Cardioprotective effects of hydrogen sulfide in attenuating myocardial ischemia‑reperfusion injury (Review)

  • Authors:
    • Dan Wu
    • Yijing Gu
    • Deqiu Zhu
  • View Affiliations

  • Published online on: October 29, 2021     https://doi.org/10.3892/mmr.2021.12515
  • Article Number: 875
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Ischemic heart disease is one of the major causes of cardiovascular‑related mortality worldwide. Myocardial ischemia can be attenuated by reperfusion that restores the blood supply. However, injuries occur during blood flow restoration that induce cardiac dysfunction, which is known as myocardial ischemia‑reperfusion injury (MIRI). Hydrogen sulfide (H2S), the third discovered endogenous gasotransmitter in mammals (after NO and CO), participates in various pathophysiological processes. Previous in vitro and in vivo research have revealed the protective role of H2S in the cardiovascular system that render it useful in the protection of the myocardium against MIRI. The cardioprotective effects of H2S in attenuating MIRI are summarized in the present review.

Introduction

Cardiovascular diseases (CVD) contribute to a high morbidity and mortality burden globally (1). In 2019, the number of patients with CVD was ~523 and ~18.6 million cases succumbed to CVD (2). Myocardial ischemia is a common clinical symptom resulting from atherosclerosis and myocardial infarction (3). Reperfusion is often used to repair myocardial structure damage and improve cardiac function following ischemia. However, reperfusion may also result in myocardial ischemia-reperfusion injury (MIRI), which aggravates cardiac dysfunction. Therapeutic strategies, such as preconditioning, postconditioning and administration of antiplatelet or antithrombotic agents, have been utilized to alleviate MIRI (4).

Hydrogen sulfide (H2S), the third discovered gaseous signaling molecule (after NO and CO), has been extensively studied in recent years (5). H2S was traditionally acknowledged as an environmental toxicant, however, it has recently gained significance as an endogenous-generated biological transmitter in mammal tissues (6). Multiple studies have revealed the physiological and pathological roles of H2S in the onset and progression of cardiac diseases (7,8). Thus, H2S is considered to be a potential treatment for MIRI. The present review has summarized the protective effects of H2S against MIRI.

Pathophysiological mechanism of MIRI

Oxidative stress

Oxygen homeostasis plays a vital role in the maintenance of physiological functions. Reactive oxygen species (ROS) are generated during the normal metabolism of oxygen and participate in signal transduction. ROS are then scavenged by various endogenous free radical scavenging enzymes, such as superoxide dismutase (SOD), catalase, glutathione peroxidase and thioredoxin (9). However, overproduction of ROS or insufficient enzyme activity may impair the equilibrium between ROS and antioxidants, resulting in damage to proteins, DNA and lipids (10). SOD1 knockout mice were shown to have excessive oxidative stress and aggravated myocardial injuries following acute myocardial ischemia (11). Moreover, excessive ROS impairs heart contraction by modifying excitation-contraction coupling proteins. Excessive ROS also activates various signaling kinases and transcription factors associated with myocardial hypertrophy. In addition, the proliferation of cardiac fibroblast and the activity of MMP are promoted by ROS (12,13).

Mitochondrial function

The mitochondria are the main source of ROS production. ROS are generated in the electron transport chain (ETC) located on the mitochondrial membrane during the process of ATP production, namely oxidative phosphorylation. Electrons are then transported by a train of proteins known as the mitochondrial complex via oxidation-reduction reactions and combine with oxygen molecules to produce water. During this process, some oxygen molecules are reduced to form ROS (14).

Mitochondria may also act as a target of ROS damage. During the early process of reperfusion, the excessive ROS generated may induce oxidative stress, leading to the abnormal opening of the mitochondrial permeability transition pore (mPTP). Opening of the mPTP leads to mitochondrial Ca2+ overload, usually accompanied by oxidative or nitrosative stress and ATP depletion. Abnormal opening of mPTP also causes loss of mitochondrial membrane potential (15), respiratory chain uncoupling and impaired ATP synthesis. The impaired mitochondrial function results in mitochondrial swelling, rupture and cell apoptosis or necrosis (16,17). Mitochondria morphological changes observed during a MIRI in rat myocardial tissues mainly manifest as mitochondrial cristae and membrane damage, disordered fiber arrangement and larger perinuclear space (18). Furthermore, inhibition of mPTP opening using pharmaceutical agents, such as cyclosporine A, has been shown to reduce myocardial infarct size in acute ischemia-reperfusion injury (IRI) animals (19).

Autophagy

Autophagy plays a key role in cell survival by transferring damaged proteins and organelles to lysosomes for degradation. However, the autophagy process is controversial in MIRI. Autophagy is activated via the AMP-activated protein kinase pathway during ischemia to promote cell survival. However, during reperfusion, autophagy exerts a harmful role via Beclin activation (20). Loos et al (21) observed the activation of autophagy in mild ischemia. However, severe ischemia did not activate autophagy. This demonstrates that autophagy induction is closely associated with the degree of MIRI.

Reperfusion injury salvage kinase (RISK) pathway

Ischemic-induced apoptosis (cell death) is accelerated by reperfusion (3). Thus, anti-apoptotic mechanisms may be exploited as potential methods to decrease reperfusion-induced cell death. Reperfusion can activate several anti-apoptotic pathways in the RISK pathway, including PI3K/Akt and ERK1/2 pathways, that regulate cell survival (22). Protein kinase C, protein kinase G and GSK-3β are also regarded as members of the RISK pathway (23). Type 2 diabetes has been shown to impair nuclear factor-erythroid factor 2-related factor 2 (Nrf2) signaling via BTB domain and CNC homolog 1 (Bach1), thereby blocking the binding of Nrf2 to the heme oxygenase-1 promoter. Moreover, db/db diabetic mice treated with Na2S for 7 days was shown to overcome this impairment by removing Bach1 from the nucleus in an ERK1/2-dependent manner (24).

Characteristics of H2S

Generation and metabolism of endogenous H2S in mammals

Endogenous H2S is produced via enzymatic or nonenzymatic pathways in mammalian tissues. Cystathionine β-synthase (CBS) and cystathionine γ-lyase (CSE) are pyridoxal-5′-phosphate-dependent enzymes expressed in the cytosol that synthesize H2S using L-cysteine or homocysteine as substrates (25). H2S may also be synthesized in a catalytic reaction by 3-mercaptopyruvate sulfurtransferase (3-MST), involving α-ketoglutarate (25). These three enzymes are tissue-specific. CSE is mainly located in the kidney, liver, heart and vessels (26). CBS is found in neurons and astrocytes of the central nervous system, while 3-MST is mainly expressed in the liver, kidney, brain and heart (Fig. 1) (27). The concentration of H2S varies in tissues, with the highest concentration observed in the heart (2831). Fig. 2 shows the concentration of H2S in tissues and plasma in mice.

In mammals, there are three main catabolic pathways of H2S: i) H2S is oxidized to thiosulfate catalyzed by mitochondrial thioquinone oxidoreductase, S-dioxygenase and S-transferase. The thiosulfate is then catalyzed by cyanide thioltransferase to sulfite, which is then oxidized by sulfite oxidase to sulfate; ii) H2S generates methyl mercaptan and dimethyl sulfide in a reaction catalyzed by cytoplasmic thiol S-methyltransferase; and iii) H2S interacts with methemoglobin to produce thiolhemoglobin (Fig. 1) (25).

H2S donors and inhibitors of H2S synthetic pathways

Various H2S donors have been employed for elucidating the physiological and pathological role of H2S. These donors are divided into the following categories: Inorganic salts, sulfur-containing organic compounds and derivatives of Allium sativum extracts (32). The H2S releasing mechanisms and protective effects of typical donors are summarized in Table I.

Table I.

Mechanism and protective effects of H2S-releasing compounds.

Table I.

Mechanism and protective effects of H2S-releasing compounds.

Donor typeTypical donor name H2S-releasing mechanismProtective effects
Inorganic saltsNaHSHydrolysisStimulating ROS scavenging (33)
Na2S Inhibiting ROS production (34)
CaS Anti-inflammation (35)
Vasodilation (36)
Promoting angiogenesis (37)
Sulfur-containing organic compoundGYY4137HydrolysisProtecting mitochondria (38)
Vasodilation (39)
Anti-inflammation (40)
Anti-oxidative stress (41)
Derivative of allium sativum extractSPRCH2S generation enzymeAnti-apoptosis (42)
S-allycysteine Inhibiting ROS production (42)
Diallyl sulfideGlutathioneAnti-inflammation (43)
Promoting angiogenesis (44)
Mitochondria-targeting compoundAP39HydrolysisProtection against mitochondrial
DNA oxidative damage (45)

[i] ROS, reactive oxygen species; SPRC, S-propargyl-cysteine; H2S, hydrogen sulfide.

The most widely-used H2S donors are sulfur-containing inorganic salts that release H2S rapidly in large amounts. The utilization of sulfur-containing inorganic salts in research may be limited by the superphysiological concentration of H2S (32). Morpholin-4-ium 4-methoxyphenyl-morpholino-phosphinodithioate (GYY4137) was synthesized to overcome this challenge (39). GYY4137 achieves lower concentrations of H2S, which can be maintained for longer period with improved efficacy and reduced cytotoxicity.

Researchers have also synthesized derivatives of naturally occurring sulfur-containing organic compounds, such as S-propargyl-cysteine, S-allycysteine and diallyl sulfide, to improve the effectiveness of the H2S donors. In contrast to conventional H2S donors that release H2S directly, Allium sativum extract derivatives increase the levels of H2S by increasing the expression and activity of CSE and CBS. This is advantageous as the levels of H2S are controlled and, thus, have a lower risk of toxicity.

Szczesny et al (46) reported a novel H2S donor, AP39, [(10-oxo-10-(4-(3-thioxo-3H-1,2-dithiol-5yl)phenoxy)decyl) triphenylphosphonium bromide] that had a preferential response in the mitochondrial regions, as triphenylphosphonium tends to accumulate in mitochondria. Exposure of cells to different concentrations of AP39 (30–300 nmol/l) revealed that the effect of AP39 on mitochondrial activity was dependent on the concentration of H2S. It was shown that lower concentrations (30–100 nmol/l) promoted mitochondrial electron transport and cellular bioenergetic functions. By contrast, higher concentrations (300 nmol/l) had an inhibitory role. Thus, the antioxidant and cytoprotective effects of AP39 against oxidative mitochondrial DNA damage have been reported.

Inhibitors blocking H2S synthesis enzymes have also been examined. In colon cancer cells, CBS inhibitor aminooxyacetic acid (AOAA) can reduce tumor growth dose-dependently (47). However, the effect of CSE inhibitor D, L-propynylglycine (PAG) on myocardial injury remains controversial. In acute myocardial infarction and heart failure animal models, PAG could upregulated oxidative stress and apoptosis by suppressing H2S generation (8,48). Nevertheless, PAG administration can exacerbated acute lung inflammation in a rat model (49).

Role of H2S in the cardiovascular system

Physiological role of H2S in the cardiovascular system

H2S has a dual biological effect in mammals. High concentrations of H2S exert pathological and toxicological effects, such as inhibition of cellular bioenergetics, pro-oxidant effects, genotoxicity, proinflammatory effects and promotion of cell death. By contrast, low H2S concentrations stimulate mitochondrial electron transport, suppress inflammation, promote physiological vasodilatation, stimulate angiogenesis and inhibit oxidative stress, which are beneficial to cell survival (50).

Therapeutic role of H2S in the cardiovascular system

In recent years, the protective role of H2S in the cardiovascular system has been confirmed. The cardioprotective effects of H2S and the possible mechanism are summarized in Table II. These studies have revealed that multiple signaling pathways are involved in the therapeutic effects of H2S in cardiovascular system (6,5167). Notably, S-sulfhydration may be the core mechanism of H2S in mediating protein function and regulating pathophysiological processes of the cardiovascular system.

Table II.

Therapeutic effects of H2S and possible mechanism.

Table II.

Therapeutic effects of H2S and possible mechanism.

AnimalDosageH2S concentrationPossible mechanismTherapeutic effects
MouseGYY4137 (133 µmol/kg/d, IP)86.36±17.78 µM in plasmaInducing Keap1 S-sulfhydrationInhibiting atherosclerosis (51)
MouseNaHS (50 µmol/kg/d, IP)~7 µM in plasmaIncreasing SIRT3 promoter activity and expressionInhibiting oxidative stress in myocardial hypertrophy (52)
MouseSPRC (10, 25 mg/kg/d, IP)~1.5, 2.5 µM in plasmaInducing CaMKII S-sulfhydrationAnti-oxidative stress and anti-apoptosis (6)
MouseNaHS (80 µmol/kg/2d, IP)N/AInducing USP8 S-sulfhydrationCardioprotection (53)
MouseNaHS (80 µmol/kg/2d, IP)N/AInducing Hrd1 Cys115 S-sulfhydrationCardioprotection (54)
MouseN/A~1.1 nmol/mg in left ventricleAlleviating pyroptosisCardioprotection (55)
MouseSG-1002 (20 mg/kg /d, PO)N/AActivating AMPK/PGC-1α pathwayPromoting cardiac mitochondrial biogenesis (56)
MouseNaHS (1 mg/kg, IP)N/ADecreasing CD11b+Gr-1+ cell numbers in blood and myocardium after MI.Anti-inflammation in chronic myocardial ischemia (57)
MouseNaHS (14 µmol/kg/d, IP)N/AInducing FoxO1 phosphorylation and nuclear exclusionCardioprotection (58)
MouseGYY4137 (50 µmol/kg/d, IP)N/ADownregulating cardiac hypertrophy, fibrosis, and apoptosis-related gene expressionCardioprotection (59)
RatNaHS (39 µmol/kg/d, IP)N/AInducing muscle RING finger-1 Cys44 S-sulfhydrationAlleviating cardiac muscle degradation (60)
RatGYY4137 (10, 25, 50 mg/kg/d, IP)N/AInducing specificity protein 1 Cys664 S-sulfhydrationInhibiting myocardial hypertrophy (61)
RatNaHS (90 µmol/kg/d, IP)~30, 42 µM in plasmaInducing TRPV1 S-sulfhydrationEnhancing carotid sinus baroreceptor sensitivity (62)
RatNaHS (56 µmol/kg/d, IP)~1.5 µM in plasmaActivating Akt/eNOS/NO pathwayCardioprotection and anti-hypertension (63)
RatGYY4137 (10, 25, 50 mg/kg/d, IP)N/AInhibiting TGF-β1/Smad2 pathway, inhibiting oxidative stress and downregulating α-SMA in cardiac fibroblastsInbihiting myocardial fibrosis (64)
RatControlled release formulation of SPRC (30 mg/kg/d, PO)6-fold compared with modelAnti-apoptosis and anti-oxidative stressCardioprotection (65)
RatNaHS (100 µM/d, IP)N/AAnti-apoptosisCardioprotection (66)
SwineNa2S (100 µg/kg bolus + 1 mg/kg/h infusion)N/AAnti-oxidative stress and anti-inflammationProtecting against MIRI (67)

[i] IP, intraperitoneal injection; PO, Oral ingestion; d, day; Keap-1, Kelch-like ECH-associated protein-1; MIRI, myocardial ischemia-reperfusion injury; SPRC, S-propargyl-cysteine; H2S, hydrogen sulfide; NO, nitric oxide; eNOS, endothelial nitric oxide synthase; TRPV1, transient receptor potential cation channel subfamily V member; PGC-1α, peroxisome proliferator-activated receptor γ coactivator-1; AMPK, AMP-activated protein kinase; Hrd1,HMG-coA reductase degradation 1 homolog (S. cerevisiae); USP8, ubiquitin specific peptidase 8; SIRT3, sirtuin3; CaMKII, calcium/calmodulin-dependent protein kinase II.

Protective effects of H2S in MIRI

During MIRI, the plasma level of H2S and activity of CSE in the myocardium are decreased, leading to a further reduction in H2S synthesis. However, the mRNA expression level of CSE is enhanced following reperfusion, which contributes to positive feedback following the depressed H2S level (68). CSE knockout mice were observed to have lower levels of H2S in the blood and heart, followed by exacerbated oxidative stress and severe MIRI (69). Furthermore, acute H2S therapy significantly reduces myocardial infarct size per area-at-risk and lowers the plasma level of troponin-I in myocardial I/R mice (69). A meta-analysis reported that preconditioning with H2S in vivo significantly decreases the infarct size by 20.25% (95% CI 25.02; 15.47), while postconditioning with H2S notably reduced the infarct size by 21.61% (95% CI 24.17; 19.05) (70). In vivo results have shown that pretreatment with H2S before MIRI resulted in improved myocardial function, ameliorated coronary microvascular reactivity and reduced infarct size (67). Apolipoprotein E knockout mice were also revealed to have enhanced plaque stability and blood lipid levels and reduced plaque formation when treated with NaHS compared with vehicle-treated controls (71).

H2S inhibits oxidative stress

Administration of H2S restores cardiac function and enhances antioxidant function. Sun et al (72) compared the effects of diallyl trisulfide-mesoporous silica nanoparticles (DATS-MSN), a long-term and slow-releasing H2S donor, with two classical donors NaHS and GYY4137. The results of this study demonstrated that these three donors preserved the levels of glutathione and the activities of SOD and catalase, while DATS-MSN had the highest antioxidant effects. This result may be attributed to the slow-release and long-term H2S effects of DATS-MSN, which mimic the generation and function of endogenous H2S. It was shown that treatment with GYY4137 for 7 days before ischemia and reperfusion decreased the serum levels of malondialdehyde and myeloperoxidase, as well as suppressed superoxide anion levels and phosphorylation of MAPKs in the myocardium (68). In a Yorkshire swine model of mid-left anterior descending coronary artery, sulfide treatment before and throughout reperfusion decreased myeloperoxidase and inflammation, thereby improving myocardial function and conferring protection against MIRI (67).

NaHS (10 µmol/l) postconditioning was revealed to decrease the myocardial infarct size of isolated rat hearts and inhibit oxidative stress by stimulating SOD activity and reducing malondialdehyde levels via the activation of the sirtuin1/peroxisome proliferator-activated receptor-γ coactivator-1α pathway in an ex vivo study (73).

On the contrary, AP39 exhibited antioxidative effects via ROS generation rather than scavenging. The alleviation of myocardial infarction induced by AP39 during MIRI partly arose from reduced production of ROS in interfibrillar and subsarcolemmal mitochondria of cardiomyocytes, which were dose-dependent (Fig. 3) (74).

H2S improves mitochondrial function

The cardioprotective effects mediated by exogenous NaHS depend on mitochondrial ETC enzymes. Hemodynamic parameters and mitochondrial ETC functional assessment revealed that the cardioprotective effects of H2S require active mitochondria (75). Following MIRI, mouse hearts showed mitochondrial swelling, disorganized cristae and lower matrix density. However, treatment with H2S during reperfusion resulted in significantly improved mitochondrial structure, stimulated mitochondrial respiration and oxygen consumption (76). Karwi et al (74) reported that AP39 inhibited ROS generation and mPTP opening during MIRI. However, inhibition of the PI3K/Akt pathway, endothelial nitric oxide (NO) synthase (eNOS) or soluble guanylyl cyclase did not reverse the protective effects of AP39. Further research is required to investigate the association of these effects to post-translation modifications mediated by H2S and the interaction with NO in mitochondria.

H2S also leads to mitochondrial ATP-sensitive K+ (KATP) channel opening. Ji et al (77) reported that treatment with NaHS before reperfusion resulted in the reduction of infarct size and inhibited creatine kinase release in isolated rat hearts. However, these observations were shown to be reversed by KATP channel blockers (glibenclamide or 5-hydroxydecanoate). Moreover, novel H2S-donor 4-carboxyphenyl isothiocyanate was reported to activate the mitochondrial KATP channel and partially depolarize the mitochondrial membrane potential (Fig. 3) (78).

H2S regulates the RISK pathway

The RISK pathway, activated at the onset of reperfusion, can be regulated by H2S, thereby protecting against MIRI. In primary cultures of neonatal cardiomyocyte damage induced by hypoxia/reoxygenation (H/R), NaHS was shown to reduce apoptosis in a dose-dependent manner. Furthermore, H2S inhibits mPTP opening at a concentration of 30 µmol/l by increasing the phosphorylation of GSK-3β at Ser9 (78). H2S administration was not shown to inhibit mPTP opening in isolated mitochondria owing to the lack of intracellular signaling elements, such as GSK-3β (79). In db/db diabetic mice, which are at an increased risk of MIRI, Na2S therapy administered at the time of reperfusion activated the ERK1/2 pathway, thereby increasing anti-apoptotic proteins and inhibiting the activation of GSK3β (79). Na2S also significantly reduced the infarct size and circulating troponin-I levels in an ERK1/2-dependent manner (80).

Kelch-like ECH-associated protein-1 (Keap-1)/Nrf2/antioxidant response elements (ARE) pathway is a primary pathway involved in the cellular defense against oxidative stress. In response to oxidative stress, H2S dissociates Nrf2 from Keap1 (81). During early preconditioning, H2S promotes the nuclear translocalization of Nrf2 and increases the phosphorylation of protein kinase C epsilon and STAT-3. Moreover, H2S increases the expression of heme oxygenase-1 and thioredoxin 1 during late preconditioning (82). As a result of Nrf2 nuclear translocation, ARE is activated and enhances the transcription of SOD, catalase and heme oxygenase-1 (83). PH domain leucine-rich repeat protein phosphatase-1 (PHLPP-1) has recently been shown to dephosphorylate Akt at Ser473, which increases infarct size and aggravates MIRI (84,85). During MIRI, the levels of cardiac malondialdehyde are increased, while the expression levels of SOD and heme oxygenase-1 are downregulated. Pretreatment with GYY4137 was shown to reverse the oxidative stress induced by MIRI. GYY4137 also increased the protein expression levels of Akt and Nrf2 by downregulating the level of PHLPP-1. Thus, the antioxidant effect of H2S in MIRI partly depended on the PHLPP-1/Akt/Nrf2 pathway (41). PI3K, an upstream factor of Akt, is considered an important molecule in the underlying mechanism of H2S protection against ischemia-reperfusion. The PI3K/Akt/Nrf2 pathway has been reported to play a major role in alleviating cerebral ischemia-reperfusion injury (86). However, to the best of our knowledge, this mechanism has not been reported in the cardiovascular system.

H2S regulates microRNA (miRNA/miR)

Several studies have reported that the expression of miRNA is influenced by H2S in MIRI (Fig. 4). In cardiomyocytes of neonatal rats, H/R injury was shown to promote the expression of miR-1. The expression of histone deacetylase 4 (HDAC4) was also observed to be decreased (at mRNA and protein levels) during H/R. Preconditioning with H2S treatment downregulated miR-1, increased HDAC4 expression and reduced caspase-3 cleavage and release of lactate dehydrogenase. However, a study showing that the protective effects of H2S could be partially reversed by transfection of cardiomyocytes with miR-1 mimic, demonstrates that H2S protected neonatal rat cardiomyocytes from apoptosis and enhanced cell viability via the miR-1/HDAC4 signaling pathway (87).

H2S reduced the activity of caspase-1, as well as the formation and activity of inflammasome in a miR-21-dependent manner. Caspase-1 is an effector enzyme of the inflammasome that is mainly responsible for the processing and release of IL-1β and IL-18 (88). Na2S administration was demonstrated to inhibit apoptosis or necrosis in cardiomyocytes in in vitro studies and reduce infarct size following MIRI in vivo by activating miR-21 (89). A potential target of interaction between miR-21 and Toll-like receptor-4 exists. For instance, in lipopolysaccharide-induced acute lung injury, miR-21 was shown to negatively regulate inflammatory responses via the Toll-like receptor-4 and NF-κB signaling pathway (90). In addition, miR-21 activates the PI3K/Akt signaling pathway to participate in rheumatoid arthritis by inhibiting PTEN expression (91). miR-21 was also shown to reduce p38 MAPK protein expression, which inhibits activation of caspase-3 via PTEN/Akt (92). However, the involvement of these mechanisms in the protection against MIRI by H2S requires further study.

Endoplasmic reticulum (ER) stress is activated to protect cells when they are exposed to hypoxia. However, sustained activation of ER stress causes apoptosis (93). Ren et al (94) reported that the expression levels of ER stress biomarkers, heat shock protein family A (Hsp70) member 5, CHOP and eukaryotic initiation factor-2α, were significantly increased during ischemia/reperfusion. However, in vitro and in vivo results revealed that pretreatment with H2S alleviated ER stress and subsequent apoptosis via the miR-133a signaling pathway by reversing the cardiomyocyte trauma induced by MIRI. The combination of H2S intervention and miR-133a overexpression notably increased the proliferation, migration and invasion of cardiomyocytes. miR-133a was also observed to promote anti-apoptotic protein Bcl-2 expression and inhibit pro-apoptotic protein Bax, caspase-3, caspase-9 and apoptotic peptidase activating factor-1 expression. Consequently, decreasing apoptosis in the cardiomyocytes (95,96).

Crosstalk between H2S and NO

Accumulating evidence has revealed that there is a crosstalk between H2S and NO. CSE knockout mice showed a reduction in NO levels due to decreased eNOS expression. Acute treatment with H2S in CSE knockout mice was found to increase NO bioavailability and restore eNOS protein expression, which consequently attenuated oxidative stress and MIRI (69). In another in vivo study, H2S, donated by diallyl trisulfide, activated eNOS protein expression and NO metabolites, reduced infarct size and restored myocardial contractile function (97). H2S was also confirmed to attenuate cardiac arrest-induced mitochondrial injury and cell death in cardiopulmonary resuscitation in mice (98). These protective effects are conferred by increasing phosphorylation of eNOS in the left ventricle and increasing serum nitrite/nitrate levels (98). However, further research is required to confirm the protective role of H2S in MIRI.

S-sulfhydration

In recent years, increased attention has been paid to S-sulfhydration, a post-translational modification between H2S and cysteine residues of proteins that modifies the structure and biological activities of protein targets (99). Pharmacological postconditioning performed at the onset of reperfusion with NaHS significantly increased S-nitrosylation of cardioprotective proteins, as well as reduced post-ischemic contractile dysfunction and infarct size (100). However, the S-sulfhydration of proteins in MIRI has not been fully studied. H2S was reported to S-sulfhydrate Keap1 in response to oxidative stress, thereby mediating the dissociation of Nrf2 from Keap1, and as a result, promoting Nrf2 translocation in sulfur mustard-induced lung injury (81). A similar mechanism was confirmed in diabetic mice, wherein, H2S attenuated diabetes-accelerated atherosclerosis by S-sulfhydrating Keap1 at Cys151, resulting in activation of Nrf2 signaling (51). These mechanisms may contribute to the potential role of H2S in MIRI.

Conclusions

In summary, H2S plays a vital protective role in attenuating MIRI via mechanisms, such as attenuation of oxidative stress, restoration of mitochondrial function, regulation of miRNA, interaction with NO and S-sulfhydration. However, while these effects have been demonstrated in cellular and animal models, they have not been replicated in humans, to the best of our knowledge. Therefore, the transition of H2S from bench to bedside is necessary. Off-target effects of H2S may result in unexpected adverse reactions, including irreversible damage. Therefore, future research should focus on maximizing the potential benefits of H2S in cardioprotection in MIRI, while minimizing the unwanted side effects.

Acknowledgements

Not applicable.

Funding

Sponsored by Shanghai Pujiang Program (grant no. 2020PJD055) and Shanghai Key Specialty Construction Project of Clinical Pharmacy (2018).

Availability of data and materials

Data sharing not applicable to this article as no datasets were generated or analyzed during the current study.

Authors' contributions

YG wrote the original manuscript and the prepared figures and tables. DW modified the manuscript according to the reviewers and editors' comments. DZ contributed to the revision of the article. All authors have read and approved the final manuscript. Data sharing not applicable.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Townsend N, Wilson L, Bhatnagar P, Wickramasinghe K, Rayner M and Nichols M: Cardiovascular disease in Europe: Epidemiological update 2016. Eur Heart J. 37:3232–3245. 2016. View Article : Google Scholar : PubMed/NCBI

2 

Roth GA, Mensah GA, Johnson CO, Addolorato G, Ammirati E, Baddour LM, Barengo NC, Beaton AZ, Benjamin EJ, Benziger CP, et al: Global burden of cardiovascular diseases and risk factors, 1990–2019: Update from the GBD 2019 Study. J Am Coll Cardiol. 76:2982–3021. 2020. View Article : Google Scholar : PubMed/NCBI

3 

Wu MY, Yiang GT, Liao WT, Tsai AP, Cheng YL, Cheng PW, Li CY and Li CJ: Current mechanistic concepts in ischemia and reperfusion injury. Cell Physiol Biochem. 46:1650–1667. 2018. View Article : Google Scholar : PubMed/NCBI

4 

Ibáñez B, Heusch G, Ovize M and Van de Werf F: Evolving therapies for myocardial ischemia/reperfusion injury. J Am Coll Cardiol. 65:1454–1471. 2015. View Article : Google Scholar : PubMed/NCBI

5 

Gorini F, Bustaffa E, Chatzianagnostou K, Bianchi F and Vassalle C: Hydrogen sulfide and cardiovascular disease: Doubts, clues, and interpretation difficulties from studies in geothermal areas. Sci Total Environ. 743:1408182020. View Article : Google Scholar : PubMed/NCBI

6 

Wu D, Hu Q, Tan B, Rose P, Zhu D and Zhu YZ: Amelioration of mitochondrial dysfunction in heart failure through S-sulfhydration of Ca2+/calmodulin-dependent protein kinase II. Redox Biol. 19:250–262. 2018. View Article : Google Scholar : PubMed/NCBI

7 

Wang ZJ, Wu J, Guo W and Zhu YZ: Atherosclerosis and the hydrogen sulfide signaling pathway-therapeutic approaches to disease prevention. Cell Physiol Biochem. 42:859–875. 2017. View Article : Google Scholar : PubMed/NCBI

8 

Donnarumma E, Trivedi RK and Lefer DJ: Protective actions of H2S in acute myocardial infarction and heart failure. Compr Physiol. 7:583–602. 2017. View Article : Google Scholar : PubMed/NCBI

9 

Xu T, Ding W, Ji X, Ao X, Liu Y, Yu W and Wang J: Oxidative stress in cell death and cardiovascular diseases. Oxid Med Cell Longev. 2019:90305632019. View Article : Google Scholar : PubMed/NCBI

10 

Zorov DB, Juhaszova M and Sollott SJ: Mitochondrial reactive oxygen species (ROS) and ROS-induced ROS release. Physiol Rev. 94:909–950. 2014. View Article : Google Scholar : PubMed/NCBI

11 

Bai YD, Yang YR, Mu XP, Lin G, Wang YP, Jin S, Chen Y, Wang MJ and Zhu YC: Hydrogen sulfide alleviates acute myocardial ischemia injury by modulating autophagy and inflammation response under oxidative stress. Oxid Med Cell Longev. 2018:34028092018. View Article : Google Scholar : PubMed/NCBI

12 

Tsutsui H, Kinugawa S and Matsushima S: Oxidative stress and heart failure. Am J Physiol Heart Circ Physiol. 301:H2181–H2190. 2011. View Article : Google Scholar : PubMed/NCBI

13 

van der Pol A, van Gilst WH, Voors AA and van der Meer P: Treating oxidative stress in heart failure: Past, present and future. Eur J Heart Fail. 21:425–435. 2019. View Article : Google Scholar : PubMed/NCBI

14 

Li X, Fang P, Mai J, Choi ET, Wang H and Yang XF: Targeting mitochondrial reactive oxygen species as novel therapy for inflammatory diseases and cancers. J Hematol Oncol. 6:192013. View Article : Google Scholar : PubMed/NCBI

15 

Briston T, Selwood DL, Szabadkai G and Duchen MR: Mitochondrial permeability transition: A molecular lesion with multiple drug targets. Trends Pharmacol Sci. 40:50–70. 2019. View Article : Google Scholar : PubMed/NCBI

16 

Bauer TM and Murphy E: Role of mitochondrial calcium and the permeability transition pore in regulating cell death. Circ Res. 126:280–293. 2020. View Article : Google Scholar : PubMed/NCBI

17 

Kwong JQ and Molkentin JD: Physiological and pathological roles of the mitochondrial permeability transition pore in the heart. Cell Metab. 21:206–214. 2015. View Article : Google Scholar : PubMed/NCBI

18 

Li HW and Xiao FY: Effect of hydrogen sulfide on cardiomyocyte apoptosis in rats with myocardial ischemia-reperfusion injury via the JNK signaling pathway. Eur Rev Med Pharmacol Sci. 24:2054–2061. 2020.PubMed/NCBI

19 

Ong S, Samangouei P, Kalkhoran SB and Hausenloy DJ: The mitochondrial permeability transition pore and its role in myocardial ischemia reperfusion injury. J Mol Cell Cardiol. 78:23–34. 2014. View Article : Google Scholar : PubMed/NCBI

20 

Matsui Y, Takagi H, Qu X, Abdellatif M, Sakoda H, Asano T, Levine B and Sadoshima J: Distinct roles of autophagy in the heart during ischemia and reperfusion: Roles of AMP-activated protein kinase and Beclin 1 in mediating autophagy. Circ Res. 100:914–922. 2007. View Article : Google Scholar : PubMed/NCBI

21 

Loos B, Genade S, Ellis B, Lochner A and Engelbrecht AM: At the core of survival: Autophagy delays the onset of both apoptotic and necrotic cell death in a model of ischemic cell injury. Exp Cell Res. 317:1437–1453. 2011. View Article : Google Scholar : PubMed/NCBI

22 

Hausenloy DJ and Yellon DM: New directions for protecting the heart against ischaemia-reperfusion injury: Targeting the Reperfusion Injury Salvage Kinase (RISK)-pathway. Cardiovasc Res. 61:448–460. 2004. View Article : Google Scholar : PubMed/NCBI

23 

Hausenloy DJ and Yellon DM: Reperfusion injury salvage kinase signalling: Taking a RISK for cardioprotection. Heart Fail Rev. 12:217–234. 2007. View Article : Google Scholar : PubMed/NCBI

24 

Peake BF, Nicholson CK, Lambert JP, Hood RL, Amin H, Amin S and Calvert JW: Hydrogen sulfide preconditions the db/db diabetic mouse heart against ischemia-reperfusion injury by activating Nrf2 signaling in an Erk-dependent manner. Am J Physiol Heart Circ Physiol. 304:H1215–H1224. 2013. View Article : Google Scholar : PubMed/NCBI

25 

Cao X, Ding L, Xie ZZ, Yang Y, Whiteman M, Moore PK and Bian JS: A review of hydrogen sulfide synthesis, metabolism, and measurement: Is modulation of hydrogen sulfide a novel therapeutic for cancer? Antioxid Redox Signal. 31:1–38. 2019. View Article : Google Scholar : PubMed/NCBI

26 

Bełtowski J and Jamroz-Wiśniewska A: Hydrogen sulfide and endothelium-dependent vasorelaxation. Molecules. 19:21183–21199. 2014. View Article : Google Scholar : PubMed/NCBI

27 

Panthi S, Chung HJ, Jung J and Jeong NY: Physiological importance of hydrogen sulfide: Emerging potent neuroprotector and neuromodulator. Oxid Med Cell Longev. 2016:90497822016. View Article : Google Scholar : PubMed/NCBI

28 

Wilinski B, Wilinski J, Somogyi E, Goralska M and Piotrowska J: Paracetamol (acetaminophen) decreases hydrogen sulfide tissue concentration in brain but increases it in the heart, liver and kidney in mice. Folia Biol (Krakow). 59:41–44. 2011. View Article : Google Scholar : PubMed/NCBI

29 

Wilinski B, Wilinski J, Somogyi E, Goralska M and Piotrowska J: Ramipril affects hydrogen sulfide generation in mouse liver and kidney. Folia Biol (Krakow). 58:177–180. 2010. View Article : Google Scholar : PubMed/NCBI

30 

Wilinski J, Wilinski B, Somogyi E, Piotrowska J, Kameczura T and Zygmunt M: Nicotine affects hydrogen sulfide concentrations in mouse kidney and heart but not in brain and liver tissues. Folia Med Cracov. 57:55–64. 2017.PubMed/NCBI

31 

Tan B, Jin S, Sun J, Gu Z, Sun X, Zhu Y, Huo K, Cao Z, Yang P, Xin X, et al: New method for quantification of gasotransmitter hydrogen sulfide in biological matrices by LC-MS/MS. Sci Rep. 7:462782017. View Article : Google Scholar : PubMed/NCBI

32 

Wu D, Hu Q and Zhu YZ; Therapeutic application of hydrogen sulfide donors, : The potential and challenges. Front Med. 10:18–27. 2016. View Article : Google Scholar : PubMed/NCBI

33 

Pei J, Wang F, Pei S, Bai R, Cong X, Nie Y and Chen X: Hydrogen sulfide promotes cardiomyocyte proliferation and heart regeneration via ROS scavenging. Oxid Med Cell Longev. 2020:14126962020. View Article : Google Scholar : PubMed/NCBI

34 

Feng A, Ling C, Xin-duo L, Bing W, San-Wu W, Yu Z, Yu-Lan H and You-En Z: Hydrogen sulfide protects human cardiac fibroblasts against H2O2-induced injury through regulating autophagy-related proteins. Cell Transplant. 27:1222–1234. 2018. View Article : Google Scholar : PubMed/NCBI

35 

Huang Z, Dong X, Zhuang X, Hu X, Wang L and Liao X: Exogenous hydrogen sulfide protects against high glucose-induced inflammation and cytotoxicity in H9c2 cardiac cells. Mol Med Rep. 14:4911–4917. 2016. View Article : Google Scholar : PubMed/NCBI

36 

Yuan C, Hou HT, Chen HX, Wang J, Wang ZQ, Chen TN, Novakovic A, Marinko M, Yang Q, Liu ZG, et al: Hydrogen sulfide-mediated endothelial function and the interaction with eNOS and PDE5A activity in human internal mammary arteries. J Int Med Res. 47:3778–3791. 2019. View Article : Google Scholar : PubMed/NCBI

37 

Wang GG and Li W: Hydrogen sulfide improves vessel formation of the ischemic adductor muscle and wound healing in diabetic db/db mice. Iran J Basic Med Sci. 22:1192–1197. 2019.PubMed/NCBI

38 

Wang CN, Liu YJ, Duan GL, Zhao W, Li XH, Zhu XY and Ni X: CBS and CSE are critical for maintenance of mitochondrial function and glucocorticoid production in adrenal cortex. Antioxid Redox Signal. 21:2192–2207. 2014. View Article : Google Scholar : PubMed/NCBI

39 

Li L, Whiteman M, Guan YY, Neo KL, Cheng Y, Lee SW, Zhao Y, Baskar R, Tan CH and Moore PK: Characterization of a novel, water-soluble hydrogen sulfide-releasing molecule (GYY4137): New insights into the biology of hydrogen sulfide. Circulation. 117:2351–2360. 2008. View Article : Google Scholar : PubMed/NCBI

40 

Castelblanco M, Lugrin J, Ehirchiou D, Nasi S, Ishii I, So A, Martinon F and Busso N: Hydrogen sulfide inhibits NLRP3 inflammasome activation and reduces cytokine production both in vitro and in a mouse model of inflammation. J Biol Chem. 293:2546–2557. 2018. View Article : Google Scholar : PubMed/NCBI

41 

Qiu Y, Wu Y, Meng M, Luo M, Zhao H, Sun H and Gao S: GYY4137 protects against myocardial ischemia/reperfusion injury via activation of the PHLPP-1/Akt/Nrf2 signaling pathway in diabetic mice. J Surg Res. 225:29–39. 2018. View Article : Google Scholar : PubMed/NCBI

42 

Yang H, Mao Y, Tan B, Luo S and Zhu Y: The protective effects of endogenous hydrogen sulfide modulator, S-propargyl-cysteine, on high glucose-induced apoptosis in cardiomyocytes: A novel mechanism mediated by the activation of Nrf2. Eur J Pharmacol. 761:135–143. 2015. View Article : Google Scholar : PubMed/NCBI

43 

Qian X, Li X, Ma F, Luo S, Ge R and Zhu Y: Novel hydrogen sulfide-releasing compound, S-propargyl-cysteine, prevents STZ-induced diabetic nephropathy. Biochem Biophys Res Commun. 473:931–938. 2016. View Article : Google Scholar : PubMed/NCBI

44 

Kan J, Guo W, Huang C, Bao G, Zhu Y and Zhu YZ: S-propargyl-cysteine, a novel water-soluble modulator of endogenous hydrogen sulfide, promotes angiogenesis through activation of signal transducer and activator of transcription 3. Antioxid Redox Signal. 20:2303–2316. 2014. View Article : Google Scholar : PubMed/NCBI

45 

Zhao FL, Fang F, Qiao PF, Yan N, Gao D and Yan Y: AP39, a mitochondria-targeted hydrogen sulfide donor, supports cellular bioenergetics and protects against Alzheimer's disease by preserving mitochondrial function in APP/PS1 mice and neurons. Oxid Med Cell Longev. 2016:83607382016. View Article : Google Scholar : PubMed/NCBI

46 

Szczesny B, Módis K, Yanagi K, Coletta C, Le Trionnaire S, Perry A, Wood ME, Whiteman M and Szabo C: AP39, a novel mitochondria-targeted hydrogen sulfide donor, stimulates cellular bioenergetics, exerts cytoprotective effects and protects against the loss of mitochondrial DNA integrity in oxidatively stressed endothelial cells in vitro. Nitric Oxide. 41:120–130. 2014. View Article : Google Scholar : PubMed/NCBI

47 

Chao C, Zatarain JR, Ding Y, Coletta C, Mrazek AA, Druzhyna N, Johnson P, Chen H, Hellmich JL, Asimakopoulou A, et al: Cystathionine-beta-synthase inhibition for colon cancer: Enhancement of the efficacy of aminooxyacetic acid via the prodrug approach. Mol Med. 22:361–379. 2016. View Article : Google Scholar : PubMed/NCBI

48 

Lilyanna S, Peh MT, Liew OW, Wang P, Moore PK, Richards AM and Martinez EC: GYY4137 attenuates remodeling, preserves cardiac function and modulates the natriuretic peptide response to ischemia. J Mol Cell Cardiol. 87:27–37. 2015. View Article : Google Scholar : PubMed/NCBI

49 

Zhou X, Tang S, Hu K, Zhang Z, Liu P, Luo Y, Kang J and Xu L: DL-Propargylglycine protects against myocardial injury induced by chronic intermittent hypoxia through inhibition of endoplasmic reticulum stress. Sleep Breath. 22:853–863. 2018. View Article : Google Scholar : PubMed/NCBI

50 

Szabo C, Ransy C, Modis K, Andriamihaja M, Murghes B, Coletta C, Olah G, Yanagi K and Bouillaud F: Regulation of mitochondrial bioenergetic function by hydrogen sulfide. Part I. Biochemical and physiological mechanisms. Br J Pharmacol. 171:2099–2122. 2014. View Article : Google Scholar : PubMed/NCBI

51 

Xie L, Gu Y, Wen M, Zhao S, Wang W, Ma Y, Meng G, Han Y, Wang Y, Liu G, et al: Hydrogen sulfide induces Keap1 S-sulfhydration and suppresses diabetes-accelerated atherosclerosis via Nrf2 activation. Diabetes. 65:3171–3184. 2016. View Article : Google Scholar : PubMed/NCBI

52 

Meng G, Liu J, Liu S, Song Q, Liu L, Xie L, Han Y and Ji Y: Hydrogen sulfide pretreatment improves mitochondrial function in myocardial hypertrophy via a SIRT3-dependent manner. Br J Pharmacol. 175:1126–1145. 2018. View Article : Google Scholar : PubMed/NCBI

53 

Sun Y, Lu F, Yu X, Wang B, Chen J, Lu F, Peng S, Sun X, Yu M, Chen H, et al: Exogenous H2S promoted USP8 sulfhydration to regulate mitophagy in the hearts of db/db mice. Aging Dis. 11:269–285. 2020. View Article : Google Scholar : PubMed/NCBI

54 

Yu M, Du H, Wang B, Chen J, Lu F, Peng S, Sun Y, Liu N, Sun X, Shiyun D, et al: Exogenous H2S induces Hrd1 S-sulfhydration and prevents CD36 translocation via VAMP3 ubiquitylation in diabetic hearts. Aging Dis. 11:286–300. 2020. View Article : Google Scholar : PubMed/NCBI

55 

Kar S, Shahshahan HR, Hackfort BT, Yadav SK, Yadav R, Kambis TN, Lefer DJ and Mishra PK: Exercise training promotes cardiac hydrogen sulfide biosynthesis and mitigates pyroptosis to prevent high-fat diet-induced diabetic cardiomyopathy. Antioxidants. 8:6382019. View Article : Google Scholar : PubMed/NCBI

56 

Shimizu Y, Polavarapu R, Eskla KL, Nicholson CK, Koczor CA, Wang R, Lewis W, Shiva S, Lefer DJ and Calvert JW: Hydrogen sulfide regulates cardiac mitochondrial biogenesis via the activation of AMPK. J Mol Cell Cardiol. 116:29–40. 2018. View Article : Google Scholar : PubMed/NCBI

57 

Wu T, Li H, Wu B, Zhang L, Wu SW, Wang JN and Zhang YE: Hydrogen sulfide reduces recruitment of CD11b+Gr-1+ cells in mice with myocardial infarction. Cell Transplant. 26:753–764. 2017. View Article : Google Scholar : PubMed/NCBI

58 

Ye P, Gu Y, Zhu YR, Chao YL, Kong XQ, Luo J, Ren XM, Zuo GF, Zhang DM and Chen SL: Exogenous hydrogen sulfide attenuates the development of diabetic cardiomyopathy via the FoxO1 pathway. J Cell Physiol. 233:9786–9798. 2018. View Article : Google Scholar : PubMed/NCBI

59 

Ellmers LJ, Templeton EM, Pilbrow AP, Frampton C, Ishii I, Moore PK, Bhatia M, Richards AM and Cameron VA: Hydrogen sulfide treatment improves post-infarct remodeling and long-term cardiac function in CSE knockout and wild-type mice. Int J Mol Sci. 21:42842020. View Article : Google Scholar : PubMed/NCBI

60 

Sun X, Zhao D, Lu F, Peng S, Yu M, Liu N, Sun Y, Du H, Wang B, Chen J, et al: Hydrogen sulfide regulates muscle RING finger-1 protein S-sulfhydration at Cys44 to prevent cardiac structural damage in diabetic cardiomyopathy. Br J Pharmacol. 177:836–856. 2020. View Article : Google Scholar : PubMed/NCBI

61 

Meng G, Xiao Y, Ma Y, Tang X, Xie L, Liu J, Gu Y, Yu Y, Park CM, Xian M, et al: Hydrogen sulfide regulates krüppel-like factor 5 transcription activity via specificity protein 1 s-sulfhydration at Cys664 to prevent myocardial hypertrophy. J Am Heart Assoc. 5:e0041602016. View Article : Google Scholar : PubMed/NCBI

62 

Yu W, Liao Y, Huang Y, Chen SY, Sun Y, Sun C, Wu Y, Tang C, Du J and Jin H: Endogenous hydrogen sulfide enhances carotid sinus baroreceptor sensitivity by activating the transient receptor potential cation channel subfamily V Member 1 (TRPV1) Channel. J Am Heart Assoc. 6:e0049712017. View Article : Google Scholar : PubMed/NCBI

63 

Jin S, Teng X, Xiao L, Xue H, Guo Q, Duan X, Chen Y and Wu Y: Hydrogen sulfide ameliorated L-NAME-induced hypertensive heart disease by the Akt/eNOS/NO pathway. Exp Biol Med (Maywood). 242:1831–1841. 2017. View Article : Google Scholar : PubMed/NCBI

64 

Meng G, Zhu J, Xiao Y, Huang Z, Zhang Y, Tang X, Xie L, Chen Y, Shao Y, Ferro A, et al: Hydrogen sulfide donor GYY4137 protects against myocardial fibrosis. Oxid Med Cell Longev. 2015:6910702015. View Article : Google Scholar : PubMed/NCBI

65 

Huang C, Kan J, Liu X, Ma F, Tran BH, Zou Y, Wang S and Zhu YZ: Cardioprotective effects of a novel hydrogen sulfide agent-controlled release formulation of S-propargyl-cysteine on heart failure rats and molecular mechanisms. PLoS One. 8:e692052013. View Article : Google Scholar : PubMed/NCBI

66 

Zhong X, Wang L, Wang Y, Dong S, Leng X, Jia J, Zhao Y, Li H, Zhang X, Xu C, et al: Exogenous hydrogen sulfide attenuates diabetic myocardial injury through cardiac mitochondrial protection. Mol Cell Biochem. 371:187–198. 2012. View Article : Google Scholar : PubMed/NCBI

67 

Sodha NR, Clements RT, Feng J, Liu Y, Bianchi C, Horvath EM, Szabo C, Stahl GL and Sellke FW: Hydrogen sulfide therapy attenuates the inflammatory response in a porcine model of myocardial ischemia/reperfusion injury. J Thorac Cardiovasc Surg. 138:977–984. 2009. View Article : Google Scholar : PubMed/NCBI

68 

Meng G, Wang J, Xiao Y, Bai W, Xie L, Shan L, Moore PK and Ji Y: GYY4137 protects against myocardial ischemia and reperfusion injury by attenuating oxidative stress and apoptosis in rats. J Biomed Res. 29:203–213. 2015.PubMed/NCBI

69 

King AL, Polhemus DJ, Bhushan S, Otsuka H, Kondo K, Nicholson CK, Bradley JM, Islam KN, Calvert JW, Tao YX, et al: Hydrogen sulfide cytoprotective signaling is endothelial nitric oxide synthase-nitric oxide dependent. Proc Natl Acad Sci USA. 111:3182–3187. 2014. View Article : Google Scholar : PubMed/NCBI

70 

Karwi QG, Bice JS and Baxter GF: Pre- and postconditioning the heart with hydrogen sulfide (H2S) against ischemia/reperfusion injury in vivo: A systematic review and meta-analysis. Basic Res Cardiol. 113:62018. View Article : Google Scholar : PubMed/NCBI

71 

Xiong Q, Wang Z, Yu Y, Wen Y, Suguro R, Mao Y and Zhu YZ: Hydrogen sulfide stabilizes atherosclerotic plaques in apolipoprotein E knockout mice. Pharmacol Res. 144:90–98. 2019. View Article : Google Scholar : PubMed/NCBI

72 

Sun X, Wang W, Dai J, Jin S, Huang J, Guo C, Wang C, Pang L and Wang Y: A Long-term and slow-releasing hydrogen sulfide donor protects against myocardial ischemia/reperfusion injury. Sci Rep. 7:35412017. View Article : Google Scholar : PubMed/NCBI

73 

Hu MZ, Zhou B, Mao HY, Sheng Q, Du B, Chen JL, Pang QF and Ji Y: Exogenous hydrogen sulfide postconditioning protects isolated rat hearts from ischemia/reperfusion injury through Sirt1/PGC-1α signaling pathway. Int Heart J. 57:477–482. 2016. View Article : Google Scholar : PubMed/NCBI

74 

Karwi QG, Bornbaum J, Boengler K, Torregrossa R, Whiteman M, Wood ME, Schulz R and Baxter GF: AP39, a mitochondria-targeting hydrogen sulfide (H2 S) donor, protects against myocardial reperfusion injury independently of salvage kinase signalling. Br J Pharmacol. 174:287–301. 2017. View Article : Google Scholar : PubMed/NCBI

75 

Nandi S, Ravindran S and Kurian GA: Role of endogenous hydrogen sulfide in cardiac mitochondrial preservation during ischemia reperfusion injury. Biomed Pharmacother. 97:271–279. 2018. View Article : Google Scholar : PubMed/NCBI

76 

Elrod JW, Calvert JW, Morrison J, Doeller JE, Kraus DW, Tao L, Jiao X, Scalia R, Kiss L, Szabo C, et al: Hydrogen sulfide attenuates myocardial ischemia-reperfusion injury by preservation of mitochondrial function. Proc Natl Acad Sci USA. 104:15560–15565. 2007. View Article : Google Scholar : PubMed/NCBI

77 

Ji Y, Pang Q, Xu G, Wang L, Wang J and Zeng Y: Exogenous hydrogen sulfide postconditioning protects isolated rat hearts against ischemia-reperfusion injury. Eur J Pharmacol. 587:1–7. 2008. View Article : Google Scholar : PubMed/NCBI

78 

Testai L, Marino A, Piano I, et al: The novel H2S-donor 4-carboxyphenyl isothiocyanate promotes cardioprotective effects against ischemia/reperfusion injury through activation of mitoKATP channels and reduction of oxidative stress. Pharmacol Res. 113:290–299. 2016. View Article : Google Scholar : PubMed/NCBI

79 

Yao LL, Huang XW, Wang YG, Cao YX, Zhang CC and Zhu YC: Hydrogen sulfide protects cardiomyocytes from hypoxia/reoxygenation-induced apoptosis by preventing GSK-3beta-dependent opening of mPTP. Am J Physiol Heart Circ Physiol. 298:H1310–H1319. 2010. View Article : Google Scholar : PubMed/NCBI

80 

Lambert JP, Nicholson CK, Amin H, Amin S and Calvert JW: Hydrogen sulfide provides cardioprotection against myocardial/ischemia reperfusion injury in the diabetic state through the activation of the RISK pathway. Med Gas Res. 4:202014. View Article : Google Scholar : PubMed/NCBI

81 

Meng W, Pei Z, Feng Y, Zhao J, Chen Y, Shi W, Xu Q, Lin F, Sun M and Xiao K: Neglected role of hydrogen sulfide in sulfur mustard poisoning: Keap1 S-sulfhydration and subsequent Nrf2 pathway activation. Sci Rep. 7:94332017. View Article : Google Scholar : PubMed/NCBI

82 

Calvert JW, Jha S, Gundewar S, Elrod JW, Ramachandran A, Pattillo CB, Kevil CG and Lefer DJ: hydrogen sulfide mediates cardioprotection through Nrf2 signaling. Circ Res. 105:365–374. 2009. View Article : Google Scholar : PubMed/NCBI

83 

Tu W, Wang H, Li S, Liu Q and Sha H: The anti-inflammatory and anti-oxidant mechanisms of the Keap1/Nrf2/ARE signaling pathway in chronic diseases. Aging Dis. 10:637–651. 2019. View Article : Google Scholar : PubMed/NCBI

84 

Gao T, Furnari F and Newton AC: PHLPP: A phosphatase that directly dephosphorylates Akt, promotes apoptosis, and suppresses tumor growth. Mol Cell. 18:13–24. 2005. View Article : Google Scholar : PubMed/NCBI

85 

Miyamoto S, Purcell NH, Smith JM, Gao T, Whittaker R, Huang K, Castillo R, Glembotski CC, Sussman MA, Newton AC and Brown JH: PHLPP-1 negatively regulates Akt activity and survival in the heart. Circ Res. 107:476–484. 2010. View Article : Google Scholar : PubMed/NCBI

86 

Ji K, Xue L, Cheng J and Bai Y: Preconditioning of H2S inhalation protects against cerebral ischemia/reperfusion injury by induction of HSP70 through PI3K/Akt/Nrf2 pathway. Brain Res Bull. 121:68–74. 2016. View Article : Google Scholar : PubMed/NCBI

87 

Kang B, Li W, Xi W, Yi Y, Ciren Y, Shen H, Zhang Y, Jiang H, Xiao J and Wang Z: Hydrogen sulfide protects cardiomyocytes against apoptosis in ischemia/reperfusion through MiR-1-regulated histone deacetylase 4 pathway. Cell Physiol Biochem. 41:10–21. 2017. View Article : Google Scholar : PubMed/NCBI

88 

Muñoz-Planillo R, Nuñez G, Franchi L and Eigenbrod T: The inflammasome: A caspase-1-activation platform that regulates immune responses and disease pathogenesis. Nat Immunol. 10:241–247. 2009. View Article : Google Scholar : PubMed/NCBI

89 

Toldo S, Das A, Mezzaroma E, Chau VQ, Marchetti C, Durrant D, Samidurai A, Van Tassell BW, Yin C, Ockaili RA, et al: Induction of microRNA-21 with exogenous hydrogen sulfide attenuates myocardial ischemic and inflammatory injury in mice. Circ Cardiovasc Genet. 7:311–320. 2014. View Article : Google Scholar : PubMed/NCBI

90 

Zhu WD, Xu J, Zhang M, Zhu TM, Zhang YH and Sun K: MicroRNA-21 inhibits lipopolysaccharide-induced acute lung injury by targeting nuclear factor-κB. Exp Ther Med. 16:4616–4622. 2018.PubMed/NCBI

91 

Yan X, Liu Y, Kong X, Ji J, Zhu H, Zhang Z, Fu T, Yang J, Zhang Z, Liu F and Gu Z: MicroRNA-21-5p are involved in apoptosis and invasion of fibroblast-like synoviocytes through PTEN/PI3K/AKT signal. Cytotechnology. 71:317–328. 2019. View Article : Google Scholar : PubMed/NCBI

92 

Huang W, Tian SS, Hang PZ, Sun C, Guo J and Du ZM: Combination of microRNA-21 and microRNA-146a attenuates cardiac dysfunction and apoptosis during acute myocardial infarction in mice. Mol Ther Nucleic Acids. 5:e2962016. View Article : Google Scholar : PubMed/NCBI

93 

Karaskov E, Scott C, Zhang L, Teodoro T, Ravazzola M and Volchuk A: Chronic palmitate but not oleate exposure induces endoplasmic reticulum stress, which may contribute to INS-1 pancreatic beta-Cell apoptosis. Endocrinology. 147:3398–3407. 2006. View Article : Google Scholar : PubMed/NCBI

94 

Ren L, Wang Q, Chen Y, Ma Y and Wang D: Involvement of MicroRNA-133a in the protective effect of hydrogen sulfide against ischemia/reperfusion-induced endoplasmic reticulum stress and cardiomyocyte apoptosis. Pharmacology. 103:1–9. 2019. View Article : Google Scholar : PubMed/NCBI

95 

He B, Xiao J, Ren AJ, Zhang YF, Zhang H, Chen M, Xie B, Gao XG and Wang YW: Role of miR-1 and miR-133a in myocardial ischemic postconditioning. J Biomed Sci. 18:222011. View Article : Google Scholar : PubMed/NCBI

96 

Dakhlallah D, Zhang J, Yu L, Marsh CB, Angelos MG and Khan M: MicroRNA-133a engineered mesenchymal stem cells augment cardiac function and cell survival in the infarct heart. J Cardiovasc Pharmacol. 65:241–251. 2015. View Article : Google Scholar : PubMed/NCBI

97 

Predmore BL, Kondo K, Bhushan S, Zlatopolsky MA, King AL, Aragon JP, Grinsfelder DB, Condit ME and Lefer DJ: The polysulfide diallyl trisulfide protects the ischemic myocardium by preservation of endogenous hydrogen sulfide and increasing nitric oxide bioavailability. Am J Physiol Heart Circ Physiol. 302:H2410–H2418. 2012. View Article : Google Scholar : PubMed/NCBI

98 

Minamishima S, Bougaki M, Sips PY, Yu JD, Minamishima YA, Elrod JW, Lefer DJ, Bloch KD and Ichinose F: Hydrogen sulfide improves survival after cardiac arrest and cardiopulmonary resuscitation via a nitric oxide synthase 3-dependent mechanism in mice. Circulation. 120:888–896. 2009. View Article : Google Scholar : PubMed/NCBI

99 

Bibli SI, Hu J, Looso M, Weigert A, Ratiu C, Wittig J, Drekolia MK, Tombor L, Randriamboavonjy V, Leisegang MS, et al: Mapping the endothelial Cell S-sulfhydrome highlights the crucial role of integrin sulfhydration in vascular function. Circulation. 143:935–948. 2021. View Article : Google Scholar : PubMed/NCBI

100 

Sun J, Aponte AM, Menazza S, Gucek M, Steenbergen C and Murphy E: Additive cardioprotection by pharmacological postconditioning with hydrogen sulfide and nitric oxide donors in mouse heart: S-sulfhydration vs. S-nitrosylation. Cardiovasc Res. 110:96–106. 2016. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

December-2021
Volume 24 Issue 6

Print ISSN: 1791-2997
Online ISSN:1791-3004

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Wu D, Gu Y and Zhu D: Cardioprotective effects of hydrogen sulfide in attenuating myocardial ischemia‑reperfusion injury (Review). Mol Med Rep 24: 875, 2021
APA
Wu, D., Gu, Y., & Zhu, D. (2021). Cardioprotective effects of hydrogen sulfide in attenuating myocardial ischemia‑reperfusion injury (Review). Molecular Medicine Reports, 24, 875. https://doi.org/10.3892/mmr.2021.12515
MLA
Wu, D., Gu, Y., Zhu, D."Cardioprotective effects of hydrogen sulfide in attenuating myocardial ischemia‑reperfusion injury (Review)". Molecular Medicine Reports 24.6 (2021): 875.
Chicago
Wu, D., Gu, Y., Zhu, D."Cardioprotective effects of hydrogen sulfide in attenuating myocardial ischemia‑reperfusion injury (Review)". Molecular Medicine Reports 24, no. 6 (2021): 875. https://doi.org/10.3892/mmr.2021.12515