Next Article in Journal
Effect of the pH on the Antibacterial Potential and Cytotoxicity of Different Plasma-Activated Liquids
Previous Article in Journal
The Role of Arrestin-1 Middle Loop in Rhodopsin Binding
Previous Article in Special Issue
A New Perspective on Cancer Therapy: Changing the Treaded Path?
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Renin–Angiotensin-Aldosterone System, Nitric Oxide, and Hydrogen Sulfide at the Crossroads of Hypertension and COVID-19: Racial Disparities and Outcomes

1
Department of Molecular, Cellular and Biomedical Sciences, Sophie Davis School of Biomedical Education, City University of New York School of Medicine, New York, NY 10031, USA
2
Graduate Program in Biology, City University of New York Graduate Center, New York, NY 10016, USA
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2022, 23(22), 13895; https://doi.org/10.3390/ijms232213895
Submission received: 22 September 2022 / Revised: 7 November 2022 / Accepted: 9 November 2022 / Published: 11 November 2022
(This article belongs to the Special Issue Nitric Oxide: Physiology, Pharmacology, and Therapeutic Applications)

Abstract

:
Coronavirus disease 2019 is caused by SARS-CoV-2 and is more severe in the elderly, racial minorities, and those with comorbidities such as hypertension and diabetes. These pathologies are often controlled with medications involving the renin–angiotensin–aldosterone system (RAAS). RAAS is an endocrine system involved in maintaining blood pressure and blood volume through components of the system. SARS-CoV-2 enters the cells through ACE2, a membrane-bound protein related to RAAS. Therefore, the use of RAAS inhibitors could worsen the severity of COVID-19’s symptoms, especially amongst those with pre-existing comorbidities. Although a vaccine is currently available to prevent and reduce the symptom severity of COVID-19, other options, such as nitric oxide and hydrogen sulfide, may also have utility to prevent and treat this virus.

1. Introduction

Coronavirus disease 2019 (COVID-19) has affected the world and is caused by severe acute respiratory coronavirus 2 (SARS-CoV-2). COVID-19 is primarily transmitted through respiratory droplets [1] and presents a broad range of symptoms. Clinical manifestations of this disease can present either asymptomatically or as flu-like symptoms, along with loss of taste and smell [2]. The most severe manifestations of the virus occur amongst the elderly and those with comorbidities, such as hypertension, diabetes mellitus, and other immunosuppressed states [3]. In addition, studies have shown that COVID-19 has disproportionately affected racial minorities [4,5]. The renin–angiotensin–aldosterone system (RAAS) is pathologically activated during these states. Usually, the RAAS plays a significant role in regulating blood pressure and volume and systemic vascular resistance [6]. Therefore, pathological activation of the RAAS results in excessive vasoconstriction and can cause hypertension and renal and cardiovascular disease [7]. One of the central RAAS regulators that plays a specific role in inducing endothelial dysfunction, vasoconstriction, and eventual fibrosis is angiotensin II [8]. To prevent angiotensin II from causing pathologies such as hypertension, angiotensin-converting enzyme (ACE) inhibitors and angiotensin receptor blockers (ARB) are prescribed [8].
It has been found that SARS-CoV-2 has access to the cells through an RAAS-related protein, ACE2. SARS-CoV-2 uses ACE2, which is present as a transmembrane and soluble form and acts as a receptor for the virus to enter the body [9]. ACE2 is often upregulated when comorbidities are present [10,11].
Even though people with comorbidities often use RAAS inhibitors to manage their underlying condition, their use could impact the severity of COVID-19 symptoms. Therefore, alternatives such as nitric oxide and hydrogen sulfide to treat and prevent COVID-19 are of significant interest.
In this article, we represent an overview of the role of RAAS in hypertension and the role of nitric oxide and hydrogen sulfide in COVID-19. We also look at racial disparities and view the clinical outcomes of COVID-19 associated with these disparities.

2. Renin–Angiotensin–Aldosterone System

The RAAS is a complex endocrine system that is critical in maintaining blood pressure through balancing electrolyte levels and blood volume [12]. The three main components of the hormonal system are renin, angiotensin II, and aldosterone [13]. Even though the baroreceptor mechanism is pertinent in managing short-term and sudden changes in blood pressure, the RAAS system is seen in more chronic, long-term scenarios [14]. In response to low blood pressure, the juxtaglomerular apparatus of the kidneys become activated and cleave the constitutively secreted proenzyme prorenin into its active form, renin [15]. Once renin is in the bloodstream, it is able to act on angiotensinogen, a protein that is produced in the liver and fat cells, and cleave it into angiotensin I [16]. Angiotensin I is then converted into angiotensin II through the angiotensin-converting enzyme (ACE), found primarily in the vascular endothelium of the lungs. Angiotensin II is considered to be the main RAAS agonist and acts on angiotensin II receptors, such as the AT1R, throughout the body [13]. Some of the roles of angiotensin II include acting as a vasoconstrictor through its effects on vascular smooth muscle, increasing heart rate and contractility through activation of the sympathetic nervous system, and stimulating the zona glomerulosa cells of the adrenal cortex to produce and secrete the hormone aldosterone [17]. The role of aldosterone is to activate the epithelial sodium channels (ENaC) found in the distal nephron and increase sodium and water reabsorption [18]. Through these mediators, the RAAS is able to regulate blood pressure. However, when the RAAS complex is pathologically activated, it results in excessive vasoconstriction, muscular hypertrophy within the cardiac and vascular systems, and fibrosis [19]. The imbalance and dysregulation of the RAAS is seen in various pathologies, such as hypertension, congestive heart failure, and diabetic nephropathy [20].

Hypertension and RAAS Inhibitors

According to the World Health Organization, hypertension is one of the most common causes of premature death globally [21]. Worldwide, hypertension affects about one billion people [22]. Hypertension is dangerous because it causes a variety of complications, including heart disease, kidney dysfunction, stroke, and transient ischemic attacks. Since the net effects of the RAAS result in vasoconstriction, increased sodium and water retention, and increased myocardial contraction, all of these effects increase the effective circulating volume and thus increase the arterial blood pressure. When consistently activated, as seen in pathological situations, it results in hypertension [23]. For that reason, RAAS inhibitors, such as direct renin inhibitors, ACE inhibitors, ARBs, and aldosterone antagonists, are often prescribed in the treatment of hypertension [20].

3. COVID-19

Human coronaviruses commonly cause the common cold and are considered to cause relatively mild infections [24]. However, there have been three major coronavirus outbreaks within the last three decades [25], the latest being coronavirus disease 2019 (COVID-19), caused by severe acute respiratory coronavirus 2 (SARS-CoV-2). The novel SARS-CoV-2 was first isolated on 7 January 2020 [26] from a series of pneumonia cases in Wuhan City, China [27]. It has infected more than 595 million people across the globe and caused 6.4 million deaths [28]. According to the World Health Organization, the virus spreads through respiratory droplets. This includes droplets of saliva and or discharge through the nose when a person coughs, sneezes, or talks. The highest viral loads were found in the nose and throat after symptom onset [29]. The clinical manifestations of COVID-19 include but are not limited to fever, cough, fatigue, muscle aches, and loss of taste and smell [1]. Even though not everyone who contracts the COVID-19 virus is symptomatic, those who are asymptomatic are found to have similar viral load contents to those of symptomatic patients [29]. According to the Centers for Disease Control and Prevention (CDC), although COVID-19 is relatively mild in most cases, the severe and fatal cases are seen among the elderly and those with underlying medical conditions, including cardiovascular disease, hypertension, and diabetes.
COVID-19 presents with flu-like symptoms in most cases and, in more severe cases, can present with respiratory complications, cytokine storms, abnormal coagulopathy, and extensive endothelial damage. Between 5 and 20% of hospitalized COVID-19 patients require ICU-level care [30]. Severe cases can lead to acute respiratory distress syndrome (ARDS), associated with acute onset hypoxemia, edema, and bilateral lung infiltrate [31]. This respiratory compromise often necessitates the use of mechanical ventilation (MV), which comes with risks of secondary infection [32] and exacerbation of lung injury [33]. Severe COVID-19 complications largely arise due to a hyperinflammatory response [34,35], which leads to extensive inflammatory damage to the pulmonary and vascular systems [36,37]. In addition to respiratory complications and uncontrolled inflammation, COVID-19 is notable for resulting in elevated markers of coagulopathy and thrombotic complications, including pulmonary embolism and disseminated intravascular coagulation in severe cases [38,39,40,41,42]. These vascular complications are reflective of extensive damage to the vascular endothelium, which results in a shift to a prothrombotic state [43]. Endothelial damage results from both direct infection of endothelial cells by SARS-CoV-2 [44,45,46] and inflammatory damage from its characteristic ‘cytokine storm’ [47]. In fact, inflammation and endothelial dysfunction are common factors between various comorbidities that place individuals at risk of poorer outcomes from COVID-19, which include diabetes and hypertension [48,49,50,51]. Thus, successful management of COVID-19 requires antiviral therapy early in the course of the infection and attention to mitigating respiratory distress, resolving inflammation, and maintaining vascular integrity.

COVID-19 and RAAS

People with diabetes or hypertension are often prescribed RAAS inhibitors to manage their diseases and prevent complications, such as diabetic nephropathy, cardiovascular disease, and mortality [52]. Even though these medications are beneficial for several indications, it is unclear whether or not they are exacerbating harm in COVID-19 patients or if they are improving their outcomes.
SARS-CoV-2 is able to enter the cells through the membrane-bound ACE2 protein in which it uses the protein as a receptor [9] (Figure 1). ACE2 is a protein that exists in two forms: a transmembrane protein and a soluble form. The transmembrane protein form of ACE2 is found and expressed mainly in the type II pneumocytes within the alveolar epithelium of the lung, as well as in the epithelial cells of the intestine, kidney, and blood vessels [10]. Meanwhile, the soluble form of ACE2 is found in very low concentrations within the body but could be increased in pathological states such as hypertension and diabetes [11]. All of these locations could be potential entry points for the SARS-CoV-2 virus [53]. This binding between SARS-CoV-2 and ACE2 is accomplished through the virus’s spike protein. The spike protein associates with the extracellular domain of ACE2, stimulating a clathrin-dependent endocytosis of the entire complex and thus allowing entry into the cell [54]. Aside from the actual binding of the virus to the ACE2 receptor, SARS-CoV-2’s spike protein is also primed by the host serine protease transmembrane, serine protease-2 (TMPRSS2) before entry into the cell [55] (Figure 1). The membranes can fuse through this interaction between the spike protein and the TMPRSS2 protease, which further permits the virus to enter the body [56]. ACE2 cleaves angiotensin II into angiotensin (1-7), which has vasodilatory, anti-inflammatory, and antifibrotic effects through its binding with the Mas receptor [57]. Ang-(1-7) demonstrates opposing effects with that of angiotensin II when bound to the G-protein-coupled receptor Mas. Through this interaction, vasodilatory mediators, such as nitric oxide and prostaglandins, are released. In addition to vasodilatory effects, Ang-(1-7) also opposes angiotensin II’s vasoconstrictor and pro-inflammatory effects, which is seen when Ang II is bound to its receptor, AT1R [58]. Therefore, Ang-(1-7) could reduce inflammatory tissue damage and has been suggested as a potential treatment for the detrimental effects demonstrated by SARS-CoV-2 [59]. Essentially, ACE2’s function opposes the physiologic role of ACE and thus maintains a balance between pro-inflammatory and pro-fibrotic activity with anti-inflammatory and antifibrotic activity [60].
RAAS inhibitors affect the balance between pro and anti-inflammation and fibrosis, and there is an increased shift towards the pro-inflammatory and pro-fibrotic effects [61]. RAAS inhibition results in the upregulation of angiotensin-converting enzyme 2 (ACE2) expression on the cell surface, which allows the entry of SARS-CoV-2 [62]. As a result of this increased expression of ACE2, it may be likely that SARS-CoV-2 could enter the body more readily when there are more ACE2 receptors present [63]. Unfortunately, COVID-19 patients with underlying conditions such as hypertension and diabetes who are treated with RAAS inhibitors have been reported to have the highest rate of fatality by the COVID-19 virus [64]. These medications increase the virus’s severity and increase mortality by twofold [65]. Since hypertensive and diabetic patients already have a hyperactive Ang II-AT1 axis due to the pathologically overactive RAAS complex, when SARS-CoV-2 binds and downregulates the production of functional ACE2, it is more likely for these patients to develop severe conditions from the virus [66]. In these situations, if the body’s immune system is unable to fight against and defeat the infection, SARS-CoV-2 will be able to replicate after binding to ACE2 and further destroy the host’s cells. This results in an inability to inactivate Ang II’s signaling with its AT1 receptor and thus exacerbates lung infection and inflammatory mechanisms associated with angiotensin II [67]. Since SARS-CoV-2 binds to and downregulates ACE2, Ang II is not metabolized to Ang-(1-7); as a result, there are fewer anti-inflammatory and vasodilatory effects and more inflammation, fibrosis, oxidative stress, and vasoconstriction present [68].
Using RAAS inhibitors while treating COVID-19 is controversial because, aside from the harmful effects associated with their usage, data also show that ACE2 could have protective measures against COVID-19. After a person gets infected with COVID-19, their ACE2 levels diminish, which results in unregulated angiotensin II destroying tissues [10]. ACE inhibitors would be protective in this case because through RAAS inhibition, there would be a decreased concentration of angiotensin II, which promotes inflammatory responses and fibrosis within the lung among SARS-CoV-2 entry and binding to ACE2 [62]. This is so because, through the downregulation of ACE2 by SARS-CoV-2, the degradation of Ang II into Ang-(1-7) is decreased, which allows further destruction of tissues due to the increased levels of Ang II present in the body [55]. In addition, ARBs have been found to be beneficial in treating COVID-19 as well. This is due to ARB’s ability to upregulate ACE2 expression, resulting in increased levels of the protective mediator Ang-(1-7). Since ARBs directly inhibit angiotensin II’s activity on its receptor, they inhibit angiotensin II from its inflammatory and vasoconstricting effects [11]. Clinical trials have indicated another way in which RAAS inhibitors could provide protective effects against COVID-19; both ACE inhibitors and ARBs decrease the viral load and prevent peripheral T-cell depletion, which could help reduce the severity of the virus [69]. Although there may be either detrimental or protective effects associated with taking RAAS inhibitors while being treated for COVID-19, multiple societies such as the American College of Cardiology, the American Heart Association, and the International Society for Hypertension all agree that due to the inconclusive information and the current data available on the effects of RAAS inhibitors and COVID-19, patients taking these medications are not advised to change therapy and should continue with their normal treatment regimen [70].

4. Nitric Oxide and Its Function in COVID-19

Nitric oxide (NO) is one of three known gasotransmitters with critical roles in cardiovascular homeostasis and the immune response, earning it significant interest as a potential management option for COVID-19 [72,73,74,75]. It is endogenously produced from L-arginine by three isoforms of the enzyme nitric oxide synthase (NOS): neuronal NOS (nNOS or NOS-1), inducible NOS (iNOS or NOS-2), and endothelial NOS (eNOS or NOS-3) [76]. nNOS is known for its role in neurotransmission and is constitutively expressed in the body alongside eNOS, which is crucial in maintaining the vasculature through vasodilation and inhibition of platelet and leukocyte adhesion [77]. iNOS is an inducible isoform that becomes critical in host defense against pathogens [77]. eNOS and iNOS have received significant attention regarding their roles in COVID-19 severity [76,78,79,80]. Dysregulation of eNOS and decreased endothelial NO bioavailability can be seen in chronic inflammatory diseases such as hypertension and may be critical in predisposing individuals to higher severity of COVID-19 [81]. Dysregulation of iNOS can occur in a hyperinflammatory response to an invading pathogen such as SARS-CoV-2 and may lead to inflammatory damage mediated by excessive production of superoxide anions and peroxynitrite [82,83,84]. SARS-CoV-2 also contributes to NO deficiency via disruption of RAAS by downregulating ACE2 and angiotensin 1–7, a metabolite that promotes NO synthesis and release [58,85]. Additionally, COVID-19 has been linked to reduced arginine bioavailability, [86,87], and arginine supplementation has been found to reduce hospitalization duration with severe COVID-19 [88]. In individuals who already have compromised eNOS function, as would be the case for people with hypertension [83,84], infection with SARS-CoV-2, which causes further endothelial damage, hyperinflammation, and RAAS imbalance, may be particularly disastrous. Therefore, restoration of NO to such individuals may be highly beneficial for management and preventing progression of symptoms.
Before the establishment of infection, NO is of great interest in its capacity for early-stage prevention of COVID-19 infection. Studies using NO donors, experimental iNOS induction, and iNOS transfection have demonstrated a broad range of antiviral activity of both exogenously administered and endogenously produced NO [89,90,91,92,93,94,95,96,97,98,99,100,101]. These antiviral actions have been confirmed with SARS-CoV-2 (Figure 2), against which NO acts to inhibit viral replication and reduce viral load, demonstrated in vitro using NO donor S-nitroso-L-acetyl penicillamine (SNAP) and in a phase 2 clinical trial of nitric oxide nasal spray (NONS) [102,103]. Of note, synthetic dinitrosyl iron complexes, the innate version of which are important as storage and transportation for endogenous nitric oxide [104], have been identified as a promising protease inhibitor with potential in suppressing SARS-CoV-2 [105,106]. In individuals at higher risk of infection, including those with hypertension, the use of NO in the early stages may provide promise for preventing or curtailing SARS-CoV-2 infection.
NO is also a vasodilator, bronchodilator, and anti-inflammatory agent, which protects the vascular endothelium, thereby holding promise for managing later stages of COVID-19. As a broncho- and vasodilator, inhaled NO (iNO) is a potential rescue treatment for respiratory compromise (Figure 2) and has been found to improve oxygenation, reduce pulmonary vascular resistance, and decrease the need for invasive respiratory support among COVID-19 patients [107,108,109]. Additionally, NO therapies may prove important in containing the inflammatory damage responsible for the respiratory and cardiovascular injury that occurs in severe COVID-19 cases. Increasing NO bioavailability was found to curtail the production of pro-inflammatory cytokines, reduce leukocytic infiltration, and decrease neutrophil activation and adhesion in sepsis-induced neutrophils and in a rabbit model of lavage-induced ARDS [110,111] (Figure 2). Exogenously administered NO both restores NO to the endothelium, where it inhibits leukocyte adhesion and prevents further endothelial damage [112], and can exert negative feedback regulation on excessive iNOS activity [113], limiting the production of damaging byproducts such as superoxide anions and peroxynitrite [114] (Figure 2). Additionally, there is an extensive interplay between the RAAS and NO (Figure 3). Ang-(1-7), a component of the non-classical RAAS pathway, has been found to increase NO bioavailability by stimulating constitutive NOS activity [58,115,116] while curtailing excessive iNOS activity [117,118] and by reducing superoxide anion production (thereby attenuating NO consumption to form damaging products such as peroxynitrite) [119,120]. On the other hand, classical RAAS metabolite Ang II reduces NO signaling by inhibiting its receptor soluble guanylyl cyclase (sGC) [121], by increasing the breakdown of its downstream signaling molecule cyclic guanosine monophosphate (cGMP) through stimulation of phosphodiesterase 1A1 (PDE1A1) [122], and by promoting NOS uncoupling through increased production of superoxide anions from nicotinamide adenine dinucleotide phosphate hydrogen (NADPH) oxidase [123,124,125]. Classical RAAS overactivation thus would result in net decrease in the protective effects of NO, an effect that would be exacerbated by infection with SARS-CoV-2. Restoration of NO may thus prove a promising approach, and, in fact, there is evidence that NO inhibits ACE [126,127,128] and downregulates Ang II type I receptors (AT1R) [129,130], thereby reducing the activity of the pro-inflammatory arm of the RAAS. These actions are especially relevant in mitigating COVID-19 severity in individuals with an imbalance in the RAAS, who are indeed already predisposed to adverse outcomes from the virus. NO is also protective towards the vascular endothelium, which is especially important both in COVID-19 and its predisposing comorbidities such as hypertension which are notable for endothelial dysfunction [131]. Endothelial damage is characterized by insufficient eNOS activity and endothelial bioavailability of NO and is a major factor both in hypertension and in COVID-19 case severity [79]. Endothelial damage results in a shift to a prothrombotic and vasoconstrictive state that can be countered by restoration of NO to the endothelium. Augmentation of NO bioavailability using NO-releasing aspirin (NCX 4215) and enhancement of eNOS activity has in fact been shown to reduce neutrophil and platelet activation and aggregation in vitro and ex vivo [110,132]. In addition, nNOS-derived NO deficiency has been implicated in cardiomyocyte arrhythmogenesis [133], as has RAAS dysfunction [134]. It has been suggested that deficient NO-mediated S-nitrosylation of ryanodine receptors potentially mediates calcium-triggered ventricular arrhythmias [133,135]. Thus, it is possible that this additional comorbidity may benefit from NO therapies in the context of RAAS dysfunction and COVID-19.
The ability of NO to prevent and limit infection, ameliorate pulmonary complications, resolve inflammation, restore RAAS balance, and protect the endothelium makes NO therapy an approach worth investigating for prevention and later-stage management of COVID-19. Hypertensive and diabetic patients, as discussed previously, are at increased risk of severe COVID-19 infection, are more susceptible to the accompanying hyperinflammatory response, and come with a history of existing endothelial damage. In these patients, using NO therapies, including iNO, NO donor molecules, and even dietary nitrates for prevention, may be a promising option.

5. Hydrogen Sulfide and Its Function in COVID-19

H2S was the last of the gasotransmitters to be discovered. It shares many of its properties with those previously discussed for NO—an antiviral, anti-inflammatory agent protective towards the lungs and vascular endothelium [136]. H2S is endogenously produced from the amino acid L-cysteine by a group of four enzymes constituting three enzymatic pathways: cystathionine γ-lyase (CSE), cystathionine β-synthase (CBS), and 3-mercaptopyruvate sulfurtransferase (3-MST) in conjunction with cysteine aminotransferase (CAT) [137]. Although underexplored in comparison to NO, the evidence suggests that H2S therapy may be an avenue worth investigating for future use.
H2S has a broad range of antiviral activity against enveloped RNA viruses belonging to orthomyxoviridae, filoviridae, flaviviridae, bunyaviridae, and paramyxoviridae families through inhibition of viral entry, replication, and assembly/release [138,139], properties that may transfer to SARS-CoV-2 (Figure 2). H2S administration using both fast and slow H2S donors has also been reported to downregulate the expression of TMPRSS2 in human airway epithelial cells [140], which is vital in the entry of SARS-CoV-2 into the host cell [56]. By this mechanism, H2S may prevent SARS-CoV-2 infection of the host. In addition, upregulation of glutathione (GSH) by H2S [141] may be another mechanism of inhibiting SARS-CoV-2 infection through both its antioxidant properties and possibly direct interference with the actions of the ACE2 and TMPRSS2 proteins [142]. N-acetylcysteine (NAC) is a compound that yields GSH [143] as well as increased H2S levels [144], and has been suggested as a therapy for the management of COVID-19 [145]. In one study, while NAC was unsuccessful in inhibiting cell fusion of SARS-CoV-2, its stronger derivative NACA inhibited S protein binding to ACE2 and viral entry in vitro and in mice transfected with human ACE2 and TMPRSS2 [146].
Later-stage management of COVID-19 symptoms may also be possible using H2S therapies, owing to its actions as a bronchodilating, anti-inflammatory, and endothelium protective agent. H2S donors have demonstrated the ability to increase oxygenation, ameliorate ALI, and reduce VILI in models of pulmonary damage [147,148] (Figure 2). These findings appear to translate to studies with COVID-19 patients administered with NAC, which have demonstrated improved oxygenation, reduced lung damage and inflammatory markers, and decreased need for respirators [149,150]. Additionally, as already mentioned, SARS-CoV-2 infection downregulates the ACE2 protein [151], which disturbs the balance between the pro- and anti-inflammatory arms of the RAAS, causing a shift to a vasoconstrictive state and resulting in pulmonary damage [152,153]. Restoration of ACE2 after infection may be significant in reducing inflammatory damage and protecting against lung injury, having demonstrated these protective actions in one murine model of sepsis-induced severe acute lung injury [154]. In fact, Ang II has been found to inhibit CSE [155]; therefore, restoring ACE2 and reducing Ang II levels may also serve to restore the protective function of H2S (Figure 3). Considering the findings of one study in which administration of NaHS upregulated ACE2 expression in the endothelial cells of atherosclerotic mice [156], it is possible that H2S administration post-infection with SARS-CoV-2 may restore the RAAS balance and enforce its own restoration and correspondingly play a role in protecting against lung injury. Additionally, there is evidence that H2S attenuates the pathological overactivity of the classical RAAS pathway [157] by inhibiting ACE activity [158], downregulating AT1R [159,160], and inhibiting renin activity [161] (Figure 3). Thus, restoration of H2S may work against the inflammatory and vasoconstrictive shift that occurs both in hypertension and in COVID-19. H2S also has a protective function in the vascular endothelium, making it important for vascular complications arising from endothelial dysfunction. H2S contributes to vasodilation, inhibits leukocyte and platelet adhesion to the endothelium, and increases NO bioavailability in the endothelium by increasing eNOS activity [162] (Figure 2). H2S depletion yields endothelial dysfunction, and restoration of H2S to the deficient endothelium can decrease thrombosis while increasing thrombolysis and reducing endothelial cell apoptosis [163,164,165,166]. H2S also demonstrates crosstalk with NO, curtailing excessive iNOS activity [167,168] while upregulating eNOS activity [169,170] and reducing superoxide levels, further increasing NO bioavailability [171] (Figure 3). This action provides further reason to investigate H2S use in COVID-19 patients, considering the dysregulation of both NOS isoforms that may play a role in severe COVID-19 cases.
Similar to NO, H2S shows promise in the prevention and in management of severe COVID-19 infection. Taking advantage of extensive feedback between the two gasotransmitters may also be a possibility when considering potential therapies. Making these therapies available to the general public may be especially important in reducing the rate of negative outcomes among populations already vulnerable to the burden of the pandemic.

6. Racial Disparities and COVID-19 Outcomes

Hypertension has been highly prevalent among the black community [172]. It is hypothesized that the black population faces a higher incidence of hypertension than other races because of increased stress and lack of compliance to medication seen within this population [173]. Although hypertension is more prominent among black people, there is data demonstrating that some antihypertensive drugs, such as ACE inhibitors and ARBs, are less effective and have increased consequences when given to black people than to individuals of other races [174]. Some reasons this is the case in blacks and not in other races could be genetic variants and mutations. For example, many ARBs are metabolized through the cytochrome P450 2C9, and for some reason, that cytochrome is less common in blacks than in whites [175]. In addition, there is data showing that blacks have naturally low renin levels due to genetic mutations, making ACE inhibitors and ARBs more resistant in this population due to RAAS deactivation through its negative feedback mechanism [176]. Due to these reasons, not only are ACE inhibitors and ARBs less effective in blacks, but they have been shown to have adverse effects as well. A recent cohort study evaluating the cardiovascular outcomes associated with ACE inhibitor use among white and black patients has demonstrated that the black patients taking the ACE inhibitors dealt with increased cardiovascular outcomes, such as nonfatal myocardial infarction and nonfatal stroke, compared to white patients [177]. Another cohort study observed the outcomes in black hypertensive patients taking ACE inhibitors and compared them to other antihypertensive drugs, such as calcium channel blockers (CCB), thiazide diuretics, and beta blockers. This study showed an increase in myocardial infarction, stroke, and death among the black hypertensive patients taking ACE inhibitors compared with the patients who took CCB, thiazide diuretics, or beta-blockers [178]. Not only are cardiovascular outcomes an adverse effect in black patients taking ACE inhibitors and ARBs, but black patients are also more likely to experience angioedema when taking ACE inhibitors than other races. Since the mechanism behind angioedema is primarily a result of increased bradykinin levels, ACE inhibitors are the drugs associated with this side effect, not ARBs [179]. A prospective study demonstrated that ACE inhibitors increase the risk of developing angioedema in blacks by threefold compared with whites [180].
Ethnic minorities have been one of the groups to have been hit the hardest by the COVID-19 virus. Several factors, such as lifestyle conditions, have contributed to this cause. Chronic diseases such as diabetes, hypertension, and obesity are more prevalent in black people than in white people, and unfortunately, these conditions have been associated with increased severity of COVID-19. Not only are chronic health conditions more common in ethnic minorities, but these populations are more likely to live in crowded areas, use more public transportation, and work jobs that pay less and do not provide sick days [181]. The ethnic minority group, especially in the United States, makes up a significant portion of “essential workers”, which includes supermarket workers, health-care workers, and public transportation workers [182]. All of these factors increase the likelihood of being exposed to the virus. Aside from these lifestyle factors that have been linked with an increase in COVID-19 exposure amongst this demographic of people, medication, such as RAAS inhibitors, could also play a role in acquiring this virus. Even though ACE inhibitors and ARBs are contraindicated as first-line monotherapy treatments in blacks, they are still used in a combination regimen with other antihypertensive drugs such as calcium channel blockers and thiazide diuretics [174]. Since RAAS inhibitors are being used as a combination therapy, they still contribute to the negative side effects seen in black people. Not only would they contribute to adverse effects, but RAAS inhibitors even in combination with other drugs, would continue to up regulate the ACE2 protein, which would increase the risk of the SARS-CoV-2 virus entering the body According to statistics from April 2020, the ethnic minority population makes up the most cases of COVID-19 in both Europe and the United States. Based on the data from the UK’s Intensive Care National Audit and Research Centre, about 5000 people were severely ill in April from COVID-19 in England, Wales, and Northern Ireland. Of those 5000 cases, 34% of the cases were from a minority background even though those ethnic groups only make up about 14% of the total population within those regions. Meanwhile, in the United States, according to the data released by the CDC from 18 April 2020, among 120,000 cases of COVID-19, 36% of those cases were non-white people, who make up around 23% of the United States population. Most cases were by the black and African American groups, who made up 30% of the cases even though they only represented 13% of the total population [183].

7. Summary and Perspectives

H2S is a gas that has various physiological functions and benefits, including cell survival and anti-inflammatory actions [184]. Studies show that H2S could portray protective properties against COVID-19. It has been demonstrated that H2S can suppress the transcription of TMPRSS2, the protease involved in the priming of the coronavirus’ spike protein to allow cell entry [185]. H2S also inhibits RNA viral replications within the lungs of several RNA viruses, such as the influenza virus and Ebola [138]. Therefore, it suggested that H2S could inhibit the coronavirus’ RNA viral replication. H2S’s mode of action for inhibiting these viral replications is through the downregulation of the viral proteins and the syncytium formation and virus assembly [139]. Exogenous H2S has protective properties in the lungs and has been effective in reversing lung damage and inflammation, as seen in COVID-19. Exogenous H2S could be given through N-acetylcysteine (NAC), a supplemental form of the amino acid cysteine. NAC is not only an antioxidant but also releases H2S and can be used to protect organs, such as the lungs, heart, and kidneys, from COVID-19’s cytokine storm [186].
NO, a vasodilator produced by endothelial cells, is another mediator that could potentially be used to treat COVID-19. NO synthesis and release are promoted by Ang-(1-7), a metabolite produced by ACE2 [187]. Since SARS-CoV-2’s spike protein binds to ACE2, it affects the production of nitric oxide through the decreased production of Ang-(1-7) [72]. Therefore, exogenous nitric oxide could benefit this situation because of its anti-inflammatory effects. NO has been associated with protective measures, especially in patients with acute respiratory distress system (ARDS), which is often a consequence of the COVID-19 virus. Not only is nitric oxide able to inhibit the cytokine storm present in the coronavirus, but it also improves oxygenation of the arteries and inhibits pulmonary hypertension through its vasodilatory actions within the lungs [188]. Supplemental nitric oxide could be administered through several routes, such as inhaled nitric oxide and donor compounds, to help combat the detrimental effects of this virus [189]. Studies have shown that the nitric oxide donor, S-nitroso-N-acetylpenicillamine (SNAP), has been able to interfere with the fusion of the SARS-CoV spike protein with its receptor as well as diminish the virus’ RNA synthesis [75].
It is crucial to have other options available for treating and potentially preventing COVID-19. Although vaccines are available since they are so new, their efficacy and long-term side effects are still unknown. Therefore, aside from unknown side effects from the COVID-19 vaccines, it would be essential to have alternative options for preventing and treating this virus, especially in individuals who refuse to receive the vaccine, to ensure their protection.

Author Contributions

Conceptualized by K.K.; literature review and article writing by T.R., P.P.O. and K.K. All authors have read and agreed to the published version of the manuscript.

Funding

Supported in part by the National Institutes of Health [R24 DA018055; R01GM123508] and the Professional Staff Congress-City University of New York (PSC-CUNY) [TRADB-49-271].

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

3-MST3-mercaptopyruvate sulfurtransferase
ACE2angiotensin-converting enzyme 2
ALIacute lung injury
Angangiotensin
ARBangiotensin receptor blockers
ARDSacute respiratory distress syndrome
CATcysteine aminotransferase
CBScystathionine β-synthase
cGMPcyclic guanosine monophosphate
COPDchronic obstructive pulmonary disease
COVID-19coronavirus disease 2019
CSEcystathionine γ-lyase
eNOSendothelial nitric oxide synthase
GSHglutathione
H2Shydrogen sulfide
iH2Sinhaled hydrogen sulfide
ILinterleukin
iNOinhaled nitric oxide
iNOSinducible nitric oxide synthase
MVmechanical ventilation
NACN-acetylcysteine
nNOSneuronal nitric oxide synthase
NOnitric oxide
PDE5phosphodiesterase 5
RAASrenin–angiotensin–aldosterone system
ROSreactive oxygen species
S proteinspike protein
SARSsevere acute respiratory syndrome
SARS-CoV-2severe acute respiratory syndrome coronavirus 2
sGCsoluble guanylyl cyclase
SNPsodium nitroprusside
SODsuperoxide dismutase
TMPRSS2transmembrane serine protease 2

References

  1. Huang, C.; Wang, Y.; Li, X.; Ren, L.; Zhao, J.; Hu, Y.; Zhang, L.; Fan, G.; Xu, J.; Gu, X.; et al. Clinical features of patients infected with 2019 novel coronavirus in Wuhan, China. Lancet 2020, 395, 497–506. [Google Scholar] [CrossRef] [Green Version]
  2. Godeau, D.; Petit, A.; Richard, I.; Roquelaure, Y.; Descatha, A. Return-to-work, disabilities and occupational health in the age of COVID-19. Scand. J. Work Environ. Health 2021, 47, 408–409. [Google Scholar] [CrossRef] [PubMed]
  3. CDC. About COVID-19: People with Certain Medical Conditions. Available online: https://www.cdc.gov/coronavirus/2019-ncov/need-extra-precautions/people-with-medical-conditions.html (accessed on 17 August 2022).
  4. Walubita, T.; Beccia, A.; Boama-Nyarko, E.; Goulding, M.; Herbert, C.; Kloppenburg, J.; Mabry, G.; Masters, G.; McCullers, A.; Forrester, S. Aging and COVID-19 in Minority Populations: A Perfect Storm. Curr. Epidemiol. Rep. 2021, 8, 63–71. [Google Scholar] [CrossRef] [PubMed]
  5. Wilder, J.M. The Disproportionate Impact of COVID-19 on Racial and Ethnic Minorities in the United States. Clin. Infect. Dis. 2021, 72, 707–709. [Google Scholar] [CrossRef] [PubMed]
  6. Cagnoni, F.; Njwe, C.A.; Zaninelli, A.; Ricci, A.R.; Daffra, D.; D’Ospina, A.; Preti, P.; Destro, M. Blocking the RAAS at different levels: An update on the use of the direct renin inhibitors alone and in combination. Vasc. Health Risk Manag. 2010, 6, 549–559. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  7. Jia, G.; Aroor, A.R.; Hill, M.A.; Sowers, J.R. Role of Renin-Angiotensin-Aldosterone System Activation in Promoting Cardiovascular Fibrosis and Stiffness. Hypertension 2018, 72, 537–548. [Google Scholar] [CrossRef]
  8. Ferrario, C.M.; Strawn, W.B. Role of the renin-angiotensin-aldosterone system and proinflammatory mediators in cardiovascular disease. Am. J. Cardiol. 2006, 98, 121–128. [Google Scholar] [CrossRef]
  9. Patel, A.B.; Verma, A. COVID-19 and Angiotensin-Converting Enzyme Inhibitors and Angiotensin Receptor Blockers: What Is the Evidence? JAMA 2020, 323, 1769–1770. [Google Scholar] [CrossRef]
  10. Vaduganathan, M.; Vardeny, O.; Michel, T.; McMurray, J.J.V.; Pfeffer, M.A.; Solomon, S.D. Renin-Angiotensin-Aldosterone System Inhibitors in Patients with COVID-19. N. Engl. J. Med. 2020, 382, 1653–1659. [Google Scholar] [CrossRef]
  11. Touyz, R.M.; Li, H.; Delles, C. ACE2 the Janus-faced protein—From cardiovascular protection to severe acute respiratory syndrome-coronavirus and COVID-19. Clin. Sci. 2020, 134, 747–750. [Google Scholar] [CrossRef]
  12. Liu, J.; Zhou, Y.; Liu, Y.; Li, L.; Chen, Y.; Liu, Y.; Feng, Y.; Yosypiv, I.V.; Song, R.; Peng, H. (Pro)renin receptor regulates lung development via the Wnt/beta-catenin signaling pathway. Am. J. Physiol. Lung Cell. Mol. Physiol. 2019, 317, L202–L211. [Google Scholar] [CrossRef] [PubMed]
  13. Fountain, J.H.; Lappin, S.L. Physiology, Renin Angiotensin System; StatPearls: Treasure Island, FL, USA, 2020. [Google Scholar]
  14. Hall, J.E.; do Carmo, J.M.; da Silva, A.A.; Wang, Z.; Hall, M.E. Obesity, kidney dysfunction and hypertension: Mechanistic links. Nat. Rev. Nephrol. 2019, 15, 367–385. [Google Scholar] [CrossRef] [PubMed]
  15. Ren, L.; Lu, X.; Danser, A.H.J. Revisiting the Brain Renin-Angiotensin System-Focus on Novel Therapies. Curr. Hypertens. Rep. 2019, 21, 28. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  16. Quinn, K.L.; Fralick, M.; Zipursky, J.S.; Stall, N.M. Renin-angiotensin-aldosterone system inhibitors and COVID-19. CMAJ 2020, 192, E553–E554. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  17. Weir, M.R.; Dzau, V.J. The renin-angiotensin-aldosterone system: A specific target for hypertension management. Am. J. Hypertens. 1999, 12, 205S–213S. [Google Scholar] [CrossRef]
  18. Shimosawa, T. Salt, the renin-angiotensin-aldosterone system and resistant hypertension. Hypertens Res 2013, 36, 657–660. [Google Scholar] [CrossRef] [Green Version]
  19. Topouchian, J.; El Feghali, R.; Pannier, B.; Wang, S.; Zhao, F.; Smetana, K.; Teo, K.; Asmar, R. Arterial stiffness and pharmacological interventions--the TRanscend arterial stiffNess Substudy (TRANS study). Vasc. Health Risk Manag. 2007, 3, 381–387. [Google Scholar]
  20. Ma, T.K.; Kam, K.K.; Yan, B.P.; Lam, Y.Y. Renin-angiotensin-aldosterone system blockade for cardiovascular diseases: Current status. Br. J. Pharmacol. 2010, 160, 1273–1292. [Google Scholar] [CrossRef]
  21. Pradhan, A.; Vishwakarma, P.; Bhandari, M.; Sethi, R.; Narain, V.S. Differential Effects of Combination of Renin-Angiotensin-Aldosterone System Inhibitors on Central Aortic Blood Pressure: A Cross-Sectional Observational Study in Hypertensive Outpatients. Cardiovasc. Ther. 2020, 2020, 4349612. [Google Scholar] [CrossRef]
  22. Kearney, P.M.; Whelton, M.; Reynolds, K.; Muntner, P.; Whelton, P.K.; He, J. Global burden of hypertension: Analysis of worldwide data. Lancet 2005, 365, 217–223. [Google Scholar] [CrossRef]
  23. Paul, M.; Poyan Mehr, A.; Kreutz, R. Physiology of local renin-angiotensin systems. Physiol. Rev. 2006, 86, 747–803. [Google Scholar] [CrossRef]
  24. Macera, M.; De Angelis, G.; Sagnelli, C.; Coppola, N.; Vanvitelli, C.-G. Clinical Presentation of COVID-19: Case Series and Review of the Literature. Int. J. Environ. Res. Public Health 2020, 17, 5062. [Google Scholar] [CrossRef] [PubMed]
  25. Chathappady House, N.N.; Palissery, S.; Sebastian, H. Corona Viruses: A Review on SARS, MERS and COVID-19. Microbiol. Insights 2021, 14, 11786361211002481. [Google Scholar] [CrossRef] [PubMed]
  26. Chaplin, S. COVID-19: A brief history and treatments in development. Prescriber 2020, 31, 23–28. [Google Scholar] [CrossRef]
  27. Zhu, N.; Zhang, D.; Wang, W.; Li, X.; Yang, B.; Song, J.; Zhao, X.; Huang, B.; Shi, W.; Lu, R.; et al. A Novel Coronavirus from Patients with Pneumonia in China, 2019. N. Engl. J. Med. 2020, 382, 727–733. [Google Scholar] [CrossRef] [PubMed]
  28. Johns-Hopkins-University-Medicine. Corona Virus Resource Center. Available online: https://coronavirus.jhu.edu/map.html (accessed on 17 August 2022).
  29. Zou, L.; Ruan, F.; Huang, M.; Liang, L.; Huang, H.; Hong, Z.; Yu, J.; Kang, M.; Song, Y.; Xia, J.; et al. SARS-CoV-2 Viral Load in Upper Respiratory Specimens of Infected Patients. N. Engl. J. Med. 2020, 382, 1177–1179. [Google Scholar] [CrossRef] [PubMed]
  30. Weinberger, J.; Rhee, C.; Klompas, M. Incidence, Characteristics, and Outcomes of Ventilator-associated Events during the COVID-19 Pandemic. Ann. Am. Thorac. Soc. 2021, 19, 82–89. [Google Scholar] [CrossRef]
  31. Batah, S.S.; Fabro, A.T. Pulmonary pathology of ARDS in COVID-19: A pathological review for clinicians. Respir. Med. 2021, 176, 106239. [Google Scholar] [CrossRef]
  32. Suarez-de-la-Rica, A.; Serrano, P.; De-la-Oliva, R.; Sánchez-Díaz, P.; Molinero, P.; Falces-Romero, I.; Ferrando, C.; Rello, J.; Maseda, E. Secondary infections in mechanically ventilated patients with COVID-19: An overlooked matter? Rev. Esp. Quimioter. 2021, 34, 330–336. [Google Scholar] [CrossRef]
  33. Yoshida, T.; Fujino, Y.; Amato, M.B.P.; Kavanagh, B.P. Fifty Years of Research in ARDS. Spontaneous Breathing during Mechanical Ventilation. Risks, Mechanisms, and Management. Am. J. Respir. Crit. Care Med. 2016, 195, 985–992. [Google Scholar] [CrossRef]
  34. Hu, B.; Huang, S.; Yin, L. The cytokine storm and COVID-19. J. Med. Virol. 2021, 93, 250–256. [Google Scholar] [CrossRef] [PubMed]
  35. Fara, A.; Mitrev, Z.; Rosalia, R.A.; Assas, B.M. Cytokine storm and COVID-19: A chronicle of pro-inflammatory cytokines. Open Biol. 2020, 10, 200160. [Google Scholar] [CrossRef] [PubMed]
  36. Sun, X.; Wang, T.; Cai, D.; Hu, Z.; Chen, J.a.; Liao, H.; Zhi, L.; Wei, H.; Zhang, Z.; Qiu, Y.; et al. Cytokine storm intervention in the early stages of COVID-19 pneumonia. Cytokine Growth Factor Rev. 2020, 53, 38–42. [Google Scholar] [CrossRef]
  37. Soy, M.; Keser, G.; Atagündüz, P.; Tabak, F.; Atagündüz, I.; Kayhan, S. Cytokine storm in COVID-19: Pathogenesis and overview of anti-inflammatory agents used in treatment. Clin. Rheumatol. 2020, 39, 2085–2094. [Google Scholar] [CrossRef] [PubMed]
  38. Klok, F.A.; Kruip, M.J.H.A.; van der Meer, N.J.M.; Arbous, M.S.; Gommers, D.A.M.P.J.; Kant, K.M.; Kaptein, F.H.J.; van Paassen, J.; Stals, M.A.M.; Huisman, M.V.; et al. Incidence of thrombotic complications in critically ill ICU patients with COVID-19. Thromb. Res. 2020, 191, 145–147. [Google Scholar] [CrossRef] [PubMed]
  39. Tang, N.; Li, D.; Wang, X.; Sun, Z. Abnormal coagulation parameters are associated with poor prognosis in patients with novel coronavirus pneumonia. J. Thromb. Haemost. 2020, 18, 844–847. [Google Scholar] [CrossRef] [Green Version]
  40. 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]
  41. Zhou, X.; Cheng, Z.; Luo, L.; Zhu, Y.; Lin, W.; Ming, Z.; Chen, W.; Hu, Y. Incidence and impact of disseminated intravascular coagulation in COVID-19 a systematic review and meta-analysis. Thromb. Res. 2021, 201, 23–29. [Google Scholar] [CrossRef]
  42. Ladikou, E.E.; Sivaloganathan, H.; Milne, K.M.; Arter, W.E.; Ramasamy, R.; Saad, R.; Stoneham, S.M.; Philips, B.; Eziefula, A.C.; Chevassut, T. Von Willebrand factor (vWF): Marker of endothelial damage and thrombotic risk in COVID-19? Clin. Med. 2020, 20, e178–e182. [Google Scholar] [CrossRef]
  43. Pearson, J.D. Endothelial cell function and thrombosis. Best Pract. Res. Clin. Haematol. 1999, 12, 329–341. [Google Scholar] [CrossRef]
  44. Melkumyants, A.; Buryachkovskaya, L.; Lomakin, N.; Antonova, O.; Serebruany, V. Mild COVID-19 and Impaired Blood Cell-Endothelial Crosstalk: Considering Long-Term Use of Antithrombotics? Thromb. Haemost. 2022, 122, 123–130. [Google Scholar] [CrossRef] [PubMed]
  45. Ackermann, M.; Verleden, S.E.; Kuehnel, M.; Haverich, A.; Welte, T.; Laenger, F.; Vanstapel, A.; Werlein, C.; Stark, H.; Tzankov, A.; et al. Pulmonary Vascular Endothelialitis, Thrombosis, and Angiogenesis in COVID-19. N. Engl. J. Med. 2020, 383, 120–128. [Google Scholar] [CrossRef] [PubMed]
  46. Varga, Z.; Flammer, A.J.; Steiger, P.; Haberecker, M.; Andermatt, R.; Zinkernagel, A.S.; Mehra, M.R.; Schuepbach, R.A.; Ruschitzka, F.; Moch, H. Endothelial cell infection and endotheliitis in COVID-19. Lancet 2020, 395, 1417–1418. [Google Scholar] [CrossRef]
  47. Sims, J.T.; Krishnan, V.; Chang, C.-Y.; Engle, S.M.; Casalini, G.; Rodgers, G.H.; Bivi, N.; Nickoloff, B.J.; Konrad, R.J.; de Bono, S.; et al. Characterization of the cytokine storm reflects hyperinflammatory endothelial dysfunction in COVID-19. J. Allergy Clin. Immunol. 2021, 147, 107–111. [Google Scholar] [CrossRef] [PubMed]
  48. Krzemińska, J.; Wronka, M.; Młynarska, E.; Franczyk, B.; Rysz, J. Arterial Hypertension—Oxidative Stress and Inflammation. Antioxidants 2022, 11, 172. [Google Scholar] [CrossRef]
  49. Lontchi-Yimagou, E.; Sobngwi, E.; Matsha, T.E.; Kengne, A.P. Diabetes Mellitus and Inflammation. Curr. Diabetes Rep. 2013, 13, 435–444. [Google Scholar] [CrossRef]
  50. Abdi, A.; Jalilian, M.; Sarbarzeh, P.A.; Vlaisavljevic, Z. Diabetes and COVID-19: A systematic review on the current evidences. Diabetes Res. Clin. Pract. 2020, 166, 108347. [Google Scholar] [CrossRef]
  51. Savoia, C.; Volpe, M.; Kreutz, R. Hypertension, a Moving Target in COVID-19. Circ. Res. 2021, 128, 1062–1079. [Google Scholar] [CrossRef]
  52. de Abajo, F.J.; Rodriguez-Martin, S.; Lerma, V.; Mejia-Abril, G.; Aguilar, M.; Garcia-Luque, A.; Laredo, L.; Laosa, O.; Centeno-Soto, G.A.; Angeles Galvez, M.; et al. Use of renin-angiotensin-aldosterone system inhibitors and risk of COVID-19 requiring admission to hospital: A case-population study. Lancet 2020, 395, 1705–1714. [Google Scholar] [CrossRef]
  53. Zhang, H.; Penninger, J.M.; Li, Y.; Zhong, N.; Slutsky, A.S. Angiotensin-converting enzyme 2 (ACE2) as a SARS-CoV-2 receptor: Molecular mechanisms and potential therapeutic target. Intensive Care Med. 2020, 46, 586–590. [Google Scholar] [CrossRef] [Green Version]
  54. Turner, A.J.; Hiscox, J.A.; Hooper, N.M. ACE2: From vasopeptidase to SARS virus receptor. Trends Pharmacol. Sci. 2004, 25, 291–294. [Google Scholar] [CrossRef] [PubMed]
  55. Bourgonje, A.R.; Abdulle, A.E.; Timens, W.; Hillebrands, J.L.; Navis, G.J.; Gordijn, S.J.; Bolling, M.C.; Dijkstra, G.; Voors, A.A.; Osterhaus, A.D.; et al. Angiotensin-converting enzyme 2 (ACE2), SARS-CoV-2 and the pathophysiology of coronavirus disease 2019 (COVID-19). J. Pathol. 2020, 251, 228–248. [Google Scholar] [CrossRef] [PubMed]
  56. Hoffmann, M.; Kleine-Weber, H.; Schroeder, S.; Kruger, N.; Herrler, T.; Erichsen, S.; Schiergens, T.S.; Herrler, G.; Wu, N.H.; Nitsche, A.; et al. SARS-CoV-2 Cell Entry Depends on ACE2 and TMPRSS2 and Is Blocked by a Clinically Proven Protease Inhibitor. Cell 2020, 181, 271–280.e8. [Google Scholar] [CrossRef] [PubMed]
  57. Sanchis-Gomar, F.; Lavie, C.J.; Perez-Quilis, C.; Henry, B.M.; Lippi, G. Angiotensin-Converting Enzyme 2 and Antihypertensives (Angiotensin Receptor Blockers and Angiotensin-Converting Enzyme Inhibitors) in Coronavirus Disease 2019. Mayo Clin. Proc. 2020, 95, 1222–1230. [Google Scholar] [CrossRef] [PubMed]
  58. 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] [Green Version]
  59. Lumpuy-Castillo, J.; Lorenzo-Almoros, A.; Pello-Lazaro, A.M.; Sanchez-Ferrer, C.; Egido, J.; Tunon, J.; Peiro, C.; Lorenzo, O. Cardiovascular Damage in COVID-19: Therapeutic Approaches Targeting the Renin-Angiotensin-Aldosterone System. Int. J. Mol. Sci. 2020, 21, 6471. [Google Scholar] [CrossRef]
  60. Hamming, I.; Cooper, M.E.; Haagmans, B.L.; Hooper, N.M.; Korstanje, R.; Osterhaus, A.D.; Timens, W.; Turner, A.J.; Navis, G.; van Goor, H. The emerging role of ACE2 in physiology and disease. J. Pathol. 2007, 212, 1–11. [Google Scholar] [CrossRef]
  61. Gupte, M.; Boustany-Kari, C.M.; Bharadwaj, K.; Police, S.; Thatcher, S.; Gong, M.C.; English, V.L.; Cassis, L.A. ACE2 is expressed in mouse adipocytes and regulated by a high-fat diet. Am. J. Physiol. Regul. Integr. Comp. Physiol. 2008, 295, R781–R788. [Google Scholar] [CrossRef] [Green Version]
  62. Sinha, S.; Sehgal, A.; Sehgal, R. Association of ACE2 receptor and ACEIs/ARBs with disease severity in COVID-19. Drug Discov. Ther. 2020, 14, 161–170. [Google Scholar] [CrossRef]
  63. Bernardi, S.; Toffoli, B.; Zennaro, C.; Tikellis, C.; Monticone, S.; Losurdo, P.; Bellini, G.; Thomas, M.C.; Fallo, F.; Veglio, F.; et al. High-salt diet increases glomerular ACE/ACE2 ratio leading to oxidative stress and kidney damage. Nephrol. Dial. Transplant. 2012, 27, 1793–1800. [Google Scholar] [CrossRef] [Green Version]
  64. Yang, G.; Tan, Z.; Zhou, L.; Yang, M.; Peng, L.; Liu, J.; Cai, J.; Yang, R.; Han, J.; Huang, Y.; et al. Effects of Angiotensin II Receptor Blockers and ACE (Angiotensin-Converting Enzyme) Inhibitors on Virus Infection, Inflammatory Status, and Clinical Outcomes in Patients With COVID-19 and Hypertension: A Single-Center Retrospective Study. Hypertension 2020, 76, 51–58. [Google Scholar] [CrossRef] [PubMed]
  65. 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] [PubMed] [Green Version]
  66. Xiao, L.; Sakagami, H.; Miwa, N. ACE2: The key Molecule for Understanding the Pathophysiology of Severe and Critical Conditions of COVID-19: Demon or Angel? Viruses 2020, 12, 491. [Google Scholar] [CrossRef] [PubMed]
  67. Hsueh, W.A.; Wyne, K. Renin-Angiotensin-aldosterone system in diabetes and hypertension. J. Clin. Hypertens. 2011, 13, 224–237. [Google Scholar] [CrossRef]
  68. South, A.M.; Diz, D.I.; Chappell, M.C. COVID-19, ACE2, and the cardiovascular consequences. Am. J. Physiol. Heart Circ. Physiol. 2020, 318, H1084–H1090. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  69. 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]
  70. Talreja, H.; Tan, J.; Dawes, M.; Supershad, S.; Rabindranath, K.; Fisher, J.; Valappil, S.; van der Merwe, V.; Wong, L.; van der Merwe, W.; et al. A consensus statement on the use of angiotensin receptor blockers and angiotensin converting enzyme inhibitors in relation to COVID-19 (corona virus disease 2019). N. Z. Med. J. 2020, 133, 85–87. [Google Scholar]
  71. Oza, P.P.; Kashfi, K. Utility of NO and H(2)S donating platforms in managing COVID-19: Rationale and promise. Nitric Oxide 2022, 128, 72–102. [Google Scholar] [CrossRef]
  72. Adusumilli, N.C.; Zhang, D.; Friedman, J.M.; Friedman, A.J. Harnessing nitric oxide for preventing, limiting and treating the severe pulmonary consequences of COVID-19. Nitric Oxide 2020, 103, 4–8. [Google Scholar] [CrossRef]
  73. Zamanian, R.T.; Pollack, C.V.; Gentile, M.A.; Rashid, M.; Fox, J.C.; Mahaffey, K.W.; de Jesus Perez, V. Outpatient Inhaled Nitric Oxide in a Patient with Vasoreactive Idiopathic Pulmonary Arterial Hypertension and COVID-19 Infection. Am. J. Respir. Crit. Care Med. 2020, 202, 130–132. [Google Scholar] [CrossRef]
  74. Martel, J.; Ko, Y.-F.; Young, J.D.; Ojcius, D.M. Could nasal nitric oxide help to mitigate the severity of COVID-19? Microbes Infect. 2020, 22, 168–171. [Google Scholar] [CrossRef] [PubMed]
  75. Pieretti, J.C.; Rubilar, O.; Weller, R.B.; Tortella, G.R.; Seabra, A.B. Nitric oxide (NO) and nanoparticles—Potential small tools for the war against COVID-19 and other human coronavirus infections. Virus Res. 2021, 291, 198202. [Google Scholar] [CrossRef] [PubMed]
  76. Bruckdorfer, R. The basics about nitric oxide. Mol. Asp. Med. 2005, 26, 3–31. [Google Scholar] [CrossRef] [PubMed]
  77. Król, M.; Kepinska, M. Human Nitric Oxide Synthase—Its Functions, Polymorphisms, and Inhibitors in the Context of Inflammation, Diabetes and Cardiovascular Diseases. Int. J. Mol. Sci. 2021, 22, 56. [Google Scholar] [CrossRef]
  78. Guan, S.P.; Seet, R.C.S.; Kennedy, B.K. Does eNOS derived nitric oxide protect the young from severe COVID-19 complications? Ageing Res. Rev. 2020, 64, 101201. [Google Scholar] [CrossRef]
  79. Vassiliou, A.G.; Zacharis, A.; Keskinidou, C.; Jahaj, E.; Pratikaki, M.; Gallos, P.; Dimopoulou, I.; Kotanidou, A.; Orfanos, S.E. Soluble Angiotensin Converting Enzyme 2 (ACE2) Is Upregulated and Soluble Endothelial Nitric Oxide Synthase (eNOS) Is Downregulated in COVID-19-induced Acute Respiratory Distress Syndrome (ARDS). Pharmaceuticals 2021, 14, 695. [Google Scholar] [CrossRef]
  80. Guimarães, L.M.F.; Rossini, C.V.T.; Lameu, C. Implications of SARS-CoV-2 infection on eNOS and iNOS activity: Consequences for the respiratory and vascular systems. Nitric Oxide 2021, 111–112, 64–71. [Google Scholar] [CrossRef]
  81. Nikolaidis, A.; Kramer, R.; Ostojic, S. Nitric Oxide: The Missing Factor in COVID-19 Severity? Med. Sci. 2022, 10, 3. [Google Scholar] [CrossRef]
  82. Guzik, T.J.; Korbut, R.; Adamek-Guzik, T. Nitric oxide and superoxide in inflammation and immune regulation. J. Physiol. Pharmacol. 2003, 54, 469–487. [Google Scholar]
  83. Li, Q.; Youn, J.-Y.; Cai, H. Mechanisms and consequences of endothelial nitric oxide synthase dysfunction in hypertension. J. Hypertens. 2015, 33, 1128–1136. [Google Scholar] [CrossRef] [Green Version]
  84. Huang, P.L. eNOS, metabolic syndrome and cardiovascular disease. Trends Endocrinol. Metab. 2009, 20, 295–302. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  85. Heitsch, H.; Brovkovych, S.; Malinski, T.; Wiemer, G. Angiotensin-(1-7)–Stimulated Nitric Oxide and Superoxide Release From Endothelial Cells. Hypertension 2001, 37, 72–76. [Google Scholar] [CrossRef] [PubMed]
  86. Rees, C.A.; Rostad, C.A.; Mantus, G.; Anderson, E.J.; Chahroudi, A.; Jaggi, P.; Wrammert, J.; Ochoa, J.B.; Ochoa, A.; Basu, R.K.; et al. Altered amino acid profile in patients with SARS-CoV-2 infection. Proc. Natl. Acad. Sci. USA 2021, 118, e2101708118. [Google Scholar] [CrossRef] [PubMed]
  87. D’Alessandro, A.; Thomas, T.; Akpan, I.J.; Reisz, J.A.; Cendali, F.I.; Gamboni, F.; Nemkov, T.; Thangaraju, K.; Katneni, U.; Tanaka, K.; et al. Biological and Clinical Factors Contributing to the Metabolic Heterogeneity of Hospitalized Patients with and without COVID-19. Cells 2021, 10, 2293. [Google Scholar] [CrossRef] [PubMed]
  88. Fiorentino, G.; Coppola, A.; Izzo, R.; Annunziata, A.; Bernardo, M.; Lombardi, A.; Trimarco, V.; Santulli, G.; Trimarco, B. Effects of adding L-arginine orally to standard therapy in patients with COVID-19: A randomized, double-blind, placebo-controlled, parallel-group trial. Results of the first interim analysis. EClinicalMedicine 2021, 40, 101125. [Google Scholar] [CrossRef] [PubMed]
  89. Saura, M.; Zaragoza, C.; McMillan, A.; Quick, R.A.; Hohenadl, C.; Lowenstein, J.M.; Lowenstein, C.J. An Antiviral Mechanism of Nitric Oxide: Inhibition of a Viral Protease. Immunity 1999, 10, 21–28. [Google Scholar] [CrossRef] [Green Version]
  90. Regev-Shoshani, G.; Vimalanathan, S.; McMullin, B.; Road, J.; Av-Gay, Y.; Miller, C. Gaseous nitric oxide reduces influenza infectivity in vitro. Nitric Oxide 2013, 31, 48–53. [Google Scholar] [CrossRef] [PubMed]
  91. Mokry, R.L.; Schumacher, M.L.; Hogg, N.; Terhune, S.S. Nitric Oxide Circumvents Virus-Mediated Metabolic Regulation during Human Cytomegalovirus Infection. mBio 2020, 11, e02630-20. [Google Scholar] [CrossRef]
  92. Xu, W.; Zheng, S.; Dweik, R.A.; Erzurum, S.C. Role of epithelial nitric oxide in airway viral infection. Free. Radic. Biol. Med. 2006, 41, 19–28. [Google Scholar] [CrossRef]
  93. Gamba, G.; Cavalieri, H.; Courreges, M.C.; Massouh, E.J.; Benencia, F. Early inhibition of nitric oxide production increases HSV-1 intranasal infection. J. Med. Virol. 2004, 73, 313–322. [Google Scholar] [CrossRef]
  94. Sanders, S.P.; Siekierski, E.S.; Porter, J.D.; Richards, S.M.; Proud, D. Nitric oxide inhibits rhinovirus-induced cytokine production and viral replication in a human respiratory epithelial cell line. J. Virol. 1998, 72, 934–942. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  95. Klingström, J.; Åkerström, S.; Hardestam, J.; Stoltz, M.; Simon, M.; Falk, K.I.; Mirazimi, A.; Rottenberg, M.; Lundkvist, Å. Nitric oxide and peroxynitrite have different antiviral effects against hantavirus replication and free mature virions. Eur. J. Immunol. 2006, 36, 2649–2657. [Google Scholar] [CrossRef] [PubMed]
  96. Zaragoza, C.; Ocampo, C.J.; Saura, M.; McMillan, A.; Lowenstein, C.J. Nitric oxide inhibition of coxsackievirus replication in vitro. J. Clin. Investig. 1997, 100, 1760–1767. [Google Scholar] [CrossRef] [PubMed]
  97. Flodström, M.; Horwitz, M.S.; Maday, A.; Balakrishna, D.; Rodriguez, E.; Sarvetnick, N. A Critical Role for Inducible Nitric Oxide Synthase in Host Survival Following Coxsackievirus B4 Infection. Virology 2001, 281, 205–215. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  98. Harris, N.; Buller, R.M.; Karupiah, G. Gamma interferon-induced, nitric oxide-mediated inhibition of vaccinia virus replication. J. Virol. 1995, 69, 910–915. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  99. Akerström, S.; Mousavi-Jazi, M.; Klingström, J.; Leijon, M.; Lundkvist, A.; Mirazimi, A. Nitric oxide inhibits the replication cycle of severe acute respiratory syndrome coronavirus. J. Virol. 2005, 79, 1966–1969. [Google Scholar] [CrossRef] [Green Version]
  100. Akerström, S.; Gunalan, V.; Keng, C.T.; Tan, Y.-J.; Mirazimi, A. Dual effect of nitric oxide on SARS-CoV replication: Viral RNA production and palmitoylation of the S protein are affected. Virology 2009, 395, 1–9. [Google Scholar] [CrossRef] [Green Version]
  101. Keyaerts, E.; Vijgen, L.; Chen, L.; Maes, P.; Hedenstierna, G.; Van Ranst, M. Inhibition of SARS-coronavirus infection in vitro by S-nitroso-N-acetylpenicillamine, a nitric oxide donor compound. Int. J. Infect. Dis. 2004, 8, 223–226. [Google Scholar] [CrossRef] [Green Version]
  102. Akaberi, D.; Krambrich, J.; Ling, J.; Luni, C.; Hedenstierna, G.; Järhult, J.D.; Lennerstrand, J.; Lundkvist, Å. Mitigation of the replication of SARS-CoV-2 by nitric oxide in vitro. Redox Biol. 2020, 37, 101734. [Google Scholar] [CrossRef]
  103. Winchester, S.; John, S.; Jabbar, K.; John, I. Clinical efficacy of nitric oxide nasal spray (NONS) for the treatment of mild COVID-19 infection. J. Infect. 2021, 83, 237–279. [Google Scholar] [CrossRef]
  104. Tsai, M.-L.; Tsou, C.-C.; Liaw, W.-F. Dinitrosyl Iron Complexes (DNICs): From Biomimetic Synthesis and Spectroscopic Characterization toward Unveiling the Biological and Catalytic Roles of DNICs. Acc. Chem. Res. 2015, 48, 1184–1193. [Google Scholar] [CrossRef] [PubMed]
  105. Pectol, D.C.; Delaney, C.R.; Zhu, J.; Mellott, D.M.; Katzfuss, A.; Taylor, Z.W.; Meek, T.D.; Darensbourg, M.Y. Dinitrosyl iron complexes (DNICs) as inhibitors of the SARS-CoV-2 main protease. Chem. Commun. 2021, 57, 8352–8355. [Google Scholar] [CrossRef] [PubMed]
  106. Vanin, A.F.; Pekshev, A.V.; Vagapov, A.B.; Sharapov, N.A.; Lakomkin, V.L.; Abramov, A.A.; Timoshin, A.A.; Kapelko, V.I. Gaseous Nitric Oxide and Dinitrosyl Iron Complexes with Thiol-Containing Ligands as Potential Medicines that Can Relieve COVID-19. Biophysics 2021, 66, 155–163. [Google Scholar] [CrossRef] [PubMed]
  107. Parikh, R.; Wilson, C.; Weinberg, J.; Gavin, D.; Murphy, J.; Reardon, C.C. Inhaled nitric oxide treatment in spontaneously breathing COVID-19 patients. Ther. Adv. Respir. Dis. 2020, 14, 1753466620933510. [Google Scholar] [CrossRef]
  108. Garfield, B.; McFadyen, C.; Briar, C.; Bleakley, C.; Vlachou, A.; Baldwin, M.; Lees, N.; Price, S.; Ledot, S.; McCabe, C.; et al. Potential for personalised application of inhaled nitric oxide in COVID-19 pneumonia. Br. J. Anaesth. 2021, 126, e72–e75. [Google Scholar] [CrossRef]
  109. Wiegand, S.B.; Safaee Fakhr, B.; Carroll, R.W.; Zapol, W.M.; Kacmarek, R.M.; Berra, L. Rescue Treatment With High-Dose Gaseous Nitric Oxide in Spontaneously Breathing Patients With Severe Coronavirus Disease 2019. Crit. Care Explor. 2020, 2, e0277. [Google Scholar] [CrossRef] [PubMed]
  110. Khan, R.; Kirschenbaum, L.A.; LaRow, C.; Berna, G.; Griffin, K.; Astiz, M.E. Augmentation of Platelet and Endothelial Cell Enos Activity Decreases Sepsis-Related Neutrophil-Endothelial Cell Interactions. Shock 2010, 33, 242–246. [Google Scholar] [CrossRef]
  111. Kosutova, P.; Kolomaznik, M.; Calkovska, A.; Mokra, D.; Mikolka, P. Nitric-Oxide-Releasing Dexamethasone Derivative NCX-1005 Improves Lung Function and Attenuates Inflammation in Experimental Lavage-Induced ARDS. Pharmaceutics 2021, 13, 2092. [Google Scholar] [CrossRef] [PubMed]
  112. Tripathi, P.; Tripathi, P.; Kashyap, L.; Singh, V. The role of nitric oxide in inflammatory reactions. FEMS Immunol. Med. Microbiol. 2007, 51, 443–452. [Google Scholar] [CrossRef] [Green Version]
  113. Connelly, L.; Palacios-Callender, M.; Ameixa, C.; Moncada, S.; Hobbs, A.J. Biphasic Regulation of NF-κB Activity Underlies the Pro- and Anti-Inflammatory Actions of Nitric Oxide. J. Immunol. 2001, 166, 3873–3881. [Google Scholar] [CrossRef] [Green Version]
  114. Yang, T.; Peleli, M.; Zollbrecht, C.; Giulietti, A.; Terrando, N.; Lundberg, J.O.; Weitzberg, E.; Carlström, M. Inorganic nitrite attenuates NADPH oxidase-derived superoxide generation in activated macrophages via a nitric oxide-dependent mechanism. Free. Radic. Biol. Med. 2015, 83, 159–166. [Google Scholar] [CrossRef] [PubMed]
  115. Zhang, Y.; Lu, J.; Shi, J.; Lin, X.; Dong, J.; Zhang, S.; Liu, Y.; Tong, Q. Central administration of angiotensin-(1–7) stimulates nitric oxide release and upregulates the endothelial nitric oxide synthase expression following focal cerebral ischemia/reperfusion in rats. Neuropeptides 2008, 42, 593–600. [Google Scholar] [CrossRef] [PubMed]
  116. Cerrato, B.D.; Frasch, A.P.; Nakagawa, P.; Longo-Carbajosa, N.; Peña, C.; Höcht, C.; Gironacci, M.M. Angiotensin-(1–7) upregulates central nitric oxide synthase in spontaneously hypertensive rats. Brain Res. 2012, 1453, 1–7. [Google Scholar] [CrossRef] [PubMed]
  117. Tsai, H.-J.; Shih, C.-C.; Chang, K.-Y.; Liao, M.-H.; Liaw, W.-J.; Wu, C.-C.; Tsao, C.-M. Angiotensin-(1–7) treatment blocks lipopolysaccharide-induced organ damage, platelet dysfunction, and IL-6 and nitric oxide production in rats. Sci. Rep. 2021, 11, 610. [Google Scholar] [CrossRef] [PubMed]
  118. Regenhardt, R.W.; Desland, F.; Mecca, A.P.; Pioquinto, D.J.; Afzal, A.; Mocco, J.; Sumners, C. Anti-inflammatory effects of angiotensin-(1-7) in ischemic stroke. Neuropharmacology 2013, 71, 154–163. [Google Scholar] [CrossRef] [Green Version]
  119. Rajendran, S.; Chirkov, Y.Y.; Horowitz, J.D. Potentiation of platelet responsiveness to nitric oxide by angiotensin-(1–7) is associated with suppression of superoxide release. Platelets 2007, 18, 158–164. [Google Scholar] [CrossRef]
  120. Tesanovic, S.; Vinh, A.; Gaspari, T.A.; Casley, D.; Widdop, R.E. Vasoprotective and Atheroprotective Effects of Angiotensin (1–7) in Apolipoprotein E–Deficient Mice. Arterioscler. Thromb. Vasc. Biol. 2010, 30, 1606–1613. [Google Scholar] [CrossRef] [Green Version]
  121. Crassous, P.-A.; Couloubaly, S.; Huang, C.; Zhou, Z.; Baskaran, P.; Kim, D.D.; Papapetropoulos, A.; Fioramonti, X.; Durán, W.N.; Beuve, A. Soluble guanylyl cyclase is a target of angiotensin II-induced nitrosative stress in a hypertensive rat model. Am. J. Physiol.-Heart Circ. Physiol. 2012, 303, H597–H604. [Google Scholar] [CrossRef] [Green Version]
  122. Kim, D.; Rybalkin, S.D.; Pi, X.; Wang, Y.; Zhang, C.; Munzel, T.; Beavo, J.A.; Berk, B.C.; Yan, C. Upregulation of Phosphodiesterase 1A1 Expression Is Associated With the Development of Nitrate Tolerance. Circulation 2001, 104, 2338–2343. [Google Scholar] [CrossRef] [Green Version]
  123. Galougahi, K.K.; Liu, C.C.; Gentile, C.; Kok, C.; Nunez, A.; Garcia, A.; Fry, N.A.S.; Davies, M.J.; Hawkins, C.L.; Rasmussen, H.H.; et al. Glutathionylation Mediates Angiotensin II–Induced eNOS Uncoupling, Amplifying NADPH Oxidase–Dependent Endothelial Dysfunction. J. Am. Heart Assoc. 2014, 3, e000731. [Google Scholar] [CrossRef] [Green Version]
  124. Yamamoto, E.; Tamamaki, N.; Nakamura, T.; Kataoka, K.; Tokutomi, Y.; Dong, Y.-F.; Fukuda, M.; Matsuba, S.; Ogawa, H.; Kim-Mitsuyama, S. Excess Salt Causes Cerebral Neuronal Apoptosis and Inflammation in Stroke-Prone Hypertensive Rats Through Angiotensin II-Induced NADPH Oxidase Activation. Stroke 2008, 39, 3049–3056. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  125. Lee, D.-Y.; Wauquier, F.; Eid, A.A.; Roman, L.J.; Ghosh-Choudhury, G.; Khazim, K.; Block, K.; Gorin, Y. Nox4 NADPH Oxidase Mediates Peroxynitrite-dependent Uncoupling of Endothelial Nitric-oxide Synthase and Fibronectin Expression in Response to Angiotensin II: ROLE OF MITOCHONDRIAL REACTIVE OXYGEN SPECIES *. J. Biol. Chem. 2013, 288, 28668–28686. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  126. Higashi, Y.; Oshima, T.; Ono, N.; Hiraga, H.; Yoshimura, M.; Watanabe, M.; Matsuura, H.; Kambe, M.; Kajiyama, G. Intravenous administration of L-arginine inhibits angiotensin-converting enzyme in humans. J. Clin. Endocrinol. Metab. 1995, 80, 2198–2202. [Google Scholar] [CrossRef] [PubMed]
  127. Ackermann, A.; Fernández-Alfonso, M.S.; Sánchez de Rojas, R.; Ortega, T.; Paul, M.; González, C. Modulation of angiotensin-converting enzyme by nitric oxide. Br. J. Pharmacol. 1998, 124, 291–298. [Google Scholar] [CrossRef] [Green Version]
  128. Kumar, K.V.; Das, U.N. Effect of cis-Unsaturated Fatty Acids, Prostaglandins, and Free Radicals on Angiotensin-Converting Enzyme Activity in Vitro. Proc. Soc. Exp. Biol. Med. 1997, 214, 374–379. [Google Scholar] [CrossRef]
  129. Ichiki, T.; Usui, M.; Kato, M.; Funakoshi, Y.; Ito, K.; Egashira, K.; Takeshita, A. Downregulation of Angiotensin II Type 1 Receptor Gene Transcription by Nitric Oxide. Hypertension 1998, 31, 342–346. [Google Scholar] [CrossRef] [Green Version]
  130. Sharma, N.M.; Zheng, H.; Li, Y.-F.; Patel, K.P. Nitric oxide inhibits the expression of AT1 receptors in neurons. Am. J. Physiol.-Cell Physiol. 2012, 302, C1162–C1173. [Google Scholar] [CrossRef] [Green Version]
  131. Nägele, M.P.; Haubner, B.; Tanner, F.C.; Ruschitzka, F.; Flammer, A.J. Endothelial dysfunction in COVID-19: Current findings and therapeutic implications. Atherosclerosis 2020, 314, 58–62. [Google Scholar] [CrossRef]
  132. Wallace, J.L.; McKnight, W.; Del Soldato, P.; Baydoun, A.R.; Cirino, G. Anti-thrombotic effects of a nitric oxide-releasing, gastric-sparing aspirin derivative. J. Clin. Investig. 1995, 96, 2711–2718. [Google Scholar] [CrossRef] [Green Version]
  133. Gonzalez, D.R.; Beigi, F.; Treuer, A.V.; Hare, J.M. Deficient ryanodine receptor S-nitrosylation increases sarcoplasmic reticulum calcium leak and arrhythmogenesis in cardiomyocytes. Proc. Natl. Acad. Sci. USA 2007, 104, 20612–20617. [Google Scholar] [CrossRef] [Green Version]
  134. Iravanian, S.; Dudley, S.C. The renin-angiotensin-aldosterone system (RAAS) and cardiac arrhythmias. Heart Rhythm 2008, 5, S12–S17. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  135. Cutler, M.J.; Plummer, B.N.; Wan, X.; Sun, Q.A.; Hess, D.; Liu, H.; Deschenes, I.; Rosenbaum, D.S.; Stamler, J.S.; Laurita, K.R. Aberrant S-nitrosylation mediates calcium-triggered ventricular arrhythmia in the intact heart. Proc. Natl. Acad. Sci. USA 2012, 109, 18186–18191. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  136. Iciek, M.; Bilska-Wilkosz, A.; Kozdrowicki, M.; Górny, M. Reactive Sulfur Compounds in the Fight against COVID-19. Antioxidants 2022, 11, 1053. [Google Scholar] [CrossRef] [PubMed]
  137. Paul, B.D.; Snyder, S.H. Gasotransmitter hydrogen sulfide signaling in neuronal health and disease. Biochem. Pharmacol. 2018, 149, 101–109. [Google Scholar] [CrossRef] [PubMed]
  138. Bazhanov, N.; Escaffre, O.; Freiberg, A.N.; Garofalo, R.P.; Casola, A. Broad-Range Antiviral Activity of Hydrogen Sulfide Against Highly Pathogenic RNA Viruses. Sci. Rep. 2017, 7, 41029. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  139. Li, H.; Ma, Y.; Escaffre, O.; Ivanciuc, T.; Komaravelli, N.; Kelley, J.P.; Coletta, C.; Szabo, C.; Rockx, B.; Garofalo, R.P.; et al. Role of hydrogen sulfide in paramyxovirus infections. J. Virol. 2015, 89, 5557–5568. [Google Scholar] [CrossRef] [Green Version]
  140. Pozzi, G.; Masselli, E.; Gobbi, G.; Mirandola, P.; Taborda-Barata, L.; Ampollini, L.; Carbognani, P.; Micheloni, C.; Corazza, F.; Galli, D.; et al. Hydrogen Sulfide Inhibits TMPRSS2 in Human Airway Epithelial Cells: Implications for SARS-CoV-2 Infection. Biomedicines 2021, 9, 1273. [Google Scholar] [CrossRef]
  141. Kimura, Y.; Goto, Y.-I.; Kimura, H. Hydrogen Sulfide Increases Glutathione Production and Suppresses Oxidative Stress in Mitochondria. Antioxid. Redox Signal. 2009, 12, 1–13. [Google Scholar] [CrossRef]
  142. Dugbartey, G.J.; Alornyo, K.K.; Ohene, B.O.; Boima, V.; Antwi, S.; Sener, A. Renal consequences of the novel coronavirus disease 2019 (COVID-19) and hydrogen sulfide as a potential therapy. Nitric Oxide 2022, 120, 16–25. [Google Scholar] [CrossRef]
  143. Rushworth, G.F.; Megson, I.L. Existing and potential therapeutic uses for N-acetylcysteine: The need for conversion to intracellular glutathione for antioxidant benefits. Pharmacol. Ther. 2014, 141, 150–159. [Google Scholar] [CrossRef]
  144. Hsu, C.-N.; Hou, C.-Y.; Chang-Chien, G.-P.; Lin, S.; Tain, Y.-L. Maternal N-Acetylcysteine Therapy Prevents Hypertension in Spontaneously Hypertensive Rat Offspring: Implications of Hydrogen Sulfide-Generating Pathway and Gut Microbiota. Antioxidants 2020, 9, 856. [Google Scholar] [CrossRef] [PubMed]
  145. De Flora, S.; Balansky, R.; La Maestra, S. Rationale for the use of N-acetylcysteine in both prevention and adjuvant therapy of COVID-19. FASEB J. 2020, 34, 13185–13193. [Google Scholar] [CrossRef] [PubMed]
  146. Manček-Keber, M.; Hafner-Bratkovič, I.; Lainšček, D.; Benčina, M.; Govednik, T.; Orehek, S.; Plaper, T.; Jazbec, V.; Bergant, V.; Grass, V.; et al. Disruption of disulfides within RBD of SARS-CoV-2 spike protein prevents fusion and represents a target for viral entry inhibition by registered drugs. FASEB J. 2021, 35, e21651. [Google Scholar] [CrossRef]
  147. Li, T.; Zhao, B.; Wang, C.; Wang, H.; Liu, Z.; Li, W.; Jin, H.; Tang, C.; Du, J. Regulatory Effects of Hydrogen Sulfide on IL-6, IL-8 and IL-10 Levels in the Plasma and Pulmonary Tissue of Rats with Acute Lung Injury. Exp. Biol. Med. 2008, 233, 1081–1087. [Google Scholar] [CrossRef] [PubMed]
  148. Faller, S.; Seiler, R.; Donus, R.; Engelstaedter, H.; Hoetzel, A.; Spassov, S.G. Pre- and posttreatment with hydrogen sulfide prevents ventilator-induced lung injury by limiting inflammation and oxidation. PLoS ONE 2017, 12, e0176649. [Google Scholar] [CrossRef] [Green Version]
  149. Gaynitdinova, V.; Avdeev, S.; Merzhoeva, Z.; Berikkhanov, Z.; Medvedeva, I.; Gorbacheva, T. N-acetylcysteine as a part of complex treatment of moderate COVID-associated pneumonia. Pulmonologiya 2021, 31, 21–29. [Google Scholar] [CrossRef]
  150. Ibrahim, H.; Perl, A.; Smith, D.; Lewis, T.; Kon, Z.; Goldenberg, R.; Yarta, K.; Staniloae, C.; Williams, M. Therapeutic blockade of inflammation in severe COVID-19 infection with intravenous N-acetylcysteine. Clin. Immunol. 2020, 219, 108544. [Google Scholar] [CrossRef]
  151. Pagliaro, P.; Penna, C. ACE/ACE2 Ratio: A Key Also in 2019 Coronavirus Disease (COVID-19)? Front. Med. 2020, 7, 335. [Google Scholar] [CrossRef]
  152. 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]
  153. Liu, Y.; Yang, Y.; Zhang, C.; Huang, F.; Wang, F.; Yuan, J.; Wang, Z.; Li, J.; Li, J.; Feng, C.; et al. Clinical and biochemical indexes from 2019-nCoV infected patients linked to viral loads and lung injury. Sci. China Life Sci. 2020, 63, 364–374. [Google Scholar] [CrossRef] [Green Version]
  154. 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]
  155. Bai, L.; Qi, Y.; Chen, S.; Wang, J.; Tang, C.; Du, J.; Jin, H.; Huang, Y. Angiotensin II downregulates vascular endothelial cell hydrogen sulfide production by enhancing cystathionine γ-lyase degradation through ROS-activated ubiquitination pathway. Biochem. Biophys. Res. Commun. 2019, 514, 907–912. [Google Scholar] [CrossRef] [PubMed]
  156. Lin, Y.; Zeng, H.; Gao, L.; Gu, T.; Wang, C.; Zhang, H. Hydrogen Sulfide Attenuates Atherosclerosis in a Partially Ligated Carotid Artery Mouse model via Regulating Angiotensin Converting Enzyme 2 Expression. Front. Physiol. 2017, 8, 782. [Google Scholar] [CrossRef]
  157. Li, Z.; Organ Chelsea, L.; Kang, J.; Polhemus David, J.; Trivedi Rishi, K.; Sharp Thomas, E.; Jenkins Jack, S.; Tao, Y.-x.; Xian, M.; Lefer David, J. Hydrogen Sulfide Attenuates Renin Angiotensin and Aldosterone Pathological Signaling to Preserve Kidney Function and Improve Exercise Tolerance in Heart Failure. JACC Basic Transl. Sci. 2018, 3, 796–809. [Google Scholar] [CrossRef]
  158. Laggner, H.; Hermann, M.; Esterbauer, H.; Muellner, M.K.; Exner, M.; Gmeiner, B.M.K.; Kapiotis, S. The novel gaseous vasorelaxant hydrogen sulfide inhibits angiotensin-converting enzyme activity of endothelial cells. J. Hypertens. 2007, 25, 2100–2104. [Google Scholar] [CrossRef]
  159. Liu, S.-Y.; Duan, X.-C.; Jin, S.; Teng, X.; Xiao, L.; Xue, H.-M.; Wu, Y.-M. Hydrogen Sulfide Improves Myocardial Remodeling via Downregulated Angiotensin Ⅱ/AT1R Pathway in Renovascular Hypertensive Rats. Am. J. Hypertens. 2017, 30, 67–74. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  160. Guo, Q.; Feng, X.; Xue, H.; Teng, X.; Jin, S.; Duan, X.; Xiao, L.; Wu, Y. Maternal Renovascular Hypertensive Rats Treatment With Hydrogen Sulfide Increased the Methylation of AT1b Gene in Offspring. Am. J. Hypertens. 2017, 30, 1220–1227. [Google Scholar] [CrossRef] [Green Version]
  161. Lu, M.; Liu, Y.-H.; Goh, H.S.; Wang, J.J.X.; Yong, Q.-C.; Wang, R.; Bian, J.-S. Hydrogen Sulfide Inhibits Plasma Renin Activity. J. Am. Soc. Nephrol. 2010, 21, 993. [Google Scholar] [CrossRef] [Green Version]
  162. Altaany, Z.; Ju, Y.; Yang, G.; Wang, R. The coordination of S-sulfhydration, S-nitrosylation, and phosphorylation of endothelial nitric oxide synthase by hydrogen sulfide. Sci. Signal. 2014, 7, ra87. [Google Scholar] [CrossRef]
  163. Qin, Y.-R.; You, S.-J.; Zhang, Y.; Li, Q.; Wang, X.-H.; Wang, F.; Hu, L.-F.; Liu, C.-F. Hydrogen sulfide attenuates ferric chloride-induced arterial thrombosis in rats. Free. Radic. Res. 2016, 50, 654–665. [Google Scholar] [CrossRef]
  164. Grambow, E.; Leppin, C.; Leppin, K.; Kundt, G.; Klar, E.; Frank, M.; Vollmar, B. The effects of hydrogen sulfide on platelet-leukocyte aggregation and microvascular thrombolysis. Platelets 2017, 28, 509–517. [Google Scholar] [CrossRef] [PubMed]
  165. Grambow, E.; Mueller-Graf, F.; Delyagina, E.; Frank, M.; Kuhla, A.; Vollmar, B. Effect of the hydrogen sulfide donor GYY4137 on platelet activation and microvascular thrombus formation in mice. Platelets 2014, 25, 166–174. [Google Scholar] [CrossRef] [PubMed]
  166. Pan, L.-L.; Liu, X.-H.; Gong, Q.-H.; Wu, D.; Zhu, Y.-Z. Hydrogen Sulfide Attenuated Tumor Necrosis Factor-α-Induced Inflammatory Signaling and Dysfunction in Vascular Endothelial Cells. PLoS ONE 2011, 6, e19766. [Google Scholar] [CrossRef] [PubMed]
  167. Benedetti, F.; Davinelli, S.; Krishnan, S.; Gallo, R.C.; Scapagnini, G.; Zella, D.; Curreli, S. Sulfur compounds block MCP-1 production by Mycoplasma fermentans-infected macrophages through NF-κB inhibition. J. Transl. Med. 2014, 145. [Google Scholar] [CrossRef] [Green Version]
  168. Oh, G.-S.; Pae, H.-O.; Lee, B.-S.; Kim, B.-N.; Kim, J.-M.; Kim, H.-R.; Jeon, S.B.; Jeon, W.K.; Chae, H.-J.; Chung, H.-T. Hydrogen sulfide inhibits nitric oxide production and nuclear factor-κB via heme oxygenase-1 expression in RAW264.7 macrophages stimulated with lipopolysaccharide. Free. Radic. Biol. Med. 2006, 41, 106–119. [Google Scholar] [CrossRef]
  169. Liu, Z.; Han, Y.; Li, L.; Lu, H.; Meng, G.; Li, X.; Shirhan, M.; Peh, M.T.; Xie, L.; Zhou, S.; et al. The hydrogen sulfide donor, GYY4137, exhibits anti-atherosclerotic activity in high fat fed apolipoprotein E(-/-) mice. Br. J. Pharmacol. 2013, 169, 1795–1809. [Google Scholar] [CrossRef] [Green Version]
  170. Lin, F.; Yang, Y.; Wei, S.; Huang, X.; Peng, Z.; Ke, X.; Zeng, Z.; Song, Y. Hydrogen Sulfide Protects Against High Glucose-Induced Human Umbilical Vein Endothelial Cell Injury Through Activating PI3K/Akt/eNOS Pathway. Drug Des. Dev. Ther. 2020, 14, 621–633. [Google Scholar] [CrossRef] [Green Version]
  171. Al-Magableh, M.R.; Kemp-Harper, B.K.; Hart, J.L. Hydrogen sulfide treatment reduces blood pressure and oxidative stress in angiotensin II-induced hypertensive mice. Hypertens. Res. 2015, 38, 13–20. [Google Scholar] [CrossRef]
  172. Merai, R.; Siegel, C.; Rakotz, M.; Basch, P.; Wright, J.; Wong, B.; Dhsc; Thorpe, P. CDC Grand Rounds: A Public Health Approach to Detect and Control Hypertension. MMWR Morb. Mortal. Wkly. Rep. 2016, 65, 1261–1264. [Google Scholar] [CrossRef] [Green Version]
  173. Bosworth, H.B.; Powers, B.; Grubber, J.M.; Thorpe, C.T.; Olsen, M.K.; Orr, M.; Oddone, E.Z. Racial differences in blood pressure control: Potential explanatory factors. J. Gen. Intern. Med. 2008, 23, 692–698. [Google Scholar] [CrossRef]
  174. Helmer, A.; Slater, N.; Smithgall, S. A Review of ACE Inhibitors and ARBs in Black Patients With Hypertension. Ann. Pharmacother. 2018, 52, 1143–1151. [Google Scholar] [CrossRef] [PubMed]
  175. Flaten, H.K.; Monte, A.A. The Pharmacogenomic and Metabolomic Predictors of ACE Inhibitor and Angiotensin II Receptor Blocker Effectiveness and Safety. Cardiovasc. Drugs Ther. 2017, 31, 471–482. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  176. Kurtz, T.W.; DiCarlo, S.E.; Pravenec, M.; Morris, R.C., Jr. An Appraisal of Methods Recently Recommended for Testing Salt Sensitivity of Blood Pressure. J. Am. Heart Assoc. 2017, 6, e005653. [Google Scholar] [CrossRef] [PubMed]
  177. Ogedegbe, G.; Shah, N.R.; Phillips, C.; Goldfeld, K.; Roy, J.; Guo, Y.; Gyamfi, J.; Torgersen, C.; Capponi, L.; Bangalore, S. Comparative Effectiveness of Angiotensin-Converting Enzyme Inhibitor-Based Treatment on Cardiovascular Outcomes in Hypertensive Blacks Versus Whites. J. Am. Coll. Cardiol. 2015, 66, 1224–1233. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  178. Bangalore, S.; Ogedegbe, G.; Gyamfi, J.; Guo, Y.; Roy, J.; Goldfeld, K.; Torgersen, C.; Capponi, L.; Phillips, C.; Shah, N.R. Outcomes with Angiotensin-converting Enzyme Inhibitors vs Other Antihypertensive Agents in Hypertensive Blacks. Am. J. Med. 2015, 128, 1195–1203. [Google Scholar] [CrossRef] [Green Version]
  179. Barreras, A.; Gurk-Turner, C. Angiotensin II receptor blockers. Bayl. Univ. Med. Cent. Proc. 2003, 16, 123–126. [Google Scholar] [CrossRef] [Green Version]
  180. Kostis, J.B.; Kim, H.J.; Rusnak, J.; Casale, T.; Kaplan, A.; Corren, J.; Levy, E. Incidence and characteristics of angioedema associated with enalapril. Arch. Intern. Med. 2005, 165, 1637–1642. [Google Scholar] [CrossRef] [Green Version]
  181. Kirby, T. Evidence mounts on the disproportionate effect of COVID-19 on ethnic minorities. Lancet Respir. Med. 2020, 8, 547–548. [Google Scholar] [CrossRef]
  182. Dorn, A.V.; Cooney, R.E.; Sabin, M.L. COVID-19 exacerbating inequalities in the US. Lancet 2020, 395, 1243–1244. [Google Scholar] [CrossRef]
  183. Liverpool, L. Why are ethnic minorities worse affected? New Sci. 2020, 246, 11. [Google Scholar] [CrossRef]
  184. Fuschillo, S.; Palomba, L.; Capparelli, R.; Motta, A.; Maniscalco, M. Nitric Oxide and Hydrogen Sulfide: A Nice Pair in the Respiratory System. Curr. Med. Chem. 2020, 27, 7136–7148. [Google Scholar] [CrossRef] [PubMed]
  185. Zhou, P.; Yang, X.L.; Wang, X.G.; Hu, B.; Zhang, L.; Zhang, W.; Si, H.R.; Zhu, Y.; Li, B.; Huang, C.L.; et al. A pneumonia outbreak associated with a new coronavirus of probable bat origin. Nature 2020, 579, 270–273. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  186. Assimakopoulos, S.F.; Marangos, M. N-acetyl-cysteine may prevent COVID-19-associated cytokine storm and acute respiratory distress syndrome. Med. Hypotheses 2020, 140, 109778. [Google Scholar] [CrossRef] [PubMed]
  187. Smyth, L.J.; Canadas-Garre, M.; Cappa, R.C.; Maxwell, A.P.; McKnight, A.J. Genetic associations between genes in the renin-angiotensin-aldosterone system and renal disease: A systematic review and meta-analysis. BMJ Open 2019, 9, e026777. [Google Scholar] [CrossRef] [Green Version]
  188. Rossaint, R.; Falke, K.J.; Lopez, F.; Slama, K.; Pison, U.; Zapol, W.M. Inhaled nitric oxide for the adult respiratory distress syndrome. N. Engl. J. Med. 1993, 328, 399–405. [Google Scholar] [CrossRef]
  189. Gonzalez-Reyes, S.; Guzman-Beltran, S.; Medina-Campos, O.N.; Pedraza-Chaverri, J. Curcumin pretreatment induces Nrf2 and an antioxidant response and prevents hemin-induced toxicity in primary cultures of cerebellar granule neurons of rats. Oxid. Med. Cell. Longev. 2013, 2013, 801418. [Google Scholar] [CrossRef]
Figure 1. SARS-CoV-2 entry into the host cell. As an enveloped virus, SARS-CoV-2 can enter the host cell through one of two pathways—pathway #1, endosomal entry and pathway #2, direct cell surface entry. In both entry forms, the S protein binds to the ACE2 receptor and must be primed and activated by proteases at the S1/S2 and S2’ site. SARS-CoV-2 infection results in the downregulation of the ACE2 receptor, leading to an increase in Ang II and decrease in its conversion to Ang-(1-7). In pathway #1, SARS-CoV-2 undergoes clathrin-mediated endocytosis, and is primed by host protease Cathepsin L present in the endolysosome, followed by membrane fusion and release of viral genomic material. In pathway #2, the S protein is cleaved by TMPRSS2 at the cell membrane, where it directly fuses and releases viral RNA without entering an endolysosome. Both pathways join in the host cytoplasm, where a viral polymerase is translated, followed by RNA replication, structural protein translation, viral assembly, and release of progeny virus that continue to spread the infection to other cells. SARS-CoV-2 infection leads to respiratory compromise by causing lung injury, results in excessive inflammation with overactivation of leukocytes and production of inflammatory mediators, results in direct and inflammation-mediated damage to the endothelium, and induces a pro-thrombotic state. Reproduced with permission from Elsevier: Nitric Oxide, 128 (2022), 72–102 [71]. Abbreviations: ACE2, angiotensin-converting enzyme 2; Ang, angiotensin; S protein, spike protein; SARS-CoV-2, severe acute respiratory syndrome coronavirus 2; TMPRSS2, transmembrane serine protease 2.
Figure 1. SARS-CoV-2 entry into the host cell. As an enveloped virus, SARS-CoV-2 can enter the host cell through one of two pathways—pathway #1, endosomal entry and pathway #2, direct cell surface entry. In both entry forms, the S protein binds to the ACE2 receptor and must be primed and activated by proteases at the S1/S2 and S2’ site. SARS-CoV-2 infection results in the downregulation of the ACE2 receptor, leading to an increase in Ang II and decrease in its conversion to Ang-(1-7). In pathway #1, SARS-CoV-2 undergoes clathrin-mediated endocytosis, and is primed by host protease Cathepsin L present in the endolysosome, followed by membrane fusion and release of viral genomic material. In pathway #2, the S protein is cleaved by TMPRSS2 at the cell membrane, where it directly fuses and releases viral RNA without entering an endolysosome. Both pathways join in the host cytoplasm, where a viral polymerase is translated, followed by RNA replication, structural protein translation, viral assembly, and release of progeny virus that continue to spread the infection to other cells. SARS-CoV-2 infection leads to respiratory compromise by causing lung injury, results in excessive inflammation with overactivation of leukocytes and production of inflammatory mediators, results in direct and inflammation-mediated damage to the endothelium, and induces a pro-thrombotic state. Reproduced with permission from Elsevier: Nitric Oxide, 128 (2022), 72–102 [71]. Abbreviations: ACE2, angiotensin-converting enzyme 2; Ang, angiotensin; S protein, spike protein; SARS-CoV-2, severe acute respiratory syndrome coronavirus 2; TMPRSS2, transmembrane serine protease 2.
Ijms 23 13895 g001
Figure 2. Interactions between RAAS, NO, and H2S. The RAAS consists of two main axes, the pro-inflammatory and vasoconstrictive ACE-Ang II pathway, and the anti-inflammatory, vasorelaxant ACE2-Ang-(1-7) pathway, both of which differently affect NO and H2S bioavailability. Ang II uses AT1R and AT2R to exert a variety of effects upon NO bioavailability; there are reports of Ang II increasing NOS activity while simultaneously increasing the production of ROS through NADPH oxidase, resulting in NOS uncoupling and a net decrease in NO bioavailability over time despite increased production and release. Ang II also inhibits sGC and increases the activity of some PDEs, diminishing the function of the NO signaling pathway. On the other hand, Ang-(1-7) augments NO bioavailability both by reducing ROS and by stimulating NOS activity. In return, NO downregulates ACE and AT1R, reducing RAAS activation. There are also reports of the RAAS affecting H2S bioavailability, where increasing Ang II decreased H2S bioavailability by downregulating CSE and CBS. Measures to increase H2S bioavailability reduce traditional RAAS activation, inhibiting or downregulating renin release, ACE, AT1R, and ROS levels while increasing ACE2 activity to increase the function of the anti-inflammatory axis of the RAAS. Both gasotransmitters also interact to reinforce the other’s increase in bioavailability. Thus, NO and H2S both act to reduce RAAS overactivation, serving as anti-inflammatory, antioxidant, and vasorelaxant mediators. Note: black arrows show effects of RAAS on NO and H2S, green arrows show effects of the gasotransmitters on components of RAAS. Abbreviations: ACE, angiotensin-converting enzyme; ACE2, angiotensin-converting enzyme 2; Ang, angiotensin; AT1R, angiotensin II type 1 receptor; AT2R, angiotensin II type 2 receptor; CBS, cystathionine β-synthase; CSE, Cystathionine γ-Lyase; H2S, hydrogen sulfide; NADPH oxidase, nicotinamide adenine dinucleotide phosphate oxidase; NEP, neutral endopeptidase; NO, nitric oxide; NOS, nitric oxide synthase; PDEs, phosphodiesterases; RAAS, renin–angiotensin–aldosterone system; ROS, reactive oxygen species; sGC, soluble guanylyl cyclase.
Figure 2. Interactions between RAAS, NO, and H2S. The RAAS consists of two main axes, the pro-inflammatory and vasoconstrictive ACE-Ang II pathway, and the anti-inflammatory, vasorelaxant ACE2-Ang-(1-7) pathway, both of which differently affect NO and H2S bioavailability. Ang II uses AT1R and AT2R to exert a variety of effects upon NO bioavailability; there are reports of Ang II increasing NOS activity while simultaneously increasing the production of ROS through NADPH oxidase, resulting in NOS uncoupling and a net decrease in NO bioavailability over time despite increased production and release. Ang II also inhibits sGC and increases the activity of some PDEs, diminishing the function of the NO signaling pathway. On the other hand, Ang-(1-7) augments NO bioavailability both by reducing ROS and by stimulating NOS activity. In return, NO downregulates ACE and AT1R, reducing RAAS activation. There are also reports of the RAAS affecting H2S bioavailability, where increasing Ang II decreased H2S bioavailability by downregulating CSE and CBS. Measures to increase H2S bioavailability reduce traditional RAAS activation, inhibiting or downregulating renin release, ACE, AT1R, and ROS levels while increasing ACE2 activity to increase the function of the anti-inflammatory axis of the RAAS. Both gasotransmitters also interact to reinforce the other’s increase in bioavailability. Thus, NO and H2S both act to reduce RAAS overactivation, serving as anti-inflammatory, antioxidant, and vasorelaxant mediators. Note: black arrows show effects of RAAS on NO and H2S, green arrows show effects of the gasotransmitters on components of RAAS. Abbreviations: ACE, angiotensin-converting enzyme; ACE2, angiotensin-converting enzyme 2; Ang, angiotensin; AT1R, angiotensin II type 1 receptor; AT2R, angiotensin II type 2 receptor; CBS, cystathionine β-synthase; CSE, Cystathionine γ-Lyase; H2S, hydrogen sulfide; NADPH oxidase, nicotinamide adenine dinucleotide phosphate oxidase; NEP, neutral endopeptidase; NO, nitric oxide; NOS, nitric oxide synthase; PDEs, phosphodiesterases; RAAS, renin–angiotensin–aldosterone system; ROS, reactive oxygen species; sGC, soluble guanylyl cyclase.
Ijms 23 13895 g002
Figure 3. NO and H2S applications in COVID-19. NO and H2S are antiviral agents with promise for inhibiting SARS-CoV-2 infection through interference with entry, replication, and release. Both gasotransmitters also have potential applications in later-stage management of COVID-19. NO and H2S demonstrate the ability to inhibit leukocyte adhesion to the endothelium, reduce levels of pro-inflammatory cytokines, curtail excessive iNOS activity, and overall reduce inflammatory damage. These properties, combined with their vaso- and bronchodilator actions in the pulmonary system, serve to reduce the need for mechanical ventilation and alleviate respiratory compromise. Additionally, NO and H2S downregulate the classical RAAS signaling pathway and its related pro-inflammatory shift by inhibiting Ang II and AT1R. As a result, NO and H2S may have significant potential in the management of COVID-19, a disease recognized for causing hyperinflammatory conditions and respiratory compromise and for having greater severity where the RAAS is imbalanced. Abbreviations: Ang II, angiotensin II; AT1R, angiotensin II type 1 receptor; COVID-19; coronavirus disease 2019; H2S, hydrogen sulfide; NO, nitric oxide; RAAS, renin–angiotensin–aldosterone system; SARS-CoV-2, severe acute respiratory syndrome coronavirus 2.
Figure 3. NO and H2S applications in COVID-19. NO and H2S are antiviral agents with promise for inhibiting SARS-CoV-2 infection through interference with entry, replication, and release. Both gasotransmitters also have potential applications in later-stage management of COVID-19. NO and H2S demonstrate the ability to inhibit leukocyte adhesion to the endothelium, reduce levels of pro-inflammatory cytokines, curtail excessive iNOS activity, and overall reduce inflammatory damage. These properties, combined with their vaso- and bronchodilator actions in the pulmonary system, serve to reduce the need for mechanical ventilation and alleviate respiratory compromise. Additionally, NO and H2S downregulate the classical RAAS signaling pathway and its related pro-inflammatory shift by inhibiting Ang II and AT1R. As a result, NO and H2S may have significant potential in the management of COVID-19, a disease recognized for causing hyperinflammatory conditions and respiratory compromise and for having greater severity where the RAAS is imbalanced. Abbreviations: Ang II, angiotensin II; AT1R, angiotensin II type 1 receptor; COVID-19; coronavirus disease 2019; H2S, hydrogen sulfide; NO, nitric oxide; RAAS, renin–angiotensin–aldosterone system; SARS-CoV-2, severe acute respiratory syndrome coronavirus 2.
Ijms 23 13895 g003
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Ranjbar, T.; Oza, P.P.; Kashfi, K. The Renin–Angiotensin-Aldosterone System, Nitric Oxide, and Hydrogen Sulfide at the Crossroads of Hypertension and COVID-19: Racial Disparities and Outcomes. Int. J. Mol. Sci. 2022, 23, 13895. https://doi.org/10.3390/ijms232213895

AMA Style

Ranjbar T, Oza PP, Kashfi K. The Renin–Angiotensin-Aldosterone System, Nitric Oxide, and Hydrogen Sulfide at the Crossroads of Hypertension and COVID-19: Racial Disparities and Outcomes. International Journal of Molecular Sciences. 2022; 23(22):13895. https://doi.org/10.3390/ijms232213895

Chicago/Turabian Style

Ranjbar, Tara, Palak P. Oza, and Khosrow Kashfi. 2022. "The Renin–Angiotensin-Aldosterone System, Nitric Oxide, and Hydrogen Sulfide at the Crossroads of Hypertension and COVID-19: Racial Disparities and Outcomes" International Journal of Molecular Sciences 23, no. 22: 13895. https://doi.org/10.3390/ijms232213895

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