenis, E.; Milligan, G.; Christopoulos, A.; Sanchez-Ferrer, C.F.; Heringer-Walther, S.; Sexton, P.M.; Gembardt, F.; Kellett, E.; Martini, L.; Vanderheyden, P.; et al. G-protein-coupled receptor Mas is a physiological antagonist of the angiotensin II type 1 receptor. Circulation 2005, 111, 1806–1813. [Google Scholar] [CrossRef]
  24. Zhao, W.; Zhao, T.; Chen, Y.; Sun, Y. Angiotensin 1-7 promotes cardiac angiogenesis following infarction. Curr. Vasc. Pharmacol. 2015, 13, 37–42. [Google Scholar] [CrossRef]
  25. Fraga-Silva, R.A.; Pinheiro, S.V.; Gonçalves, A.C.; Alenina, N.; Bader, M.; Santos, R.A. The antithrombotic effect of angiotensin-(1-7) involves mas-mediated NO release from platelets. Mol. Med. 2008, 14, 28–35. [Google Scholar] [CrossRef]
  26. Tetzner, A.; Gebolys, K.; Meinert, C.; Klein, S.; Uhlich, A.; Trebicka, J.; Villacañas, Ó.; Walther, T. G-Protein-Coupled Receptor MrgD Is a Receptor for Angiotensin-(1-7) Involving Adenylyl Cyclase, cAMP, and Phosphokinase A. Hypertension 2016, 68, 185–194. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  27. Tallant, E.A.; Ferrario, C.M.; Gallagher, P.E. Angiotensin-(1-7) inhibits growth of cardiac myocytes through activation of the mas receptor. Am. J. Physiol. Heart Circ. Physiol. 2005, 289, H1560–H1566. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  28. Ferreira, A.J.; Castro, C.H.; Guatimosim, S.; Almeida, P.W.; Gomes, E.R.; Dias-Peixoto, M.F.; Alves, M.N.; Fagundes-Moura, C.R.; Rentzsch, B.; Gava, E.; et al. Attenuation of isoproterenol-induced cardiac fibrosis in transgenic rats harboring an angiotensin-(1-7)-producing fusion protein in the heart. Ther. Adv. Cardiovasc. Dis. 2010, 4, 83–96. [Google Scholar] [CrossRef] [PubMed]
  29. Dias-Peixoto, M.F.; Santos, R.A.; Gomes, E.R.; Alves, M.N.; Almeida, P.W.; Greco, L.; Rosa, M.; Fauler, B.; Bader, M.; Alenina, N.; et al. Molecular mechanisms involved in the angiotensin-(1-7)/Mas signaling pathway in cardiomyocytes. Hypertension 2008, 52, 542–548. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  30. Costa, M.A.; Lopez Verrilli, M.A.; Gomez, K.A.; Nakagawa, P.; Peña, C.; Arranz, C.; Gironacci, M.M. Angiotensin-(1-7) upregulates cardiac nitric oxide synthase in spontaneously hypertensive rats. Am. J. Physiol. Heart Circ. Physiol. 2010, 299, H1205–H1211. [Google Scholar] [CrossRef] [PubMed]
  31. Zhong, J.; Basu, R.; Guo, D.; Chow, F.L.; Byrns, S.; Schuster, M.; Loibner, H.; Wang, X.H.; Penninger, J.M.; Kassiri, Z.; et al. Angiotensin-converting enzyme 2 suppresses pathological hypertrophy, myocardial fibrosis, and cardiac dysfunction. Circulation 2010, 122, 717–728. [Google Scholar] [CrossRef] [PubMed]
  32. Qi, Y.; Shenoy, V.; Wong, F.; Li, H.; Afzal, A.; Mocco, J.; Sumners, C.; Raizada, M.K.; Katovich, M.J. Lentivirus-mediated overexpression of angiotensin-(1-7) attenuated ischaemia-induced cardiac pathophysiology. Exp. Physiol. 2011, 96, 863–874. [Google Scholar] [CrossRef] [PubMed]
  33. Crackower, M.A.; Sarao, R.; Oudit, G.Y.; Yagil, C.; Kozieradzki, I.; Scanga, S.E.; Oliveira-dos-Santos, A.J.; da Costa, J.; Zhang, L.; Pei, Y.; et al. Angiotensin-converting enzyme 2 is an essential regulator of heart function. Nature 2002, 417, 822–828. [Google Scholar] [CrossRef]
  34. Gurley, S.B.; Allred, A.; Le, T.H.; Griffiths, R.; Mao, L.; Philip, N.; Haystead, T.A.; Donoghue, M.; Breitbart, R.E.; Acton, S.L.; et al. Altered blood pressure responses and normal cardiac phenotype in ACE2-null mice. J. Clin. Investig. 2006, 116, 2218–2225. [Google Scholar] [CrossRef] [Green Version]
  35. Yamamoto, K.; Ohishi, M.; Katsuya, T.; Ito, N.; Ikushima, M.; Kaibe, M.; Tatara, Y.; Shiota, A.; Sugano, S.; Takeda, S.; et al. Deletion of angiotensin-converting enzyme 2 accelerates pressure overload-induced cardiac dysfunction by increasing local angiotensin II. Hypertension 2006, 47, 718–726. [Google Scholar] [CrossRef] [Green Version]
  36. Wong, D.W.; Oudit, G.Y.; Reich, H.; Kassiri, Z.; Zhou, J.; Liu, Q.C.; Backx, P.H.; Penninger, J.M.; Herzenberg, A.M.; Scholey, J.W. Loss of angiotensin-converting enzyme-2 (Ace2) accelerates diabetic kidney injury. Am. J. Pathol. 2007, 171, 438–451. [Google Scholar] [CrossRef] [Green Version]
  37. Patel, V.B.; Bodiga, S.; Fan, D.; Das, S.K.; Wang, Z.; Wang, W.; Basu, R.; Zhong, J.; Kassiri, Z.; Oudit, G.Y. Cardioprotective effects mediated by angiotensin II type 1 receptor blockade and enhancing angiotensin 1-7 in experimental heart failure in angiotensin-converting enzyme 2-null mice. Hypertension 2012, 59, 1195–1203. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  38. Souza, Á.P.; Sobrinho, D.B.; Almeida, J.F.; Alves, G.M.; Macedo, L.M.; Porto, J.E.; Vêncio, E.F.; Colugnati, D.B.; Santos, R.A.; Ferreira, A.J.; et al. Angiotensin II type 1 receptor blockade restores angiotensin-(1-7)-induced coronary vasodilation in hypertrophic rat hearts. Clin. Sci. 2013, 125, 449–459. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  39. Tallant, E.A.; Clark, M.A. Molecular mechanisms of inhibition of vascular growth by angiotensin-(1-7). Hypertension 2003, 42, 574–579. [Google Scholar] [CrossRef] [Green Version]
  40. Sousa-Lopes, A.; de Freitas, R.A.; Carneiro, F.S.; Nunes, K.P.; Allahdadi, K.J.; Webb, R.C.; Tostes, R.C.; Giachini, F.R.; Lima, V.V. Angiotensin (1-7) Inhibits Ang II-mediated ERK1/2 Activation by Stimulating MKP-1 Activation in Vascular Smooth Muscle Cells. Int. J. Mol. Cell Med. 2020, 9, 50–61. [Google Scholar] [PubMed]
  41. Kalinowski, L.; Matys, T.; Chabielska, E.; Buczko, W.; Malinski, T. Angiotensin II AT1 receptor antagonists inhibit platelet adhesion and aggregation by nitric oxide release. Hypertension 2002, 40, 521–527. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  42. Shariat-Madar, Z.; Mahdi, F.; Warnock, M.; Homeister, J.W.; Srikanth, S.; Krijanovski, Y.; Murphey, L.J.; Jaffa, A.A.; Schmaier, A.H. Bradykinin B2 receptor knockout mice are protected from thrombosis by increased nitric oxide and prostacyclin. Blood 2006, 108, 192–199. [Google Scholar] [CrossRef]
  43. Fraga-Silva, R.A.; Costa-Fraga, F.P.; De Sousa, F.B.; Alenina, N.; Bader, M.; Sinisterra, R.D.; Santos, R.A. An orally active formulation of angiotensin-(1-7) produces an antithrombotic effect. Clinics 2011, 66, 837–841. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Silva, D.M.; Gomes-Filho, A.; Olivon, V.C.; Santos, T.M.; Becker, L.K.; Santos, R.A.; Lemos, V.S. Swimming training improves the vasodilator effect of angiotensin-(1-7) in the aorta of spontaneously hypertensive rat. J. Appl. Physiol. 2011, 111, 1272–1277. [Google Scholar] [CrossRef] [PubMed]
  45. Fang, C.; Stavrou, E.; Schmaier, A.A.; Grobe, N.; Morris, M.; Chen, A.; Nieman, M.T.; Adams, G.N.; LaRusch, G.; Zhou, Y.; et al. Angiotensin 1-7 and Mas decrease thrombosis in Bdkrb2−/− mice by increasing NO and prostacyclin to reduce platelet spreading and glycoprotein VI activation. Blood 2013, 121, 3023–3032. [Google Scholar] [CrossRef]
  46. Marshall, R.P. The pulmonary renin-angiotensin system. Curr. Pharm. Des. 2003, 9, 715–722. [Google Scholar] [CrossRef]
  47. Wiener, R.S.; Cao, Y.X.; Hinds, A.; Ramirez, M.I.; Williams, M.C. Angiotensin converting enzyme 2 is primarily epithelial and is developmentally regulated in the mouse lung. J. Cell Biochem. 2007, 101, 1278–1291. [Google Scholar] [CrossRef]
  48. Xie, X.; Chen, J.; Wang, X.; Zhang, F.; Liu, Y. Age- and gender-related difference of ACE2 expression in rat lung. Life Sci. 2006, 78, 2166–2171. [Google Scholar] [CrossRef]
  49. Hamming, I.; Timens, W.; Bulthuis, M.L.; Lely, A.T.; 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. 2004, 203, 631–637. [Google Scholar] [CrossRef]
  50. Zhang, H.; Li, Y.; Zeng, Y.; Wu, R.; Ou, J. Endothelin-1 downregulates angiotensin-converting enzyme-2 expression in human bronchial epithelial cells. Pharmacology 2013, 91, 297–304. [Google Scholar] [CrossRef]
  51. Lukassen, S.; Chua, R.L.; Trefzer, T.; Kahn, N.C.; Schneider, M.A.; Muley, T.; Winter, H.; Meister, M.; Veith, C.; Boots, A.W.; et al. SARS-CoV-2 receptor ACE2 and TMPRSS2 are primarily expressed in bronchial transient secretory cells. EMBO J. 2020, 39, e105114. [Google Scholar] [CrossRef]
  52. Jia, H.P.; Look, D.C.; Shi, L.; Hickey, M.; Pewe, L.; Netland, J.; Farzan, M.; Wohlford-Lenane, C.; Perlman, S.; McCray, P.B., Jr. ACE2 receptor expression and severe acute respiratory syndrome coronavirus infection depend on differentiation of human airway epithelia. J. Virol. 2005, 79, 14614–14621. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  53. Magalhães, G.S.; Rodrigues-Machado, M.G.; Motta-Santos, D.; Silva, A.R.; Caliari, M.V.; Prata, L.O.; Abreu, S.C.; Rocco, P.R.; Barcelos, L.S.; Santos, R.A.; et al. Angiotensin-(1-7) attenuates airway remodelling and hyperresponsiveness in a model of chronic allergic lung inflammation. Br. J. Pharmacol. 2015, 172, 2330–2342. [Google Scholar] [CrossRef] [Green Version]
  54. Zhang, Y.H.; Zhang, Y.H.; Dong, X.F.; Hao, Q.Q.; Zhou, X.M.; Yu, Q.T.; Li, S.Y.; Chen, X.; Tengbeh, A.F.; Dong, B.; et al. ACE2 and Ang-(1-7) protect endothelial cell function and prevent early atherosclerosis by inhibiting inflammatory response. Inflamm. Res. 2015, 64, 253–260. [Google Scholar] [CrossRef]
  55. Huang, W.; Cao, Y.; Liu, Y.; Ping, F.; Shang, J.; Zhang, Z.; Li, Y. Activating Mas receptor protects human pulmonary microvascular endothelial cells against LPS-induced apoptosis via the NF-kB p65/P53 feedback pathways. J. Cell Physiol. 2019, 234, 12865–12875. [Google Scholar] [CrossRef] [PubMed]
  56. Marshall, R.P.; Gohlke, P.; Chambers, R.C.; Howell, D.C.; Bottoms, S.E.; Unger, T.; McAnulty, R.J.; Laurent, G.J. Angiotensin II and the fibroproliferative response to acute lung injury. Am. J. Physiol. Lung Cell Mol. Physiol. 2004, 286, L156–L164. [Google Scholar] [CrossRef]
  57. Senchenkova, E.Y.; Russell, J.; Almeida-Paula, L.D.; Harding, J.W.; Granger, D.N. Angiotensin II-mediated microvascular thrombosis. Hypertension 2010, 56, 1089–1095. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  58. Mollnau, H.; Wendt, M.; Szöcs, K.; Lassègue, B.; Schulz, E.; Oelze, M.; Li, H.; Bodenschatz, M.; August, M.; Kleschyov, A.L.; et al. Effects of angiotensin II infusion on the expression and function of NAD(P)H oxidase and components of nitric oxide/cGMP signaling. Circ. Res. 2002, 90, E58–E65. [Google Scholar] [CrossRef] [Green Version]
  59. Morrell, N.W.; Upton, P.D.; Kotecha, S.; Huntley, A.; Yacoub, M.H.; Polak, J.M.; Wharton, J. Angiotensin II activates MAPK and stimulates growth of human pulmonary artery smooth muscle via AT1 receptors. Am. J. Physiol. 1999, 277, L440–L448. [Google Scholar] [CrossRef]
  60. Mei, D.; Tan, W.S.D.; Liao, W.; Heng, C.K.M.; Wong, W.S.F. Activation of angiotensin II type-2 receptor protects against cigarette smoke-induced COPD. Pharmacol. Res. 2020, 161, 105223. [Google Scholar] [CrossRef]
  61. Uhal, B.D.; Li, X.; Xue, A.; Gao, X.; Abdul-Hafez, A. Regulation of alveolar epithelial cell survival by the ACE-2/angiotensin 1-7/Mas axis. Am. J. Physiol. Lung Cell Mol. Physiol. 2011, 301, L269–L274. [Google Scholar] [CrossRef] [Green Version]
  62. Li, Y.; Cao, Y.; Zeng, Z.; Liang, M.; Xue, Y.; Xi, C.; Zhou, M.; Jiang, W. Angiotensin-converting enzyme 2/angiotensin-(1-7)/Mas axis prevents lipopolysaccharide-induced apoptosis of pulmonary microvascular endothelial cells by inhibiting JNK/NF-κB pathways. Sci. Rep. 2015, 5, 8209. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. El-Hashim, A.Z.; Renno, W.M.; Raghupathy, R.; Abduo, H.T.; Akhtar, S.; Benter, I.F. Angiotensin-(1-7) inhibits allergic inflammation, via the MAS1 receptor, through suppression of ERK1/2- and NF-κB-dependent pathways. Br. J. Pharmacol. 2012, 166, 1964–1976. [Google Scholar] [CrossRef] [Green Version]
  64. Gopallawa, I.; Uhal, B.D. Angiotensin-(1-7)/mas inhibits apoptosis in alveolar epithelial cells through upregulation of MAP kinase phosphatase-2. Am. J. Physiol. Lung Cell Mol. Physiol. 2016, 310, L240–L248. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Uhal, B.D.; Nguyen, H.; Dang, M.; Gopallawa, I.; Jiang, J.; Dang, V.; Ono, S.; Morimoto, K. Abrogation of ER stress-induced apoptosis of alveolar epithelial cells by angiotensin 1-7. Am. J. Physiol. Lung Cell Mol. Physiol. 2013, 305, L33–L41. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  66. Ma, X.; Xu, D.; Ai, Y.; Zhao, S.; Zhang, L.; Ming, G.; Liu, Z. Angiotensin-(1-7)/Mas Signaling Inhibits Lipopolysaccharide-Induced ADAM17 Shedding Activity and Apoptosis in Alveolar Epithelial Cells. Pharmacology 2016, 97, 63–71. [Google Scholar] [CrossRef] [PubMed]
  67. Meng, Y.; Li, T.; Zhou, G.S.; Chen, Y.; Yu, C.H.; Pang, M.X.; Li, W.; Li, Y.; Zhang, W.Y.; Li, X. The angiotensin-converting enzyme 2/angiotensin (1-7)/Mas axis protects against lung fibroblast migration and lung fibrosis by inhibiting the NOX4-derived ROS-mediated RhoA/Rho kinase pathway. Antioxid. Redox Signal. 2015, 22, 241–258. [Google Scholar] [CrossRef]
  68. Meng, Y.; Yu, C.H.; Li, W.; Li, T.; Luo, W.; Huang, S.; Wu, P.S.; Cai, S.X.; 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. 2014, 50, 723–736. [Google Scholar] [CrossRef] [PubMed]
  69. Zambelli, V.; Bellani, G.; Borsa, R.; Pozzi, F.; Grassi, A.; Scanziani, M.; Castiglioni, V.; Masson, S.; Decio, A.; Laffey, J.G.; et al. Angiotensin-(1-7) improves oxygenation, while reducing cellular infiltrate and fibrosis in experimental Acute Respiratory Distress Syndrome. Intensive Care Med. Exp. 2015, 3, 44. [Google Scholar] [CrossRef] [Green Version]
  70. Samuel, J.; Franklin, C. Hypoxemia and Hypoxia. In Common Surgical Diseases: An Algorithmic Approach to Problem Solving; Myers, J.A., Millikan, K.W., Saclarides, T.J., Eds.; Springer: New York, NY, USA, 2008; pp. 391–394. [Google Scholar]
  71. Hobler, K.E.; Carey, L.C. Effect of acute progressive hypoxemia on cardiac output and plasma excess lactate. Ann. Surg. 1973, 177, 199–202. [Google Scholar] [CrossRef]
  72. Fulda, S.; Gorman, A.M.; Hori, O.; Samali, A. Cellular stress responses: Cell survival and cell death. Int. J. Cell Biol. 2010, 2010, 214074. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  73. Michiels, C. Physiological and pathological responses to hypoxia. Am. J. Pathol. 2004, 164, 1875–1882. [Google Scholar] [CrossRef] [Green Version]
  74. Wu, M.Y.; Yiang, G.T.; Liao, W.T.; Tsai, A.P.; Cheng, Y.L.; Cheng, P.W.; Li, C.Y.; Li, C.J. Current Mechanistic Concepts in Ischemia and Reperfusion Injury. Cell Physiol. Biochem. 2018, 46, 1650–1667. [Google Scholar] [CrossRef] [PubMed]
  75. Acute Respiratory Distress Syndrome. Available online: https://www.nhlbi.nih.gov/health-topics/acute-respiratory-distress-syndrome (accessed on 23 November 2021).
  76. Bayer, C.; Shi, K.; Astner, S.T.; Maftei, C.A.; Vaupel, P. Acute versus chronic hypoxia: Why a simplified classification is simply not enough. Int. J. Radiat. Oncol. Biol. Phys. 2011, 80, 965–968. [Google Scholar] [CrossRef]
  77. Peinado, V.I.; Santos, S.; Ramírez, J.; Roca, J.; Rodriguez-Roisin, R.; Barberà, J.A. Response to hypoxia of pulmonary arteries in chronic obstructive pulmonary disease: An in vitro study. Eur. Respir. J. 2002, 20, 332–338. [Google Scholar] [CrossRef] [Green Version]
  78. Nathan, S.D.; Barbera, J.A.; Gaine, S.P.; Harari, S.; Martinez, F.J.; Olschewski, H.; Olsson, K.M.; Peacock, A.J.; Pepke-Zaba, J.; Provencher, S.; et al. Pulmonary hypertension in chronic lung disease and hypoxia. Eur. Respir. J. 2019, 53, 1801914. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  79. Benowitz, N.L.; Meister, W. Pharmacokinetics in patients with cardiac failure. Clin. Pharmacokinet. 1976, 1, 389–405. [Google Scholar] [CrossRef] [PubMed]
  80. Fitridge, R.; Thompson, M. (Eds.) Mechanisms of Vascular Disease: A Reference Book for Vascular Specialists; University of Adelaide Press: Adelaide, SA, Australia, 2011. [Google Scholar]
  81. Go, A.S.; Hsu, C.Y.; Yang, J.; Tan, T.C.; Zheng, S.; Ordonez, J.D.; Liu, K.D. Acute Kidney Injury and Risk of Heart Failure and Atherosclerotic Events. Clin. J. Am. Soc. Nephrol. 2018, 13, 833–841. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  82. Dargie, H.J.; Byrne, J. Pathophysiological aspects of the renin-angiotensin-aldosterone system in acute myocardial infarction. J. Cardiovasc. Risk 1995, 2, 389–395. [Google Scholar] [CrossRef]
  83. Sharma, N.; Gaikwad, A.B. Ameliorative effect of AT2R and ACE2 activation on ischemic renal injury associated cardiac and hepatic dysfunction. Environ. Toxicol. Pharmacol. 2020, 80, 103501. [Google Scholar] [CrossRef]
  84. Kim, M.A.; Yang, D.; Kida, K.; Molotkova, N.; Yeo, S.J.; Varki, N.; Iwata, M.; Dalton, N.D.; Peterson, K.L.; Siems, W.E.; et al. Effects of ACE2 inhibition in the post-myocardial infarction heart. J. Card. Fail. 2010, 16, 777–785. [Google Scholar] [CrossRef] [Green Version]
  85. Kassiri, Z.; Zhong, J.; Guo, D.; Basu, R.; Wang, X.; Liu, P.P.; Scholey, J.W.; Penninger, J.M.; Oudit, G.Y. Loss of angiotensin-converting enzyme 2 accelerates maladaptive left ventricular remodeling in response to myocardial infarction. Circ. Heart Fail. 2009, 2, 446–455. [Google Scholar] [CrossRef] [Green Version]
  86. Qi, Y.; Zhang, J.; Cole-Jeffrey, C.T.; Shenoy, V.; Espejo, A.; Hanna, M.; Song, C.; Pepine, C.J.; Katovich, M.J.; Raizada, M.K. Diminazene aceturate enhances angiotensin-converting enzyme 2 activity and attenuates ischemia-induced cardiac pathophysiology. Hypertension 2013, 62, 746–752. [Google Scholar] [CrossRef] [PubMed]
  87. Burrell, L.M.; Risvanis, J.; Kubota, E.; Dean, R.G.; MacDonald, P.S.; Lu, S.; Tikellis, C.; Grant, S.L.; Lew, R.A.; Smith, A.I.; et al. Myocardial infarction increases ACE2 expression in rat and humans. Eur. Heart J. 2005, 26, 369–375. [Google Scholar] [CrossRef] [Green Version]
  88. Zhao, Y.X.; Yin, H.Q.; Yu, Q.T.; Qiao, Y.; Dai, H.Y.; Zhang, M.X.; Zhang, L.; Liu, Y.F.; Wang, L.C.; Liu, D.S.; et al. ACE2 overexpression ameliorates left ventricular remodeling and dysfunction in a rat model of myocardial infarction. Hum. Gene Ther. 2010, 21, 1545–1554. [Google Scholar] [CrossRef] [PubMed]
  89. De Mello, W.C. Angiotensin (1-7) re-establishes impulse conduction in cardiac muscle during ischaemia-reperfusion. The role of the sodium pump. J. Renin-Angiotensin-Aldosterone Syst. 2004, 5, 203–208. [Google Scholar] [CrossRef] [Green Version]
  90. Badae, N.M.; El Naggar, A.S.; El Sayed, S.M. Is the cardioprotective effect of the ACE2 activator diminazene aceturate more potent than the ACE inhibitor enalapril on acute myocardial infarction in rats? Can. J. Physiol. Pharmacol. 2019, 97, 638–646. [Google Scholar] [CrossRef]
  91. Chen, J.; Cui, L.; Yuan, J.; Zhang, S.; Ma, R.; Sang, H.; Liu, Q.; Shan, L. Protective effect of diminazene attenuates myocardial infarction in rats via increased inflammation and ACE2 activity. Mol. Med. Rep. 2017, 16, 4791–4796. [Google Scholar] [CrossRef] [PubMed]
  92. Castardeli, C.; Sartório, C.L.; Pimentel, E.B.; Forechi, L.; Mill, J.G. The ACE 2 activator diminazene aceturate (DIZE) improves left ventricular diastolic dysfunction following myocardial infarction in rats. Biomed. Pharmacother. 2018, 107, 212–218. [Google Scholar] [CrossRef] [PubMed]
  93. Wang, M.; Zhang, W.; Zhou, Y.; Zhou, X. Association between serum angiotensin-converting enzyme 2 levels and postoperative myocardial infarction following coronary artery bypass grafting. Exp. Ther. Med. 2014, 7, 1721–1727. [Google Scholar] [CrossRef] [Green Version]
  94. Chen, L.N.; Yang, X.H.; Nissen, D.H.; Chen, Y.Y.; Wang, L.J.; Wang, J.H.; Gao, J.L.; Zhang, L.Y. Dysregulated renin-angiotensin system contributes to acute lung injury caused by hind-limb ischemia-reperfusion in mice. Shock 2013, 40, 420–429. [Google Scholar] [CrossRef]
  95. Li, S.M.; Wang, X.Y.; Liu, F.; Yang, X.H. ACE2 agonist DIZE alleviates lung injury induced by limb ischemia-reperfusion in mice. Sheng Li Xue Bao 2018, 70, 175–183. [Google Scholar] [PubMed]
  96. Yang, X.H.; Wang, Y.H.; Wang, J.J.; Liu, Y.C.; Deng, W.; Qin, C.; Gao, J.L.; Zhang, L.Y. Role of angiotensin-converting enzyme (ACE and ACE2) imbalance on tourniquet-induced remote kidney injury in a mouse hindlimb ischemia-reperfusion model. Peptides 2012, 36, 60–70. [Google Scholar] [CrossRef]
  97. Da Silveira, K.D.; Pompermayer Bosco, K.S.; Diniz, L.R.; Carmona, A.K.; Cassali, G.D.; Bruna-Romero, O.; de Sousa, L.P.; Teixeira, M.M.; Santos, R.A.; e Silva, A.C.S.; et al. ACE2-angiotensin-(1-7)-Mas axis in renal ischaemia/reperfusion injury in rats. Clin. Sci. 2010, 119, 385–394. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  98. Fang, F.; Liu, G.C.; Zhou, X.; Yang, S.; Reich, H.N.; Williams, V.; Hu, A.; Pan, J.; Konvalinka, A.; Oudit, G.Y.; et al. Loss of ACE2 exacerbates murine renal ischemia-reperfusion injury. PLoS ONE 2013, 8, e71433. [Google Scholar]
  99. Zhang, X.; Gao, F.; Yan, Y.; Ruan, Z.; Liu, Z. Combination therapy with human umbilical cord mesenchymal stem cells and angiotensin-converting enzyme 2 is superior for the treatment of acute lung ischemia-reperfusion injury in rats. Cell Biochem. Funct. 2015, 33, 113–120. [Google Scholar] [CrossRef]
  100. Fan, E.; Brodie, D.; Slutsky, A.S. Acute Respiratory Distress Syndrome: Advances in Diagnosis and Treatment. JAMA 2018, 319, 698–710. [Google Scholar] [CrossRef] [PubMed]
  101. Ferrando, C.; Suarez-Sipmann, F.; Mellado-Artigas, R.; Hernández, M.; Gea, A.; Arruti, E.; Aldecoa, C.; Martínez-Pallí, G.; Martínez-González, M.A.; Slutsky, A.S.; et al. Clinical features, ventilatory management, and outcome of ARDS caused by COVID-19 are similar to other causes of ARDS. Intensive Care Med. 2020, 46, 2200–2211. [Google Scholar] [CrossRef]
  102. Banu, N.; Panikar, S.S.; Leal, L.R.; Leal, A.R. Protective role of ACE2 and its downregulation in SARS-CoV-2 infection leading to Macrophage Activation Syndrome: Therapeutic implications. Life Sci. 2020, 256, 117905. [Google Scholar] [CrossRef]
  103. Glowacka, I.; Bertram, S.; Herzog, P.; Pfefferle, S.; Steffen, I.; Muench, M.O.; Simmons, G.; Hofmann, H.; Kuri, T.; Weber, F.; et al. Differential downregulation of ACE2 by the spike proteins of severe acute respiratory syndrome coronavirus and human coronavirus NL63. J. Virol. 2010, 84, 1198–1205. [Google Scholar] [CrossRef] [Green Version]
  104. Yang, P.; Gu, H.; Zhao, Z.; Wang, W.; Cao, B.; Lai, C.; Yang, X.; Zhang, L.; Duan, Y.; Zhang, S.; et al. Angiotensin-converting enzyme 2 (ACE2) mediates influenza H7N9 virus-induced acute lung injury. Sci. Rep. 2014, 4, 7027. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  105. Reddy, R.; Asante, I.; Liu, S.; Parikh, P.; Liebler, J.; Borok, Z.; Rodgers, K.; Baydur, A.; Louie, S.G. Circulating angiotensin peptides levels in Acute Respiratory Distress Syndrome correlate with clinical outcomes: A pilot study. PLoS ONE 2019, 14, e0213096. [Google Scholar] [CrossRef]
  106. Yilin, Z.; Yandong, N.; Faguang, J. Role of angiotensin-converting enzyme (ACE) and ACE2 in a rat model of smoke inhalation induced acute respiratory distress syndrome. Burns 2015, 41, 1468–1477. [Google Scholar] [CrossRef] [PubMed]
  107. Smith, J.C.; Sausville, E.L.; Girish, V.; Yuan, M.L.; Vasudevan, A.; John, K.M.; Sheltzer, J.M. Cigarette Smoke Exposure and Inflammatory Signaling Increase the Expression of the SARS-CoV-2 Receptor ACE2 in the Respiratory Tract. Dev. Cell 2020, 53, 514–529.e3. [Google Scholar] [CrossRef] [PubMed]
  108. Liu, A.; Zhang, X.; Li, R.; Zheng, M.; Yang, S.; Dai, L.; Wu, A.; Hu, C.; Huang, Y.; Xie, M.; et al. Overexpression of the SARS-CoV-2 receptor ACE2 is induced by cigarette smoke in bronchial and alveolar epithelia. J. Pathol. 2021, 253, 17–30. [Google Scholar] [CrossRef] [PubMed]
  109. Imai, Y.; Kuba, K.; Rao, S.; Huan, Y.; Guo, F.; Guan, B.; Yang, P.; Sarao, R.; Wada, T.; Leong-Poi, H.; et al. Angiotensin-converting enzyme 2 protects from severe acute lung failure. Nature 2005, 436, 112–116. [Google Scholar] [CrossRef] [PubMed]
  110. Li, Y.; Zeng, Z.; Cao, Y.; Liu, Y.; Ping, F.; Liang, M.; Xue, Y.; Xi, C.; Zhou, M.; Jiang, W. Angiotensin-converting enzyme 2 prevents lipopolysaccharide-induced rat acute lung injury via suppressing the ERK1/2 and NF-κB signaling pathways. Sci. Rep. 2016, 6, 27911. [Google Scholar] [CrossRef] [Green Version]
  111. Zhou, F.; Yu, T.; Du, R.; Fan, G.; Liu, Y.; Liu, Z.; Xiang, J.; Wang, Y.; Song, B.; Gu, X.; et al. Clinical course and risk factors for mortality of adult inpatients with COVID-19 in Wuhan, China: A retrospective cohort study. Lancet 2020, 395, 1054–1062. [Google Scholar] [CrossRef]
  112. Feng, Y.; Ling, Y.; Bai, T.; Xie, Y.; Huang, J.; Li, J.; Xiong, W.; Yang, D.; Chen, R.; Lu, F.; et al. COVID-19 with Different Severities: A Multicenter Study of Clinical Features. Am. J. Respir. Crit. Care Med. 2020, 201, 1380–1388. [Google Scholar] [CrossRef] [PubMed]
  113. Wang, B.; Li, R.; Lu, Z.; Huang, Y. Does comorbidity increase the risk of patients with COVID-19: Evidence from meta-analysis. Aging 2020, 12, 6049–6057. [Google Scholar] [CrossRef]
  114. Kuba, K.; Imai, Y.; Rao, S.; Gao, H.; Guo, F.; Guan, B.; Huan, Y.; Yang, P.; Zhang, Y.; Deng, W.; et al. A crucial role of angiotensin converting enzyme 2 (ACE2) in SARS coronavirus-induced lung injury. Nat. Med. 2005, 11, 875–879. [Google Scholar] [CrossRef] [PubMed]
  115. Oudit, G.Y.; Kassiri, Z.; Jiang, C.; Liu, P.P.; Poutanen, S.M.; Penninger, J.M.; Butany, J. SARS-coronavirus modulation of myocardial ACE2 expression and inflammation in patients with SARS. Eur. J. Clin. Investig. 2009, 39, 618–625. [Google Scholar] [CrossRef]
  116. Menachery, V.D.; Eisfeld, A.J.; Schäfer, A.; Josset, L.; Sims, A.C.; Proll, S.; Fan, S.; Li, C.; Neumann, G.; Tilton, S.C.; et al. Pathogenic influenza viruses and coronaviruses utilize similar and contrasting approaches to control interferon-stimulated gene responses. MBio 2014, 5, e01174-14. [Google Scholar] [CrossRef] [Green Version]
  117. Lee, J.; Jo, S.J.; Cho, Y.; Lee, J.H.; Oh, I.Y.; Park, J.J.; Cho, Y.S.; Choi, D.J. Effects of renin-angiotensin system blockers on the risk and outcomes of severe acute respiratory syndrome coronavirus 2 infection in patients with hypertension. Korean J. Intern. Med. 2021, 36 (Suppl. S1), S123–S131. [Google Scholar] [CrossRef]
  118. Hippisley-Cox, J.; Young, D.; Coupland, C.; Channon, K.M.; Tan, P.S.; Harrison, D.A.; Rowan, K.; Aveyard, P.; Pavord, I.D.; Watkinson, P.J. Risk of severe COVID-19 disease with ACE inhibitors and angiotensin receptor blockers: Cohort study including 8.3 million people. Heart 2020, 106, 1503–1511. [Google Scholar] [CrossRef]
  119. Xie, Q.; Tang, S.; Li, Y. The divergent protective effects of angiotensin-converting enzyme inhibitors and angiotensin receptor blockers on clinical outcomes of coronavirus disease 2019 (COVID-19): A systematic review and meta-analysis. Ann. Palliat. Med. 2021, 4, apm-21-972. [Google Scholar] [CrossRef]
  120. Jakovac, H. COVID-19: Is the ACE2 just a foe? Am. J. Physiol. Lung Cell Mol. Physiol. 2020, 318, L1025–L1026. [Google Scholar] [CrossRef] [Green Version]
  121. Kommoss, F.K.F.; Schwab, C.; Tavernar, L.; Schreck, J.; Wagner, W.L.; Merle, U.; Jonigk, D.; Schirmacher, P.; Longerich, T. The Pathology of Severe COVID-19-Related Lung Damage. Dtsch. Arztebl. Int. 2020, 117, 500–506. [Google Scholar] [CrossRef]
  122. Vizcaychipi, M.P.; Shovlin, C.L.; McCarthy, A.; Howard, A.; Brown, A.; Hayes, M.; Singh, S.; Christie, L.; Sisson, A.; Davies, R.; et al. Development and implementation of a COVID-19 near real-time traffic light system in an acute hospital setting. Emerg. Med. J. 2020, 37, 630–636. [Google Scholar] [CrossRef] [PubMed]
  123. Simões, E.S.A.C.; Teixeira, M.M. ACE inhibition, ACE2 and angiotensin-(1-7) axis in kidney and cardiac inflammation and fibrosis. Pharmacol. Res. 2016, 107, 154–162. [Google Scholar] [CrossRef] [PubMed]
  124. Fraga-Silva, R.A.; Sorg, B.S.; Wankhede, M.; Dedeugd, C.; Jun, J.Y.; Baker, M.B.; Li, Y.; Castellano, R.K.; Katovich, M.J.; Raizada, M.K.; et al. ACE2 activation promotes antithrombotic activity. Mol. Med. 2010, 16, 210–215. [Google Scholar] [CrossRef] [PubMed]
  125. Khan, A.; Benthin, C.; Zeno, B.; Albertson, T.E.; Boyd, J.; Christie, J.D.; Hall, R.; Poirier, G.; Ronco, J.J.; Tidswell, M.; et al. A pilot clinical trial of recombinant human angiotensin-converting enzyme 2 in acute respiratory distress syndrome. Crit. Care 2017, 21, 234. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  126. Recombinant Human Angiotensin-converting Enzyme 2 (rhACE2) as a Treatment for Patients With COVID-19. Available online: https://ClinicalTrials.gov/show/NCT04287686 (accessed on 23 November 2021).
  127. Imai, Y.; Kuba, K.; Penninger, J.M. The discovery of angiotensin-converting enzyme 2 and its role in acute lung injury in mice. Exp. Physiol. 2008, 93, 543–548. [Google Scholar] [CrossRef] [PubMed]
  128. Rey-Parra, G.J.; Vadivel, A.; Coltan, L.; Hall, A.; Eaton, F.; Schuster, M.; Loibner, H.; Penninger, J.M.; Kassiri, Z.; Oudit, G.Y.; et al. Angiotensin converting enzyme 2 abrogates bleomycin-induced lung injury. J. Mol. Med. 2012, 90, 637–647. [Google Scholar] [CrossRef]
  129. Wösten-van Asperen, R.M.; Lutter, R.; Specht, P.A.; Moll, G.N.; van Woensel, J.B.; van der Loos, C.M.; van Goor, H.; Kamilic, J.; Florquin, S.; Bos, A.P. Acute respiratory distress syndrome leads to reduced ratio of ACE/ACE2 activities and is prevented by angiotensin-(1-7) or an angiotensin II receptor antagonist. J. Pathol. 2011, 225, 618–627. [Google Scholar] [CrossRef]
  130. Wösten-van Asperen, R.M.; Bos, A.P.; Bem, R.A.; Dierdorp, B.S.; Dekker, T.; van Goor, H.; Kamilic, J.; van der Loos, C.M.; van den Berg, E.; Bruijn, M.; et al. Imbalance between pulmonary angiotensin-converting enzyme and angiotensin-converting enzyme 2 activity in acute respiratory distress syndrome. Pediatr. Crit. Care Med. 2013, 14, e438–e441. [Google Scholar] [CrossRef]
  131. Navarrete-Opazo, A.; Mitchell, G.S. Therapeutic potential of intermittent hypoxia: A matter of dose. Am. J. Physiol. Regul. Integr. Comp. Physiol. 2014, 307, R1181–R1197. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  132. Perry, J.C.; Bergamaschi, C.T.; Campos, R.R.; Andersen, M.L.; Casarini, D.E.; Tufik, S. Differential sympathetic activation induced by intermittent hypoxia and sleep loss in rats: Action of angiotensin (1-7). Auton. Neurosci. 2011, 160, 32–36. [Google Scholar] [CrossRef] [Green Version]
  133. Punjabi, N.M.; Caffo, B.S.; Goodwin, J.L.; Gottlieb, D.J.; Newman, A.B.; O’Connor, G.T.; Rapoport, D.M.; Redline, S.; Resnick, H.E.; Robbins, J.A.; et al. Sleep-disordered breathing and mortality: A prospective cohort study. PLoS Med. 2009, 6, e1000132. [Google Scholar] [CrossRef] [Green Version]
  134. Zhou, J.P.; Lin, Y.N.; Li, N.; Sun, X.W.; Ding, Y.J.; Yan, Y.R.; Zhang, L.; Li, Q.Y. Angiotensin-(1-7) Rescues Chronic Intermittent Hypoxia-Aggravated Transforming Growth Factor-β-Mediated Airway Remodeling in Murine and Cellular Models of Asthma. J. Pharmacol. Exp. Ther. 2020, 375, 268–275. [Google Scholar] [CrossRef] [PubMed]
  135. Xiang, Y.H.; Su, X.L.; Hu, C.P.; Luo, Y.Q.; He, R.X. A study of the related pathways of oxidative stress in chronic intermittent hypoxia rats and the effect of N-acetylcysteine. Zhonghua Jie He He Hu Xi Za Zhi 2010, 33, 912–916. [Google Scholar] [PubMed]
  136. Xiang, Y.H.; Su, X.L.; He, R.X.; Hu, C.P. Effects of different patterns of hypoxia on renin angiotension system in serum and tissues of rats. Zhonghua Jie He He Hu Xi Za Zhi 2012, 35, 33–36. [Google Scholar] [PubMed]
  137. Lu, W.; Kang, J.; Hu, K.; Tang, S.; Zhou, X.; Yu, S.; Xu, L. Angiotensin-(1-7) relieved renal injury induced by chronic intermittent hypoxia in rats by reducing inflammation, oxidative stress and fibrosis. Braz. J. Med. Biol. Res. 2017, 50, e5594. [Google Scholar] [CrossRef] [Green Version]
  138. Wang, W.; Song, A.; Zeng, Y.; Chen, X.; Zhang, Y.; Shi, Y.; Lin, Y.; Luo, W. Telmisartan protects chronic intermittent hypoxic mice via modulating cardiac renin-angiotensin system activity. BMC Cardiovasc. Disord. 2018, 18, 133. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  139. Wohlrab, P.; Johann Danhofer, M.; Schaubmayr, W.; Tiboldi, A.; Krenn, K.; Markstaller, K.; Ullrich, R.; Ulrich Klein, K.; Tretter, V. Oxygen conditions oscillating between hypoxia and hyperoxia induce different effects in the pulmonary endothelium compared to constant oxygen conditions. Physiol. Rep. 2021, 9, e14590. [Google Scholar] [CrossRef]
  140. Kou, Y.L.; Zhang, P.P.; Wang, H.Y.; Zhang, J.B.; Tan, X.S.; Huang, C.; Zhang, M. Protective Effect of Angiotensin Converting Enzyme 2 (ACE2) Against Chronic Intermittent Hypoxia-induced Pulmonary Oxidative Stress Injury in Rats. Sichuan Da Xue Xue Bao Yi Xue Ban 2016, 47, 43–48. [Google Scholar]
  141. Lu, W.; Kang, J.; Hu, K.; Tang, S.; Zhou, X.; Yu, S.; Li, Y.; Xu, L. Angiotensin-(1-7) inhibits inflammation and oxidative stress to relieve lung injury induced by chronic intermittent hypoxia in rats. Braz. Med. Biol. Res. 2016, 49, e5431. [Google Scholar] [CrossRef] [Green Version]
  142. Papakonstantinou, E.; Roth, M.; Tamm, M.; Eickelberg, O.; Perruchoud, A.P.; Karakiulakis, G. Hypoxia differentially enhances the effects of transforming growth factor-beta isoforms on the synthesis and secretion of glycosaminoglycans by human lung fibroblasts. J. Pharmacol. Exp. Ther. 2002, 301, 830–837. [Google Scholar] [CrossRef] [Green Version]
  143. Papakonstantinou, E.; Aletras, A.J.; Roth, M.; Tamm, M.; Karakiulakis, G. Hypoxia modulates the effects of transforming growth factor-beta isoforms on matrix-formation by primary human lung fibroblasts. Cytokine 2003, 24, 25–35. [Google Scholar] [CrossRef]
  144. Braun, R.K.; Broytman, O.; Braun, F.M.; Brinkman, J.A.; Clithero, A.; Modi, D.; Pegelow, D.F.; Eldridge, M.; Teodorescu, M. Chronic intermittent hypoxia worsens bleomycin-induced lung fibrosis in rats. Respir. Physiol. Neurobiol. 2018, 256, 97–108. [Google Scholar] [CrossRef]
  145. Gille, T.; Didier, M.; Rotenberg, C.; Delbrel, E.; Marchant, D.; Sutton, A.; Dard, N.; Haine, L.; Voituron, N.; Bernaudin, J.F.; et al. Intermittent Hypoxia Increases the Severity of Bleomycin-Induced Lung Injury in Mice. Oxid. Med. Cell Longev. 2018, 2018, 1240192. [Google Scholar] [CrossRef]
  146. Kang, H.H.; Kim, I.K.; Yeo, C.D.; Kim, S.W.; Lee, H.Y.; Im, J.H.; Kwon, H.Y.; Lee, S.H. The Effects of Chronic Intermittent Hypoxia in Bleomycin-Induced Lung Injury on Pulmonary Fibrosis via Regulating the NF-κB/Nrf2 Signaling Pathway. Tuberc. Respir. Dis. 2020, 83 (Suppl. S1), S63–S74. [Google Scholar] [CrossRef] [PubMed]
  147. Cooke, M.; Cruttenden, R.; Mellor, A.; Lumb, A.; Pattman, S.; Burnett, A.; Boot, C.; Burnip, L.; Boos, C.; O’Hara, J.; et al. A pilot investigation into the effects of acute normobaric hypoxia, high altitude exposure and exercise on serum angiotensin-converting enzyme, aldosterone and cortisol. J. Renin-Angiotensin-Aldosterone Syst. 2018, 19, 1470320318782782. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  148. Osculati, G.; Revera, M.; Branzi, G.; Faini, A.; Malfatto, G.; Bilo, G.; Giuliano, A.; Gregorini, F.; Ciambellotti, F.; Lombardi, C.; et al. Effects of hypobaric hypoxia exposure at high altitude on left ventricular twist in healthy subjects: Data from HIGHCARE study on Mount Everest. Eur. Heart J. Cardiovasc. Imaging 2016, 17, 635–643. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  149. Revera, M.; Salvi, P.; Faini, A.; Giuliano, A.; Gregorini, F.; Bilo, G.; Lombardi, C.; Mancia, G.; Agostoni, P.; Parati, G. Renin-Angiotensin-Aldosterone System Is Not Involved in the Arterial Stiffening Induced by Acute and Prolonged Exposure to High Altitude. Hypertension 2017, 70, 75–84. [Google Scholar] [CrossRef]
  150. Parati, G.; Bilo, G.; Faini, A.; Bilo, B.; Revera, M.; Giuliano, A.; Lombardi, C.; Caldara, G.; Gregorini, F.; Styczkiewicz, K.; et al. Changes in 24 h ambulatory blood pressure and effects of angiotensin II receptor blockade during acute and prolonged high-altitude exposure: A randomized clinical trial. Eur. Heart J. 2014, 35, 3113–3122. [Google Scholar] [CrossRef] [Green Version]
  151. Li, Y.Y.; Luo, D.C.; Xiao, Q. Clinical significance of changes in plasma renin-angiotensin aldosterone system in patients with high altitude pulmonary edema. Zhonghua Nei Ke Za Zhi 1993, 32, 232–234. [Google Scholar] [PubMed]
  152. Giordano, F.J. Oxygen, oxidative stress, hypoxia, and heart failure. J. Clin. Investig. 2005, 115, 500–508. [Google Scholar] [CrossRef]
  153. Tyrankiewicz, U.; Olkowicz, M.; Skórka, T.; Jablonska, M.; Orzylowska, A.; Bar, A.; Gonet, M.; Berkowicz, P.; Jasinski, K.; Zoladz, J.A.; et al. Activation pattern of ACE2/Ang-(1-7) and ACE/Ang II pathway in course of heart failure assessed by multiparametric MRI in vivo in Tgαq*44 mice. J. Appl. Physiol. 2018, 124, 52–65. [Google Scholar] [CrossRef]
  154. Cohen-Segev, R.; Francis, B.; Abu-Saleh, N.; Awad, H.; Lazarovich, A.; Kabala, A.; Aronson, D.; Abassi, Z. Cardiac and renal distribution of ACE and ACE-2 in rats with heart failure. Acta Histochem. 2014, 116, 1342–1349. [Google Scholar] [CrossRef]
  155. Liang, B.; Li, Y.; Han, Z.; Xue, J.; Zhang, Y.; Jia, S.; Wang, C. ACE2-Ang (1-7) axis is induced in pressure overloaded rat model. Int. Clin. Exp. Pathol. 2015, 8, 1443–1450. [Google Scholar]
  156. Wang, J.; He, W.; Guo, L.; Zhang, Y.; Li, H.; Han, S.; Shen, D. The ACE2-Ang (1-7)-Mas receptor axis attenuates cardiac remodeling and fibrosis in post-myocardial infarction. Mol. Med. Rep. 2017, 16, 1973–1981. [Google Scholar] [CrossRef] [PubMed]
  157. Gomes-Santos, I.L.; Fernandes, T.; Couto, G.K.; Ferreira-Filho, J.C.; Salemi, V.M.; Fernandes, F.B.; Casarini, D.E.; Brum, P.C.; Rossoni, L.V.; de Oliveira, E.M.; et al. Effects of exercise training on circulating and skeletal muscle renin-angiotensin system in chronic heart failure rats. PLoS ONE 2014, 9, e98012. [Google Scholar]
  158. Larouche-Lebel, É.; Loughran, K.A.; Oyama, M.A.; Solter, P.F.; Laughlin, D.S.; Sánchez, M.D.; Assenmacher, C.A.; Fox, P.R.; Fries, R.C. Plasma and tissue angiotensin-converting enzyme 2 activity and plasma equilibrium concentrations of angiotensin peptides in dogs with heart disease. J. Vet. Intern. Med. 2019, 33, 1571–1584. [Google Scholar] [CrossRef] [Green Version]
  159. Yu, J.; Wu, Y.; Zhang, Y.; Zhang, L.; Ma, Q.; Luo, X. Role of ACE2-Ang (1-7)-Mas receptor axis in heart failure with preserved ejection fraction with hypertension. Zhong Nan Da Xue Xue Bao Yi Xue Ban 2018, 43, 738–746. [Google Scholar] [PubMed]
  160. Cernecka, H.; Doka, G.; Srankova, J.; Pivackova, L.; Malikova, E.; Galkova, K.; Kyselovic, J.; Krenek, P.; Klimas, J. Ramipril restores PPARβ/δ and PPARγ expressions and reduces cardiac NADPH oxidase but fails to restore cardiac function and accompanied myosin heavy chain ratio shift in severe anthracycline-induced cardiomyopathy in rat. Eur. J. Pharmacol. 2016, 791, 244–253. [Google Scholar] [CrossRef]
  161. Ma, H.; Kong, J.; Wang, Y.L.; Li, J.L.; Hei, N.H.; Cao, X.R.; Yang, J.J.; Yan, W.J.; Liang, W.J.; Dai, H.Y.; et al. Angiotensin-converting enzyme 2 overexpression protects against doxorubicin-induced cardiomyopathy by multiple mechanisms in rats. Oncotarget 2017, 8, 24548–24563. [Google Scholar] [CrossRef] [Green Version]
  162. Huentelman, M.J.; Grobe, J.L.; Vazquez, J.; Stewart, J.M.; Mecca, A.P.; Katovich, M.J.; Ferrario, C.M.; Raizada, M.K. Protection from angiotensin II-induced cardiac hypertrophy and fibrosis by systemic lentiviral delivery of ACE2 in rats. Exp. Physiol. 2005, 90, 783–790. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  163. Patel, V.B.; Takawale, A.; Ramprasath, T.; Das, S.K.; Basu, R.; Grant, M.B.; Hall, D.A.; Kassiri, Z.; Oudit, G.Y. Antagonism of angiotensin 1-7 prevents the therapeutic effects of recombinant human ACE2. J. Mol. Med. 2015, 93, 1003–1013. [Google Scholar] [CrossRef] [Green Version]
  164. Grobe, J.L.; Mecca, A.P.; Lingis, M.; Shenoy, V.; Bolton, T.A.; Machado, J.M.; Speth, R.C.; Raizada, M.K.; Katovich, M.J. Prevention of angiotensin II-induced cardiac remodeling by angiotensin-(1-7). Am. J. Physiol. Heart Circ. Physiol. 2007, 292, H736–H742. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  165. Wang, X.; Ye, Y.; Gong, H.; Wu, J.; Yuan, J.; Wang, S.; Yin, P.; Ding, Z.; Kang, L.; Jiang, Q.; et al. 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. 2016, 97, 180–190. [Google Scholar] [CrossRef]
  166. Epelman, S.; Tang, W.H.; Chen, S.Y.; Van Lente, F.; Francis, G.S.; Sen, S. Detection of soluble angiotensin-converting enzyme 2 in heart failure: Insights into the endogenous counter-regulatory pathway of the renin-angiotensin-aldosterone system. J. Am. Coll. Cardiol. 2008, 52, 750–754. [Google Scholar] [CrossRef] [Green Version]
  167. Úri, K.; Fagyas, M.; Mányiné Siket, I.; Kertész, A.; Csanádi, Z.; Sándorfi, G.; Clemens, M.; Fedor, R.; Papp, Z.; Édes, I.; et al. New perspectives in the renin-angiotensin-aldosterone system (RAAS) IV: Circulating ACE2 as a biomarker of systolic dysfunction in human hypertension and heart failure. PLoS ONE 2014, 9, e87845. [Google Scholar] [CrossRef]
  168. Shao, Z.; Schuster, A.; Borowski, A.G.; Thakur, A.; Li, L.; Tang, W.H.W. Soluble angiotensin converting enzyme 2 levels in chronic heart failure is associated with decreased exercise capacity and increased oxidative stress-mediated endothelial dysfunction. Transl. Res. 2019, 212, 80–88. [Google Scholar] [CrossRef]
  169. Wang, J.; Li, N.; Gao, F.; Song, R.; Zhu, S.; Geng, Z. Balance between angiotensin converting enzyme and angiotensin converting enzyme 2 in patients with chronic heart failure. J. Renin-Angiotensin-Aldosterone Syst. 2015, 16, 553–558. [Google Scholar] [CrossRef] [PubMed]
  170. Messmann, R.; Dietl, A.; Wagner, S.; Domenig, O.; Jungbauer, C.; Luchner, A.; Maier, L.S.; Schopka, S.; Hirt, S.; Schmid, C.; et al. Alterations of the renin angiotensin system in human end-stage heart failure before and after mechanical cardiac unloading by LVAD support. Mol. Cell Biochem. 2020, 472, 79–94. [Google Scholar] [CrossRef] [PubMed]
  171. Goulter, A.B.; Goddard, M.J.; Allen, J.C.; Clark, K.L. ACE2 gene expression is up-regulated in the human failing heart. BMC Med. 2004, 2, 19. [Google Scholar] [CrossRef]
  172. Batlle, M.; Roig, E.; Perez-Villa, F.; Lario, S.; Cejudo-Martin, P.; García-Pras, E.; Ortiz, J.; Roqué, M.; Orús, J.; Rigol, M.; et al. Increased expression of the renin-angiotensin system and mast cell density but not of angiotensin-converting enzyme II in late stages of human heart failure. J. Heart Lung Transplant. 2006, 25, 1117–1125. [Google Scholar] [CrossRef] [PubMed]
  173. Walters, T.E.; Kalman, J.M.; Patel, S.K.; Mearns, M.; Velkoska, E.; Burrell, L.M. Angiotensin converting enzyme 2 activity and human atrial fibrillation: Increased plasma angiotensin converting enzyme 2 activity is associated with atrial fibrillation and more advanced left atrial structural remodelling. Europace 2017, 19, 1280–1287. [Google Scholar] [CrossRef]
  174. Murray, C.J.; Barber, R.M.; Foreman, K.J.; Ozgoren, A.A.; Abd-Allah, F.; Abera, S.F.; Aboyans, V.; Abraham, J.P.; Abubakar, I.; Abu-Raddad, L.J.; et al. Global, regional, and national age-sex specific all-cause and cause-specific mortality for 240 causes of death, 1990–2013: A systematic analysis for the Global Burden of Disease Study 2013. Lancet 2015, 385, 117–171. [Google Scholar] [CrossRef]
  175. Lala, A.; Desai, A.S. The role of coronary artery disease in heart failure. Heart Fail. Clin. 2014, 10, 353–365. [Google Scholar] [CrossRef]
  176. Knuuti, J.; Wijns, W.; Saraste, A.; Capodanno, D.; Barbato, E.; Funck-Brentano, C.; Prescott, E.; Storey, R.F.; Deaton, C.; Cuisset, T.; et al. 2019 ESC Guidelines for the diagnosis and management of chronic coronary syndromes: The Task Force for the diagnosis and management of chronic coronary syndromes of the European Society of Cardiology (ESC). Eur. Heart J. 2019, 41, 407–477. [Google Scholar] [CrossRef] [PubMed]
  177. Bangalore, S.; Fakheri, R.; Wandel, S.; Toklu, B.; Wandel, J.; Messerli, F.H. Renin angiotensin system inhibitors for patients with stable coronary artery disease without heart failure: Systematic review and meta-analysis of randomized trials. BMJ 2017, 356, j4. [Google Scholar] [CrossRef] [Green Version]
  178. Zhou, X.; Zhang, P.; Liang, T.; Chen, Y.; Liu, D.; Yu, H. Relationship between circulating levels of angiotensin-converting enzyme 2-angiotensin-(1-7)-MAS axis and coronary heart disease. Heart Vessel. 2020, 35, 153–161. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  179. Ramchand, J.; Patel, S.K.; Srivastava, P.M.; Farouque, O.; Burrell, L.M. Elevated plasma angiotensin converting enzyme 2 activity is an independent predictor of major adverse cardiac events in patients with obstructive coronary artery disease. PLoS ONE 2018, 13, e0198144. [Google Scholar] [CrossRef]
  180. Campbell, D.J.; Zeitz, C.J.; Esler, M.D.; Horowitz, J.D. Evidence against a major role for angiotensin converting enzyme-related carboxypeptidase (ACE2) in angiotensin peptide metabolism in the human coronary circulation. J. Hypertens. 2004, 22, 1971–1976. [Google Scholar] [CrossRef] [PubMed]
  181. Tikoo, K.; Patel, G.; Kumar, S.; Karpe, P.A.; Sanghavi, M.; Malek, V.; Srinivasan, K. Tissue specific up regulation of ACE2 in rabbit model of atherosclerosis by atorvastatin: Role of epigenetic histone modifications. Biochem. Pharmacol. 2015, 93, 343–351. [Google Scholar] [CrossRef]
  182. Pernomian, L.; do Prado, A.F.; Gomes, M.S.; Pernomian, L.; da Silva, C.; Gerlach, R.F.; de Oliveira, A.M. MAS receptors mediate vasoprotective and atheroprotective effects of candesartan upon the recovery of vascular angiotensin-converting enzyme 2-angiotensin-(1-7)-MAS axis functionality. Eur. J. Pharmacol. 2015, 764, 173–188. [Google Scholar] [CrossRef]
  183. Sluimer, J.C.; Gasc, J.M.; Hamming, I.; van Goor, H.; Michaud, A.; van den Akker, L.H.; Jütten, B.; Cleutjens, J.; Bijnens, A.P.; Corvol, P.; et al. Angiotensin-converting enzyme 2 (ACE2) expression and activity in human carotid atherosclerotic lesions. J. Pathol. 2008, 215, 273–279. [Google Scholar] [CrossRef]
  184. Thatcher, S.E.; Zhang, X.; Howatt, D.A.; Lu, H.; Gurley, S.B.; Daugherty, A.; Cassis, L.A. 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, 758–765. [Google Scholar] [CrossRef] [Green Version]
  185. Sahara, M.; Ikutomi, M.; Morita, T.; Minami, Y.; Nakajima, T.; Hirata, Y.; Nagai, R.; Sata, M. Deletion of angiotensin-converting enzyme 2 promotes the development of atherosclerosis and arterial neointima formation. Cardiovasc. Res. 2014, 101, 236–246. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  186. Lovren, F.; Pan, Y.; Quan, A.; Teoh, H.; Wang, G.; Shukla, P.C.; Levitt, K.S.; Oudit, G.Y.; Al-Omran, M.; Stewart, D.J.; et al. Angiotensin converting enzyme-2 confers endothelial protection and attenuates atherosclerosis. Am. J. Physiol. Heart Circ. Physiol. 2008, 295, H1377–H1384. [Google Scholar] [CrossRef] [Green Version]
  187. Dong, B.; Zhang, C.; Feng, J.B.; Zhao, Y.X.; Li, S.Y.; Yang, Y.P.; Dong, Q.L.; Deng, B.P.; Zhu, L.; Yu, Q.T.; et al. Overexpression of ACE2 enhances plaque stability in a rabbit model of atherosclerosis. Arterioscler. Thromb. Vasc. Biol. 2008, 28, 1270–1276. [Google Scholar] [CrossRef] [PubMed]
  188. Guo, Y.J.; Li, W.H.; Wu, R.; Xie, Q.; Cui, L.Q. ACE2 overexpression inhibits angiotensin II-induced monocyte chemoattractant protein-1 expression in macrophages. Arch. Med. Res. 2008, 39, 149–154. [Google Scholar] [CrossRef] [PubMed]
  189. Zhang, C.; Zhao, Y.X.; Zhang, Y.H.; Zhu, L.; Deng, B.P.; Zhou, Z.L.; Li, S.Y.; Lu, X.T.; Song, L.L.; Lei, X.M.; et al. Angiotensin-converting enzyme 2 attenuates atherosclerotic lesions by targeting vascular cells. Proc. Natl. Acad. Sci. USA 2010, 107, 15886–15891. [Google Scholar] [CrossRef] [Green Version]
  190. Fraga-Silva, R.A.; Montecucco, F.; Costa-Fraga, F.P.; Nencioni, A.; Caffa, I.; Bragina, M.E.; Mach, F.; Raizada, M.K.; Santos, R.A.S.; da Silva, R.F.; et al. Diminazene enhances stability of atherosclerotic plaques in ApoE-deficient mice. Vascul. Pharmacol. 2015, 74, 103–113. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  191. Stachowicz, A.; Wiśniewska, A.; Kuś, K.; Białas, M.; Łomnicka, M.; Totoń-Żurańska, J.; Kiepura, A.; Stachyra, K.; Suski, M.; Bujak-Giżycka, B.; et al. Diminazene Aceturate Stabilizes Atherosclerotic Plaque and Attenuates Hepatic Steatosis in apoE-Knockout Mice by Influencing Macrophages Polarization and Taurine Biosynthesis. Int. J. Mol. Sci. 2021, 22, 5861. [Google Scholar] [CrossRef]
  192. Yang, J.; Yang, X.; Meng, X.; Dong, M.; Guo, T.; Kong, J.; Zhang, K.; Zhang, Y.; Zhang, C. Endogenous activated angiotensin-(1-7) plays a protective effect against atherosclerotic plaques unstability in high fat diet fed ApoE knockout mice. Int. J. Cardiol. 2015, 184, 645–652. [Google Scholar] [CrossRef] [PubMed]
  193. Stewart, A.G.; Waterhouse, J.C.; Billings, C.G.; Baylis, P.; Howard, P. Effects of angiotensin converting enzyme inhibition on sodium excretion in patients with hypoxaemic chronic obstructive pulmonary disease. Thorax 1994, 49, 995–998. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  194. Gu, Y.Q.; Huang, D.J.; Hu, B.X. Influence of acute respiratory failure on plasma renin activity and plasma angiotensin II level in advanced chronic obstructive pulmonary disease and chronic cor pulmonale. Zhonghua Nei Ke Za Zhi 1989, 28, 347–350. [Google Scholar]
  195. Hemlin, M.; Ljungman, S.; Carlson, J.; Maljukanovic, S.; Mobini, R.; Bech-Hanssen, O.; Skoogh, B.E. The effects of hypoxia and hypercapnia on renal and heart function, haemodynamics and plasma hormone levels in stable COPD patients. Clin. Respir. J. 2007, 1, 80–90. [Google Scholar] [CrossRef] [PubMed]
  196. Raff, H.; Levy, S.A. Renin-angiotensin II-aldosterone and ACTH-cortisol control during acute hypoxemia and exercise in patients with chronic obstructive pulmonary disease. Am. Rev. Respir. Dis. 1986, 133, 396–399. [Google Scholar] [PubMed]
  197. Muñoz, J.; González, J.M.; de Ramón, A.; García, J.L.; Ortega, B.; Martínez, I.; Madero, R.; Sánchez, F.; Villamor, J. Variations of angiotensin converting enzyme in chronic obstructive pulmonary disease and chronic respiratory insufficiency. Rev. Clin. Esp. 1994, 194, 1018–1022. [Google Scholar] [PubMed]
  198. Zeng, G.B.; Duan, S.F. Role of the renin-angiotensin-aldosterone system in the pathogenesis of edema formation in chronic obstructive pulmonary disease. Zhonghua Nei Ke Za Zhi 1989, 28, 283–285. [Google Scholar]
  199. Sandoval, J.; Del Valle-Mondragón, L.; Masso, F.; Zayas, N.; Pulido, T.; Teijeiro, R.; Gonzalez-Pacheco, H.; Olmedo-Ocampo, R.; Sisniega, C.; Paez-Arenas, A.; et al. Angiotensin converting enzyme 2 and angiotensin (1-7) axis in pulmonary arterial hypertension. Eur. Respir. J. 2020, 56, 1902416. [Google Scholar] [CrossRef]
  200. Pan, X.; Shao, Y.; Wu, F.; Wang, Y.; Xiong, R.; Zheng, J.; Tian, H.; Wang, B.; Wang, Y.; Zhang, Y.; et al. FGF21 Prevents Angiotensin II-Induced Hypertension and Vascular Dysfunction by Activation of ACE2/Angiotensin-(1-7) Axis in Mice. Cell Metab. 2018, 27, 1323–1337.e5. [Google Scholar] [CrossRef] [Green Version]
  201. Muñoz, M.C.; Burghi, V.; Miquet, J.G.; Giani, J.F.; Banegas, R.D.; Toblli, J.E.; Fang, Y.; Wang, F.; Bartke, A.; Dominici, F.P. Downregulation of the ACE2/Ang-(1-7)/Mas axis in transgenic mice overexpressing GH. J. Endocrinol. 2014, 221, 215–227. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  202. Srivastava, P.; Badhwar, S.; Chandran, D.S.; Jaryal, A.K.; Jyotsna, V.P.; Deepak, K.K. Imbalance between Angiotensin II-Angiotensin (1-7) system is associated with vascular endothelial dysfunction and inflammation in type 2 diabetes with newly diagnosed hypertension. Diabetes Metab. Syndr. 2019, 13, 2061–2068. [Google Scholar] [CrossRef]
  203. Han, W.; Wei, Z.; Dang, R.; Guo, Y.; Zhang, H.; Geng, C.; Wang, C.; Feng, Q.; Jiang, P. Angiotensin-Ⅱ and angiotensin-(1-7) imbalance affects comorbidity of depression and coronary heart disease. Peptides 2020, 131, 170353. [Google Scholar] [CrossRef]
  204. Tikellis, C.; Pickering, R.; Tsorotes, D.; Du, X.J.; Kiriazis, H.; Nguyen-Huu, T.P.; Head, G.A.; Cooper, M.E.; Thomas, M.C. Interaction of diabetes and ACE2 in the pathogenesis of cardiovascular disease in experimental diabetes. Clin. Sci. 2012, 123, 519–529. [Google Scholar] [CrossRef] [Green Version]
  205. Moritani, T.; Iwai, M.; Kanno, H.; Nakaoka, H.; Iwanami, J.; Higaki, T.; Ishii, E.; Horiuchi, M. ACE2 deficiency induced perivascular fibrosis and cardiac hypertrophy during postnatal development in mice. J. Am. Soc. Hypertens. 2013, 7, 259–266. [Google Scholar] [CrossRef] [PubMed]
  206. Hemnes, A.R.; Rathinasabapathy, A.; Austin, E.A.; Brittain, E.L.; Carrier, E.J.; Chen, X.; Fessel, J.P.; Fike, C.D.; Fong, P.; Fortune, N.; et al. A potential therapeutic role for angiotensin-converting enzyme 2 in human pulmonary arterial hypertension. Eur. Respir. J. 2018, 51, 1702638. [Google Scholar] [CrossRef]
  207. Xue, T.; Wei, N.; Xin, Z.; Qingyu, X. Angiotensin-converting enzyme-2 overexpression attenuates inflammation in rat model of chronic obstructive pulmonary disease. Inhal. Toxicol. 2014, 26, 14–22. [Google Scholar] [CrossRef]
  208. Zhang, Y.; Li, Y.; Shi, C.; Fu, X.; Zhao, L.; Song, Y. Angiotensin-(1-7)-mediated Mas1 receptor/NF-κB-p65 signaling is involved in a cigarette smoke-induced chronic obstructive pulmonary disease mouse model. Environ. Toxicol. 2018, 33, 5–15. [Google Scholar] [CrossRef] [PubMed]
  209. Higham, A.; Singh, D. Increased ACE2 Expression in Bronchial Epithelium of COPD Patients who are Overweight. Obesity 2020, 28, 1586–1589. [Google Scholar] [CrossRef] [PubMed]
  210. Cai, G.; Bossé, Y.; Xiao, F.; Kheradmand, F.; Amos, C.I. Tobacco Smoking Increases the Lung Gene Expression of ACE2, the Receptor of SARS-CoV-2. Am. J. Respir. Crit. Care Med. 2020, 201, 1557–1559. [Google Scholar] [CrossRef] [PubMed]
  211. Leung, J.M.; Yang, C.X.; Tam, A.; Shaipanich, T.; Hackett, T.L.; Singhera, G.K.; Dorscheid, D.R.; Sin, D.D. ACE-2 expression in the small airway epithelia of smokers and COPD patients: Implications for COVID-19. Eur. Respir. J. 2020, 55, 2000688. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  212. Lebek, S.; Tafelmeier, M.; Messmann, R.; Provaznik, Z.; Schmid, C.; Maier, L.S.; Birner, C.; Arzt, M.; Wagner, S. Angiotensin-converting enzyme inhibitor/angiotensin II receptor blocker treatment and haemodynamic factors are associated with increased cardiac mRNA expression of angiotensin-converting enzyme 2 in patients with cardiovascular disease. Eur. J. Heart Fail. 2020, 22, 2248–2257. [Google Scholar] [CrossRef]
  213. Sinha, S.; Cheng, K.; Schäffer, A.A.; Aldape, K.; Schiff, E.; Ruppin, E. In vitro and in vivo identification of clinically approved drugs that modify ACE2 expression. Mol. Syst. Biol. 2020, 16, e9628. [Google Scholar] [CrossRef]
  214. Klimas, J.; Olvedy, M.; Ochodnicka-Mackovicova, K.; Kruzliak, P.; Cacanyiova, S.; Kristek, F.; Krenek, P.; Ochodnicky, P. Perinatally administered losartan augments renal ACE2 expression but not cardiac or renal Mas receptor in spontaneously hypertensive rats. J. Cell Mol. Med. 2015, 19, 1965–1974. [Google Scholar] [CrossRef] [PubMed]
  215. Finney, L.J.; Glanville, N.; Farne, H.; Aniscenko, J.; Fenwick, P.; Kemp, S.V.; Trujillo-Torralbo, M.B.; Loo, S.L.; Calderazzo, M.A.; Wedzicha, J.A.; et al. Inhaled corticosteroids downregulate the SARS-CoV-2 receptor ACE2 in COPD through suppression of type I interferon. J. Allergy Clin. Immunol. 2021, 147, 510–519.e5. [Google Scholar] [CrossRef]
  216. Peters, M.C.; Sajuthi, S.; Deford, P.; Christenson, S.; Rios, C.L.; Montgomery, M.T.; Woodruff, P.G.; Mauger, D.T.; Erzurum, S.C.; Johansson, M.W.; et al. COVID-19-related Genes in Sputum Cells in Asthma. Relationship to Demographic Features and Corticosteroids. Am. J. Respir. Crit. Care Med. 2020, 202, 83–90. [Google Scholar] [CrossRef]
  217. Aliee, H.; Massip, F.; Qi, C.; de Biase, M.S.; van Nijnatten, J.L.; Kersten, E.T.G.; Kermani, N.Z.; Khuder, B.; Vonk, J.M.; Vermeulen, R.C.H.; et al. Determinants of SARS-CoV-2 receptor gene expression in upper and lower airways. medRxiv 2020. [Google Scholar]
  218. Shin, Y.H.; Min, J.J.; Lee, J.H.; Kim, E.H.; Kim, G.E.; Kim, M.H.; Lee, J.J.; Ahn, H.J. The effect of fluvastatin on cardiac fibrosis and angiotensin-converting enzyme-2 expression in glucose-controlled diabetic rat hearts. Heart Vessels 2017, 32, 618–627. [Google Scholar] [CrossRef]
  219. Li, Y.H.; Wang, Q.X.; Zhou, J.W.; Chu, X.M.; Man, Y.L.; Liu, P.; Ren, B.B.; Sun, T.R.; An, Y. Effects of rosuvastatin on expression of angiotensin-converting enzyme 2 after vascular balloon injury in rats. J. Geriatr. Cardiol. 2013, 10, 151–158. [Google Scholar] [PubMed]
  220. Chen, J.Y.; Lin, C.H.; Chen, B.C. Hypoxia-induced ADAM 17 expression is mediated by RSK1-dependent C/EBPβ activation in human lung fibroblasts. Mol. Immunol. 2017, 88, 155–163. [Google Scholar] [CrossRef]
  221. Zheng, D.Y.; Zhao, J.; Yang, J.M.; Wang, M.; Zhang, X.T. Enhanced ADAM17 expression is associated with cardiac remodeling in rats with acute myocardial infarction. Life Sci. 2016, 151, 61–69. [Google Scholar] [CrossRef] [PubMed]
  222. Ge, J.; Tang, L.; Mu, P.; Zhu, F.; Xie, L.; Tang, Y. Association of ADAM17 Expression Levels in Patients with Interstitial Lung Disease. Immunol. Invest. 2020, 49, 134–145. [Google Scholar] [CrossRef] [PubMed]
  223. Ottolenghi, S.; Zulueta, A.; Caretti, A. Iron and Sphingolipids as Common Players of (Mal)Adaptation to Hypoxia in Pulmonary Diseases. Int. J. Mol. Sci. 2020, 21, 307. [Google Scholar] [CrossRef] [Green Version]
  224. Lettieri, C.J.; Nathan, S.D.; Barnett, S.D.; Ahmad, S.; Shorr, A.F. Prevalence and outcomes of pulmonary arterial hypertension in advanced idiopathic pulmonary fibrosis. Chest 2006, 129, 746–752. [Google Scholar] [CrossRef] [PubMed]
  225. Terzano, C.; Conti, V.; Di Stefano, F.; Petroianni, A.; Ceccarelli, D.; Graziani, E.; Mariotta, S.; Ricci, A.; Vitarelli, A.; Puglisi, G.; et al. Comorbidity, hospitalization, and mortality in COPD: Results from a longitudinal study. Lung 2010, 188, 321–329. [Google Scholar] [CrossRef] [PubMed]
  226. Li, X.; Molina-Molina, M.; Abdul-Hafez, A.; Uhal, V.; Xaubet, A.; Uhal, B.D. Angiotensin converting enzyme-2 is protective but downregulated in human and experimental lung fibrosis. Am. J. Physiol. Lung Cell Mol. Physiol. 2008, 295, L178–L185. [Google Scholar] [CrossRef] [Green Version]
  227. Oarhe, C.I.; Dang, V.; Dang, M.; Nguyen, H.; Gopallawa, I.; Gewolb, I.H.; Uhal, B.D. Hyperoxia downregulates angiotensin-converting enzyme-2 in human fetal lung fibroblasts. Pediatr. Res. 2015, 77, 656–662. [Google Scholar] [CrossRef] [Green Version]
  228. Mohamed, T.L.; Nguyen, H.T.; Abdul-Hafez, A.; Dang, V.X.; Dang, M.T.; Gewolb, I.H.; Uhal, B.D. Prior hypoxia prevents downregulation of ACE-2 by hyperoxia in fetal human lung fibroblasts. Exp. Lung Res. 2016, 42, 121–130. [Google Scholar] [CrossRef] [Green Version]
  229. Liu, M.L.; Xing, S.J.; Liang, X.Q.; Luo, Y.; Zhang, B.; Li, Z.C.; Dong, M.Q. Reversal of Hypoxic Pulmonary Hypertension by Hypoxia-Inducible Overexpression of Angiotensin-(1-7) in Pulmonary Endothelial Cells. Mol. Ther. Methods Clin. Dev. 2020, 17, 975–985. [Google Scholar] [CrossRef]
  230. Hu, H.H.; Zhang, R.F.; Dong, L.L.; Chen, E.G.; Ying, K.J. Overexpression of ACE2 prevents hypoxia-induced pulmonary hypertension in rats by inhibiting proliferation and immigration of PASMCs. Eur. Rev. Med. Pharmacol. Sci. 2020, 24, 3968–3980. [Google Scholar]
  231. Kleinsasser, A.; Pircher, I.; Treml, B.; Schwienbacher, M.; Schuster, M.; Janzek, E.; Loibner, H.; Penninger, J.M.; Loeckinger, A. Recombinant angiotensin-converting enzyme 2 suppresses pulmonary vasoconstriction in acute hypoxia. Wilderness Environ. Med. 2012, 23, 24–30. [Google Scholar] [CrossRef] [Green Version]
  232. Dang, Z.; Su, S.; Jin, G.; Nan, X.; Ma, L.; Li, Z.; Lu, D.; Ge, R. Tsantan Sumtang attenuated chronic hypoxia-induced right ventricular structure remodeling and fibrosis by equilibrating local ACE-AngII-AT1R/ACE2-Ang1-7-Mas axis in rat. J. Ethnopharmacol. 2020, 250, 112470. [Google Scholar] [CrossRef] [PubMed]
  233. Zhang, R.; Wu, Y.; Zhao, M.; Liu, C.; Zhou, L.; Shen, S.; Liao, S.; Yang, K.; Li, Q.; Wan, H. Role of HIF-1alpha in the regulation ACE and ACE2 expression in hypoxic human pulmonary artery smooth muscle cells. Am. J. Physiol. Lung Cell Mol. Physiol. 2009, 297, L631–L640. [Google Scholar] [CrossRef] [PubMed]
  234. Joshi, S.; Wollenzien, H.; Leclerc, E.; Jarajapu, Y.P. Hypoxic regulation of angiotensin-converting enzyme 2 and Mas receptor in human CD34(+) cells. J. Cell Physiol. 2019, 234, 20420–20431. [Google Scholar] [CrossRef] [PubMed]
  235. Wang, Y.X.; Liu, M.L.; Zhang, B.; Fu, E.Q.; Li, Z.C. Fasudil alleviated hypoxia-induced pulmonary hypertension by stabilizing the expression of angiotensin-(1-7) in rats. Eur. Rev. Med. Pharmacol. Sci. 2016, 20, 3304–3312. [Google Scholar] [PubMed]
  236. Zhang, R.; Su, H.; Ma, X.; Xu, X.; Liang, L.; Ma, G.; Shi, L. MiRNA let-7b promotes the development of hypoxic pulmonary hypertension by targeting ACE2. Am. J. Physiol. Lung Cell Mol. Physiol. 2019, 316, L547–L557. [Google Scholar] [CrossRef] [PubMed]
  237. Hampl, V.; Herget, J.; Bíbová, J.; Baňasová, A.; Husková, Z.; Vaňourková, Z.; Jíchová, Š.; Kujal, P.; Vernerová, Z.; Sadowski, J.; et al. Intrapulmonary activation of the angiotensin-converting enzyme type 2/angiotensin 1-7/G-protein-coupled Mas receptor axis attenuates pulmonary hypertension in Ren-2 transgenic rats exposed to chronic hypoxia. Physiol. Res. 2015, 64, 25–38. [Google Scholar] [CrossRef] [PubMed]
  238. Shang, Y.; Xu, C.; Jiang, F.; Huang, R.; Li, Y.; Zhou, Y.; Xu, F.; Dai, H. Clinical characteristics and changes of chest CT features in 307 patients with common COVID-19 pneumonia infected SARS-CoV-2: A multicenter study in Jiangsu, China. Int. J. Infect. Dis. 2020, 96, 157–162. [Google Scholar] [CrossRef] [PubMed]
  239. Wang, C.; Shi, B.; Wei, C.; Ding, H.; Gu, J.; Dong, J. Initial CT features and dynamic evolution of early-stage patients with COVID-19. Radiol. Infect. Dis. 2020, 7, 195–203. [Google Scholar] [CrossRef]
  240. Francone, M.; Iafrate, F.; Masci, G.M.; Coco, S.; Cilia, F.; Manganaro, L.; Panebianco, V.; Andreoli, C.; Colaiacomo, M.C.; Zingaropoli, M.A.; et al. Chest CT score in COVID-19 patients: Correlation with disease severity and short-term prognosis. Eur. Radiol. 2020, 30, 6808–6817. [Google Scholar] [CrossRef] [PubMed]
  241. Salehi, S.; Abedi, A.; Balakrishnan, S.; Gholamrezanezhad, A. Coronavirus Disease 2019 (COVID-19): A Systematic Review of Imaging Findings in 919 Patients. Am. J. Roentgenol. 2020, 215, 87–93. [Google Scholar] [CrossRef] [PubMed]
  242. Wu, J.; Feng, L.C.; Xian, X.Y.; Qiang, J.; Zhang, J.; Mao, Q.X.; Kong, S.F.; Chen, Y.C.; Pan, J.P. Novel coronavirus pneumonia (COVID-19) CT distribution and sign features. Zhonghua Jie He He Hu Xi Za Zhi 2020, 43, 321–326. [Google Scholar] [PubMed]
  243. Xu, J.; Xu, X.; Jiang, L.; Dua, K.; Hansbro, P.M.; Liu, G. SARS-CoV-2 induces transcriptional signatures in human lung epithelial cells that promote lung fibrosis. Respir. Res. 2020, 21, 182. [Google Scholar] [CrossRef] [PubMed]
  244. Dai, H.; Zhang, X.; Xia, J.; Zhang, T.; Shang, Y.; Huang, R.; Liu, R.; Wang, D.; Li, M.; Wu, J.; et al. High-resolution Chest CT Features and Clinical Characteristics of Patients Infected with COVID-19 in Jiangsu, China. Int. J. Infect. Dis. 2020, 95, 106–112. [Google Scholar] [CrossRef]
  245. Shi, H.; Han, X.; Jiang, N.; Cao, Y.; Alwalid, O.; Gu, J.; Fan, Y.; Zheng, C. Radiological findings from 81 patients with COVID-19 pneumonia in Wuhan, China: A descriptive study. Lancet Infect. Dis. 2020, 20, 425–434. [Google Scholar] [CrossRef]
  246. Yu, M.; Liu, Y.; Xu, D.; Zhang, R.; Lan, L.; Xu, H. Prediction of the Development of Pulmonary Fibrosis Using Serial Thin-Section CT and Clinical Features in Patients Discharged after Treatment for COVID-19 Pneumonia. Korean J. Radiol. 2020, 21, 746–755. [Google Scholar] [CrossRef]
  247. Humbert, M.; Sitbon, O.; Simonneau, G. Treatment of pulmonary arterial hypertension. N. Engl. J. Med. 2004, 351, 1425–1436. [Google Scholar] [CrossRef] [PubMed]
  248. Galiè, N.; Humbert, M.; Vachiery, J.L.; Gibbs, S.; Lang, I.; Torbicki, A.; Simonneau, G.; Peacock, A.; Vonk Noordegraaf, A.; Beghetti, M.; et al. 2015 ESC/ERS Guidelines for the diagnosis and treatment of pulmonary hypertension: The Joint Task Force for the Diagnosis and Treatment of Pulmonary Hypertension of the European Society of Cardiology (ESC) and the European Respiratory Society (ERS): Endorsed by: Association for European Paediatric and Congenital Cardiology (AEPC), International Society for Heart and Lung Transplantation (ISHLT). Eur. Heart J. 2016, 37, 67–119. [Google Scholar] [PubMed]
  249. Ni, Z.; Bemanian, S.; Kivlighn, S.D.; Vaziri, N.D. Role of endothelin and nitric oxide imbalance in the pathogenesis of hypoxia-induced arterial hypertension. Kidney Int. 1998, 54, 188–192. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  250. Shimoda, L.A.; Laurie, S.S. Vascular remodeling in pulmonary hypertension. J. Mol. Med. 2013, 91, 297–309. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  251. Shenoy, V.; Ferreira, A.J.; Qi, Y.; Fraga-Silva, R.A.; Díez-Freire, C.; Dooies, A.; Jun, J.Y.; Sriramula, S.; Mariappan, N.; Pourang, D.; et al. 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. 2010, 182, 1065–1072. [Google Scholar] [CrossRef] [Green Version]
  252. Yuan, Y.M.; Luo, L.; Guo, Z.; Yang, M.; Ye, R.S.; Luo, C. Activation of renin-angiotensin-aldosterone system (RAAS) in the lung of smoking-induced pulmonary arterial hypertension (PAH) rats. J. Renin-Angiotensin-Aldosterone Syst. 2015, 16, 249–253. [Google Scholar] [CrossRef]
  253. Shen, H.; Zhang, J.; Wang, C.; Jain, P.P.; Xiong, M.; Shi, X.; Lei, Y.; Chen, S.; Yin, Q.; Thistlethwaite, P.A.; et al. MDM2-Mediated Ubiquitination of Angiotensin-Converting Enzyme 2 Contributes to the Development of Pulmonary Arterial Hypertension. Circulation 2020, 142, 1190–1204. [Google Scholar] [CrossRef]
  254. Dai, H.L.; Guo, Y.; Guang, X.F.; Xiao, Z.C.; Zhang, M.; Yin, X.L. The changes of serum angiotensin-converting enzyme 2 in patients with pulmonary arterial hypertension due to congenital heart disease. Cardiology 2013, 124, 208–212. [Google Scholar] [CrossRef]
  255. Dai, H.; Gong, Y.; Xiao, Z.; Guang, X.; Yin, X. Decreased levels of serum Angiotensin-(1-7) in patients with pulmonary arterial hypertension due to congenital heart disease. Int. J. Cardiol. 2014, 176, 1399–1401. [Google Scholar] [CrossRef]
  256. Malikova, E.; Galkova, K.; Vavrinec, P.; Vavrincova-Yaghi, D.; Kmecova, Z.; Krenek, P.; Klimas, J. Local and systemic renin-angiotensin system participates in cardiopulmonary-renal interactions in monocrotaline-induced pulmonary hypertension in the rat. Mol. Cell Biochem. 2016, 418, 147–157. [Google Scholar] [CrossRef] [PubMed]
  257. Martiniuc, C.; Braniste, A.; Braniste, T. Angiotensin converting enzyme inhibitors and pulmonary hypertension. Rev. Med. Chir. Soc. Med. Nat. Iasi. 2012, 116, 1016–1020. [Google Scholar]
  258. Zhang, J.; Dong, J.; Martin, M.; He, M.; Gongol, B.; Marin, T.L.; Chen, L.; Shi, X.; Yin, Y.; Shang, F.; et al. AMP-activated Protein Kinase Phosphorylation of Angiotensin-Converting Enzyme 2 in Endothelium Mitigates Pulmonary Hypertension. Am. J. Respir. Crit. Care Med. 2018, 198, 509–520. [Google Scholar] [CrossRef] [PubMed]
  259. Yamazato, Y.; Ferreira, A.J.; Hong, K.H.; Sriramula, S.; Francis, J.; Yamazato, M.; Yuan, L.; Bradford, C.N.; Shenoy, V.; Oh, S.P.; et al. Prevention of pulmonary hypertension by Angiotensin-converting enzyme 2 gene transfer. Hypertension 2009, 54, 365–371. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  260. Li, G.; Liu, Y.; Zhu, Y.; Liu, A.; Xu, Y.; Li, X.; Li, Z.; Su, J.; Sun, L. ACE2 activation confers endothelial protection and attenuates neointimal lesions in prevention of severe pulmonary arterial hypertension in rats. Lung 2013, 191, 327–336. [Google Scholar] [CrossRef]
  261. Ferreira, A.J.; Shenoy, V.; Yamazato, Y.; Sriramula, S.; Francis, J.; Yuan, L.; Castellano, R.K.; Ostrov, D.A.; Oh, S.P.; Katovich, M.J.; et al. Evidence for angiotensin-converting enzyme 2 as a therapeutic target for the prevention of pulmonary hypertension. Am. J. Respir. Crit. Care. 2009, 179, 1048–1054. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  262. Li, G.; Zhang, H.; Zhao, L.; Zhang, Y.; Yan, D.; Liu, Y. Angiotensin-converting enzyme 2 activation ameliorates pulmonary endothelial dysfunction in rats with pulmonary arterial hypertension through mediating phosphorylation of endothelial nitric oxide synthase. J. Am. Soc. Hypertens. 2017, 11, 842–852. [Google Scholar] [CrossRef]
  263. Yan, D.; Li, G.; Zhang, Y.; Liu, Y. Angiotensin-converting enzyme 2 activation suppresses pulmonary vascular remodeling by inducing apoptosis through the Hippo signaling pathway in rats with pulmonary arterial hypertension. Clin. Exp. Hypertens. 2019, 41, 589–598. [Google Scholar] [CrossRef]
  264. Haga, S.; Tsuchiya, H.; Hirai, T.; Hamano, T.; Mimori, A.; Ishizaka, Y. A novel ACE2 activator reduces monocrotaline-induced pulmonary hypertension by suppressing the JAK/STAT and TGF-β cascades with restored caveolin-1 expression. Exp. Lung Res. 2015, 41, 21–31. [Google Scholar] [CrossRef] [PubMed]
  265. Breitling, S.; Krauszman, A.; Parihar, R.; Walther, T.; Friedberg, M.K.; Kuebler, W.M. Dose-dependent, therapeutic potential of angiotensin-(1-7) for the treatment of pulmonary arterial hypertension. Pulm. Circ. 2015, 5, 649–657. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  266. Chen, L.; Xiao, J.; Li, Y.; Ma, H. Ang-(1-7) might prevent the development of monocrotaline induced pulmonary arterial hypertension in rats. Eur. Rev. Med. Pharmacol. Sci. 2011, 15, 1–7. [Google Scholar] [PubMed]
  267. A Dose-Escalation Study in Subjects With Pulmonary Arterial Hypertension (PAH). Available online: https://ClinicalTrials.gov/show/NCT03177603 (accessed on 23 November 2021).
  268. Recombinant Human Angiotensin-Converting Enzyme 2 (rhACE2) as a Treatment for Patients With COVID-19 (APN01-COVID-19). Available online: https://ClinicalTrials.gov/show/NCT04335136 (accessed on 23 November 2021).
  269. Combination of Recombinant Bacterial ACE2 Receptors -Like Enzyme of B38-CAP and Isotretinoin Could be Promising Treatment for COVID-19 Infection- and Its Inflammatory Complications. Available online: https://ClinicalTrials.gov/show/NCT04382950 (accessed on 23 November 2021).
  270. Recombinant Bacterial ACE2 Receptors -Like Enzyme of B38-CAP Could be Promising Treatment for COVID-19 Infection- and Its Inflammatory Complications Better Than Recombinant Human ACE2. Available online: https://ClinicalTrials.gov/show/NCT04375046 (accessed on 23 November 2021).
  271. Procko, E. The sequence of human ACE2 is suboptimal for binding the S spike protein of SARS coronavirus 2. bioRxiv 2020. [Google Scholar]
  272. A Study of SI-F019 in Healthy Participants. Available online: https://ClinicalTrials.gov/show/NCT04851444 (accessed on 23 November 2021).
  273. Angiotensin-(1,7) Treatment in COVID-19: The ATCO Trial. Available online: https://ClinicalTrials.gov/show/NCT04332666 (accessed on 23 November 2021).
  274. Angiotensin 1-7 as a Therapy in the Treatment of COVID-19. Available online: https://ClinicalTrials.gov/show/NCT04605887 (accessed on 23 November 2021).
  275. Angiotensin (1-7) for the Treatment of COVID-19 in Hospitalized Patients. Available online: https://ClinicalTrials.gov/show/NCT04570501 (accessed on 23 November 2021).
  276. Use of Angiotensin-(1-7) in COVID-19. Available online: https://ClinicalTrials.gov/show/NCT04633772 (accessed on 23 November 2021).
  277. Angiotensin-(1-7) in Peripheral Arterial Disease. Available online: https://ClinicalTrials.gov/show/NCT03240068 (accessed on 23 November 2021).
  278. Cardiovascular Effects of Angiotensin (1-7) in Essential Hypertension. Available online: https://ClinicalTrials.gov/show/NCT02245230 (accessed on 23 November 2021).
  279. Angiotensin 1-7 in Obesity Hypertension. Available online: https://ClinicalTrials.gov/show/NCT03604289 (accessed on 23 November 2021).
  280. Wysocki, J.; Ye, M.; Rodriguez, E.; González-Pacheco, F.R.; Barrios, C.; Evora, K.; Schuster, M.; Loibner, H.; Brosnihan, K.B.; Ferrario, C.M.; et al. Targeting the degradation of angiotensin II with recombinant angiotensin-converting enzyme 2: Prevention of angiotensin II-dependent hypertension. Hypertension 2010, 55, 90–98. [Google Scholar] [CrossRef]
  281. Hay, M.; Polt, R.; Heien, M.L.; Vanderah, T.W.; Largent-Milnes, T.M.; Rodgers, K.; Falk, T.; Bartlett, M.J.; Doyle, K.P.; Konhilas, J.P. A Novel Angiotensin-(1-7) Glycosylated Mas Receptor Agonist for Treating Vascular Cognitive Impairment and Inflammation-Related Memory Dysfunction. J. Pharmacol. Exp. Ther. 2019, 369, 9–25. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  282. Santos, S.H.; Giani, J.F.; Burghi, V.; Miquet, J.G.; Qadri, F.; Braga, J.F.; Todiras, M.; Kotnik, K.; Alenina, N.; Dominici, F.P.; et al. Oral administration of angiotensin-(1-7) ameliorates type 2 diabetes in rats. J. Mol. Med. 2014, 92, 255–265. [Google Scholar] [CrossRef]
  283. Fraga-Silva, R.A.; Costa-Fraga, F.P.; Savergnini, S.Q.; De Sousa, F.B.; Montecucco, F.; da Silva, D.; Sinisterra, R.D.; Mach, F.; Stergiopulos, N.; da Silva, R.F.; et al. An oral formulation of angiotensin-(1-7) reverses corpus cavernosum damages induced by hypercholesterolemia. J. Sex. Med. 2013, 10, 2430–2442. [Google Scholar] [CrossRef]
  284. Verma, A.; Xu, K.; Du, T.; Kulkarni, M.; Zhu, P.; Prasad, T.; Daniell, H.; Li, Q. Oral delivery of Angiotensin-(1-7) bioencapsulated in plant cells protect against diabetes-induced retinopathy and other complications. Investig. Ophthalmol. Vis. Sci. 2017, 58, 5213. [Google Scholar]
  285. Shenoy, V.; Kwon, K.C.; Rathinasabapathy, A.; Lin, S.; Jin, G.; Song, C.; Shil, P.; Nair, A.; Qi, Y.; Li, Q.; et al. Oral delivery of Angiotensin-converting enzyme 2 and Angiotensin-(1-7) bioencapsulated in plant cells attenuates pulmonary hypertension. Hypertension 2014, 64, 1248–1259. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  286. Shil, P.K.; Kwon, K.C.; Zhu, P.; Verma, A.; Daniell, H.; Li, Q. Oral delivery of ACE2/Ang-(1-7) bioencapsulated in plant cells protects against experimental uveitis and autoimmune uveoretinitis. Mol. Ther. 2014, 22, 2069–2082. [Google Scholar] [CrossRef] [Green Version]
  287. Dilauro, M.; Burns, K.D. Angiotensin-(1-7) and its effects in the kidney. Sci. World J. 2009, 9, 522–535. [Google Scholar] [CrossRef] [Green Version]
  288. Alghamri, M.S.; Morris, M.; Meszaros, J.G.; Elased, K.M.; Grobe, N. Novel role of aminopeptidase-A in angiotensin-(1-7) metabolism post myocardial infarction. Am. J. Physiol. Heart Circ. Physiol. 2014, 306, H1032–H1040. [Google Scholar] [CrossRef] [Green Version]
  289. Bodineau, L.; Frugière, A.; Marc, Y.; Inguimbert, N.; Fassot, C.; Balavoine, F.; Roques, B.; Llorens-Cortes, C. Orally active aminopeptidase A inhibitors reduce blood pressure: A new strategy for treating hypertension. Hypertension 2008, 51, 1318–1325. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  290. Ozhan, O.; Parlakpinar, H.; Acet, A. Comparison of the effects of losartan, captopril, angiotensin II type 2 receptor agonist compound 21, and MAS receptor agonist AVE 0991 on myocardial ischemia-reperfusion necrosis in rats. Fundam. Clin. Pharmacol. 2020. [Google Scholar] [CrossRef] [PubMed]
  291. Hong, L.; Wang, Q.; Chen, M.; Shi, J.; Guo, Y.; Liu, S.; Pan, R.; Yuan, X.; Jiang, S. Mas receptor activation attenuates allergic airway inflammation via inhibiting JNK/CCL2-induced macrophage recruitment. Biomed. Pharmacother. 2021, 137, 111365. [Google Scholar] [CrossRef] [PubMed]
Figure 1. RAS cleavage cascade and the principal receptors of Ang II and Ang(1-7).
Figure 1. RAS cleavage cascade and the principal receptors of Ang II and Ang(1-7).
Ijms 22 12800 g001
Figure 2. Selected molecular pathways affected by ACE2/Ang(1-7)/Mas axis in cardiovascular and pulmonary systems.
Figure 2. Selected molecular pathways affected by ACE2/Ang(1-7)/Mas axis in cardiovascular and pulmonary systems.
Ijms 22 12800 g002
Figure 3. The main conclusions of the overall ACE2/Ang(1-7)/Mas axis activity in cardiovascular and pulmonary hypoxic conditions.
Figure 3. The main conclusions of the overall ACE2/Ang(1-7)/Mas axis activity in cardiovascular and pulmonary hypoxic conditions.
Ijms 22 12800 g003
Table 1. Overview of ACE2/Ang(1-7)/Mas parameters alteration in CVS and pulmonary hypoxia models.
Table 1. Overview of ACE2/Ang(1-7)/Mas parameters alteration in CVS and pulmonary hypoxia models.
Hypoxic Condition Model Increased Parameter Decreased Parameter Species Reference
Acute hypoxia I/R injury (after ≤4 h) ↑ pulmonary Ang(1-7) and ACE2 mRNA mouse [94]
↑ renal ACE2 and Mas mRNA↓ rat [97]
I/R injury (after >4 h) ↓ plasma Ang(1-7), pulmonary ACE2 mouse [94]
↓ pulmonary Ang(1-7) mouse [95]
↑ renal Ang (1-7) ↓ renal ACE2, serum Ang(1-7) mouse [96]
↓ cardiac and hepatic ACE2 rat [83]
Myocardial infarction (after 4 weeks) ↑ cardiac (infarct zone) ACE2 and Mas rat [24]
↑ cardiac (infarct zone) ACE2 activity rat [84]
↑ cardiac (infarct zone) ACE2 ↓ cardiac (infarct and non-infarct) ACE2 mRNA mouse [85]
↑ cardiac (infarct zone) ACE2 mRNA ↓ plasma ACE2 and peri-infarct tissue ACE2 activity rat [86]
↑ cardiac (infarct zone) ACE2 protein and mRNA rat [87]
ARDS (SARS-CoV) ↓ ACE2 Vero E6 cells [103]
ARDS (H7N9 influenza A virus) ↓ pulmonary ACE2 mouse [104]
ARDS (various) ↑ plasma Ang(1-10)/Ang(1-9) ratio ↓ plasma ACE2, Ang(1-9), Ang (1-7)/Ang(1-9) ratio human [105]
ARDS (RSV) ↓ pulmonary ACE2 expression and activity human (age ≈ 2 years) [130]
Intermittent hypoxia Obstructive sleep apnoe ↓ plasma Ang(1-7) rat [132]
↓ plasma Ang(1-7), renal arteriole ACE2/ACE rat [135]
↓ plasma and renal Ang(1-7), renal arteriole ACE2/ACE rat [136]
↓ cardiac ACE2 mouse [138]
Subchronic intermittent lung hypoxia ↓ ACE2 mRNA MLECs [139]
Chronic intermittent lung hypoxia ↑ pulmonary ACE2 mRNA rat [140]
Chronic hypoxia Heart failure (early stage) ↑ plasma and cardiac Ang(1-7), Ang(1-9), cardiac ACE2 activity mouse [153]
↑ cardiac and renal ACE2 rat [154]
↑ plasma ACE and Ang(1-7), cardiac ACE2 mRNA rat [155]
↑ plasma Ang(1-7) rat [156]
↑ plasma ACE2 human [159]
↑ cardiac ACE2 mRNA rat [160]
↑ cardiac ACE2/ACE mRNA ratio human [169]
Heart failure (late stage) ↓ cardiac ACE2 activty mouse [153]
↑ plasma ACE2 human [159,167,168]
↑ cardiac ACE2 and Mas rat [159]
↓ plasma ACE2 rat [157]
↑ plasma ACE2 activity dog [158]
↓ cardiac ACE2 mRNA rat [160]
↑ cardiac ACE2 protein and mRNA ↓ cardiac ACE2/ACE mRNA ratio human [169]
↑ cardiac ACE2 mRNA ↓ cardiac Mas mRNA human [170]
↑ cardiac ACE2 mRNA human [171]
Atherosclerosis ↓ cardiac and renal ACE2 rabbit [181]
↓ aortic ACE2 activity mouse [182]
↓ carotid aortic ACE2 activity human [183]
Coronary artery disease ↑ plasma ACE2 human [178,179]
Chronic obstructive pulmonary disease/Pulmonary fibrosis ↓ pulmonary ACE2 mRNA Rat [207]
↑ bronchial ACE2 mRNA human [209]
↑ pulmonary ACE2 mRNA human [210]
↑ pulmonary ACE2 human [211]
↓ pulmonary ACE2 mRNA and enzyme activity human, mouse [226]
Pulmonary hypertension ↓ pulmonary ACE2 mRNA rat [251]
↓ pulmonary ACE2 rat [252]
↓ pulmonary ACE2 protein and mRNA human, mouse [253]
↓ plasma ACE2 human [254]
↓ plasma Ang(1-7) human [255]
↑ pulmonary ACE2/ACE mRNA rat [256]
↓ plasma Ang(1-7), ACE2 human [199]
↓ plasma ACE2 activity and Ang(1-7)/Ang II ratio human [206]
↑ increased parameter; ↓decreased parameter.
Table 2. Gene modulation and experimental ACE2/Ang(1-7)/Mas pharmacological treatment in hypoxic conditions.
Table 2. Gene modulation and experimental ACE2/Ang(1-7)/Mas pharmacological treatment in hypoxic conditions.
Hypoxic Condition Model/
Disease
Gene Modulation Model Pharmacological Treatment Species Ref.
Downregu-lation Upregulation Enzyme ACE2 Activator Mas Agonist ACE2 Inhibitor Mas Ant-Agonist
Acute hypoxia I/R injury (after 4 h) ACE2-Tg DIZE mouse [95]
I/R injury (after >4 h) ACE2-KO ACE2-Tg mouse [96]
ACE2-KO mouse [98]
Myocardial infarction (after 4 weeks) A779 rat [24]
compound 16 rat [84]
ACE2-KO mouse [85]
DIZE compound 16 rat [86]
ACE2-Tg A779 rat [88]
Ang(1-7) rat [89]
DIZE rat [90,91,92]
Acute respiratory distress syndrome rACE2 mouse [109]
rACE2 rat [110,129]
rACE2 human [125]
ACE2 shRNA rACE2 mouse [127]
ACE2-KO rACE2 mouse [128]
COVID-19 rhACE2 * human [126,268]
Intermittent hypoxia Obstructive sleep apnoe Ang(1-7) rat [137]
Subchronic intermittent lung hypoxia (MLECs) Ang(1-7) mouse [134]
Chronic intermittent lung hypoxia (in vivo) Ang(1-7) mouse [134]
Ang(1-7) rat [141]
Chronic hypoxia Heart failure (early stage) rhACE2 A779 mouse [163]
Heart failure (late stage) ACE2-Tg rat [161,162]
Atherosclerosis ACE2-Tg HUVECs [54]
ACE2-KO, Mas-KO mouse [184]
ACE2 siRNA rat [185]
ACE2-Tg A779 mouse [186]
ACE2-Tg A779 rabbit [187]
ACE2-Tg A779 THP-1 cells [188]
ACE2-Tg VSMCs [189]
DIZE mouse [190,191]
Ang(1-7) A779 mouse [192]
Chronic obstructive pulmonary disease/Pulmonary fibrosis ACE2-Tg rat [207]
Ang(1-7) mouse [208]
ACE2 mouse [226]
Pulmonary hypertension rhACE2 human [206]
Ang(1-7)-Tg PMVECs [229]
ACE2-Tg PASMCs [230]
rACE2 pig [231]
ACE2-Tg A779 rat [251]
ACE2-KO ACE2-Tg mouse [258]
ACE2-Tg mouse [259]
Resorcinol-naphthalein A779 rat [260]
XNT rat [261]
Resorcinol-
naphthalein
MLN4760 rat [262]
Resorcinol-
naphthalein
MLN4760 A779 rat [263]
NCP-2454 rat [264]
Ang(1-7) rat [25,265]
rhACE2 human [267]
* ongoing clinical trials.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Rajtik, T.; Galis, P.; Bartosova, L.; Paulis, L.; Goncalvesova, E.; Klimas, J. Alternative RAS in Various Hypoxic Conditions: From Myocardial Infarction to COVID-19. Int. J. Mol. Sci. 2021, 22, 12800. https://doi.org/10.3390/ijms222312800

AMA Style

Rajtik T, Galis P, Bartosova L, Paulis L, Goncalvesova E, Klimas J. Alternative RAS in Various Hypoxic Conditions: From Myocardial Infarction to COVID-19. International Journal of Molecular Sciences. 2021; 22(23):12800. https://doi.org/10.3390/ijms222312800

Chicago/Turabian Style

Rajtik, Tomas, Peter Galis, Linda Bartosova, Ludovit Paulis, Eva Goncalvesova, and Jan Klimas. 2021. "Alternative RAS in Various Hypoxic Conditions: From Myocardial Infarction to COVID-19" International Journal of Molecular Sciences 22, no. 23: 12800. https://doi.org/10.3390/ijms222312800

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop