Next Article in Journal
Lactococcus lactis Delivery of Surface Layer Protein A Protects Mice from Colitis by Re-Setting Host Immune Repertoire
Next Article in Special Issue
Endothelial Dysfunction: From Pathophysiology to Novel Therapeutic Approaches
Previous Article in Journal
Impact of Frailty on Hippocampal Volume in Patients with Chronic Obstructive Pulmonary Disease
Previous Article in Special Issue
17β-Estradiol Increases APE1/Ref-1 Secretion in Vascular Endothelial Cells and Ovariectomized Mice: Involvement of Calcium-Dependent Exosome Pathway
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

H2S as a Bridge Linking Inflammation, Oxidative Stress and Endothelial Biology: A Possible Defense in the Fight against SARS-CoV-2 Infection?

1
Institute of Clinical Physiology, National Research Council, 56124 Pisa, Italy
2
Fondazione CNR-Regione Toscana G. Monasterio, 56124 Pisa, Italy
*
Authors to whom correspondence should be addressed.
Biomedicines 2021, 9(9), 1107; https://doi.org/10.3390/biomedicines9091107
Submission received: 14 July 2021 / Revised: 12 August 2021 / Accepted: 26 August 2021 / Published: 28 August 2021

Abstract

:
The endothelium controls vascular homeostasis through a delicate balance between secretion of vasodilators and vasoconstrictors. The loss of physiological homeostasis leads to endothelial dysfunction, for which inflammatory events represent critical determinants. In this context, therapeutic approaches targeting inflammation-related vascular injury may help for the treatment of cardiovascular disease and a multitude of other conditions related to endothelium dysfunction, including COVID-19. In recent years, within the complexity of the inflammatory scenario related to loss of vessel integrity, hydrogen sulfide (H2S) has aroused great interest due to its importance in different signaling pathways at the endothelial level. In this review, we discuss the effects of H2S, a molecule which has been reported to demonstrate anti-inflammatory activity, in addition to many other biological functions related to endothelium and sulfur-drugs as new possible therapeutic options in diseases involving vascular pathobiology, such as in SARS-CoV-2 infection.

1. Introduction

The endothelium is the inner lining that covers all blood vessels with a very large spatial distribution, and consequently with potentially different characteristics depending on its position in the body. Not just an inert barrier, the endothelium is recognized as a biologically active tissue that regulates vascular tone and structure through autocrine, paracrine, and hormone-like mechanisms [1]. In fact, it may respond to various stimuli (e.g., shear stress on the endothelial surface) and release numerous vasoactive substances with vasodilator or vasoconstrictor properties [1]. Loss of physiological homeostasis leads to endothelial dysfunction, a condition underlying micro- and macrovascular diseases, characterized by reduced capacity for vasodilation, recruitment of neutrophils, enhanced inflammation and oxidative stress, prothrombotic properties, impaired cell growth and vessel permeability [1]. Inflammatory processes remain critical determinants which can provoke increases in prothrombotic and pro-oxidative events. Therefore, endothelial dysfunction has been observed associated with different clinical diseases, such as chronic kidney disease, liver failure, atherosclerosis, hypertension, dyslipidemia, diabetes, and obesity [2]. This implies that endothelial dysfunction can have important repercussions on health and on the onset and development of practically all diseases. In particular, endothelial dysfunction can be promoted or exacerbated by severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) infection, either by direct (interaction of SARS-CoV-2 virus with endothelial angiotensin-converting enzyme 2 (ACE2)) or indirect mechanisms (e.g., systemic inflammation, leukocyte recruitment, immune dysregulation, procoagulant state, impaired fibrinolysis, or activation of the complement system) [3,4,5,6]. Conversely, preexisting endothelial dysfunction underlies cardiovascular risk factors (e.g., hypertension, diabetes, obesity, and aging), all conditions which may favor the risk and severity of novel coronavirus disease 2019 (COVID-19) [4]. Increasing experimental, clinical, and translational findings suggest that many common drugs, such as lipid-lowering, antihypertensive, and antidiabetic drugs, and also antioxidants (e.g., vitamin C and E, N-acetylcysteine-NAC) and anti-inflammatories (e.g., cyclooxygenase-2 inhibitors, and glucocorticoids) may target the endothelium [2]. Consequently, further knowledge of endothelium pathophysiology may greatly help in patient care, given its enormous diagnostic and therapeutic potential.
In this context, the role of hydrogen sulfide (H2S), defined as the third endogenous gaseous signaling molecule besides nitric oxide (NO) and carbon monoxide (CO), in recent years has aroused a great interest due to its importance on different signaling pathways at the endothelial level [5]. In particular, given the close interaction between reduced H2S levels and endothelial dysfunction and inflammation, and the relationship between NO and H2S, the therapeutic release of H2S may represent a new and intriguing development in the prevention and treatment of inflammatory-related endothelial dysfunction conditions [6].
In this review, we discuss the effects of H2S and the use of sulfur-drugs as a new possible therapeutic option in diseases involving vascular pathobiology, such as in the SARS-CoV-2 infection.

2. Hydrogen Sulfide: An Additive Key Factor in Vascular Homeostasis

In the last few years, the role of H2S as an interesting novel mediator involved in inflammation and endothelial function has emerged [7], in terms of the relaxation of blood vessels, regulation of blood pressure, reduction of inflammatory response, and induction of antioxidant defense [8].
This gasotransmitter is mostly produced through the reverse trans-sulfuration pathway by three different enzymes. Two enzymes, cystathionine beta synthase (CBS) and cystathionine gamma lyase (CSE), use piridoxal 5′ phosphate (PLP) as a cofactor, while 3-mercaptopiruvate sulfur transferase (3-MST), primarily located in the mitochondria, is not dependent on PLP [9,10].
The protective effect of H2S is mediated by many different cellular and molecular mechanisms (Figure 1).
S-sulfhydration, a chemical modification on specific cysteine residues of target proteins to form a persulfide group (–SSH), is considered a primary mechanism through which H2S alters the function of signaling proteins [11,12]. A striking example is the S-sulfhydration of endothelial nitric oxide synthase (eNOS), which promotes eNOS dimer stability, NO production, and consequent vasorelaxation [13]. Indeed, sulfhydration of the Kir 6.1 subunit of ATP-sensitive potassium (KATP) channels activates the channel, causing vascular endothelial and smooth muscle cell hyperpolarization and vasorelaxation [14]. Accordingly, exogenous administration of H2S attenuates the rise of blood pressure in both spontaneously hypertensive rats [15] and in mice rendered hypertensive with angiotensin II (AngII) [16].
Although H2S may directly inactivate reactive oxygen species (ROS), e.g., through the inhibition of peroxynitrite-mediated processes in vivo [17], it also protects cells via the upregulation of antioxidant defense systems [6,8]. Specifically, sodium hydrosulfide (NaHS, an H2S donor) induces the sulfhydration of Kelch-like ECH-associating protein 1 (Keap1), a repressor of nuclear-factor-E2-related factor-2 (Nrf2), which is the main regulator of the antioxidant response. This action results in Keap1/Nrf2 disassociation, Nrf2 nuclear translocation, and increased mRNA expression of Nrf2-targeted downstream genes [18,19]. In human umbilical vein endothelial cells (HUVECs) exposed to H2O2, H2S upregulates a wide range of enzymes attenuating oxidative stress, such as catalase (CAT), superoxide dismutase (SOD), glutathione peroxidase, and glutathione-S-transferase [20]. Moreover, in endothelial cells (ECs) and fibroblasts, H2S-induced S-sulfhydration and activation of mitogen-activated extracellular signal-regulated kinase 1 are followed by phosphorylated ERK1/2 translocation into the nucleus to stimulate activity of PARP-1, a nuclear protein that exerts an important role in DNA damage repair [21]. H2S is further implicated in the regulation of the other major cellular inflammatory signaling pathway, namely the nuclear factor-κB (NF-κB) pathway [22]. The multifunctional pro-inflammatory cytokine, tumor necrosis factor α (TNF-α), stimulates the transcription of CSE. Consequently, the H2S-induced sulfhydration of the p65 subunit of NF-κB, promotes transcription of anti-apoptotic genes by enhancing its ability to bind the co-activator ribosomal protein S3 [23]. On the other hand, NaHS can exert an anti-inflammatory effect in ECs by inhibiting the expression of adhesion molecules (i.e., intercellular adhesion molecule-1 (ICAM-1), vascular cell adhesion molecule-1, P-selectin, and E-selectin), an early marker of endothelial activation and dysfunction, by upregulating the cytoprotective enzyme heme oxygenase-1 (HO-1), and decreasing TNF-α-induced NF-kB activation and ROS production [24]. Exogenous H2S also attenuates the inflammation and cytotoxicity induced by AngII in HUVECs via inhibition of the NF-κB/endothelin-I signaling pathway [25]. The pretreatment of HUVECs with NaHS can inhibit high-glucose-induced ICAM-1 expression at both the protein and mRNA levels, leading to a reduction of ROS production and NF-kB activity [26]. Furthermore, the administration of NaHS to high-glucose-treated ECs attenuates both the apoptosis and impairment of the CAT and SOD expression and activities induced by type 2 diabetes [27]. Interestingly, both diabetic rats [27] and patients with type 2 diabetes display markedly decreased plasma H2S levels [28,29].
H2S has also been ascertained to interact with the metal centers of proteins [30]. The interaction of H2S with the oxygenated form of human hemoglobin and myoglobin produces a sulfheme protein complex that participates in the catabolism of H2S [31]. Of interest is that H2S affects the function of soluble guanylyl cyclase (sGC), a receptor for NO, through the reduction of the ferric sGC heme into a ferrous state, facilitating NO-dependent vasodilation and thus providing an additional level of cross-talk between NO and H2S [32].
H2S stimulates EC proliferation, adhesion and migration in vitro and in vivo [6,11]. The mechanism whereby H2S regulates the key steps of angiogenesis can be driven by the increase in intracellular calcium levels in vascular ECs through the activation of many calcium-dependent signaling pathways and enzymes [9,33,34]. Also, H2S donors activate the PI-3K/Akt axis and enhance the phosphorylation of components of the MAPK pathway (p38 and ERK1/2) in EC models in vitro, with a subsequent pro-angiogenic effect [35]. Other mechanisms in H2S-induced angiogenesis in vascular ECs could also involve the opening of KATP channels by persulfidation of sulfonylurea receptor 1 subunit [36] and the activation of vascular endothelial growth factor receptor 2 (VEGFR2), a receptor tyrosine kinase that mediates most of the biological effects of the vascular endothelial growth factor through the breakage of a cysteine-cysteine disulfide bond within VEGFR2 [37].
Importantly, altered H2S metabolism is likely to be involved in the initiation and progression of atherosclerosis [9]. The H2S donors NaHS and GYY4137 may decrease aortic atherosclerotic plaque formation, macrophage infiltration, and aortic inflammation, and may partially restore endothelium-dependent relaxation in the aorta of apolipoprotein E (ApoE) gene-knockout mice [38,39]. In addition, CSE/H2S treatment directly sulfhydrated sirtuin-1, a histone deacetylase with a crucial role in longevity, increasing its activity and stability, thereby reducing atherosclerotic plaque formation in the aorta of these animals [39].
A few studies finally have investigated the anti-aggregatory and anticoagulatory effects of H2S [40]. In mice, GYY4137 seems to act as an anti-thrombotic and to regulate thrombogenesis by reducing platelet activation and adhesion molecule-mediated aggregation [41]. In mice treated with the H2S donor sodium sulfide (Na2S), thrombus formation induced using a phototoxic light/dye-injury model is significantly delayed compared to controls, due to the up-regulation of eNOS and inducible NOS (iNOS) [40]. Likewise, in an in vitro study on human whole blood, GYY4137 was observed to reduce platelet-leukocyte aggregation provoked by the thrombin-receptor activating peptide, and consequently facilitate microvascular thrombolysis [42].
It is important to note that H2S, in biological systems, co-exists with the sulfane sulfur species, i.e., uncharged sulfur atoms carrying six valence electrons being able to attach reversibly to other sulfur atoms [43]. Sulfane sulfur does not exist in the free form and can be considered as a sort of H2S storage, releasing H2S under reducing conditions, following a physiological signal [44,45]. Inorganic hydrogen polysulfides (H2Sn, n ≥ 2), in particular, are endogenously produced through several enzymatic routes involving 3-MST [46], copper/zinc SOD [47], or the direct reaction between H2S and NO [48]. H2Sn is greatly reactive, and has recently emerged as a potential signaling molecule that immediately reacts with intracellular cysteine, glutathione (GSH), and protein cysteine residues [49].

2.1. Sulfur-Drugs as New Therapeutic Options in Endothelial Dysfunction

The use of sulfur moieties as therapeutic agents in a wide array of applications (e.g., arterial hypertension, atherosclerosis, myocardial hypertrophy, heart failure, ischemia-reperfusion, diabetic nephron- and retinopathy, and chronic inflammatory diseases), has attracted growing attention in the last few years [50,51] (Table 1).

2.1.1. H2S Donors

Inorganic sulfide salts such as NaHS and Na2S are cheap and readily available, and have been largely employed in vitro and in animal models with the main effects of protecting ECs from inflammation, oxidative stress, damage induced by hyperglycemia, and promoting vasorelaxation and neovascularization, as illustrated above [6,52]. Nevertheless, they are unsuitable for clinical use, owing to the fast increase in H2S concentration to supraphysiological concentration, the change of intracellular pH if used in unbuffered solution, and possible toxicity characterized by pro-inflammatory effects [50,53]. In fact, in experimental studies, sulfide salts are frequently used at a very high concentration (100 μM to 10 mM), in great excess of the levels of H2S measured in vivo [52].
Unlike sulfide salts, the phosphorodithioate GYY4137 belongs to the class of organic slow-release H2S compounds, being able to release H2S over 3–4 h after dissolving [50]. GYY4137 has been demonstrated to exert vasodilator, antihypertensive, anti-atherosclerotic, and anti-thrombotic activities [38,41]. Whereas NaHS increases the synthesis of the interleukins (IL)-1β and -6, NO and prostaglandin E2 at a high concentration, GYY4137 inhibits the release of these pro-inflammatory mediators in a dose-dependent manner and also promotes the production of the anti-inflammatory chemokine IL-10 in lipopolysaccharide (LPS)-treated macrophages [53].
Diphosphorothioates such as AP67 and AP105 derive from structural modifications of the phosphorodithioate core and, if compared to GYY4137, have the advantages of being able to be employed at a lower concentration, showing an even enhanced activity [52]. The novel mitochondria-targeted AP39 and AP123 were found to reduce the high-glucose-induced hyperpolarization of the mitochondrial membrane and inhibit ROS production in microvascular ECs, with a long-lasting effect suggesting their application in the treatment of diabetic vascular complications [54]. The H2S prodrug sodium polysulthionate (SG1002) has been successfully used in both a swine model of acute limb ischemia [55] and in heart failure patients [56], in which it promotes an increase in circulating H2S and NO and, consequently, coronary artery vasorelaxation.
Garlic, which has been associated with multiple health beneficial effects in folk medicine for centuries, is rich in organosulfur compounds considered responsible for most of its pharmacological activities [57]. In particular, garlic-derived organic polysulfides like diallyl disulfide and diallyl trisulfide, as well as their analogs, act as H2S donors in the presence of GSH, and promote vasorelaxation (NO bioavailability) [57], lowering of arterial blood pressure [58], decreasing apoptosis and oxidative stress [59], and improved angiogenesis [60]. Similarly, the natural isothiocyanates commonly present in the Brassicaceae (e.g., broccoli, mustard, horseradish, rocket salad), can be considered as potential slow and long-lasting H2S donors able to release H2S in cell environments with high concentration of GSH and cysteine [61,62]. Sulforaphane and other isothiocyanates (i.e., benzyl isothiocyanate and phenethyl isocyanate) have anti-inflammatory properties mediated through the upregulation of HO-1 [63,64] and the glutamine cysteine ligase that plays a critical role in maintaining GSH homeostasis [63]. A diet based on sulforaphane-enriched foods was reported to significantly reduce oxidative stress related to improved endothelial-dependent relaxation of the aorta and lower blood pressure in spontaneously hypertensive rats [65].
Although L-cysteine is frequently employed in experimental studies to increase the production of endogenous H2S, it is not suitable for clinical use due to its unstable nature, being metabolized in a number of pathways including GSH synthesis, taurine synthesis and oxidation to sulfate [51]. Conversely, NAC, a well-tolerated compound used in clinical settings to enhance cellular levels of GSH, could represent a promising compound able to generate H2S [6], since the administration of NAC prevents the development of hypertension in rodents [66] and humans (Clinical Trial NCT01232257, 2011). Supplementation of taurine (2-aminoethanesulfonic acid), a metabolite of cysteine, has the ability to moderately lower blood pressure in prehypertensive subjects via endothelium-dependent and endothelium-independent vasodilation [67,68]. Like NAC, these antihypertensive effects are associated with increased expression of CSE and CBS, as observed both in the aorta of spontaneously hypertensive rats and in human vascular tissue cultures [67].
Synthetic cysteine derivatives such as S-propyl-cysteine, S-allyl-cysteine, and S-propargyl-cysteine (SPRC) are also enzymatically converted to H2S by CSE and CBS [50]. SPRC, though not available for clinical use [68], can attenuate the LPS-induced inflammatory response in cardiac myocytes by reducing the mRNA expression of TNF-α, ICAM-1 and iNOS [69], increase H2S levels through the upregulation of CSE, and protect HUVECs from TNF-α-induced dysfunction [70].

2.1.2. H2S-Hybrid Drugs

In addition to H2S donors, there is a group of compounds known as H2S-hybrid drugs that, while able to release H2S, have a mechanism of action independent of H2S-mediated properties [61]. The group of ACE inhibitors represents one of the fundamental drug classes for the antihypertensive treatment [52]. Many of them, e.g., Omaprilat, Remikiren, Macitentan, Bosentan, Vardenafil, Sildenafil, have a sulfonil moiety in their structure [51]. Captocapril contains a thiol that, in plasma, can react with other thiol-containing compounds (cysteine, GSH) to form mixed disulfides, however it remains to be clarified whether the drug’s effects depend on sulfide signaling [6,51]. On the other hand, the pro-angiogenic [71], anti-inflammatory [72], and anti-apoptotic [73] activities of Zofenopril in ECs are at least partially mediated by its ability to increase H2S availability [72]. Of interest, Zofenopril, but not Enalapril (a non-thiol ACE inhibitor), also improves the vascular response to acetylcholine in spontaneously hypertensive rats, which is accompanied by increased H2S concentration in the plasma and the vascular wall [74].
Non-steroidal anti-inflammatory drugs (NSAIDs) are among the most widely used classes of medicines [75]. H2S-releasing derivatives of NSAIDs, synthetized by the conjugation of the parent NSAID with a dithiolethione moiety, show improved efficacy and reduced toxicity (i.e., gastrolesivity), which are mainly attributable to intracellular H2S/GSH formation in comparison to the non-releasing H2S compounds [75,76,77]. In particular, S-aspirin (ACS14), which exerts anti-platelet [78] and antithrombotic [79] activity in vivo and in vitro, prevents the formation and development of atherosclerosis in ApoE-deficient mice [80] and attenuates the oxidative stress caused by methylglyoxal (a chemically active metabolite of glucose and fructose) and high glucose in vascular smooth cultured cells, indicating a possible future use in the treatment of diabetic patients [81]. Likewise, S-diclofenac (ACS15) produces marked anti-inflammatory effects but significantly less gastric toxicity than diclofenac [82]. Notably, this drug can inhibit the smooth muscle cell growth that has been recognized as a fundamental event in vascular injury in diseases such as atherosclerosis [83].

2.2. H2S-Producing Compounds: A Further Tool against COVID-19

A growing body of evidence supports the potential role of H2S as an effective host defense factor against SARS-CoV-2 [84,85] (Figure 2).
In a SARS-CoV-2 infection, a pro-inflammatory cytokine storm is a primary event characterized by increases in IL-1β, IL-6, and TNF-α [85], and even moderately elevated IL-6 levels have been associated with a high risk of respiratory failure in COVID-19 patients [86]. Interestingly, the 4-day change in ratio of IL-6 to IL-10 (a cytokine with anti-inflammatory effects involved both in innate and adaptive immunity), named the Dublin–Boston score, has proven to be a more reliable tool than IL-6 alone in predicting clinical progression and poor outcome in COVID-19 patients [87,88].
H2S can significantly downregulate the IL-6/STAT3 signaling pathway that is implicated in inflammatory responses and cell apoptosis [89]. Of note, serum H2S levels were found to inversely correlate with IL-6 as well as with the severity and final outcome of pneumonia in a cohort of patients with COVID-19, suggesting that the reduction of H2S bioavailability may be considered a biomarker of enhanced pro-inflammatory response, whereas exogenous administration of H2S could represent a valuable strategy to counteract severe manifestations of the infection [90,91]. In addition to decreasing IL-6 and IL-8 levels and the infiltration of polymorphonuclear cells, NaHS increases plasma levels of IL-10 in an animal model of induced acute lung injury [92], and administering IL-10 to IL-10 deficient mice in turn restores H2S production and homocysteine metabolism [93]. H2S has also been reported to play a role in enhancing T cell activation [94] and, among the variety of cell types capable to produce IL-10, IL-10 generation by CD4(+) T cells is crucial to preventing early mortality caused by excessive inflammation [87].
The anti-inflammatory effects of H2S further encompass the regulation of the ACE/ACE2 balance, although the underlying mechanisms remain elusive [95]. Specifically, NaHS may promote, dose-dependently, the expression of ACE2, a key enzymatic component of the renin-angiotensin-aldosterone system that is recognized to have vasodilating, anti-inflammatory, antioxidant, and antifibrotic effects, by catalyzing the generation of Ang (1–7) from AngII in ECs [96,97]. Conversely, NaHS treatment reduces ACE expression in spontaneously hypertensive rats [98], consistent with the recognized enhanced anti-hypertensive role of ACE-inhibitors containing a sulfur moiety in their chemical structure (see the previous section).
The role of H2S in acute and chronic inflammatory pulmonary diseases has been extensively investigated [91]. Besides anti-inflammatory activities (e.g., reduced mRNA expression of NF-kB, macrophage inflammatory protein-2, and interferon regulatory factor 3 [99,100]), H2S donors increase endogenous antioxidant defenses (e.g., SOD, glutathione peroxidase, and glutathione reductase) and inhibit leukocyte recruitment and transmigration through the inflamed endothelium [100,101]. Notably, there is evidence indicating that higher levels of GSH may improve an individual’s responsiveness to viral infections, protecting host cells from oxidative damage of the lung [102]. De Flora et al. observed that a 6-month preventive administration of NAC, a known precursor of GSH, provides a significant attenuation of influenza and influenza-like episodes, especially in the elderly [103]. The addition of NAC to a conventional therapy ameliorates oxidative stress and inflammation parameters (i.e., a decrease of malondialdehyde and TNF-α, and an increase of total antioxidant capacity) in patients with pneumonia [104]. NAC can act both as a direct scavenger of free radicals [105] and as a mucolytic agent capable of reducing disulphide bonds in heavily cross-linked mucus glycoproteins [106]. Moreover, the efficacy of NAC in the treatment of patients with chronic bronchitis and chronic obstructive pulmonary disease has been documented in several clinical trials and meta-analyses [107].
The levels of GSH are also negatively related with COVID-19 severity, and patients with moderate and severe illnesses show increased levels of ROS and a higher ROS/GSH ratio than subjects with mild symptoms [102]. Hence, it is sensible to hypothesize that replenishing intracellular GSH could be a useful strategy against SARS-CoV-2, as shown in two cases in which a glutathione-based therapy (GSH and NAC) combined with antioxidants (alpha-lipoic acid and vitamin C) was immediately effective in relieving symptoms of dyspnea [108]. In another case report, despite treatment with antibiotics, antiviral, and antibacterial medications, a 64-year-old male COVID-19 patient developed respiratory failure on the 13th day of admission. The patient’s conditions improved following NAC supplementation, and discharge occurred after 46 days of hospitalization [109]. In particular, on the basis of its mucolytic and antioxidant properties, it has been proposed that inclusion of 1200 mg/d oral NAC in the therapeutic schemes of patients with COVID-19 could be an effective measure to prevent a cytokine storm and the associated acute respiratory distress syndrome [110].
A case-control study found that both H2S and NO were significantly higher in expired COVID-19 patients compared to those who survived, emphasizing the complex interaction between these two gasotransmitters and a more synergistic role in this context [111]. The reasons for this increase can hint a compensatory response of sicker patients to the detrimental effects of COVID-19 infection or, alternatively, an underutilization of NO and H2S, which results in fatal outcomes [111].
Finally, it has recently been proposed that another mechanism of defense against COVID-19 could involve the activation of the HO-1/CO/H2S system [112]. Indeed, HO-1 metabolizes the heme group of a variety of hemeproteins with the release of CO which, under endoplasmic reticulum stress conditions, inhibits CBS with decreased production of GSH, while the SH groups are enzymatically converted to H2S by CSE [112,113]. Liu and Li have hypothesized that ORF8 and the surface glycoprotein of SARS-CoV-2 attack and destroy the hemes of heme proteins, resulting in the suppression of HO-1 activation and H2S signaling [114,115]. Subjects with a long promoter for the HMOX1 gene (associated with decreased HO-1 anticoagulant activity) present an increased risk of recurrent venous thromboembolism [116], whilst HO-1 has been demonstrated to exert a significant antiviral activity against a wide variety of viruses [117]. Therefore, activation of the HO-1/CO/H2S axis has the potential to improve the clinical manifestations of COVID-19, and pharmacological treatments based on H2S delivery could once again represent an effective strategy in the treatment of patients with COVID-19 [112].
In addition to counteracting the inflammatory response in COVID-19, H2S may interfere with viral replication [96]. In fact, there is some well-established evidence demonstrating that H2S inhibits the replications of many other highly pathogenic RNA viruses in lungs [118], both by decreasing the expression of viral proteins and mRNA and by inhibiting syncytium formation and virus assembly/release [95,99].
Overall, a large array of data indicate that H2S could be a potential target for attenuating viral replication, inflammation development and progression, and organ damage, which needs to be further explored in preclinical models of viral infections [95].

3. Discussion

Amid the complexity of events and effectors underlying processes leading to endothelial dysfunction, H2S has emerged as one of the crucial determinants of endothelial homeostasis. Hence, sulfur drugs can represent advanced tools in the prevention and treatment of the numerous diseases involving endothelial dysfunction. For these reasons, several compounds are currently being investigated in clinical studies with promising results.
On the other hand, it has been recognized that a high H2S concentration (i.e., >250 ppm/~350 mg/m3) is harmful to health, and is associated with increased oxidative stress and inflammation [119]. Less well known are the long-term effects of chronic low-dose H2S exposures in light of controversial results and due to differences between and limitations of the available studies [119].
In addition to endogenous production, it is also important to consider the intake of H2S from exogenous sources. Indeed, H2S may originate from a variety of natural sources, including volcanoes, sulfur springs, undersea vents, swamps, bogs, crude oil and natural gas, or from man-made activities, such as oil refineries, tanneries, natural gas, products petrochemicals, ovens, food processing plants, municipal sewage and wastewater treatment plants, fertilization processes, and paper mills. Occupational exposure to H2S is therefore greater than that from environmental sources [119]. Furthermore, smokers have low serum H2S levels, while chronic alcohol users have high levels of H2S in their breath, suggesting that smoking and alcohol consumption may modulate endogenous H2S concentration [120].
Dietary intake can also affect endogenous levels of H2S. In addition to garlic, broccoli, mustard, etc., a high consumption of proteins and fats or a high carbohydrate content in the diet also appears to increase and reduce H2S levels, respectively [121]. In particular, the intake of sulfur in the diet, together with the presence and different composition of sulfate-reducing bacteria in the gastrointestinal tract, represent the most significant modulators of H2S production [121].
In this context, the discovery that H2S can protect the mucus layer and reduce inflammation when produced at nanomolar to low micromolar levels, exerting adverse effects when released at a higher concentration (from high micromolar to millimolar) by the local microbiota, is interesting because it has highlighted the double face of H2S in the balance between beneficial and harmful effects [122].
Although H2S donors may represent valuable tools to protect the endothelium, further clinical studies targeting their effect on the endothelium in terms of reducing or slowing the progression of dysfunction in specific diseases (e.g., COVID-19) would be desirable. Furthermore, it is currently unknown whether any drugs added to endogenous levels resulting from environmental exposure, lifestyle habits (cigarette smoking or alcohol consumption and diet) and microbiota activity could reach toxic concentrations, and this requires further refinement work towards a more personalized and targeted therapy for each patient.

Author Contributions

Conceptualization, F.G., S.D.T. and C.V.; writing—original draft preparation, F.G., S.D.T. and C.V.; writing—review and editing, L.S. and M.G. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Medina-Leyte, D.J.; Zepeda-García, O.; Domínguez-Pérez, M.; González-Garrido, A.; Villarreal-Molina, T.; Jacobo-Albavera, L. Endothelial Dysfunction, Inflammation and Coronary Artery Disease: Potential Biomarkers and Promising Therapeutical Approaches. Int. J. Mol. Sci. 2021, 22, 3850. [Google Scholar] [CrossRef]
  2. Xu, S.; Ilyas, I.; Little, P.J.; Li, H.; Kamato, D.; Zheng, X.; Luo, S.; Li, Z.; Liu, P.; Han, J.; et al. Endothelial Dysfunction in Atherosclerotic Cardiovascular Diseases and Beyond: From Mechanism to Pharmacotherapies. Pharmacol Rev. 2021, 73, 924–967. [Google Scholar] [CrossRef]
  3. Dou, Q.; Wei, X.; Zhou, K.; Yang, S.; Jia, P. Cardiovascular Manifestations and Mechanisms in Patients with COVID-19. Trends Endocrinol. Metab. 2020, 31, 893–904. [Google Scholar] [CrossRef]
  4. Guzik, T.J.; Mohiddin, S.A.; Dimarco, A.; Patel, V.; Savvatis, K.; Marelli-Berg, F.M.; Madhur, M.S.; Tomaszewski, M.; Maffia, P.; D’Acquisto, F.; et al. COVID-19 and the cardiovascular system: Implications for risk assessment, diagnosis, and treatment options. Cardiovasc. Res. 2020, 116, 1666–1687. [Google Scholar] [CrossRef] [PubMed]
  5. Pan, L.L.; Qin, M.; Liu, X.H.; Zhu, Y.Z. The Role of Hydrogen Sulfide on Cardiovascular Homeostasis: An Overview with Update on Immunomodulation. Front. Pharmacol. 2017, 8, 686. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  6. Ciccone, V.; Genah, S.; Morbidelli, L. Endothelium as a Source and Target of H2S to Improve Its Trophism and Function. Antioxidants (Basel) 2021, 10, 486. [Google Scholar] [CrossRef]
  7. Wang, R.; Szabo, C.; Ichinose, F.; Ahmed, A.; Whiteman, M.; Papapetropoulos, A. The role of H2S bioavailability in endothelial dysfunction. Trends Pharmacol. Sci. 2015, 36, 568–578. [Google Scholar] [CrossRef] [Green Version]
  8. Xiao, Q.; Ying, J.; Xiang, L.; Zhang, C. The biologic effect of hydrogen sulfide and its function in various diseases. Medicine (Baltimore) 2018, 97, e13065. [Google Scholar] [CrossRef]
  9. Wallace, J.L.; Wang, R. Hydrogen sulfide-based therapeutics: Exploiting a unique but ubiquitous gasotransmitter. Nat. Rev. Drug Discov. 2015, 14, 329–345. [Google Scholar] [CrossRef]
  10. Majtan, T.; Krijt, J.; Sokolová, J.; Křížková, M.; Ralat, M.A.; Kent, J.; Gregory, J.F., 3rd; Kožich, V.; Kraus, J.P. Biogenesis of Hydrogen Sulfide and Thioethers by Cystathionine Beta-Synthase. Antioxid. Redox Signal. 2018, 8, 311–323. [Google Scholar] [CrossRef]
  11. Kanagy, N.L.; Szabo, C.; Papapetropoulos, A. Vascular biology of hydrogen sulfide. Am. J. Physiol. Cell Physiol. 2017, 312, C537–C549. [Google Scholar] [CrossRef]
  12. Meng, G.; Zhao, S.; Xie, L.; Han, Y.; Ji, Y. Protein S-sulfhydration by hydrogen sulfide in cardiovascular system. Br. J. Pharmacol. 2018, 175, 1146–1156. [Google Scholar] [CrossRef] [PubMed]
  13. 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] [PubMed]
  14. Mustafa, A.K.; Sikka, G.; Gazi, S.K.; Steppan, J.; Jung, S.M.; Bhunia, A.K.; Barodka, V.M.; Gazi, F.K.; Barrow, R.K.; Wang, R.; et al. Hydrogen sulfide as endothelium-derived hyperpolarizing factor sulfhydrates potassium channels. Circ. Res. 2011, 109, 1259–1268. [Google Scholar] [CrossRef]
  15. Yan, H.; Du, J.; Tang, C. The possible role of hydrogen sulfide on the pathogenesis of spontaneous hypertension in rats. Biochem. Biophys Res. Commun. 2004, 313, 22–27. [Google Scholar] [CrossRef]
  16. 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] [PubMed]
  17. Whiteman, M.; Armstrong, J.S.; Chu, S.H.; Jia-Ling, S.; Wong, B.S.; Cheung, N.S.; Halliwell, B.; Moore, P.K. The novel neuromodulator hydrogen sulfide: An endogenous peroxynitrite ‘scavenger’? J. Neurochem. 2004, 90, 765–768. [Google Scholar] [CrossRef]
  18. Yang, G.; Zhao, K.; Ju, Y.; Mani, S.; Cao, Q.; Puukila, S.; Khaper, N.; Wu, L.; Wang, R. Hydrogen sulfide protects against cellular senescence via S-sulfhydration of Keap1 and activation of Nrf2. Antioxid. Redox Signal. 2013, 18, 1906–1919. [Google Scholar] [CrossRef] [PubMed]
  19. Guo, C.; Liang, F.; Shah Masood, W.; Yan, X. Hydrogen sulfide protected gastric epithelial cell from ischemia/reperfusion injury by Keap1 s-sulfhydration, MAPK dependent anti-apoptosis and NF-κB dependent anti-inflammation pathway. Eur. J. Pharmacol. 2014, 725, 70–78. [Google Scholar] [CrossRef]
  20. Wen, Y.D.; Wang, H.; Kho, S.H.; Rinkiko, S.; Sheng, X.; Shen, H.M.; Zhu, Y.Z. Hydrogen sulfide protects HUVECs against hydrogen peroxide induced mitochondrial dysfunction and oxidative stress. PLoS ONE 2013, 8, e53147. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  21. Zhao, K.; Ju, Y.; Li, S.; Altaany, Z.; Wang, R.; Yang, G. S-sulfhydration of MEK1 leads to PARP-1 activation and DNA damage repair. EMBO Rep. 2014, 15, 792–800. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  22. Li, L.; Rose, P.; Moore, P.K. Hydrogen sulfide and cell signaling. Annu. Rev. Pharmacol. Toxicol. 2011, 51, 169–187. [Google Scholar] [CrossRef] [Green Version]
  23. Sen, N.; Paul, B.D.; Gadalla, M.M.; Mustafa, A.K.; Sen, T.; Xu, R.; Kim, S.; Snyder, S.H. Hydrogen sulfide-linked sulfhydration of NF-κB mediates its antiapoptotic actions. Mol. Cell. 2012, 45, 13–24. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. 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] [Green Version]
  25. Hu, H.J.; Jiang, Z.S.; Zhou, S.H.; Liu, Q.M. Hydrogen sulfide suppresses angiotensin II-stimulated endothelin-1 generation and subsequent cytotoxicity-induced endoplasmic reticulum stress in endothelial cells via NF-κB. Mol. Med. Rep. 2016, 14, 4729–4740. [Google Scholar] [CrossRef]
  26. Guan, Q.; Wang, X.; Gao, L.; Chen, J.; Liu, Y.; Yu, C.; Zhang, N.; Zhang, X.; Zhao, J. Hydrogen sulfide suppresses high glucose-induced expression of intercellular adhesion molecule-1 in endothelial cells. J. Cardiovasc. Pharmacol. 2013, 62, 278–284. [Google Scholar] [CrossRef] [PubMed]
  27. Liu, J.; Wu, J.; Sun, A.; Sun, Y.; Yu, X.; Liu, N.; Dong, S.; Yang, F.; Zhang, L.; Zhong, X.; et al. Hydrogen sulfide decreases high glucose/palmitate-induced autophagy in endothelial cells by the Nrf2-ROS-AMPK signaling pathway. Cell Biosci. 2016, 6, 33. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  28. Jain, S.K.; Bull, R.; Rains, J.L.; Bass, P.F.; Levine, S.N.; Reddy, S.; McVie, R.; Bocchini, J.A. Low levels of hydrogen sulfide in the blood of diabetes patients and streptozotocin-treated rats causes vascular inflammation? Antioxid. Redox Signal. 2010, 12, 1333–1337. [Google Scholar] [CrossRef]
  29. Suzuki, K.; Sagara, M.; Aoki, C.; Tanaka, S.; Aso, Y. Clinical Implication of Plasma Hydrogen Sulfide Levels in Japanese Patients with Type 2 Diabetes. Intern. Med. 2017, 56, 17–21. [Google Scholar] [CrossRef] [Green Version]
  30. Boubeta, F.M.; Bieza, S.A.; Bringas, M.; Palermo, J.C.; Boechi, L.; Estrin, D.A.; Bari, S.E. Hemeproteins as Targets for Sulfide Species. Antioxid. Redox Signal. 2020, 32, 247–257. [Google Scholar] [CrossRef]
  31. Ríos-González, B.B.; Román-Morales, E.M.; Pietri, R.; López-Garriga, J. Hydrogen sulfide activation in hemeproteins: The sulfheme scenario. J. Inorg. Biochem. 2014, 133, 78–86. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  32. Zhou, Z.; Martin, E.; Sharina, I.; Esposito, I.; Szabo, C.; Bucci, M.; Cirino, G.; Papapetropoulos, A. Regulation of soluble guanylyl cyclase redox state by hydrogen sulfide. Pharmacol. Res. 2016, 111, 556–562. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Bauer, C.C.; Boyle, J.P.; Porter, K.E.; Peers, C. Modulation of Ca(2+) signalling in human vascular endothelial cells by hydrogen sulfide. Atherosclerosis 2010, 209, 374–380. [Google Scholar] [CrossRef] [PubMed]
  34. Moccia, F.; Bertoni, G.; Pla, A.F.; Dragoni, S.; Pupo, E.; Merlino, A.; Mancardi, D.; Munaron, L.; Tanzi, F. Hydrogen sulfide regulates intracellular Ca2+ concentration in endothelial cells from excised rat aorta. Curr. Pharm. Biotechnol. 2011, 12, 1416–1426. [Google Scholar] [CrossRef] [PubMed]
  35. Szabó, C.; Papapetropoulos, A. Hydrogen sulphide and angiogenesis: Mechanisms and applications. Br. J. Pharmacol. 2011, 164, 853–865. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  36. Jiang, B.; Tang, G.; Cao, K.; Wu, L.; Wang, R. Molecular mechanism for H2S-induced activation of K(ATP) channels. Antioxid. Redox Signal. 2010, 12, 1167–1178. [Google Scholar] [CrossRef]
  37. Tao, H.; Shi, K.H.; Yang, J.J. Vascular endothelial growth factor: A novel potential therapeutic target for hypertension. J. Clin. Hypertens. (Greenwich) 2013, 15, 514. [Google Scholar] [CrossRef]
  38. 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]
  39. Du, C.; Lin, X.; Xu, W.; Zheng, F.; Cai, J.; Yang, J.; Cui, Q.; Tang, C.; Cai, J.; Xu, G.; et al. Sulfhydrated Sirtuin-1 Increasing Its Deacetylation Activity Is an Essential Epigenetics Mechanism of Anti-Atherogenesis by Hydrogen Sulfide. Antioxid. Redox Signal. 2019, 30, 184–197. [Google Scholar] [CrossRef]
  40. Kram, L.; Grambow, E.; Mueller-Graf, F.; Sorg, H.; Vollmar, B. The anti-thrombotic effect of hydrogen sulfide is partly mediated by an upregulation of nitric oxide synthases. Thromb. Res. 2013, 132, e112–e117. [Google Scholar] [CrossRef] [Green Version]
  41. 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]
  42. 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]
  43. Kharma, A.; Grman, M.; Misak, A.; Domínguez-Álvarez, E.; Nasim, M.J.; Ondrias, K.; Chovanec, M.; Jacob, C. Inorganic Polysulfides and Related Reactive Sulfur–Selenium Species from the Perspective of Chemistry. Molecules 2019, 24, 1359. [Google Scholar] [CrossRef] [Green Version]
  44. Kimura, H. Hydrogen sulfide and polysulfides as signaling molecules. Proc. Jpn. Acad. Ser. B Phys. Biol. Sci. 2015, 91, 131–159. [Google Scholar] [CrossRef] [Green Version]
  45. Toohey, J.I. Sulfur signaling: Is the agent sulfide or sulfane? Anal. Biochem. 2011, 413, 1–7. [Google Scholar] [CrossRef]
  46. Kimura, Y.; Toyofuku, Y.; Koike, S.; Shibuya, N.; Nagahara, N.; Lefer, D.; Ogasawara, Y.; Kimura, H. Identification of H2S3 and H2S produced by 3-mercaptopyruvate sulfurtransferase in the brain. Sci. Rep. 2015, 5, 14774. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  47. Olson, K.R.; Gao, Y.; Arif, F.; Arora, K.; Patel, S.; DeLeon, E.R.; Sutton, T.R.; Feelisch, M.; Cortese-Krott, M.M.; Straub, K.D. Metabolism of hydrogen sulfide (H2S) and Production of Reactive Sulfur Species (RSS) by superoxide dismutase. Redox. Biol. 2018, 15, 74–85. [Google Scholar] [CrossRef] [PubMed]
  48. Kimura, H. Hydrogen polysulfide (H2Sn) signaling along with hydrogen sulfide (H2S) and nitric oxide (NO). J. Neural Transm. (Vienna) 2016, 123, 1235–1245. [Google Scholar] [CrossRef] [PubMed]
  49. Kimura, Y.; Koike, S.; Shibuya, N.; Lefer, D.; Ogasawara, Y.; Kimura, H. 3-Mercaptopyruvate sulfurtransferase produces potential redox regulators cysteine- and glutathione-persulfide (Cys-SSH and GSSH) together with signaling molecules H2S2, H2S3 and H2S. Sci. Rep. 2017, 7, 10459. [Google Scholar] [CrossRef] [PubMed]
  50. Bełtowski, J. Hydrogen sulfide in pharmacology and medicine—An update. Pharmacol. Rep. 2015, 67, 647–658. [Google Scholar] [CrossRef] [PubMed]
  51. Zaorska, E.; Tomasova, L.; Koszelewski, D.; Ostaszewski, R.; Ufnal, M. Hydrogen Sulfide in Pharmacotherapy, Beyond the Hydrogen Sulfide-Donors. Biomolecules 2020, 10, 323. [Google Scholar] [CrossRef] [Green Version]
  52. Whiteman, M.; Perry, A.; Zhou, Z.; Bucci, M.; Papapetropoulos, A.; Cirino, G.; Wood, M.E. Phosphinodithioate and Phosphoramidodithioate Hydrogen Sulfide Donors. Handb. Exp. Pharmacol. 2015, 230, 337–363. [Google Scholar] [CrossRef]
  53. Whiteman, M.; Li, L.; Rose, P.; Tan, C.H.; Parkinson, D.B.; Moore, P.K. The effect of hydrogen sulfide donors on lipopolysaccharide-induced formation of inflammatory mediators in macrophages. Antioxid. Redox Signal. 2010, 12, 1147–1154. [Google Scholar] [CrossRef] [PubMed]
  54. Gerő, D.; Torregrossa, R.; Perry, A.; Waters, A.; Le-Trionnaire, S.; Whatmore, J.L.; Wood, M.; Whiteman, M. The novel mitochondria-targeted hydrogen sulfide (H2S) donors AP123 and AP39 protect against hyperglycemic injury in microvascular endothelial cells in vitro. Pharmacol. Res. 2016, 113 Pt A, 186–198. [Google Scholar] [CrossRef]
  55. Rushing, A.M.; Donnarumma, E.; Polhemus, D.J.; Au, K.R.; Victoria, S.E.; Schumacher, J.D.; Li, Z.; Jenkins, J.S.; Lefer, D.J.; Goodchild, T.T. Effects of a novel hydrogen sulfide prodrug in a porcine model of acute limb ischemia. J. Vasc. Surg. 2019, 69, 1924–1935. [Google Scholar] [CrossRef]
  56. Polhemus, D.J.; Li, Z.; Pattillo, C.B.; Gojon, G., Sr.; Gojon, G., Jr.; Giordano, T.; Krum, H. A novel hydrogen sulfide prodrug, SG1002, promotes hydrogen sulfide and nitric oxide bioavailability in heart failure patients. Cardiovasc. Ther. 2015, 33, 216–226. [Google Scholar] [CrossRef] [PubMed]
  57. Benavides, G.A.; Squadrito, G.L.; Mills, R.W.; Patel, H.D.; Isbell, T.S.; Patel, R.P.; Darley-Usmar, V.M.; Doeller, J.E.; Kraus, D.W. Hydrogen sulfide mediates the vasoactivity of garlic. Proc. Natl. Acad. Sci. USA 2007, 104, 17977–17982. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  58. Sharma, D.K.; Manral, A.; Saini, V.; Singh, A.; Srinivasan, B.P.; Tiwari, M. Novel diallyldisulfide analogs ameliorate cardiovascular remodeling in rats with L-NAME-induced hypertension. Eur. J. Pharmacol. 2012, 691, 198–208. [Google Scholar] [CrossRef]
  59. Hayashida, R.; Kondo, K.; Morita, S.; Unno, K.; Shintani, S.; Shimizu, Y.; Calvert, J.W.; Shibata, R.; Murohara, T. Diallyl Trisulfide Augments Ischemia-Induced Angiogenesis via an Endothelial Nitric Oxide Synthase-Dependent Mechanism. Circ. J. 2017, 81, 870–878. [Google Scholar] [CrossRef] [Green Version]
  60. Yang, H.B.; Liu, H.M.; Yan, J.C.; Lu, Z.Y. Effect of Diallyl Trisulfide on Ischemic Tissue Injury and Revascularization in a Diabetic Mouse Model. J. Cardiovasc. Pharmacol. 2018, 71, 367–374. [Google Scholar] [CrossRef]
  61. Calderone, V.; Martelli, A.; Testai, L.; Citi, V.; Breschi, M.C. Using hydrogen sulfide to design and develop drugs. Expert Opin. Drug. Discov. 2016, 11, 163–175. [Google Scholar] [CrossRef]
  62. Martelli, A.; Piragine, E.; Citi, V.; Testai, L.; Pagnotta, E.; Ugolini, L.; Lazzeri, L.; Di Cesare Mannelli, L.; Manzo, O.L.; Bucci, M.; et al. Erucin exhibits vasorelaxing effects and antihypertensive activity by H2 S-releasing properties. Br. J. Pharmacol. 2020, 177, 824–835. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Huang, C.S.; Lin, A.H.; Liu, C.T.; Tsai, C.W.; Chang, I.S.; Chen, H.W.; Lii, C.K. Isothiocyanates protect against oxidized LDL-induced endothelial dysfunction by upregulating Nrf2-dependent antioxidation and suppressing NFκB activation. Mol. Nutr. Food Res. 2013, 57, 1918–1930. [Google Scholar] [CrossRef]
  64. Kashfi, K.; Olson, K.R. Biology and therapeutic potential of hydrogen sulfide and hydrogen sulfide-releasing chimeras. Biochem. Pharmacol. 2013, 85, 689–703. [Google Scholar] [CrossRef] [Green Version]
  65. Wu, L.; Noyan Ashraf, M.H.; Facci, M.; Wang, R.; Paterson, P.G.; Ferrie, A.; Juurlink, B.H. Dietary approach to attenuate oxidative stress, hypertension, and inflammation in the cardiovascular system. Proc. Natl. Acad. Sci. USA 2004, 101, 7094–7099. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  66. Nagababu, E.; Steppan, J.; Santhanam, L.; Frank, S.M.; Berkowitz, D.E. Effect of Nitrite and N-acetylcysteine Treatment on Blood Pressure, Arterial Stiffness and Vascular Function in Spontaneously Hypertensive Rats. Free Radic. Biol. Med. 2017, 112, 118–119. [Google Scholar] [CrossRef]
  67. Sun, Q.; Wang, B.; Li, Y.; Sun, F.; Li, P.; Xia, W.; Zhou, X.; Li, Q.; Wang, X.; Chen, J.; et al. Taurine Supplementation Lowers Blood Pressure and Improves Vascular Function in Prehypertension: Randomized, Double-Blind, Placebo-Controlled Study. Hypertension 2016, 67, 541–549. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  68. DiNicolantonio, J.J.; OKeefe, J.H.; McCarty, M.F. Boosting endogenous production of vasoprotective hydrogen sulfide via supplementation with taurine and N-acetylcysteine: A novel way to promote cardiovascular health. Open Heart 2017, 4, e000600. [Google Scholar] [CrossRef] [PubMed]
  69. Pan, L.L.; Liu, X.H.; Gong, Q.H.; Zhu, Y.Z. S-Propargyl-cysteine (SPRC) attenuated lipopolysaccharide-induced inflammatory response in H9c2 cells involved in a hydrogen sulfide-dependent mechanism. Amino Acids 2011, 41, 205–215. [Google Scholar] [CrossRef] [PubMed]
  70. Pan, L.L.; Liu, X.H.; Zheng, H.M.; Yang, H.B.; Gong, Q.H.; Zhu, Y.Z. S-propargyl-cysteine, a novel hydrogen sulfide-modulated agent, attenuated tumor necrosis factor-α-induced inflammatory signaling and dysfunction in endothelial cells. Int. J. Cardiol. 2012, 155, 327–332. [Google Scholar] [CrossRef]
  71. Terzuoli, E.; Monti, M.; Vellecco, V.; Bucci, M.; Cirino, G.; Ziche, M.; Morbidelli, L. Characterization of zofenoprilat as an inducer of functional angiogenesis through increased H2 S availability. Br. J. Pharmacol. 2015, 172, 2961–2973. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  72. Monti, M.; Terzuoli, E.; Ziche, M.; Morbidelli, L. H2S dependent and independent anti-inflammatory activity of zofenoprilat in cells of the vascular wall. Pharmacol. Res. 2016, 113, 426–437. [Google Scholar] [CrossRef]
  73. Monti, M.; Terzuoli, E.; Ziche, M.; Morbidelli, L. The sulphydryl containing ACE inhibitor Zofenoprilat protects coronary endothelium from Doxorubicin-induced apoptosis. Pharmacol. Res. 2013, 76, 171–181. [Google Scholar] [CrossRef]
  74. Bucci, M.; Vellecco, V.; Cantalupo, A.; Brancaleone, V.; Zhou, Z.; Evangelista, S.; Calderone, V.; Papapetropoulos, A.; Cirino, G. Hydrogen sulfide accounts for the peripheral vascular effects of zofenopril independently of ACE inhibition. Cardiovasc. Res. 2014, 102, 138–147. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  75. Wallace, J.L. Hydrogen sulfide-releasing anti-inflammatory drugs. Trends Pharmacol. Sci. 2007, 28, 501–505. [Google Scholar] [CrossRef]
  76. Sparatore, A.; Perrino, E.; Tazzari, V.; Giustarini, D.; Rossi, R.; Rossoni, G.; Erdmann, K.; Schröder, H.; Del Soldato, P. Pharmacological profile of a novel H2S-releasing aspirin. Free Radic. Biol. Med. 2009, 46, 586–592. [Google Scholar] [CrossRef]
  77. Liu, L.; Cui, J.; Song, C.J.; Bian, J.S.; Sparatore, A.; Soldato, P.D.; Wang, X.Y.; Yan, C.D. H2S-releasing aspirin protects against aspirin-induced gastric injury via reducing oxidative stress. PLoS ONE 2012, 7, e46301. [Google Scholar] [CrossRef]
  78. Gao, L.; Cheng, C.; Sparatore, A.; Zhang, H.; Wang, C. Hydrogen sulfide inhibits human platelet aggregation in vitro in part by interfering gap junction channels: Effects of ACS14, a hydrogen sulfide-releasing aspirin. Heart Lung Circ. 2015, 24, 77–85. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  79. Pircher, J.; Fochler, F.; Czermak, T.; Mannell, H.; Kraemer, B.F.; Wörnle, M.; Sparatore, A.; Del Soldato, P.; Pohl, U.; Krötz, F. Hydrogen sulfide-releasing aspirin derivative ACS14 exerts strong antithrombotic effects in vitro and in vivo. Arterioscler. Thromb. Vasc. Biol. 2012, 32, 2884–2891. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  80. Zhang, H.; Guo, C.; Zhang, A.; Fan, Y.; Gu, T.; Wu, D.; Sparatore, A.; Wang, C. Effect of S-aspirin, a novel hydrogen-sulfide-releasing aspirin (ACS14), on atherosclerosis in apoE-deficient mice. Eur. J. Pharmacol. 2012, 697, 106–116. [Google Scholar] [CrossRef]
  81. Huang, Q.; Sparatore, A.; Del Soldato, P.; Wu, L.; Desai, K. Hydrogen sulfide releasing aspirin, ACS14, attenuates high glucose-induced increased methylglyoxal and oxidative stress in cultured vascular smooth muscle cells. PLoS ONE 2014, 9, e97315. [Google Scholar] [CrossRef] [PubMed]
  82. Li, L.; Rossoni, G.; Sparatore, A.; Lee, L.C.; Del Soldato, P.; Moore, P.K. Anti-inflammatory and gastrointestinal effects of a novel diclofenac derivative. Free Radic. Biol. Med. 2007, 42, 706–719. [Google Scholar] [CrossRef] [PubMed]
  83. Baskar, R.; Sparatore, A.; Del Soldato, P.; Moore, P.K. Effect of S-diclofenac, a novel hydrogen sulfide releasing derivative inhibit rat vascular smooth muscle cell proliferation. Eur. J. Pharmacol. 2008, 594, 1–8. [Google Scholar] [CrossRef] [PubMed]
  84. 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]
  85. Bourgonje, A.R.; Offringa, A.K.; van Eijk, L.E.; Abdulle, A.E.; Hillebrands, J.L.; van der Voort, P.H.J.; van Goor, H.; van Hezik, E.J. N-Acetylcysteine and Hydrogen Sulfide in Coronavirus Disease 2019. Antioxid. Redox Signal. 2021. [Google Scholar] [CrossRef] [PubMed]
  86. Herold, T.; Jurinovic, V.; Arnreich, C.; Hellmuth, J.C.; von Bergwelt-Baildon, M.; Klein, M.; Weinberger, T. Level of IL-6 Predicts Respiratory Failure in Hospitalized Symptomatic COVID-19 Patients. medRxiv 2020. [Google Scholar] [CrossRef]
  87. Trinchieri, G. Interleukin-10 production by effector T cells: Th1 cells show self control. J. Exp. Med. 2007, 204, 239–243. [Google Scholar] [CrossRef]
  88. McElvaney, O.J.; Hobbs, B.D.; Qiao, D.; McElvaney, O.F.; Moll, M.; McEvoy, N.L.; Clarke, J.; O’Connor, E.; Walsh, S.; Cho, M.H.; et al. A linear prognostic score based on the ratio of interleukin-6 to interleukin-10 predicts outcomes in COVID-19. EBioMedicine 2020, 61, 103026. [Google Scholar] [CrossRef]
  89. Fouad, A.A.; Hafez, H.M.; Hamouda, A. Hydrogen sulfide modulates IL-6/STAT3 pathway and inhibits oxidative stress, inflammation, and apoptosis in rat model of methotrexate hepatotoxicity. Hum. Exp. Toxicol. 2020, 39, 77–85. [Google Scholar] [CrossRef]
  90. Renieris, G.; Katrini, K.; Damoulari, C.; Akinosoglou, K.; Psarrakis, C.; Kyriakopoulou, M.; Dimopoulos, G.; Lada, M.; Koufargyris, P.; Giamarellos-Bourboulis, E.J. Serum Hydrogen Sulfide and Outcome Association in Pneumonia by the SARS-CoV-2 Coronavirus. Shock 2020, 54, 633–637. [Google Scholar] [CrossRef]
  91. Citi, V.; Martelli, A.; Gorica, E.; Brogi, S.; Testai, L.; Calderone, V. Role of hydrogen sulfide in endothelial dysfunction: Pathophysiology and therapeutic approaches. J. Adv. Res. 2020, 27, 99–113. [Google Scholar] [CrossRef] [PubMed]
  92. 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. (Maywood) 2008, 233, 1081–1087. [Google Scholar] [CrossRef] [PubMed]
  93. Flannigan, K.L.; Agbor, T.A.; Blackler, R.W.; Kim, J.J.; Khan, W.I.; Verdu, E.F.; Ferraz, J.G.; Wallace, J.L. Impaired hydrogen sulfide synthesis and IL-10 signaling underlie hyperhomocysteinemia-associated exacerbation of colitis. Proc. Natl. Acad. Sci. USA 2014, 111, 13559–13564. [Google Scholar] [CrossRef] [Green Version]
  94. Miller, T.W.; Wang, E.A.; Gould, S.; Stein, E.V.; Kaur, S.; Lim, L.; Amarnath, S.; Fowler, D.H.; Roberts, D.D. Hydrogen sulfide is an endogenous potentiator of T cell activation. J. Biol. Chem. 2012, 287, 4211–4221. [Google Scholar] [CrossRef] [Green Version]
  95. Yang, G. H2S as a potential defense against COVID-19? Am. J. Physiol. Cell Physiol. 2020, 319, C244–C249. [Google Scholar] [CrossRef] [PubMed]
  96. Danser, A.H.J.; Epstein, M.; Batlle, D. Renin-Angiotensin System Blockers and the COVID-19 Pandemic: At Present There Is No Evidence to Abandon Renin-Angiotensin System Blockers. Hypertension 2020, 75, 1382–1385. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  97. 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] [Green Version]
  98. Tain, Y.L.; Hsu, C.N.; Lu, P.C. Early short-term treatment with exogenous hydrogen sulfide postpones the transition from prehypertension to hypertension in spontaneously hypertensive rat. Clin. Exp. Hypertens. 2018, 40, 58–64. [Google Scholar] [CrossRef]
  99. 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] [PubMed] [Green Version]
  100. Faller, S.; Hausler, F.; Goeft, A.; von Itter, M.A.; Gyllenram, V.; Hoetzel, A.; Spassov, S.G. Hydrogen sulfide limits neutrophil transmigration, inflammation, and oxidative burst in lipopolysaccharide-induced acute lung injury. Sci Rep. 2018, 8, 14676. [Google Scholar] [CrossRef] [Green Version]
  101. Benetti, L.R.; Campos, D.; Gurgueira, S.A.; Vercesi, A.E.; Guedes, C.E.; Santos, K.L.; Wallace, J.L.; Teixeira, S.A.; Florenzano, J.; Costa, S.K.; et al. Hydrogen sulfide inhibits oxidative stress in lungs from allergic mice in vivo. Eur. J. Pharmacol. 2013, 698, 463–469. [Google Scholar] [CrossRef] [Green Version]
  102. Polonikov, A. Endogenous Deficiency of Glutathione as the Most Likely Cause of Serious Manifestations and Death in COVID-19 Patients. ACS Infect. Dis. 2020, 6, 1558–1562. [Google Scholar] [CrossRef] [PubMed]
  103. De Flora, S.; Grassi, C.; Carati, L. Attenuation of influenza-like symptomatology and improvement of cell-mediated immunity with long-term N-acetylcysteine treatment. Eur. Respir. J. 1997, 10, 1535–1541. [Google Scholar] [CrossRef] [PubMed]
  104. Zhang, Q.; Ju, Y.; Ma, Y.; Wang, T. N-acetylcysteine improves oxidative stress and inflammatory response in patients with community acquired pneumonia: A randomized controlled trial. Medicine (Baltimore) 2018, 97, e13087. [Google Scholar] [CrossRef] [PubMed]
  105. Benrahmoune, M.; Thérond, P.; Abedinzadeh, Z. The reaction of superoxide radical with N-acetylcysteine. Free Radic. Biol. Med. 2000, 29, 775–782. [Google Scholar] [CrossRef]
  106. Aldini, G.; Altomare, A.; Baron, G.; Vistoli, G.; Carini, M.; Borsani, L.; Sergio, F. N-Acetylcysteine as an antioxidant and disulphide breaking agent: The reasons why. Free Radic. Res. 2018, 52, 751–762. [Google Scholar] [CrossRef]
  107. Santus, P.; Corsico, A.; Solidoro, P.; Braido, F.; Di Marco, F.; Scichilone, N. Oxidative stress and respiratory system: Pharmacological and clinical reappraisal of N-acetylcysteine. COPD 2014, 11, 705–717. [Google Scholar] [CrossRef]
  108. Horowitz, R.I.; Freeman, P.R.; Bruzzese, J. Efficacy of glutathione therapy in relieving dyspnea associated with COVID-19 pneumonia: A report of 2 cases. Respir. Med. Case Rep. 2020, 30, 101063. [Google Scholar] [CrossRef]
  109. Liu, Y.; Wang, M.; Luo, G.; Qian, X.; Wu, C.; Zhang, Y.; Chen, B.; Leung, E.L.; Tang, Y. Experience of N-acetylcysteine airway management in the successful treatment of one case of critical condition with COVID-19: A case report. Medicine (Baltimore) 2020, 99, e22577. [Google Scholar] [CrossRef]
  110. 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]
  111. Dominic, P.; Ahmad, J.; Bhandari, R.; Pardue, S.; Solorzano, J.; Jaisingh, K.; Watts, M.; Bailey, S.R.; Orr, A.W.; Kevil, C.G.; et al. Decreased availability of nitric oxide and hydrogen sulfide is a hallmark of COVID-19. Redox Biol. 2021, 43, 101982. [Google Scholar] [CrossRef]
  112. Dattilo, M. The role of host defences in Covid 19 and treatments thereof. Mol. Med. 2020, 26, 90. [Google Scholar] [CrossRef]
  113. Kabil, O.; Yadav, V.; Banerjee, R. Heme-dependent Metabolite Switching Regulates H2S Synthesis in Response to Endoplasmic Reticulum (ER) Stress. J. Biol. Chem. 2016, 291, 16418–16423. [Google Scholar] [CrossRef] [Green Version]
  114. Liu, W.; Li, H. COVID-19 Disease: ORF8 and Surface Glycoprotein Inhibit Heme Metabolism by Binding to Porphyrin. ChemRxiv Preprint 2020. [Google Scholar] [CrossRef] [Green Version]
  115. Liu, W.; Li, H. COVID-19: Attacks the 1-Beta chain of hemoglobin and captures the Porphyrin to inhibit human Heme metabolism. ChemRxiv Preprint 2020. [Google Scholar] [CrossRef]
  116. Mustafa, S.; Weltermann, A.; Fritsche, R.; Marsik, C.; Wagner, O.; Kyrle, P.A.; Eichinger, S. Genetic variation in heme oxygenase 1 (HMOX1) and the risk of recurrent venous thromboembolism. J. Vasc. Surg. 2008, 47, 566–570. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  117. Espinoza, J.A.; González, P.A.; Kalergis, A.M. Modulation of Antiviral Immunity by Heme Oxygenase-1. Am. J. Pathol. 2017, 187, 487–493. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  118. 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]
  119. Gorini, F.; Bustaffa, E.; Chatzianagnostou, K.; Bianchi, F.; Vassalle, C. Hydrogen sulfide and cardiovascular disease: Doubts, clues, and interpretation difficulties from studies in geothermal areas. Sci. Total Environ. 2020, 743, 140818. [Google Scholar] [CrossRef]
  120. Read, E.; Zhu, J.; Yang, G. Disrupted HmS Signaling by Cigarette Smoking and Alcohol Drinking: Evidence from Cellular, Animal, and Clinical Studies. Antioxidants (Basel) 2021, 10, 49. [Google Scholar] [CrossRef]
  121. Teigen, L.M.; Geng, Z.; Sadowsky, M.J.; Vaughn, B.P.; Hamilton, M.J.; Khoruts, A. Dietary Factors in Sulfur Metabolism and Pathogenesis of Ulcerative Colitis. Nutrients 2019, 11, 931. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  122. Blachier, F.; Beaumont, M.; Kim, E. Cysteine-derived hydrogen sulfide and gut health: A matter of endogenous or bacterial origin. Curr. Opin. Clin. Nutr. Metab. Care 2019, 22, 68–75. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Main molecular, cellular, and systemic actions of hydrogen sulfide. H2S attenuates apoptosis and decreases oxidative stress by a direct action, upregulating the cellular antioxidant system. H2S also produces an anti-inflammatory response through the inhibition or induction of specific pathways. At the vascular level, in addition to promoting cell proliferation and migration, it induces angiogenesis, vasodilation, represses aggregation and coagulation, and reduces aortic atherosclerotic plaque formation. The mechanisms underlying the effects of H2S include the phosphorylation and addition of cysteine residues to target proteins, interaction with the metal protein centers of proteins, and gene regulation (for more details see text). Abbreviations: eNOS: Endothelial nitric oxide synthase; H2S: Hydrogen sulfide; KATP: ATP-sensitive potassium channels; Keap1: Kelch-like ECH-associating protein 1.
Figure 1. Main molecular, cellular, and systemic actions of hydrogen sulfide. H2S attenuates apoptosis and decreases oxidative stress by a direct action, upregulating the cellular antioxidant system. H2S also produces an anti-inflammatory response through the inhibition or induction of specific pathways. At the vascular level, in addition to promoting cell proliferation and migration, it induces angiogenesis, vasodilation, represses aggregation and coagulation, and reduces aortic atherosclerotic plaque formation. The mechanisms underlying the effects of H2S include the phosphorylation and addition of cysteine residues to target proteins, interaction with the metal protein centers of proteins, and gene regulation (for more details see text). Abbreviations: eNOS: Endothelial nitric oxide synthase; H2S: Hydrogen sulfide; KATP: ATP-sensitive potassium channels; Keap1: Kelch-like ECH-associating protein 1.
Biomedicines 09 01107 g001
Figure 2. Primary biological mechanisms involving H2S (donors) in the protective response against SARS-CoV-2. Following viral infection, H2S counteracts inflammation by decreasing levels of IL-6 and IL-8, inducing an increase in IL-10 concentration and inhibiting the recruitment of leukocytes to the endothelium. It also represses ACE expression, which in turn leads to vasodilation, decreased inflammation and oxidative stress, and reduced fibrosis. H2S donors enhance endogenous antioxidant defenses and play a mucolytic role. The activity of H2S also results in the activation of the HO-1/CO/H2S system and inhibition of virus replication. Abbreviations: ACE: Angiotensin-converting enzyme; CO: Carbon monoxide; H2S: Hydrogen sulfide; HO-1: Heme oxygenase-1; IL: Interleukin.
Figure 2. Primary biological mechanisms involving H2S (donors) in the protective response against SARS-CoV-2. Following viral infection, H2S counteracts inflammation by decreasing levels of IL-6 and IL-8, inducing an increase in IL-10 concentration and inhibiting the recruitment of leukocytes to the endothelium. It also represses ACE expression, which in turn leads to vasodilation, decreased inflammation and oxidative stress, and reduced fibrosis. H2S donors enhance endogenous antioxidant defenses and play a mucolytic role. The activity of H2S also results in the activation of the HO-1/CO/H2S system and inhibition of virus replication. Abbreviations: ACE: Angiotensin-converting enzyme; CO: Carbon monoxide; H2S: Hydrogen sulfide; HO-1: Heme oxygenase-1; IL: Interleukin.
Biomedicines 09 01107 g002
Table 1. Summary of the main biological effects of sulfur drugs.
Table 1. Summary of the main biological effects of sulfur drugs.
Sulfur DrugsEffectsReference
Inorganic sulfide salts NaHS and Na2SReduction of inflammation, oxidative stress and damage induced by hyperglycemia; promotion of vasorelaxation and neovascularization[6,52]
Organic “slow-release” H2S compounds
GYY4137
Vasodilator, antihypertensive, anti-atherosclerotic, anti-thrombotic and anti-inflammatory effects[38,41]
Diphosphorothioates AP67 and AP105Promotion of high-glucose-induced hyperpolarization of the mitochondrial membrane and inhibition of ROS production in microvascular ECs,[54]
H2S prodrug sodium polysulthionate (SG1002)Promotion of increase in circulating H2S and NO and the consequent endothelial-dependent coronary artery vasorelaxation[55,56]
Natural organosulfur compound
Garlic
Natural isothiocyanates
Promotion of vasorelaxation, lower arterial blood pressure, decreased apoptosis and oxidative stress, angiogenesis
Anti-inflammatory and antioxidant effects
[57,58,59,60]
[63,64,65]
N-acetyl-Cysteine (NAC)
and taurine
Anti-hypertensive and anti-inflammatory effects[66,67,68]
Synthetic cysteine derivatives
(S-propyl-cysteine, S-allyl-cysteine and S-propargyl-cysteine)
Increase in H2S levels, anti-inflammatory effects[69,70]
H2S-hybrid drug
ACE inhibitors: Omaprilat, Remikiren, Macitentan, Bosentan, Vardenafil, Sildenafil
Pro-angiogenic, anti-inflammatory and anti-apoptotic activities
Zofenopril: increase in H2S concentration in plasma and vascular wall
[71,72,73]
[74]
H2S-releasing derivatives of NSAIDsAnti-platelet, antithrombotic and antioxidant effects
S-diclofenac (ACS15): anti-inflammatory and antiproliferative effects
[78,79,81]
[82,83]
The classes of sulfur drugs are in bold.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Gorini, F.; Del Turco, S.; Sabatino, L.; Gaggini, M.; Vassalle, C. H2S as a Bridge Linking Inflammation, Oxidative Stress and Endothelial Biology: A Possible Defense in the Fight against SARS-CoV-2 Infection? Biomedicines 2021, 9, 1107. https://doi.org/10.3390/biomedicines9091107

AMA Style

Gorini F, Del Turco S, Sabatino L, Gaggini M, Vassalle C. H2S as a Bridge Linking Inflammation, Oxidative Stress and Endothelial Biology: A Possible Defense in the Fight against SARS-CoV-2 Infection? Biomedicines. 2021; 9(9):1107. https://doi.org/10.3390/biomedicines9091107

Chicago/Turabian Style

Gorini, Francesca, Serena Del Turco, Laura Sabatino, Melania Gaggini, and Cristina Vassalle. 2021. "H2S as a Bridge Linking Inflammation, Oxidative Stress and Endothelial Biology: A Possible Defense in the Fight against SARS-CoV-2 Infection?" Biomedicines 9, no. 9: 1107. https://doi.org/10.3390/biomedicines9091107

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