Abstract
Hydrogen sulfide can signal through 3 distinct mechanisms: 1) reduction and/or direct binding of metalloprotein heme centers, 2) serving as a potent antioxidant through reactive oxygen species/reactive nitrogen species scavenging, or 3) post-translational modification of proteins by addition of a thiol (-SH) group onto reactive cysteine residues: a process known as persulfidation. Below toxic levels, hydrogen sulfide promotes mitochondrial biogenesis and function, thereby conferring protection against cellular stress. For these reasons, increases in hydrogen sulfide and hydrogen sulfide–producing enzymes have been implicated in several human disease states. This review will first summarize our current understanding of hydrogen sulfide production and metabolism, as well as its signaling mechanisms; second, this work will detail the known mechanisms of hydrogen sulfide in the mitochondria and the implications of its mitochondrial-specific impacts in several pathologic conditions.—Murphy, B., Bhattacharya, R., Mukherjee, P. Hydrogen sulfide signaling in mitochondria and disease.
Keywords: H2S, metabolism, ROS, apoptosis, persulfidation
Hydrogen sulfide (H2S) is a colorless, poisonous, and corrosive gas that gives rotting eggs their characteristic odor. Structurally, it is the sulfur analog of a water molecule, and it can be oxidized into elemental sulfur, sulfate (SO42−), thiosulfate (S2O3−), and sulfur dioxide (SO2) (1). Ecologically, the sulfur cycle is intricately intertwined with the carbon cycle through the activity of microorganisms known as SO42−-reducing bacteria (SRBs), which are key for the anaerobic breakdown of organic material. SRBs thrive in a vast array of anoxic habitats, including shallow marine and freshwater sediments, mud volcanoes, and hydrothermal vents. They are unique in their ability to perform anaerobic respiration through utilizing SO42− as their terminal electron acceptors rather than oxygen (O2), generating H2S as the main respiratory by-product; SRBs can breathe without oxygen (2, 3). They have a symbiotic relationship with aerobic sulfide-oxidizing bacteria, which can oxidize H2S back into SO42− to be continually utilized by SRBs. This is particularly noteworthy given the endosymbiotic hypothesis of mitochondrial origin, which suggests that all mitochondria originate from the integration of an endosymbiotic alphaproteobacterium into a host cell related to Asgard Archaea (4). For these reasons, it is not surprising that one of the main targets of H2S signaling in eukaryotic cells is the mitochondria.
The study of H2S began in the 1700s after the Italian physician and “father of occupational medicine,” Bernardino Ramazzini, noted the irritating effects of sewer gas on the eyes of sewer workers in his book De Morbis Artificum Diatriba (5). However, it was not until 1775–1776 that the responsible compound was identified as H2S (6). This discovery catalyzed early research on H2S as an environmental toxin. Nearly 166 yr later, in 1942, an American biochemist named Vincent Du Vigneaud crudely demonstrated the production of H2S in mammalian tissue homogenates. However, it was not until 48 yr after that, in 1989/1990, that H2S was truly considered to be a biologic mediator. During this time, 3 independent groups [Warenycia et al. (7), Goodwin et al. (8), and Savage and Gould (9)] reported the endogenous presence of H2S in the animal and human brain. Abe and Kimura’s (10) pioneering work in the late 1990s demonstrated the ability of H2S to act as a neuromodulator and vasorelaxant, confirming H2S as the third physiologic signaling gas, alongside carbon monoxide (CO) and nitric oxide (NO). Since these landmark studies, the field of sulfide research has grown rapidly, and H2S signaling has been implicated in several physiologic conditions including, but not limited to, promoting vasorelaxation, altering lipid metabolism, inducing angiogenesis, inhibiting monocyte adhesion, promoting tissue repair, and evading apoptosis (11–16). Moreover, H2S enzymatic generation and signaling is conserved in many organisms beyond human beings, including other mammals, bacteria, and plants, and may play a protective role in all (17). In a 2005 landmark paper, Blackstone et al. (18) reported that H2S could induce a reversible suspended animation–like state in mice. They posited that H2S-mediated induction of suspended animation could have beneficial medical applications, such as following ischemia-reperfusion (I/R) injury or organ preservation following trauma. Another landmark paper by Shatalin et al. (19) suggested that H2S has a key role in the development of antibiotic resistance in bacteria and showed that inhibition of the H2S-producing enzymes cystathionine β synthase (CBS) or 3-mercaptopyruvate sulfurtransferase (3-MST) rendered many pathogens sensitive to antibiotic treatment. Finally, in plants, H2S contributes to plant growth and development, plays a key role in the stress response, and may aid adaptation to stressful environments (20). Although H2S signaling is physiologically far reaching because of its gaseous properties, one of the main intracellular compartments where it exerts its effects in mammalian cells is the mitochondria. This review will summarize the current literature on H2S production, metabolism, and signaling, and then focus on H2S signaling in the mitochondria of humans and preclinical models of disease.
HYDROGEN SULFIDE BIOGENESIS AND METABOLISM
Localization of H2S-producing enzymes: CBS, cystathionine γ lyase, and 3-MST
H2S is endogenously produced in mammalian tissues through both enzymatic and nonenzymatic pathways. Although the former is generally considered to be the primary source of H2S, a 2019 report by Yang et al. (21) demonstrated that nonenzymatic H2S production predominates in all human tissues besides the liver and kidneys. Nonenzymatic generation of H2S occurs via the reaction of thiol or thiol-containing compounds with other molecules, such as the reduction of dietary inorganic polysulfides by glutathione (GSH) or the hydrolysis of inorganic sulfide salts (i.e., sodium sulfide [Na2S] or sodium hydrogen sulfide [NaHS] with water) (22–24). Yang et al. (21) reported that cysteine is the preferred substrate for nonenzymatic generation of H2S and that such a process is catalyzed by iron and vitamin B6. They emphasize that the nonenzymatic generation of H2S is underappreciated in the field and suggest that a better understanding of these processes is critical for the treatment of potentially several pathologies. Moreover, H2S produced by sulfur-reducing gut bacteria has also been reported to supply nonnegligible amounts of H2S systemically (25).
Enzymatic H2S production, on the other hand, has been extensively researched and occurs primarily through the actions of 3 enzymes: CBS, cystathionine γ lyase (CSE), and 3-MST. These enzymes are differentially expressed in tissues and can be found in various cellular compartments (26). For example, 3-MST may be more significant for H2S generation in tissues in which cysteine concentration is high (like the kidney) because it may play a role in sulfur amino acid catabolism (27). 3-MST expression has been reported in human kidney, liver, cardiac, and brain cells (specifically the neurons), with its predominant expression being in the gastrointestinal tract (28). It has also been shown to localize to both the cytosol and mitochondrial cellular compartments (29). CBS is predominantly expressed in the pancreas, male and female reproductive organs, and the astrocytes of the brain (30–34). CBS is also expressed in the liver, but its expression does not appear to be necessary for H2S production because CBS−/− mouse livers do not display reduced H2S levels when compared with wild-type (WT) mouse tissues (35). CSE, on the other hand, is highly expressed in the liver, kidney, neurons, macrophages, and smooth muscle cells, with its highest expression in the liver (36–40). However, CSE expression and activity is generally considered to be negligible in the heart and spleen (1, 41). Furthermore, CBS and CSE exist extracellularly as circulating enzymes, secreted by microvascular endothelial cells and hepatocytes; however, the recent report by Yang et al. (21) suggests circulating H2S may actually be due to nonenzymatic processes (42). Active 3-MST has been identified in red blood cells, although CBS and CSE expression was undetected (43). Although there is apparent overlap in tissue localization, these enzymes appear to perform nonredundant functions, and one enzyme may be more responsible for a particular tissue’s source of H2S. Therefore, H2S signaling may be enzyme and/or tissue specific; this specificity is discussed in later sections.
Our understanding of where these enzymes are located intracellularly is evolving. 3-MST has been reported to be found in both the mitochondria and cytosol, whereas CBS and CSE have been reported to be primarily cytosolic (44). However, our group and others have shown CBS in the mitochondria of both ovarian and colon cancer cells, and CSE can translocate from the cytosol to the mitochondria under conditions of cellular stress, such as increases in intracellular Ca2+ levels (45–47). Moreover, CBS and CSE are both capable of undergoing a post-translational modification by small ubiquitin-like modifier (SUMO) protein; SUMO is typically considered to be a nuclear localizing signal (48, 49). Indeed, sumoylation of CBS correlates with its localization to the nucleus and loss of catalytic activity and appears to be driven by levels of its substrates and products in the transsulfuration pathway. The consequences of CSE sumoylation are unknown, but it is hypothesized to be a nuclear localizing signal when nuclear GSH demand is high (49). How these H2S-producing enzymes are regulated to maintain their spatiotemporal production of H2S, as well as their crosstalk regulation of one another within mammalian cells, remains underexplored.
CBS and CSE function and regulation
CBS and CSE are key enzymes in the transsulfuration pathway, which fluxes methionine metabolism away from proteinogenesis and toward cysteine production and catabolism (Fig. 1) (50). Primarily, CBS catalyzes the first irreversible step in this pathway by driving the β-replacement reaction of homocysteine with serine to form the asymmetric thioether cystathionine and water. Interestingly, CBS can also carry out this reaction with a homocysteine and cysteine to directly generate cystathionine and H2S. Nevertheless, serine is believed to be the primary substrate for homocysteine condensation (36, 51). This is because human CBS shows a lower Michaelis constant for serine (2.76 mM) than for cysteine (6.8 mM) (52). The Michaelis constant is defined as the substrate concentration required to reach half the maximum enzymatic rate (Vmax); CBS requires less serine to reach half its maximum enzymatic rate compared with cysteine. Additionally, serine is reported to be more abundant within a cell than cysteine (36, 53, 54). It is not known whether or how the substrate preference for CBS can be switched, calling into question the physiologic relevance of homocysteine-cysteine condensation. For these reasons, the main mechanisms by which CBS produces H2S are via a β-replacement reaction with water and cysteine to form serine and through another β-replacement reaction between 2 cysteines to generate lanthionine (36, 55, 56).
Figure 1.
H2S generation via the transsulfuration pathway. The transsulfuration pathway plays a central role in sulfur metabolism and redox regulation in cells. It involves the transfer of sulfur from homocysteine to cysteine through an intermediate (cystathionine) and is the only way in which mammals generate cysteine endogenously. Methionine is converted into homocysteine in a reversible 2-step process involving the enzymatic activity of SAM and S-adenosylhomocysteine (S-AdoCys) (not pictured). Homocysteine is then converted to cystathionine through CBS-mediated condensation of homocysteine and serine or cysteine, which can generate H2O or H2S, respectively. Cystathionine is then acted upon by CSE to generate cysteine and α-ketobutyrate (not pictured). Cysteine can be further acted upon by CBS, CSE, or 3-MST/cysteine aminotransferase (CAT) to generate H2S, as well as other byproducts, such as GSH, pyruvate, and lanthionine. Notably, CSE is responsible for the physiologic clearance of homocysteine and can act on it alone to generate H2S.
The main role of the other H2S-producing enzyme in the transsulfuration pathway, CSE, is to convert cystathionine into cysteine and α-ketobutyrate. CSE can also produce H2S by 2 main mechanisms: via an α, β-elimination reaction with cysteine or through a γ-replacement reaction between 2 molecules of homocysteine. Under high homocysteine levels (hyperhomocysteinemia), the latter reaction is dominant and is essential for the rapid clearance of toxic homocysteine from the system (44, 52, 55). Therefore, CBS and CSE activity are key drivers of the transsulfuration pathway, as well as cysteine catabolism and H2S generation.
CBS is a homotetrameric enzyme of about 63-kDa subunits, and it binds 2 cofactors, pyridoxal 5′-phosphate (PLP) and heme (57). Although it can be regulated at the transcriptional level by nuclear factor (erythroid-derived 2)–like 2 (Nrf2), specific protein (SP)1, and SP3 in response to intrinsic and extrinsic stimuli, it is primarily constitutively expressed and regulated post-translationally (55). The noncatalytic N-terminal heme moiety in CBS acts as a redox sensor and exists in 2 oxidation states: ferric (Fe3+) and ferrous (Fe2+). Fe2+-CBS can bind CO and NO, resulting in the inhibition of its catalytic activity, implicating CBS as a crucial crossroads for all 3 gaseous signaling systems (58). In fact, a CBS variant commonly found in patients with homocystinuria, p.P49L, has altered spectral properties and an increased propensity to bind CO. This inhibition-prone CBS mutant may present a potential pathogenetic mechanism for the development of homocystinuria (59). CO/NO-induced inhibition of CBS can be rescued via exposure to air and the subsequent oxidation and recovery of the Fe3+ heme state (43, 58, 60–62). More recently, CBS has been reported to further demonstrate redox sensitivity through a CXXC motif in its central domain, which harbors a redox-active disulfide bond (Cys272–Cys275) (63). Niu et al. (63) demonstrated that this motif is essential to increase CBS activity and amplify CBS-dependent generation of H2S in human embryonic kidney (HEK) 293 cells under reductive-stress conditions. Additionally, CBS can be regulated by the allosteric activation of S-adenosylmethionine (SAM), a universal methyl donor (64). The C-terminal regulatory domain of CBS exerts intrasteric regulation through partial inhibition of its own catalytic domain. SAM can bind this C-terminal regulatory domain and induce a conformational change, which clears the way for substrate binding and alleviates this inhibitory effect; SAM binding to CBS enhances the β-replacement reaction of homocysteine with serine 2–5-fold (53, 55, 58, 65). CBS is also modulated by post-translational modifications, such as sumoylation, glutathionylation, and phosphorylation. Sumoylation was the first reported post-translational modification of CBS; Agrawal and Banerjee (49) reported that sumoylation of CBS by human polycomb group protein 2 correlated with its localization to the nucleus and subsequent loss of catalytic activity. Furthermore, Niu et al. (66) later showed that CBS could be S-glutathionylated at Cys346; this modification was induced by oxidative stress in HEK293 cells (challenged with peroxide) and resulted in an ∼3-fold increase in CBS activity. In this way, CBS activity can be propagated by one of the products of the transsulfuration pathway in an indirect positive-feedback mechanism (Fig. 1) (66). Finally, a 2016 report by d’Emmanuele di Villa Bianca et al. (67) demonstrated that activation of muscarinic receptors, M1 and M3, induced phosphorylation of CBSSer227 and subsequently enhanced H2S production in the urothelium, the epithelium that lines much of the urinary tract. Although considered to be constitutively expressed and rarely regulated at the transcriptional level, CBS activity is tightly regulated via its redox-sensitive nature, and post-translational modifications and can be super activated by the universal methyl donor, SAM.
CSE is a homotetramer composed of PLP-bound 45-kDa subunits and is the second H2S-generating enzyme in the transsulfuration pathway (68). CSE primarily breaks down the CBS by-product cystathionine into cysteine, α-ketobutyrate, and ammonia. Like CBS, CSE can also catabolize cysteine into various other byproducts to generate H2S (Fig. 1). The PLP-CSE interaction is required for enzymatic activity; the catalytic activity of CSEN187A, a mutant enzyme that cannot bind PLP, is zero. Unfortunately, we know little about the post-translational regulation of CSE, despite several known phosphorylation sites that may modulate CSE activity. Renga et al. (69) reported that CSE can be phosphorylated, and therefore activated, by the Akt pathway in response to G protein-coupled bile acid receptor 1 (GPBAR1) activation (a bile acid–activated receptor), leading ultimately to H2S-dependent vasodilation. Another group has shown that CSE can undergo persulfidation (a post-translational modification of cysteine discussed in detail later), but the significance of this modification is unknown (55, 70). Finally, CSE also undergoes sumoylation and the resulting nuclear localization (49). Unlike CBS, CSE is not constitutively expressed within cells; its expression is highly inducible in cells by a range of stimuli including endoplasmic reticulum (ER) stress, oxidative stress, nutrient deprivation, and hyperhomocysteinemia (55). CSE is encoded by the cystathionine gamma lyase (cth) gene, and its transcription is regulated under basal conditions by the transcription factor SP1 (71, 72). In the liver, where CSE is predominately expressed, farnesoid X receptor binds the CSE promotor and induces CSE expression and H2S production upon activation by bile acid (73). CSE can also be induced by inflammation; in macrophages, LPS and TNF-α stimulation lead to increased CSE expression and H2S production, as well as subsequent NF-κB persulfidation and recruitment to antiapoptotic gene promoters (74). CSE expression can be additionally induced by oxidative stress because the CSE promoter has been shown to contain a binding site for Nrf2, a master regulator of the oxidative stress response (75). Nrf2 has been reported to have binding sites within the sequences 5′-GTGATCTAGCA-3′ and 5′-ATGAGG CAGCT-3′ for CBS and CSE, respectively, implicating Nrf2 in the observed enhanced expression of both CBS and CSE under conditions of oxidative stress (76, 77). Surprisingly, H2S can regulate Nrf2 expression as a means of positive feedback, and this is further discussed in the persulfidation signaling section of this review. Nutrient deprivation and amino acid starvation have also been reported to up-regulate CSE expression via activating transcription factor 4 (78). Additionally, hormones can regulate CSE expression; reduced signaling of growth hormone and thyroid stimulating hormone, or depleted growth hormone receptor expression can globally increase CSE and CBS expression via a mechanism involving the hypothalamic-pituitary axis (79). Finally, Nandi et al. (80) recently showed that increasing levels of H2S itself can decrease CSE expression in HL1 cardiomyocytes. Mechanistically, they determined that H2S inhibits SP1 activation and CSE transcription, resulting in a loss of CSE expression. Additionally, they showed that increasing homocysteine levels conversely resulted in an increase in CSE expression, but the exact mechanism for this observation remains unknown. Surprisingly, increasing homocysteine and H2S levels had the opposite effect on CBS expression decreasing and increasing CBS, respectively, indicating an inverse relationship and potential novel feedback mechanism between CBS, CSE, and substrates of the transsulfuration pathway (80). This inverse relationship between CBS and CSE expression has not been adequately explored beyond this study, but it has notably been observed by several independent groups, including our own (12, 45, 46, 81, 82). Further exploration into the regulation and feedback of CBS and CSE is imperative to understanding their seemingly linear, nonredundant functions within the cell. Although CSE can be regulated through similar mechanisms to CBS, unlike CBS, it is predominantly controlled at the transcriptional level in response to cellular stress.
3-MST function and regulation
3-MST is a 33-kDa, cysteine-catabolizing enzyme that exists as either a monomer or an inactive disulfide-linked homodimer. It is primarily located in the mitochondria and cytosol of cells in most mammalian tissues, although its subcellular location seems dependent on its splice variant (29, 83). The human mecaptopyruvate sulfurtransferase (MPST) gene contains 595- and 299-bp exons divided by a 4405-bp intron, and the promoter region has features of a typical housekeeping gene promoter (84). Unlike CBS and CSE, 3-MST is primarily regulated via its redox-sensitive characteristics rather than post-translational modifications or transcriptional regulation. Its homodimer is formed through the redox-sensitive disulfide bond formation between 2 surface cysteines (Cys154 and Cys263) of two 3-MST molecules. Three other cysteine residues are present (Cys64, Cys247, and Cys254), with Cys247 being within the catalytically active site (85, 86). 3-MST can act as an antioxidant via oxidation of its Cys247 (3-MST-S) to cysteine sulfenate (3-MST-SO) or through oxidant-driven homodimerization. Both of these redox-sensing actions serve as enzymatic switches, thus ablating 3-MST activity; however, enzymatic activity is restored upon reduction by thioredoxin (Trx) of either 3-MST-SO or the disulfide-linked homodimer (83).
3-MST generates H2S and pyruvate through the breakdown of cysteine in a 2-step manner. First, cysteine aminotransferase converts l-cysteine and α-ketoglutarate to 3-mercaptopyruvate (3-MP) and glutamate. Next, 3-MST transfers the sulfur group from 3-MP to its sulfur-accepting nucleophilic Cys247 to generate a 3-MST–bound persulfide and pyruvate. This MST-persulfide can then react with thiols or be reduced by Trx to generate H2S (87, 88). Also of note, 3-MST can transfer the sulfur group from 3-MP to a cyanide molecule to form thiocyanate, which is safely metabolized and excreted (83). Because of this, 3-MST activity has inspired the development of a potential sulfanegen for treating cyanide poisoning (89). Like CBS and CSE, 3-MST can generate H2S through the catabolism of cysteine; however, unlike CBS and CSE, 3-MST regulation is primarily redox driven.
H2S metabolism
The intracellular levels of H2S have been reported to range from undetectable to >100 μM, yet the steady-state concentration of H2S in most tissues is estimated to be in the low nanomolar range (90). Kinetic studies have shown that sulfur fluxes more rapidly into H2S than to GSH, which, considering the low steady-state concentration of H2S, indicates a high clearance and/or consumption of H2S in tissues (91). Indeed, when administered at sublethal doses, H2S is rapidly oxidized and excreted as S2O3− and SO42− at a rate that is tissue specific (92, 93). This elimination of H2S occurs in a tightly regulated enzymatic fashion through a mitochondrial sulfide oxidation pathway, which is associated with the mitochondrial electron transport chain (ETC) at the level of complex III (Fig. 2). Briefly, H2S is oxidized in the mitochondrial matrix by sulfide quinone oxidoreductase (SQR) to generate a persulfide. This persulfide is further oxidized by ethylmalonic encephalopathy 1 protein (ETHE1), also known as persulfide dioxygenase, to produce sulfite. Sulfite is then further oxidized to SO42− by sulfite oxidase, or to S2O3− by rhodenase (36). Electrons released by SQR are captured by ubiquinone and transferred to the ETC at complex III, indicating that H2S oxidation and elimination can promote ATP synthesis.
Figure 2.
H2S metabolism. H2S can be metabolized via oxidation or methylation, occurring in the mitochondria and cytoplasm, respectively. However, H2S oxidation is thought to be the primary clearance pathway. During H2S oxidation, H2S is oxidized by SQR, thus forming a persulfide. This persulfide is further oxidized by ETHE1 to generate SO32−, which is converted to SO42− and S2O32− by sulfite oxidase and rhodanese, respectively. SO42− and S2O32− are then excreted into the urine and cleared from the body. Electrons released by H2S oxidation by SQR are captured by ubiquinone and transferred to the ETC at the levels of complex III. During H2S methylation, H2S is converted into CH4S and (CH3)2S via S-methyltransferase (TMT). (CH3)2S can also be further metabolized by rhodanese to generate SO42−, which is excreted as waste through the urine. Cyt c, cytochrome c.
Methylation of H2S is a secondary mechanism of metabolism and clearance. However, this mechanism is considered to be less important than oxidation and, unlike oxidation, occurs primarily in the cytoplasm of the cell (44). Through this mechanism, H2S is converted into methanethiol and dimethyl sulfide via thiol S-methyltransferase. Dimethyl sulfide is another substrate of rhodenase, which is oxidized to thiocynate and SO42− to be excreted in the urine.
Finally, H2S can be scavenged by the metalloproteins (methemoglobin, metmyoglobin, and metneuroglobin) by forming sulfheme products or metallo-/disulfide-containing molecules like oxidized GSH (GSSG) (43, 44, 94, 95). This catabolic pathway is particularly interesting when considering the signaling balance of the other bioactive gases, CO and NO, because hemoglobin (Hb) is also a common sink for these gases. Therefore, if this sink is exhausted by any of these gases, the other two are unable to bind, ultimately leading to ineffective quenching and altered target activities.
HYDROGEN SULFIDE SIGNALING
Chemically, H2S is a weak acid that directly ionizes in aqueous solution and freely diffuses through membranes (96). In biologic systems, H2S is known to signal through 3 distinct pathways: 1) scavenging reactive oxygen species (ROS) and reactive nitrogen species (RNS), 2) binding to and/or reduction of the metal centers of iron-heme proteins, and 3) post-translationally modifying protein cysteine residues in a process known as persulfidation (26).
Redox role of H2S
Several studies have shown that H2S can readily scavenge ROS and RNS, like superoxide (⋅O2), at higher rates than other classic antioxidants, such as cysteine and GSH (97). However, it should be noted that the capacity of H2S to serve as a physiologic antioxidant is questionable given that its physiologic concentration is much lower than other canonical antioxidants. H2S is accepted to be in the low nanomolar range, whereas GSH is present in the millimolar range at steady state (26). Nonetheless, exogenous H2S has demonstrated profound antioxidant and cytoprotective capabilities in physiologic systems exposed to ROS and RNS. Moreover, the small size of H2S, as well as its ability to freely diffuse across cellular membranes, may make it a more effective antioxidant in its immediate microenvironment than the larger molecule GSH, despite the dramatic concentration differences (98). Even so, it is more readily accepted that the observed antioxidant effects of H2S are the result of broader indirect signaling rather than direct ROS/RNS quenching. As such, H2S treatment can exert profound and prolonged antioxidant protection in cells.
One such way H2S may exert its antioxidant effects is through modulating the expression and activity of classic antioxidants, like GSH and Trx. GSH is a tripeptide made up of glycine, glutamate, and cysteine, one of the major antioxidants in cells and the primary building block in GSH synthesis. As previously described, cysteine is synthesized from methionine through the transsulfuration pathway, wherein the key H2S-producing enzymes CBS and CSE catalyze each step. Interestingly, several studies have shown that H2S can promote GSH production and subsequent protection against oxidative stress in the brain, spinal cord, heart, gastrointestinal tract, liver, kidney, and lung (99). This indicates potential positive feedback, whereby the H2S generated through the synthesis of GSH further promotes GSH production; several studies over the years have demonstrated this feedback. Lu et al. (100) illustrated this potential early on by showing that an H2S donor, NaHS, promoted glutamate uptake into astrocytes through the enhanced trafficking of glial glutamate transporter [glutamate transporter 1 (GLT-1), also known as excitatory amino acid transporter 2 (EAAT2)], which, in turn, increased the cystine/glutamate antiporter system, which pumps oxidized cystine in from the extracellular matrix. The now intracellular cystine could then be reduced into cysteine and converted to GSH through the concerted actions of 2 enzymes, glutamate-cysteine ligase and GSH synthetase (100). Kimura et al. (101) also described a different means by which H2S can promote cellular GSH production. They showed that H2S produced intracellularly diffuses out of the cell to reduce cystine into cysteine in the extracellular space. Cysteine would then be taken up in the cell via a cysteine transporter and utilized for GSH synthesis (101). Jain et al. (102) further reported in 2014 that H2S was able to increase cellular GSH through the up-regulation of glutamate-cysteine ligase catalytic subunit and modifier subunit. Most recently, however, this same group reported that treatment of C2C12 mouse myotubes with the H2S-donor NaHS increased the expression of GSH biosynthesis genes and that the silencing of endogenous CSE expression by small interfering RNA attenuated this expression (103). In these ways, H2S activity may indirectly contribute to redox homeostasis through the modulation of cellular GSH levels.
Trx is a small molecule of ∼12 kDa that contains a cysteine-glycine-proline-cysteine motif in its catalytic site. The 2 cysteine residues are the major sites of Trx oxidation, whereby reduction of ROS is facilitated via thiol-disulfide exchange. Oxidized Trx is reduced by Trx reductase, which is then further reduced by NADPH. Trx activity has been shown to exert intra- and extracellular functions in scavenging ROS to protect against oxidative stress, evade apoptosis, induce gene expression, and promote cellular proliferation (104). One of the earliest reports of H2S signaling on Trx came in 2008 when Jha et al. (105) demonstrated the protective effect of H2S against I/R injury in the mouse liver. This H2S-mediated protection was associated with an improved GSH/GSSG balance, reduced lipid hydroperoxide formation, and an increase in Trx levels (105). A more recent study from 2013 showed that treatment of H2S as Na2S not only increased Trx expression in a model of ischemic-induced heart failure but also attenuated high-fat–induced left ventricle remodeling of the heart (106). The cardioprotective effects of H2S against this model of heart failure were determined to be Trx dependent because mice that expressed a dominant negative mutant of Trx did not respond to Na2S treatment. Finally, H2S has also been shown to modulate the expression of Trx-interacting protein (TXNIP) in endothelial cells. TXNIP can bind to and inhibit Trx activity (107). Tian et al. (108) recently reported that endogenous H2S is necessary for the preservation of endothelial cell function via suppressing the MAPK/TXNIP cascade. CSE−/− knockout mice exhibited impaired endothelial-dependent relaxation, which was associated with an increase in MAPK phosphorylation and consequent up-regulation of TXNIP. Treatment of these knockout mice with the H2S donor NaHS rescued these results and restored endothelial function (108). Taken together, one way in which H2S signaling may contribute to redox homeostasis is through the regulation of classic antioxidants, like GSH and Trx.
As mentioned previously, H2S readily scavenges ROS/RNS, and such interactions may serve a greater purpose than simple redox balance. Indeed, some studies have shown that H2S can directly interact with several ROS and RNS, including hypochloric acid (HClO) (which is produced by neutrophils) and peroxynitrite (ONO2−), to not only protect against harmful oxidative stress but also propagate further H2S signaling and synergy with another signaling gas, NO (26).
Nagy et al. (109) showed that H2S readily reacts with HClO to form polysulfides, chemical compounds that contain a chain of sulfur atoms. Interestingly, hydrogen polysulfides (H2Sn) have been recently suggested to have signaling properties of their own, including regulating ion channels, tumor suppressors, and protein kinases (110). Additionally, some effects previously attributed to H2S signaling, such as persulfidation are now considered to be potentially regulated by H2Sn rather than directly through H2S, although the physiologic relevance of this is unclear (50, 111, 112). This is because H2S cannot react directly with a protein thiolate, because of thermodynamic constraints, but a polysulfide can (26, 111). Indeed, direct incubation of H2S with proteins that can undergo persulfidation, like glyceraldehyde 3-phosphate dehydrogenase (GAPDH) or bovine serum albumin (BSA), was unable to cause persulfide formation (97, 113, 114). However, the internal sulfurs in H2Sn are more oxidized and can therefore react with cysteine thiols and yield protein persulfidation in vitro (115). However, the physiologic relevance of persulfidation by polysulfides is controversial because persulfides are notoriously unstable and are readily reduced. Therefore, it is unclear how polysulfides survive endogenously when continuously at risk of reduction via intracellular molecules, such as GSH (50).
Of further interest is the interaction of H2S with ONO2−, a powerful oxidant and inducer of cell death that is formed by the reaction of free radical superoxide (⋅O2) with the free radical NO (⋅NO) in vivo. Inhibition of ONO2− formation or promotion of its decomposition has been reported to be cytoprotective in several pathologies, including vascular diseases, I/R injury, circulatory shock, inflammation, pain, and neurodegeneration (116). Thionitrate (HSNO2), a physiologic NO donor, is generated upon the direct interaction of H2S with ONO2− (98). Filipovic et al. (117) first showed this formation of HSNO2 and consequential promotion of NO signaling under physiologically relevant conditions in cells treated with H2S. Therefore, H2S has been shown to not only mitigate the toxicity of ONO2− through its active scavenging and clearance but also promote NO synergy through the formation of HSNO2. H2S can further modulate NO signaling pathways by reacting with S-nitrosothiols to form the small trans-nitrosylating thionitrous acid (HSNO) (117) and by reacting directly with NO to yield the signaling molecule nitroxyl (HNO), which has implications in cardiovascular health (118, 119).
Metalloprotein and H2S interactions
Another way in which H2S can exert its effects is through its reduction and/or direct binding of the heme centers of metalloproteins. The mechanism and consequences of these reactions are influenced by several aspects including: 1) the redox state of the heme-iron [(Fe3+)-heme or (Fe2+)-heme], 2) the environment in the heme pocket, 3) the protonation state of bound sulfide, and 4) the presence or absence of oxygen or reducing agents in solution (1). For example, Fe3+-heme can bind H2S or HS− directly, but the resulting moiety of such interactions is dependent on the residues in the heme pocket. Specifically, if there is a strong presence of basic residues within the pocket that can adequately accept protons from bound H2S, the resulting sulfide-reacted heme moiety is (Fe3+)-heme-HS−. On the other hand, if there are several nonpolar residues in the pocket, the fully protonated H2S interaction with the Fe3+-heme is stabilized, yielding (Fe3+)-heme-H2S. A comprehensive review of the chemistry of H2S heme-iron interactions are beyond the scope of the current review; readers are directed to other articles for additional details (106, 114, 120). This section will continue by examining several reported specific H2S-metalloprotein interactions and their physiologic consequences.
It is well established that heme proteins can also interact with NO, CO, and O2, illustrating another layer of crosstalk between the signaling gases. Cytochrome c oxidase (COX), the final structural complex of the ETC, is one such protein that can interact with all 3 signaling gases. Functionally, COX drives the final steps of the ETC by accepting 4 electrons from cytochrome c and transferring them to 1 molecule of oxygen, thereby converting 1 molecular oxygen (O2) into 2 molecules of water (H2O). This process of transferring electrons fuels the pumping of 4 protons (H+) across the inner mitochondrial membrane to establish an electrochemical proton gradient, which drives ATP synthesis by ATP synthase. The binding of H2S to COX has been reported to have a biphasic effect on cellular respiration, promoting it at low levels and completely ablating it at higher levels (121). This effect may be due to the 2 main mechanisms by which H2S can modulate COX: by binding the Fe3+-a3 and CuB subunits of COX, thereby inhibiting its enzymatic activity, or directly donating electrons to (i.e., reducing) COX and ostensibly serving as the only known inorganic substrate of the respiratory chain (122–124). Pietri et al. (125) suggest that the polarity of the environment around the heme a3 subunit of COX can explain this observed biphasic effect. They posit that the polar environment around the heme a3 subunit, which contains a tyrosine residue and CuB center, promotes heme a3 reduction to stimulate ETC activity at low H2S concentrations. However, at higher concentrations, H2S can directly bind both the a3 and CuB centers, thereby forming stable H2S-CuB and unstable heme H2S-Fe2+ inhibitory moieties. Because of the instability of the heme H2S-Fe2+ inhibitory complex, the reduced activity of COX at moderate levels of H2S is reversible. In fact, the displacement of H2S by O2 can completely restore COX activity at this stage (126). However, if H2S is in excess, it can push the further reduction of heme a3 to a stable heme a3 H2S-Fe3+ inhibitory complex, thereby decreasing heme affinity for O2 and rendering the inhibition of COX irreversible.
Interestingly, COX is not the only component of the ETC that can interact with H2S. Vitvitsky et al. (127) reported that the interaction between H2S and cytochrome c can lead to a plethora of complex signaling outcomes and protective effects. They suggest that the reduction of cytochrome c by H2S leads to the initial formation of a HS⋅/S⋅− radical. This radical can then either interact with a protein thiol to generate a persulfidated protein and superoxide (a process which will be discussed in detail in the next section) or promote further cytochrome c reduction and sulfite/S2O3− production. Reduction of cytochrome c by H2S can adequately promote ETC activity by bypassing complex III, which could lead to an increase in ATP synthesis and an accumulation of reactive sulfur species. Moreover, cytochrome c–mediated protein persulfidation could have major cytoprotective implications in and out of the mitochondria. For example, persulfidation of ATP synthase in the mitochondria would increase ATP synthesis, whereas persulfidation of procaspase 9 by cytoplasmic cytochrome c would abort cellular apoptosis postinitiation. Both scenarios lend credence to the cytoprotective effects of H2S.
Finally, as mentioned previously, interactions of H2S with methemoglobin, metmyoglobin, and metneuroglobin can serve as a scavenging mechanism and sink for excess H2S. Hb is composed of 4 globular chains, all of which harbor an oxygen-binding heme group. Myoglobin and neuroglobin, on the other hand, are monomeric globular proteins with only 1 heme group, both of which have higher affinities for oxygen than Hb (125). Interactions of these heme proteins with H2S form sulfheme derivatives, although the mechanism by which these are formed remains unknown. These sulfheme products have a much lower affinity for O2 and cannot be converted back into normal hemeproteins (125, 128–130). Because sulfheme proteins cannot adequately bind O2, high levels can be potentially toxic (as in the case of sulfhemoglobinemia), although low to moderate concentrations can be tolerated (131). This tolerance can be simply explained due to the physiologic turnover of red blood cells and implicates erythrocyte destruction as another means of H2S degradation (125, 132). More recently, Potor et al. (133) demonstrated that H2S can attenuate Hb oxidation and the subsequent lipid peroxidation in atherosclerotic lesions. Oxidized Hb can be both prooxidant and proinflammatory and may play a crucial role in atherogenesis, illustrating another means by which H2S is cardioprotective.
Persulfidation detection
The third and final way that H2S can signal is through a process known as persulfidation, whereby a thiol (-SH) is added to a reactive and accessible cysteine (-SSH) within a protein to alter its function. Persulfidation can be identified in many ways, but the most common assay is the modified biotin-switch assay, which was originally designed to identify S-nitrosylation in cell and tissue lysates (Fig. 3A) (134). Simply stated, free cysteines within a protein are blocked via methyl methanethiosulfonate, therefore leaving reactive persulfidated residues alone to be attacked by a biotinylated nucleophile (biotin-HPDP), leaving any disulfide bonds within the protein undisturbed. This biotinylated protein can then be precipitated using streptavidin beads for identification by Western blot and/or mass spectrometry (MS) analysis. Using this method, persulfidation was first identified in 2009 in a report by Mustafa et al. (70), wherein it was suggested that nearly 10–25% of proteins in the liver undergo persulfidation, including actin, tubulin, and GAPDH. This early report showed that persulfidation appeared to enhance cysteine reactivity, whereas S-nitrosylation diminished it. This observed trend of S-nitrosylation and persulfidation acting inversely to one another has held up in many reports following (134). Additionally, this early report identified 2 mechanistic consequences of persulfidation: it enhanced the catalytic activity of GAPDH, and it induced actin polymerization in liver.
Figure 3.
Methods of persulfide detection. A) Thiols are presumed to be selectively blocked by MMTS, which leaves persulfides free to be readily biotinylated by nucleophilic attack. These proteins can then be precipitated on streptavidin beads for analysis; however, this method is chemically problematic. B) In the modified tag-switch assay, both free thiols and persulfides are blocked by methylsulfonyl benzothiazole (MSBT), but the resulting persulfide-mediated mixed aromatic disulfide can selectively react with a cyanoacetic acid–based probe. Differential fluorescence tagging involves the tagging of both thiols and persulfides with a fluorescent maleimide derivative. C) The resulting disulfide from the tagged persulfide can then be reduced by DTT, and fluorescence is lost. D) Persulfidated peptides can be directly identified by MS following persulfide stabilization by maleimide. E) Persulfides can also be stabilized and biotinylated via biotin-tagged maleimide, immobilized on streptavidin beads, and eluted by DTT for detection and analysis. BT, benzothiazole; CN, cyanide; HPDP, pyridyldithiol; Mal, maleimide; MMTS, S-methyl methanethiosulfonate; SMe, methylated sulfur; S-SMe, dithiomethane.
It is worth mentioning that, despite continued wide use in the field, the modified biotin-switch method for persulfide identification is chemically flawed and therefore not considered to be completely selective for persulfides (50, 135). Its predominant flaw is that methyl methanethiosulfonate readily reacts with thiols and persulfides. This is the major hurdle for any method seeking to selectively tag persulfides. Given this flaw, other methods of persulfide identification have been developed, and use of the modified biotin-switch method is discouraged. However, Zhang et al. (113) refined this method and proposed a modified tag-switch assay, which is based on the notion that the disulfide bonds generated from reacting a persulfide with a thiol-blocking reagent are more sensitive to nucleophilic attack than other protein disulfides or thioethers (, 136). They proposed that persulfides could then be tagged using a nucleophile coupled to a reporter molecule following thiol and persulfide blocking. Specifically, they blocked thiols and persulfides using methylsulfonyl benzothiazole and used a biotinylated methyl cyanoacetate to tag the persulfides (Fig. 3B). The selectivity of this method was demonstrated using glutathionylated, sulfenylated, and unmodified BSA (which were untagged and contained intramolecular disulfides) and persulfidated BSA (which was successfully tagged). Two fluorescent cyanoacetic acid derivatives, which are considered to be more specific, have since been developed and now allow for detection of persulfides by fluorescence microscopy or in gels (137). Importantly, sulfenic acids are known to react with cyanoacetic acids, but this issue is easily circumvented by blocking sulfenic acids with dimedone. This method is currently the most selective and sensitive persulfide-tagging method for identification.
Another assay for persulfide tagging exploits the similarity in thiol and persulfide reactivity with electrophiles by labeling both with the fluorescent cyanine 5 (Cy5)-maleimide (74). The resulting R-S-S-maleimideCy5 adduct from the reaction with the protein persulfide is then subsequently reduced using a reducing agent, such as DTT, resulting in a loss of red fluorescent signal (Fig. 3C). Unfortunately, this method of detection is also not without issue. First, the presence of persulfidation is identified by a loss of signal, so it is not easily coupled to methods of proteomic analysis, which require a positive signal, like MS. Next, the ability of Cy5-maleimide to react with amines can also generate a lot of background signal, which could obscure any possible change in signal if persulfides are low in concentration. Finally, it is not clear how reducing agents differentiate between other DTT-reducible residues, like S-nitrosothiols and dilsufides, and persulfides (113).
The final common method of persulfide identification utilizes a proteomics MS analysis approach. Although MS can technically be used to directly identify the presence of persulfides in proteins, the shift in mass due to the addition of a sulfur atom is almost the same as the addition of 2 oxygen atoms (70). This makes it very difficult to differentiate these shifts in whole proteins. Additionally, persulfides are inherently unstable, which further adds to the difficulty of their direct detection by MS. Therefore, methods have been developed to stabilize peptide persulfides prior to MS analysis with N-ethylmaleimide or iodoacetamide (135, 138, 139) (Fig. 3D). A method that also utilizes persulfide trapping and MS analysis is the biotin thiol assay (Fig. 3E). In this assay, biotin maleimide, like Cy5-maleimide, reacts with both thiols and persulfides prior to protein fragmenting. The biotinylated peptides are bound to streptavidin beads, and persulfidated peptides are eluted from the beads via a reducing agent and analyzed via MS. Thiolated peptides, which are not held by a disulfide bond, remain on the beads.
Identified persulfidated proteins
Since the discovery of persulfidation, several other proteins have been shown to undergo this modification. We briefly discuss the following proteins, while leaving several others undescribed: transcription factors NF-κB, SP1, kelch-like ECH-associated protein 1 (Keap1), IFN regulatory factor 1 (IRF-1); enzymes MEK1, protein tyrosine phosphatase 1B (PTP-1b), ATP synthase F1 subunit α (ATP5A1), and lactate dehydrogenase (LDH) A; as well as ion channels such as transient receptor potential (TRP) channels and ATP-sensitive K+ (KATP) channels.
There are several transcription factors that can be persulfidated. As briefly touched upon in a previous section, the persulfidation of NF-κB, a crucial transcription factor for several prosurvival genes within a cell, is crucial for its antiapoptotic activity (74). Mechanistically, H2S persulfidates the p65 subunit of NF-kB at Cys38, which promotes binding to its coactivator ribosomal protein S3, therefore enhancing NF-kB transcription activity. Additionally, our group showed that H2S can persulfidate the transcription factor SP1 in endothelial cells at Cys68 and Cys755. Persulfidation of these residues stabilizes SP1 and regulates its transcription of VEGF receptor 2 and neurolipin-1, 2 major regulators of endothelial cell function (140). Interestingly, the previously mentioned antioxidant effects of H2S may be credited to persulfidation because increases in Trx, as well as GSH, may be attributed to impact of H2S on Nrf2 transcriptional activity. Nrf2 is mostly found inactive in the cytoplasm in a complex with Keap1. However, it has been reported that Keap1 can undergo persulfidation at Cys151, which induces a conformational change in Keap1 and leads to the dissociation of Nrf2 from the complex (141, 142). The newly liberated Nrf2 translocates to the nucleus, where it binds to promoters containing the antioxidant response element sequence and induces antioxidant response element–dependent genes, such as Trx and GSH. Finally, IRF-1, a transcription repressor, has been shown to undergo persulfidation to maintain mitochondrial biogenesis (143). H2S-induced persulfidation of IRF-1 enhances both its binding to the DNA methyltransferase 3a (Dnmt3a) promoter and the resulting repression of Dnmt3a. Dnmt3a is crucial for the methylation and inhibition of mitochondrial transcription factor A (TFAM) promoter. Therefore, upon IRF-1 persulfidation, Dnmt3a is bound up and unable to methylate TFAM, which enhances mitochondria biogenesis. Clearly, H2S has far-reaching cellular impacts at the level of protein transcription via regulation of transcription factors.
Cellular enzymatic activity can also be impacted by persulfidation. For example, MEK1 (also known as MAP2K1) has been shown to be persulfidated at Cys341 in human endothelial cells and fibroblasts (144). Persulfidation of MEK1 leads to the phosphorylation of ERK1/2 and translocation of ERK1/2 into the nucleus, where it can activate poly [ADP-ribose] polymerase 1 (PARP-1) and induce DNA damage repair. This persulfidation appeared to be crucial because mutations of Cys341 inhibited ERK1/2 phosphorylation and attenuated the DNA damage repair response. Moreover, PTP-1b, one of the earliest-identified protein tyrosine phosphatases and classic negative regulator of the insulin signaling pathway, has also been shown to be persulfidated. A 2011 report by Krishnan et al. (145) showed that H2S-mediated persulfidation of PTP-1b at Cys215 inhibited its enzymatic activity. This subsequently resulted in the phosphorylation and activation of downstream ER kinases, which facilitated the restoration of ER homeostasis under conditions of ER stress (145). In this way, persulfidation of PTP-1b may explain how H2S signaling can protect against ER stress and may therefore be an attractive therapeutic target in several disease states. Another enzyme that has been identified as a target of persulfidation is ATP synthase, which is driven by the proton gradient developed via the ETC. It is located in the inner mitochondrial membrane and is responsible for the generation of ATP by cellular respiration. Módis et al. (146) demonstrated that increasing H2S levels increased persulfidation of ATP5A1 at 2 cysteine residues (Cys244 and Cys294) in HEK293 and HepG2 cells. ATP synthase mutants containing cysteine-to-serine mutations at both Cys244 and Cys294 had significantly reduced enzymatic activity. Additionally, liver tissues taken from CSE−/− mice displayed lower ATP5A1 persulfidation and attenuated ATP synthase activity, indicating 1 way in which H2S signaling can maintain mitochondrial energy production and cellular respiration. Interestingly, H2S has been suggested to impact mitochondrial respiration beyond just ATP synthase persulfidation, but these additional mechanisms will be discussed in a later section concerning H2S impacts on mitochondria. Finally, one of the 2 LDH subunits, LDHA, has also been shown to undergo persulfidation (147). LDH is a tetramer that resides at the crossroads of aerobic and anaerobic glycolysis; there are 5 LDH isoenzymes composed of different arrangements of its 2 subunits, LDHA and LDHB. Isoenzymes rich in LDHA tend to assist in pyruvate-to-lactate conversion, whereas isoenzymes rich in LDHB conversely facilitate lactate-to-pyruvate conversion, which can feed into the Krebs cycle and promote oxidative phosphorylation. LDHA expression and activity is particularly pronounced in various cancers, shunting pyruvate away from feeding the Krebs cycle in the mitochondria and promoting the production of lactate and NAD+ in the cytosol. This shunting process, known as anaerobic glycolysis, is typically observed in normal cells in hypoxic conditions and is only viable for short amounts of time. However, despite the presence of oxygen, this process has been repeatedly observed in cancer cells (a canonical characteristic of cancer known as aerobic glycolysis or the Warburg effect). Because CBS, H2S, and LDHA are up-regulated in colon cancer, Untereiner et al. (147) wanted to see whether there was a link between H2S signaling and LDHA activity. Indeed, they discovered that LDHA activity is increased by H2S in a dose-dependent manner. Additionally, they showed that LDHA is persulfidated at Cys163, which is at least partially responsible for the observed increase in LDHA activity upon treatment with H2S donor, GYY4137.
The final group of proteins this review will briefly describe are ion channels, specifically TRP channels and KATP channels. TRP channels are ion channels located on the plasma membranes of most cells. There are about 30 different kinds of TRP channels, which can mediate a host of sensations like heat, pressure, taste, and pain. The first TRP channel to be identified to undergo persulfidation was reported in 2014 by Liu et al. (148). TRP channel TRPV6 is a membrane Ca2+ channel that is expressed in bone marrow mesenchymal stem cells (BMMSCs). Liu et al. (148) demonstrated that BMMSCs produce H2S as a means to propagate their self-renewal and osteogenic differentiation. They suggested that this self-sustaining process was due to persulfidation of TRPV6 channels at Cys172 and Cys329. Indeed, loss of H2S-dependent TRP cation channel subfamily V member 6 (TRPV6) persulfidation led to aberrant intracellular Ca2+ flux, which attenuated PKC/Erk-mediated Wnt/β-catenin signaling, and consequently inhibited osteogenic differentiation of BMMSCs. Although they only looked at the mechanism of TRPV6 in their report, Liu et al. (148) also suggested that several other TRP channels could undergo persulfidation, including TRPV3 and TRPM4, yet there has not been any additional research into the consequence of persulfidation on these channels. However, another group more recently identified another TRP channel, TRPV4, as another target of persulfidation. Naik et al. (149) demonstrated in 2016 that persulfidation stimulates TRPV4 activity in aortic endothelial cells to enhance Ca2+ and K+ influx and dilate vessels. Finally, the Kir6.1 subunit of the KATP channels was reported to be persulfidated at Cys43 in endothelial cells. Surprisingly, persulfidation led to a reduction in ATP association but an increase in phosphatidylinositol 4,5 bisphosphate binding to Kir6.1, which resulted in increased KATP channel activity and enhanced vasodilation (150). Moreover, both H2S-mediated KATP persulfidation and vasodilation were attenuated in Kir6.1-Cys43 mutants, suggesting that persulfidation of KATP may be the crucial mechanism by which H2S mediates smooth muscle relaxation and vasodilation.
Persulfidation formation and deformation
Despite the intense interest in H2S signaling by persulfidation, very little is actually understood about the process by which proteins undergo this modification. Originally, it was believed that thiolate on a protein could react directly with H2S to form a protein persulfide; however, this reaction is not possible, because of thermodynamic constraints. This lack of reaction was confirmed because incubation of proteins, like GAPDH, with H2S led to no detectable protein persulfidation (26, 97, 113). There are several working hypotheses to explain how persulfidation can occur (enzymatically or nonenzymatically) (Fig. 4).
Figure 4.
Mechanisms of protein persulfidation. A) H2S cannot directly act on a free thiol to produce a protein persulfide. B, C) However, H2S has been shown to directly act on preexisting modifications like S-sulfenylation (B) and S-nitrosylation (C), although reacting H2S with an S-nitrosothiol residue to generate a protein persulfide has only been demonstrated in silico. It has been reported that H2S may react with a disulfide bond (D), but the persulfide products are unstable, and the reaction rate is quite slow. E) Finally, persulfidation may not arise from H2S signaling directly but, rather, through polysulfide (HSx-, x = 2–7) signaling on a protein cysteine. Reactions in red are impossible, yellow are physiologically questionable, and green are potential true mechanisms.
Reactions with preexisting protein modifications is one proposed means by which proteins can acquire the addition of a thiol group onto its cysteine. For example, S-sulfenylation (S-OH) is an important oxidative post-translational modification that generates sulfenic acids and has been identified in nearly 1000 proteins, including several proteins that have been identified to be persulfidated as well, such as phosphatase and tensin homolog (PTEN) and PTP-1b (151). Sulfenic acids react readily with thiols to generate disulfide bonds and therefore have been suggested to react similarly with H2S. Indeed, Zhang et al. (113) demonstrated in a 2014 report that GAPDH can form a sulfenic acid that can be acted upon by H2S to generate a persulfide in its place. Moreover, they were able to generate persulfidated BSA utilizing its relatively stable sulfenic acid, as well as demonstrate that intracellular persulfidation colocalizes with the ER, an organelle that is rich with sulfenic acids. Notably, it is well established that sulfenic acids can be formed on cysteines simply in the presence of oxygen or metal ions in buffers, which can then react readily with H2S to form S-SH residues. This could offer one explanation as to why it was originally believed that H2S could directly react with thiols, a process which is now known to be thermodynamically unfavorable (26). As previously described, Vitvitsky et al. (127) demonstrated a means by which the direct interaction of H2S with cytochrome c generates a reactive HS⋅/S⋅− radical, which can tag proteins, such as procaspase 9, with a persulfide and further generate H2O2. H2O2 can also induce sulfenic acid (S-OH) formation to further propagate persulfidation and H2S signaling. The researchers posit that such reactivity may be particularly relevant under hypoxic conditions, when H2S metabolism is inhibited, or under apoptotic conditions, when cytochrome c is released into the cytoplasm.
S-nitrosylation is another important protein post-translational modification that can react directly with H2S. The reaction of H2S with S-nitrosothiols can lead to the removal of the -NO group from the protein cysteine to form the smallest S-nitrosothiol, thionitrous acid (HSNO), which can freely diffuse across membranes to propagate NO signaling (117). Although, the generation of a persulfide from this interaction is thermodynamically unfavorable, a 2013 in silico study did indicate that the surrounding molecular environment of the S-NO could feasibly impact the thermodynamics enough to allow for certain proteins to be directly persulfidated (152).
H2S interaction with an inter- or intramolecular disulfide bond is another attractive means by which H2S may generate persulfides without the requirement of enzymatic intervention; however, this mechanism’s physiologic relevance is questionable. Francoleon et al. (138) were able to show that GSSH could, in fact, be generated from mixing GSSG and H2S; however, the product was incredibly unstable. Moreover, Filipovic et al. (117) determined that between the bond energies of GSH disulfide (GSSG) and GSH-persulfide (GSSH), GSSH was lower; therefore indicating that the reaction of H2S with oxidized thiols is a slow reaction. Additionally, 2 other groups were unable to identify persulfidation of BSA or IgG as a consequence of reacting H2S with disulfides at physiologically relevant H2S levels.
Protein persulfidation as an intermediate is a process that has been known to be carried out in several biochemical processes by enzymes known as sulfurtransferases and cysteine desulfurases (153). Two examples of this enzymatic generation of persulfides have already been previously discussed through the examination of 3-MST and SQR activity. Even more interesting, work done by Ida et al. (154) on reactive cysteine persulfides has introduced a new way of thinking of CBS and CSE activity. They suggest that cysteine persulfide (Cys-SSH) arises from the respective CBS and CSE generation and cleavage of Cys-S-S-Cys (cystine) and is the true by-product of the transsulfuration pathway (H2S being a product of C-SSH decomposition) (154). They found that the generation of the reactive intermediate, Cys-SSH, was sufficient to lead to dramatic increases in per- and polysulfidated proteins rather than H2S. This offers insight into CBS/CSE/3-MST persulfidation specificity but does not exactly answer why exogenous H2S or H2S donors could rescue persulfidation in several studies described above. Nonetheless, such research offers new insights into the complexity of persulfide formation and H2S signaling.
More intriguing still is that persulfidation may not directly involve H2S at all but, rather, molecules derived from H2S, the previously mentioned polysulfides. Polysulfides (HSx−) are products of incomplete H2S oxidation and can range from x = 2 to x = 7; H2S2 is extremely unstable under biologic conditions and readily breaks down into H2S and elemental sulfur, whereas longer polysulfides are more stable, and HSx−, where x = 4 or 5 are the most abundant species (26). Surprisingly, a 2013 report by Kimura et al. (115) suggested that polysulfides have a 300-times-higher capacity to activate TRP channels than H2S. Additionally, another group suggested that many experimental H2S donors, such as NaHS, are contaminated with polysulfides, which may ultimately be responsible for experimentally observed persulfide formations (155). Mechanistically, the sulfane sulfur atoms (sulfur atoms that are linearly covalently bound only to other sulfur atoms) within these polysulfides are believed to attack the free thiol within a protein, thereby leading to persulfidated proteins.
Like other post-translational modifications, cellular mechanisms exist that can remove a persulfide from a protein to shut down the signal. As mentioned previously, Trx is a disulfide oxioreductase, which plays a critical role in redox homeostasis within the body. It has 2 cysteines in its catalytic site that are the primary regions of ROS reduction. However, beyond this redox role, Trx and other Trx-related proteins are also key players for the removal of persulfide modifications (50, 137, 156). Indeed, Trx successfully reduces HAS persulfide and Cys-SSH; auranofin, a Trx reductase inhibitor, increases total levels of intracellular persulfidation (137). Moreover, silencing of Trx-related protein of 14 kDa also was shown to increase intracellular persulfidation, which may be a relevant depersulfidase under conditions in which Trx activity is focused to other systems, like the peroxiredoxin system during conditions of oxidative stress (156). Mechanistically, it is thought that the outer sulfur of the protein persulfide is transferred to the nucleophilic cysteine in Trx, resulting in a transient Trx-SSH, which is rapidly resolved into H2S and oxidized Trx. Alternatively, the nucleophilic cysteine of Trx may react with the inner sulfur of the persulfide, therefore resulting in the elimination of H2S and a Trx disulfide complex, which is subsequently resolved. Interestingly, another canonical redox system may also act to depersulfidate proteins. The glutaredoxin system (GSH/GSH reductase/glutaredoxin), which maintains low levels of GSSG to maintain redox homeostasis, has also been reported to reduce persulfides in vitro (156). However, GSH reductase knockout mice did not show any difference in persulfidation when compared with their WT counterparts; the physiologic contribution of the glutaredoxin system to depersulfidation remains unclear.
Although there is still much to learn regarding the specifics of H2S physiologic signaling, H2S has promise as a therapeutic agent in a range of disease states. Several were touched upon in earlier sections, but the rest of this review will take a more in-depth look at the role of H2S in disease and specifically how H2S effects on the mitochondria may be the central link.
HYDROGEN SULFIDE, MITOCHONDRIA, AND DISEASE
Hydrogen sulfide and mitochondria
The mitochondria are considered one of the primary targets of H2S signaling, having impacts on mitochondrial respiration, mitochondrion-dependent apoptosis, biogenesis, and morphology (Fig. 5). Like the other signaling gases, NO and CO, H2S is known to directly impact the activity of the ETC, acting as both a substrate and COX antagonist depending on concentration (122, 157). At concentrations <20 μM, H2S can actually act as the only inorganic substrate, donating an electron to the ETC via its own metabolism by SQR (158–160). The electrons released from H2S oxidation by SQR are captured by ubiquinone and transferred to the ETC at complex III to be further propagated down the chain. This activity can stimulate oxidative phosphorylation and ATP production within the cell. In a 2019 report, Libiad et al. (161) demonstrated the importance of this mitochondrial sulfide oxidation pathway in colon cell metabolism. The colon epithelium is harbored in a unique environment in that it is regularly exposed to high levels of H2S generated from the gut microbiome. Because of this, the sulfide oxidation pathway in colon cells is exquisitely efficient. The researchers report that the sulfide oxidation pathway enzymes are located at the host-microbiome interface and that SQR expression is critical to shield the epithelium from H2S poisoning and effectively promote oxidative phosphorylation. On the other hand, high levels of H2S were reported to inhibit the ETC, perturb redox balance, shift metabolism toward reductive carboxylation, and inhibit cellular proliferation (161). Additionally, H2S generated inside the mitochondria by 3-MST has been reported to be sufficient to enhance mitochondrial electron transport and cellular bioenergetics at low levels in a manner that is SQR-dependent (124). Moreover, a mitochondrion-specific H2S donor, AP39, which consists of a mitochondrion-targeting motif (triphenylphosphonium) coupled to an H2S-donating moiety (dithiolethione), was shown to stimulate mitochondrial bioenergetics (162). Furthermore, direct persulfidation and consequential enhancement of ATP synthase is yet another means in which H2S can stimulate ETC activity within the mitochondria (146).
Figure 5.
Impacts of H2S on mitochondria. Green arrows indicate persulfidation signaling, blue arrows are antioxidant effects, and red arrows are metalloprotein interactions by H2S. H2S can persulfidate a number of transcription factors, such as Nf-KB, IRF-1, and PGC-1a, to promote transcription of several antiapoptotic and mitochondrial biogenesis genes. Moreover, H2S can increase PGC-1a expression via cAMP/PKA/AMPK activation, which enhances its transcriptional stimulation of mitochondrial biogenesis, as well as coactivates Nrf2, which promotes cellular survival pathways. Nrf2 can also be activated by H2S through its persulfidation-induced disassociation from its complex with Keap1. Furthermore, H2S activation of mitoKATP channels via persulfidation has also been shown to directly inhibit apoptosis. Finally, H2S can prevent apoptosis after its induction via binding to and sequestering the cytochrome c (Cyt c) released into the cytosol from the mitochondria. H2S can also stimulate mitochondria bioenergetics. Persulfidation of ATP synthase and direct H2S electron transfer to the ETC via SQR/ubiquinone and COX interactions stimulate ATP production within the mitochondria. Lastly, H2S production has been shown to maintain mitochondrial fusion and prevent mitochondrial fission. One mechanism by which it can do this is through the redox-sensitive maintenance of the outer mitochondrial membrane fusion protein MFN2.
As H2S levels increase, its inhibitory effects on COX overpower its stimulatory effects until the complex is completely inhibited. In an in vivo lugworm model, Arenicola marina, complete inhibition is achieved at 50 µM; however, the concentration required for full COX inhibition in other models is dependent on factors such as cell type and cell environment (158, 163). At these levels, H2S is highly cytotoxic, shutting down ETC activity, decreasing intracellular ATP levels, enhancing ROS generation, and inducing mitochondrial depolarization and apoptosis (158, 164). Moreover, H2S has been shown to further inhibit ATP production in high doses via up-regulation of uncoupling protein 2, which dissipates the proton gradient to disrupt ATP synthesis, and down-regulation of COX subunits I and II (165). The researchers suggest that through these processes, the cells can adapt to H2S onslaught. Indeed, up-regulation of uncoupling protein 2 can protect the cells from oxidative stress via reduction of ROS production and down-regulation of COX subunits, which helps to conserve precious energy while ATP synthesis is disrupted.
H2S can also elicit its effects on mitochondrion-dependent apoptosis. At toxic levels, H2S induces apoptosis via translocation of Bcl-2 associated x protein (BAX) to the mitochondria, which assists in the formation and opening of the mitochondrial permeability transition pore (mPTP), subsequent release of cytochrome c, activation of proapoptotic proteins (p21), and down-regulation of antiapoptotic proteins [B-cell lymphoma 2 (Bcl-2)] (164, 166, 167). In rat cerebellums with an ETHE1 deficiency, representative of the rare autosomal disorder ethylmalonic encephalopathy, H2S accumulation led to mitochondrial swelling, decreased mitochondrial membrane potential (ΔΨm), and enhanced mPTP induction (168). Moreover, a 2019 report by George et al. (169) suggested that CSE/H2S signaling mediated LPS-induced late-apoptotic events in murine macrophages.
However, under certain conditions, and at nontoxic levels, H2S has actually been demonstrated to play an antiapoptotic role. An early report in 2009 by Hu et al. (170) showed that H2S attenuated rotenone-induced apoptosis in neuroblastoma cells. The researchers found that H2S elicited precisely the opposite effects on ΔΨm, cytochrome c release, and BAX levels, respective to earlier reports, thereby explaining the observed antiapoptotic phenotype. They suggested that H2S achieved this via inhibition of the proapoptotic MAPK pathway, which is mediated by mitochondrial KATP, which, as previously established, can directly undergo persulfidation at Cys43 to induce vasorelaxation. Blockage of these channels ablated H2S-mediated protection, therefore confirming that the cytoprotective role of H2S is, at least partially, KATP dependent. Another study by Kimura et al. (171) also demonstrated the cytoprotective effects of H2S-dependent KATP activation in cortical neuronal cells. Furthermore, a recent report by Wang et al. (15) indicated that persulfidation of mitochondrial ATP5A1 at Cys244 conferred resistance to mitochondrion-mediated apoptosis in the adrenal glands of male mice challenged with LPS as well as CBSb/− mice (15). Even more recently, Vitvitsky et al. (127) demonstrated a mechanism by which H2S can directly prevent apoptosis, even after an apoptotic event has been triggered. They suggest that under apoptotic conditions, when cytochrome c is released into the cytoplasm, oxidation of H2S by cytochrome c can propagate the persulfidation of nearby target proteins, such as procaspase 9. Specifically, they showed that persulfidation of procaspase 9 resulted in its inhibition and consequential inhibition of the caspase cascade and mitochondrion-dependent apoptosis. Finally, H2S can exert its antiapoptotic effects through direct persulfidation of NF-κB, which enhances its binding to the promoters of several antiapoptotic genes (74).
H2S has also been shown to regulate mitochondrial biogenesis, or the generation of new mitochondria. This process is controlled by its master regulator, peroxisome proliferator-activated receptor γ 1α (PGC-1α), which coactivates Nrf2, to turn on TFAM and induce the transcription of nuclear-encoded mitochondrial proteins (172). H2S impacts on Nrf2 and TFAM were already discussed in a previous section of this review, where it was reported to indirectly enhance activity of both via persulfidation of upstream proteins (141–143). Interestingly, 2 more recent reports by Untereiner et al. (173, 174) demonstrated a more direct impact of H2S signaling on mitochondrial biogenesis. In their first report, they showed that endogenous CSE-dependent H2S production up-regulated PGC-1α in primary hepatocytes via activation of the cAMP/PKA pathway. Moreover, they reported an increase in PGC-1α activity due to H2S-driven persulfidation. They postulated that such impacts would enhance mitochondrial biogenesis and went on to confirm this speculation in their following report. They further reported H2S impact on another regulator of mitochondrial biogenesis, peroxisome proliferator–activated receptor-γ coactivator-related protein (PPRC), which was shown to be a target of persulfidation and whose expression was also regulated by CSE-derived H2S. Recently, another independent group showed H2S regulation of mitochondrial biogenesis via AMPK regulation of PGC-1α in primary murine cardiac tissue (175). Oral administration of an active H2S-releasing prodrug, SG-1002, enhanced cardiac mitochondrial content and localization of PGC-1α to the nucleus in a dose-dependent manner. Perhaps unsurprisingly, lower levels of SG-1002 had a positive influence on PGC-1α activity, whereas higher doses imparted a negative influence. Finally, a 2019 report indicated the propensity of H2S to maintain mitochondrial biogenesis in osteoblasts challenged with elevated levels of homocysteine, which was able to restore osteoblast function, ATP synthesis, and redox homeostasis (176).
Finally, and most recently, H2S has been reported to impact mitochondrial morphogenesis, the balance maintained between mitochondrial fission and fusion processes within the cell. Although once thought to be static organelles, mitochondria are now recognized to undergo dynamic fission and fusion processes, which can impact mitochondrial function and spatial placement within the cell (177). These processes are regulated by a family of GTPases known as dynamin-related proteins (Drps). Drp1 is the primary driver of mitochondrial fission, whereas mitofusins 1 and 2 (MFN1/MFN2) drive outer mitochondrial membrane fusion, and optic atrophy 1 (OPA1) drives inner mitochondrial membrane fusion (178–181). Fission 1 (Fis1) regulates Drp1 recruitment to the mitochondria and can further assist in the fission process as well (182). Interestingly, H2S can also regulate these GTPase expression levels. The first account of H2S regulation of mitochondrial morphogenesis machineries came from Liu et al. (183) in 2017; they demonstrated that H2S reduced mitochondrial fragmentation in endothelial cells in a high-glucose/high-palmitate environment by down-regulating phosphorylated Drp1/Drp1 levels, and this in turn prevented ROS accumulation and apoptosis (183). An earlier report by Qiao et al. (184) demonstrated H2S-driven mitochondrial fusion; utilizing transmission electron microscopy, they showed that the H2S donor NaHS inhibited mitochondrial fission in a dose- and time-dependent manner in vitro in mouse neuroblastoma cells (neuro-2a). They determined that the impact on mitochondrial morphology was due to an ERK1/2-dependent loss of Drp1 at the mRNA and protein level and that Drp1 overexpression was sufficient to reverse mitochondrial elongation seen in H2S-treated cells. Two 2018 independent studies further implicated H2S signaling activity on mitochondrial morphogenesis (185, 186). Both studies observed a sirtuin 3 (SIRT3)-dependent up-regulation in OPA1 and down-regulation in DRP1, whereas Meng et al. (185) also specifically noted an increase in MFN1/MFN2 and decrease in Fis1 expression levels. Both groups noted enhanced mitochondrial function upon H2S treatment in mouse models of cardiac hypertrophy, which was partly due to its regulation on mitochondrial morphogenesis. A 2018 report by our group further reported the necessity of H2S promotion of mitochondrial fusion, showing that CBS-dependent H2S stabilized MFN2 in a redox-sensitive manner in ovarian cancer cells (187). Upon CBS knockdown, mitochondria become hyperfragmented, which leads to an increase in oxidative stress and induces MFN2 degradation through the activation of JNK/HUWE1 pathway. Finally, a 2019 study by Xu et al. (188) on wound healing demonstrated that exogenous H2S increased OPA1 and decreased Drp1 expression levels, which consequently enhanced ΔΨm, inhibited ROS production, and enhanced wound-healing capacity in skin fibroblasts.
Clearly, H2S signaling is far reaching, but the importance of its impact on mitochondrial function and mitochondrion-dependent processes cannot be overstated. Mitochondria lie firmly at the crossroads of cell life and death, serving as major redox organelles and a key source of ATP generation. Therefore, it is not surprising that H2S has shown therapeutic promise in a host of disease states, due in no small part to its regulation of the mitochondria. The following sections will briefly examine the recent and impactful reports of H2S signaling in several diseases from a mitochondrion-focused perspective. Several reviews exist that focus on the role of H2S in specific diseases beyond its mitochondrial impact. The reader is encouraged to read these other sources for more in-depth information on H2S in specific disease states (13, 44, 189–198).
Hydrogen sulfide and cardiovascular disease
Although the role of H2S as a signaling gas was first demonstrated in the brain, its potential role as a cardioprotective agent was immediately appreciated. This is because it has been shown to be endogenously produced in vascular smooth muscle cells, as well as to induce vasorelaxation in rat aortic tissues, much like another signaling gas, NO (10, 11). Moreover, because of the roles CBS and CSE play in metabolizing homocysteine through the transsulfuration pathway, defective CBS/CSE function has been linked to hyperhomocysteinemia, a medical condition and serious cardiovascular disease risk characterized by excessive levels of homocysteine (>15 µM) in the blood (51, 199). Additionally, homocystinuria, a debilitating multisystem disorder that targets the cardiovascular system as well as connective tissues and the nervous system, is caused by a genetic deficiency of CBS (199). For these reasons, and others, H2S was considered as a potential cardioprotective agent and potential therapeutic against a multitude of cardiovascular diseases.
Endothelial cells line the interior of blood and lymphatic vessels, and their dysfunction has been suggested to contribute to a host of cardiovascular diseases, including atherosclerosis and hypertension (200, 201). Hydrogen sulfide has been reported to protect endothelial cell function from various stressors (such as hyperglycemic, hyperlipidemic, and high-salt insult) via the mitochondria in several studies. In one such study, Liu et al. (183) reported that rat aortic endothelial cells treated with high glucose and high palmitate (a fatty acid) had significantly less CSE expression and H2S production. However, treatment of these cells with exogenous H2S inhibited mitochondrion-dependent apoptosis, which was associated with a reduction in expression of mitochondrial fission proteins, mitochondrial fragmentation, and ROS levels, as well as increased mitophagy activity (183). An earlier study also demonstrated that the mitochondrion-targeted H2S donors AP39 and AP123 successfully decreased mitochondrial membrane hyperpolarization and ROS production and additionally increased ETC complex III activity and mitochondrial metabolism in endothelial cells challenged by glucose onslaught (202). A high-salt diet, a contributing factor to clinical hypertension, induces vascular endothelial cell apoptosis (203). A 2015 report by Zong et al. (203) demonstrated that high salt decreased expression of CSE in HUVECs and that H2S may protect endothelial cells from apoptosis by attenuating oxidative stress and maintaining mitochondrial health. Moreover, AP39 pretreatment also protected mitochondrial DNA integrity and bioenergetics in endothelial cells under oxidative stress, thus conferring a cytoprotective effect (162).
Atherosclerosis is defined as the hardening and narrowing of arteries due to the buildup of fatty plaque along the arterial walls. It is the leading risk factor in a host of cardiovascular diseases, including stroke, heart attack, and coronary artery disease, and develops from chronic pathophysiological processes such as vascular inflammation, endothelial dysfunction, smooth muscle cell proliferation and migration, lipoprotein accumulation, macrophage differentiation, and monocyte adhesion (204). Over the last several years, H2S has been demonstrated to have effects in many of these processes and is therefore considered to be an attractive therapeutic against atherosclerosis. For example, H2S administration has been shown to inhibit lipid hydroperoxide formation in LDL and protect against oxidized (oxidized LDL) cytotoxicity via inhibition of cytotoxic lipid accumulation and foam cell formation, which are key steps in atherogenesis (14, 142, 205, 206). Moreover, oxidative stress-induced mitochondrial dysfunction and apoptosis in endothelial cells is considered to play a pathogenic role in the development of atherosclerosis (207, 208). Wen et al. (209) demonstrated that CSE-dependent H2S signaling prevented HUVECs from undergoing apoptosis upon exposure to H2O2. This effect was credited to H2S-mediated protection of mitochondrial function from oxidative damage, as determined by analysis of ATP synthesis, ΔΨm, and cytochrome c release. But H2S mitigation of atherogenesis goes beyond redox restoration. Mitochondrial DNA damage has been reported as a risk factor independent of ROS to accelerate atherosclerosis by acting on vascular smooth muscle cells and monocytes (210). As previously discussed, MEK1 persulfidation by H2S induces PARP-1–mediated DNA damage repair (144). A 2018 report by Cheung et al. (211) further elucidated the preventative nature of H2S against atherosclerosis via persulfidation. They identified 70 persulfidated aortic proteins, 23 of which were localized to the mitochondria, such as superoxide dismutase and medium-chain specific acyl-CoA dehydrogenase, which play key roles in mitochondrial lipid metabolism and redox maintenance. GSH peroxidase, which is key in reducing lipid peroxidation, was also found to be persulfidated, which consequently reduced lipid peroxidation and enhanced antioxidant defense. Finally, H2S was shown to inhibit macrophage-derived foam cell formation, another key event in atherogenesis, via activation of the KATP/ERK1/2 pathway (14).
Untreated atherosclerosis and the resulting blockage of coronary arteries can lead to a life-threatening condition known as acute myocardial infarction, or heart attack. H2S has been shown to reduce myocardial ischemia injury through protection of mitochondrial function and potentially via mitochondrion-mediated macrophage M2 polarization (212, 213). In the first scenario, rat hearts were perfused and subjected to ischemic conditions both with and without the H2S donor NaHS. H2S treatment successfully reduced infarct volumes following ischemia injury (212). The researchers suggested that this protection was mediated by H2S antioxidant signaling and protection of mitochondrial function. In the latter scenario, the researchers reported reduced pathologic cardiac remodeling following myocardial infarction in WT and CSE−/− mice treated with H2S supplementation (213). H2S treatment significantly shrunk infarct size and mortality, which correlated with an increase in the proresolution M2 subset of macrophages. Interestingly, transplanting NaHS-treated bone marrow–derived macrophages into macrophage-depleted WT and CSE−/− mice was able to attenuate cardiac damage following myocardial infarction. The researchers determined that this H2S-dependent M2 polarization was driven by enhanced mitochondrial biogenesis and increased fatty acid oxidation in the macrophages.
Tissue damage caused by the reintroduction of blood into the tissue following periods of ischemia (lack of oxygen/hypoxia) is known as I/R injury, or reoxygenation injury. During the prolonged ischemic period, such as during cardiac arrest, tissues acutely adapt to the hypoxic environment in such a way that upon rapid oxygen reperfusion, oxidative stress and oxidative damage can damage the tissue. In myocardial I/R injury, rapid pH normalization, and Ca2+ overload, as well as ROS/RNS onslaught at reperfusion due to loss of proper mitochondrial function, result in the opening of the mPTP and propagation of the apoptotic pathway (214, 215). There is growing evidence that H2S protects mitochondria during I/R to improve respiration and promote biogenesis. Indeed, treatment of isolated mitochondria with an H2S donor, Na2S, induced a greater recovery of posthypoxic respiration than in those mitochondria that went untreated (216). Furthermore, the mitochondrion-targeted H2S donor, AP39, distributed to rats undergoing I/R at the time of reperfusion, was shown to protect against myocardial I/R injury by inhibiting mPTP in a cyclophilin d–dependent manner (217). AP39 has also shown reproducible protection against I/R in brain and kidney injury (218–220).
Finally, H2S has also been shown to have promise as a potential therapeutic agent against hypertension and systemic hypertensive damage. Physiologically, although CSE expression is dominant over CBS expression in the vasculature, pharmacological inhibition of CSE by dl-propargylglycine or CBS by aminooxyacetic acid (AOAA) alone was not sufficient to alter blood pressure in rats. However, utilizing both drugs in combination with one another was able to induce hypotension (221). It is worth mentioning that a purely selective pharmacological inhibitor for CBS does not exist on the market. In fact, AOAA, which is commonly used as a CBS inhibitor, actually inhibits both CBS and CSE and surprisingly preferentially inhibits CSE over CBS (222). Therefore, we speculate that perhaps the observed hypotensive phenotype observed by dl-propargylglycine/AOAA treatment was due to intense CSE inhibition rather than CSE/CBS dual inhibition. Regardless, several studies have shown that H2S deficiency may contribute to the development of hypertension in humans, with reported plasma H2S levels being lower in patients with grade 2 and 3 hypertension when compared with patients with normal BP (223). Indeed, H2S intervention in several studies utilizing murine models of hypertension has shown therapeutic promise (224–228).
Hydrogen sulfide and neurodegenerative disease
Before H2S was assigned any other functional role in humans, it was first established as a neuromodulator (10). All 3 H2S-generating enzymes have been thoroughly implicated in the CNS, with CBS being the predominant source in astrocytes and CSE/3-MST predominantly in the neurons (192). Therefore, it is not surprising that H2S signaling has been extensively studied in the brain and neurologic diseases, including Alzheimer’s disease (AD), Parkinson disease (PD), and Huntington’s disease (HD).
The most prevalent neurodegenerative disorder is AD, a devastating condition associated with aging that impacts the cerebral cortex and hippocampus and dramatically impairs an individual’s cognitive function and memory (229). A key characteristic of AD cases is the presence of mutations in the amyloid precursor protein (APP), which undergoes sequential proteolysis by α, β, and γ secretases. Another hallmark of AD is the aggregation of β-amyloid (Aβ), which is formed by the aforementioned dysfunctional processing of APP and Tau proteins, which generate amyloid plaques and neurofibrillary tangles, respectively. Many murine AD models have APP mutations and display pronounced amyloid plaques (230). It has been suggested that soluble Aβ can deregulate H2S signaling through inhibition of the neuronal cysteine transporter EAAT3/EAAC1, which results in oxidative stress and neurodegeneration (231, 232). This inhibition could impact H2S generation and GSH production by CBS and CSE acting on cysteine in the brain. Indeed, H2S levels in the plasma and brain, as well as neuronal expression of SAM, an allosteric activator of CBS, were both diminished in patients with AD when compared with healthy controls (233–235). Treatment of H2S in the APP/PS1 mouse model of AD has been reported to reduce cognitive impairment as well as mitigate oxidative stress (236). Moreover, H2S impact on redox homeostasis and mitochondrial biogenesis through Nrf2 activity via Keap1 persulfidation may also offer another explanation of therapeutic protection that H2S confers against AD, given that NaHS enhances Nrf2 expression (77, 236). Moreover, a recent study reported that H2S treatment inhibits β-secretase 1 (BACE1), the β secretase that processes APP and is responsible for Aβ generation, via regulation of the PI3K/Akt pathway in the APP/PS1 model of AD (237). Finally, AP39, a mitochondrion-targeted H2S donor, was shown to enhance H2S in the mitochondrial intracellular compartments and ameliorate spatial memory defects and inhibit brain atrophy in the APP/PS1 mouse model of AD (238). The researchers surmise that H2S protection was achieved through the preservation of mitochondrial function and bioenergetic metabolism.
PD is the second most prevalent neurodegenerative disease behind AD, and it is characterized by debilitating motor dysfunction. It affects the substantia nigra of the brain and is associated with misfolding and aggregation of the protein α-synuclein (192). Persulfidation of the E3-ligase, Parkin, has been shown to be important for the clearance of such misfolded proteins, and that dysregulated H2S signaling has been reported in the disease (239). Indeed, post mortem analysis of patient striata indicated decreased levels of Parkin persulfidation and reduced enzymatic activity. Moreover, H2S supplementation by pharmacological H2S donors confers protection against neurodegeneration in cultured cells and animal models of PD. Hu et al. (170) determined that apoptosis induced by rotenone challenge, which is commonly used to model PD in vitro and in vivo, was ameliorated upon NaHs treatment in the human-derived dopaminergic neuroblastoma cell line (SH-SY5Y). They attributed the protective effects of H2S to maintenance of mitochondrial health via regulation of the ATP-sensitive potassium channel from the inner mitochondrial membrane (mitoKATP)/p38 and Jnk/MAPK pathway. This same group later demonstrated H2S protection against PD in 2 mouse models of PD (240). They determined that H2S impacted neuroinflammation associated with PD by attenuating microglial activation, as well as conferred protection against oxidative stress. Another group treated a mouse model of PD with inhaled H2S for 8 h/d for 7 d (241). Inhalation of H2S protected against the PD-induced movement dysfunction and prevented neuronal apoptosis and microglia activation in the nigrostriatal region. Finally, CBS overexpression has also been reported to protect against the 6 hydroxydopamine–induced model of PD (242).
HD, another neurodegenerative disease that affects cognitive and motor function, is characterized by aggregation of the huntingtin protein, which is induced via polyglutamine repeats on the protein (192). Cysteine metabolism and H2S signaling dysfunction has recently been identified in in vitro and in vivo models of HD, as well as in human HD brains (243). This loss of cysteine and H2S activity in HD models was attributed to decreased CSE expression due to inhibition of its physiologic transcription factor, SP1, by mutated huntingtin. Unsurprisingly, loss of CSE, cysteine, and H2S levels contributed to pathophysiological redox imbalance, elevated ROS levels, and impaired stress response. It is worth noting that CSE can be alternatively transcribed by activating transcription factor 4 in response to cellular stressors like amino acid deprivation and ER stress. Interestingly, this pathway has also been reported to be dysregulated and a contributing factor to neuronal death in HD (244). Surprisingly, no study to date has attempted to treat HD with H2S donor or in models of CSE overexpression. However, H2S may still be an attractive therapy against HD given that cysteine resupplementation in models of HD have shown therapeutic promise (245). Moreover, our group has shown that persulfidation of SP1 by H2S can enhance its transcriptional activity, thereby indicating a potential mechanism by which HD may be treated by H2S (140).
H2S signaling has wider implications in the field of neuronal health beyond the 3 major neurodegenerative diseases described above. Its regulation of mitoKATP channels has been reported to protect against traumatic brain injury in a rat model treated with exogenous H2S (246). A toxic buildup of H2S was found in the cerebrospinal fluid of 37 patients with amyotrophic lateral sclerosis when compared with healthy controls. Further examination in in vitro and in vivo models revealed that these enhanced H2S levels are due to astrocyte and microglia dysfunction and lead to characteristic cell death in motor neurons (54). Additionally, H2S can restore cognitive function in 2 diabetic murine models (streptozotocin-induced diabetes in rats and db/db mice) by ameliorating ER stress and mitochondrion-dependent apoptosis, respectively, in the hippocampus (247, 248). Finally, CSE expression has been reported to be decreased in patients with spinocerebellar ataxia type 3 (SCA3) (249). SCA3 is a disorder driven by a CAG repeat expansion in the ATXN3 gene, which leads to toxic protein aggregation and consequential tissue damage and impairments in coordination, balance, and movement. Overexpression of CSE in a Drosophila model of SCA3 was reported to be protective against protein aggregation–mediated damage by promoting protein persulfidation, reducing oxidative stress, reducing immune response, and attenuating tissue damage (249).
More broadly, H2S may also regulate neuronal health via its impacts on mitochondrial morphogenesis. As discussed above, H2S signaling can alter mitochondrial shape and function through regulation of the mitochondrial morphogenesis GTPases (Drp1, Fis1, MFN2, MFN1, and OPA1). Abnormal mitochondrial morphology has been linked to several neurodegenerative disease states, such as PD, but H2S signaling and therapy in this context have yet to be examined (250, 251).
Hydrogen sulfide and cancer
The most recent disease of interest in the H2S therapeutics field is cancer. The impact of H2S in cancer was first described in 2013 by 2 independent research groups, one [Szabo et al. (46)] working on colorectal cancer and ourselves working on ovarian cancer (45, 46). Szabo et al. (46) were the first to show selective up-regulation of the H2S-producing enzyme CBS in colon cancer tissues when compared with healthy patient-matched mucosa tissues or the surrounding noncancerous peritumor tissue. CBS was also up-regulated in several colon adenocarcinoma-derived cell lines as compared with the nonmalignant colonic epithelial cell line NCM356 (46). Our group showed that CBS was selectively and significantly up-regulated in several ovarian cancer cell lines and primary epithelial tumor tissues when compared with healthy controls. Moreover, within the patient samples, CBS expression was found to be particularly overexpressed in serous carcinoma (the most commonly diagnosed ovarian cancer type) and higher-grade tumors (45). Both studies surprisingly demonstrated CBS localization in the mitochondria and elucidated the positive effects of CBS/H2S on mitochondrial bioenergetics and cancer cell malignancy. Our laboratory further showed that CBS expression conferred resistance against platinum-based chemotherapy in ovarian cancer cells. Silencing CBS via small interfering RNA or small-molecule inhibition attenuated cancer cell proliferation, invasion, and migration in colon and ovarian cancer, as well as resensitized ovarian cancer to chemotherapy. Moreover, CBS activation by SAM increased cellular proliferation and mitochondrial bioenergetics in colon cancer cells (252).
Our group has since published 2 more articles elucidating the role of CBS function on lipid metabolism and mitochondrial morphogenesis in ovarian cancer cells. Our 2015 report found that CBS expression was essential to maintain a process known as lipogenesis, or de novo lipid synthesis (12). Elevated lipid generation is recognized as a hallmark of many cancers because an increased lipid content is required for building new lipid membranes in rapidly dividing cells, as well as for oxidation in the mitochondria for ATP synthesis to meet the high energy demand of cancer. We found that CBS silencing greatly attenuated the expression of several key lipogenic enzymes, like fatty acid synthase (FASN) and acetyl-CoA carboxylase 1 (ACC1), through the repression of their transcription factors sterol regulatory element-binding protein 1 and 2 (SREBP1/2), and inhibited cell migration and invasion as well as tumor growth in vivo. Moreover, SREBP1/2 expression was able to rescue CBS silencing phenotype, implicating CBS regulation of lipid metabolism as essential for ovarian cancer malignancy. Interestingly, H2S control of mitochondrial morphogenesis may also have implications in cancer because mitochondrial dynamics are strikingly deregulated in cancer (253). Our most recently published study discovered the CBS/H2S-dependent maintenance of mitochondrial fusion via promotion of redox-sensitive MFN2 stabilization in ovarian cancer (187). Silencing of CBS induced mitochondrial fragmentation and subsequent bioenergetic dysfunction in ovarian cancer cells. Notably, mitochondrial fusion has been shown to be abundant in chemotherapy-resistant ovarian cancer and supportive of cell survival, which can reconcile our initial observation of chemoresistance with our observation of CBS promotion of mitochondrial fusion (254).
Szabo et al. (147, 255, 256) have published several additional studies of H2S-generating enzymes in colorectal cancer since their initial 2013 report. Phillips et al. (255) notably showed that forced CBS overexpression in an adenoma-like colonic epithelial cell line was sufficient to induce carcinogenesis, during which these “cancer-like” cells develop metabolic and gene signatures that are characteristic of colorectal cancer. CBS overexpression also increased cellular bioenergetics, proliferation, invasion, tumor formation in vivo, and resistance to loss-of-attachment-induced cell death (anoikis). Also, Untereiner et al. (147) reported that H2S-induced persulfidation of LDHA stimulated mitochondrial respiration in HCT116 colon cancer cells. Another more recent report suggested, strikingly, that acquired resistance to the chemotherapeutic agent 5-FU actually induced CBS up-regulation in HCT116 colon cancer cells, which accompanied a glycolysis-to-oxidative-phosphorylation switch and increased cytochrome p450 enzymes CYP1A2 and CYP2A6, which are the major enzymes involved in drug metabolism (256). Finally, as previously discussed, a report by Libiad et al. (161) reported the importance of the SQR/sulfide oxidation pathway in colon cells. In this same study, they showed that high levels of H2S had an antiproliferative impact on colon epithelial cells. Colorectal cancer cell lines and tissues were shown to circumvent this attenuation of proliferation by up-regulating the mitochondrial sulfide oxidation pathway enzymes, like SQR (161). Taken together, targeting H2S biogenesis and/or metabolism may offer a new therapeutic strategy against colorectal cancers.
H2S signaling and biogenesis have been reported in several other cancer types beyond ovarian and colorectral. Szczesny et al. (257) also demonstrated the up-regulation of all 3 H2S-producing enzymes (CBS, CSE, and 3-MST) in lung adenocarcinoma. This increase in H2S production was shown to stimulate mitochondrial DNA repair via persulfidation of exonuclease G, a mitochondrial enzyme that catalyzes the hydrolysis of ester linkages at the 5′ end of a nucleic acid chain and may play a role in apoptosis. Human breast cancer invasion and epithelial-mesenchymal transition has been shown to be inhibited by H2S sequestration of phosphorylated p38 expression, as well as decreased proliferation, G0/G1 phase cell cycle arrest, and increased apoptosis (258). Zhao et al. (259) reported that CSE/H2S signaling can repress androgen receptor transactivation through direct persulfidation at Cys611 and Cys614 and can inhibit cellular proliferation in both androgen-dependent and antiandrogen-resistant prostate cancer cell lines. Furthermore, H2S, as well as CBS, CSE, and 3-MST, was shown to be up-regulated in patient tissues of oral squamous cell carcinoma when compared with surrounding benign oral mucosa, and CBS has been reported as specifically up-regulated in thyroid carcinomas (260, 261). Given that H2S at elevated, yet nontoxic, levels confers cytoprotection in a host of disease states, it is not surprising that cancer cells would exploit H2S signaling, especially to achieve therapy resistance.
Hydrogen sulfide and metabolic disease
Diabetes mellitus (DM) is a chronic condition that involves a dysfunction in production and/or utilization of the hormone insulin. Insulin is secreted by β cells from the pancreatic islets to assist the body in the storage and utilization of dietary sugars and fats. After eating, insulin levels spike and stimulate glucose uptake and metabolism in the liver, adipose tissue, and skeletal muscle, where it is metabolized or stored as glycogen for later use. The one exception is in adipose tissue, where glucose is partially metabolized into and stored as fatty acids. DM can occur when the pancreas secretes little to no insulin, or if the body is incapable of responding to insulin signaling. DM itself is not necessarily life threatening; however, it is associated with several other chronic complications, like cardiac myopathy, atherosclerosis, peripheral neuropathy, chronic kidney disease, and stroke (198). H2S has protective implications in many, if not all, of the previously listed chronic complications (some of which have already been reviewed) primarily through its cytoprotective properties. However, H2S has also been shown to potentially play a role in the regulation of insulin secretion and sensitivity and may therefore find purpose in the treatment of DM itself, rather than simply the management of symptoms. Although, admittedly, the data are contradictory, this section will briefly focus on a few key studies that elucidate the role of H2S signaling on insulin secretion and resistance with a small focus on mitochondrial impacts.
H2S has been shown to be produced in pancreatic β cells primarily through the actions of CBS and CSE, although there have been reported species-specific preferences between the two with little to no evidence of 3-MST activity (262–264). Surprisingly, H2S signaling has been reported to inhibit insulin secretion from β cells in the pancreas in conditions of hyperglycemia but elicit no effect in the presence of low or no glucose (262, 265, 266). CBS overexpression induced the same effect on insulin secretion in a high-glucose environment, and glucose-stimulated release of insulin was inhibited in CSE−/− mice when compared with WT (266, 267). Altogether, it appears that endogenous H2S generated in pancreatic β cells inhibits glucose-dependent release of insulin. The mechanistic explanation for how H2S achieves this inhibitory effect is somewhat complex. Early hypotheses suggested that H2S signaling impacted insulin secretion via regulation of KATP channels, given that they are targets for persulfidation. Indeed, Yang et al. (268) reported that H2S could effectively stimulate KATP in hyperglycemic conditions but was incapable of such stimulation under normoglycemia. Furthermore, another study reported that NaHS treatment could reduce intracellular [K+] and [Ca2+] as well as increase membrane potential in nearly all HIT-T15 insulinoma cells, which all indicated KATP stimulation (266). However, other mechanisms have been reported as well because H2S has also been shown to prevent insulin secretion in models that do not rely on KATP stimulation, like KCl- and tolbutamide-induced insulin secretion, and was even shown to still inhibit insulin secretion in the presence of a KATP opener, suggesting that H2S may act in a post-KATP manner (262). Indeed, another report suggested that H2S may actually inhibit insulin secretion at the level of voltage-dependent Ca2+ channels, which prevents an influx of Ca2+ into the cell (the go signal for release of insulin from intracellular stores) (267). Surprisingly, a recent article by Takahashi et al. (269) demonstrated the role of H2S on proinsulin synthesis and argued that H2S actually stimulates insulin release. Proinsulin synthesis requires 2-methylthiolation (so-called ms2 modification) of an adenosine residue in lysine-transporting tRNA molecule, a process which has been reported to be tightly regulated by CBS/CSE activity.
Conflicting reports exist with regard to the impact of glucose on H2S levels and signaling. For example, Yang et al. (268) showed that incubation of INS-1E cells in high-glucose medium for 24 h reduced H2S production by 46% when compared with cells cultured in normoglycemic conditions, and Zhang et al. (72) demonstrated similar results in INS-1E cells and freshly cultured rat pancreatic islets via CSE inhibition. On the other hand, Kaneko et al. (264) reported that a hyperglycemic environment actually stimulated CSE expression as well as increased H2S production in isolated mouse islets and cultured MIN6 mouse pancreatic β cells. This discrepancy remains unresolved to date. Moreover, contradictory reports also exist with regard to the pro- and antiapoptotic nature of H2S on pancreatic β cells. Yang et al. (265) demonstrated that both exogenous H2S and CSE overexpression decreased cellular viability accompanied by morphologic feature of apoptosis, positive TUNEL staining, and DNA fragmentation. The researchers showed that H2S-induced apoptosis occurred through p38 MAPK stimulation and ER stress. In contrast, Kaneko et al. (264) showed that NaHS treatment attenuated glucose-stimulated apoptosis in mouse β cells. Taken together with their observation that glucose induced H2S/CSE expression, the researchers postulate that H2S/CSE may be up-regulated in hyperglycemic conditions to limit β cell glucotoxicity. This same group reconfirmed their findings in another report 2 years later, demonstrating that NaHS protected isolated mouse pancreatic β cells from apoptosis induced by palmitate, a cytokine mixture of TNF-α, IFN-γ, and IL-1β or hydrogen peroxide (28). In all 3 scenarios, apoptosis was inhibited upon H2S treatment, as evidenced by reduced DNA fragmentation, TUNEL staining, and caspase activity. The researchers hypothesized that in mouse β cells, which constitutively express CBS, H2S inhibits insulin under normal conditions. However, under conditions of stress, CSE expression is induced, and CSE-derived H2S confers cytoprotection. It is worth noting that the discrepancies between studies cannot be attributed to H2S levels because both groups utilized a similar dose for their respective experiments (50–200 and 100 μM, respectively). Unfortunately, because H2S regulation of insulin secretion and β cell apoptosis remains controversial, it is unclear whether H2S signaling contributes to or prevents the development of DM.
H2S thus far has been suggested to play a therapeutic role in several disease states previously described; however, in the case of DM, it may be disease exacerbating. Yet, again, it remains unclear whether H2S treatment is helpful or detrimental. It has been reported that H2S can promote insulin resistance in the liver. NaHS was able to mimic the effects of high insulin/high glucose–induced insulin resistance in hepatocytes and high insulin–induced CSE expression (270). These results indicate that up-regulation of H2S/CSE contributes to high glucose and high insulin–induced liver insulin resistance. However, in contrast, H2S promotion of mitochondrial biogenesis suggests a potential therapeutic benefit to patients with DM. Mitochondrial dysfunction contributes greatly to the pathogenesis of insulin resistance. Indeed, reduced mitochondrial content and impaired oxidative phosphorylation have been reported in diabetic mouse models, and human patients and may contribute to pathologic glucose metabolism (271). As previously described, H2S can elicit its effects for the maintenance and promotion of mitochondria biogenesis. Indeed, CSE expression appears to regulate mitochondrial biogenesis in the liver because CSE−/− mice had ∼33% lower mitochondrial DNA when compared with WT (174). This loss in mitochondrial content was accompanied by a decrease in biogenesis transcription factors, such as Nrf1/2, PGC-1α, and PPRC. Moreover, NaHS increased ATP synthase, PGC-1α, and PPRC in both WT and CSE−/− hepatocytes. Additionally, moderate exercise, which is well known to restore insulin sensitivity, has been reported to up-regulate CBS, CSE, and 3-MST levels in the livers of mice kept on a high-fat diet, thereby indicating yet another way H2S bioavailability may be therapeutically viable in DM (272).
Unfortunately, studies in which in vivo models of insulin resistance are treated with H2S donor remain just as unclear and contradictory as any other H2S diabetes study. Intraperitoneal administration of NaHS to adult Wistar rats induced a fleeting increase in blood glucose levels and decreased insulin concentration, indicating H2S-mediated inhibition of insulin secretion, but did not have an effect on glucose tolerance (273, 274). Moreover, NaHS continuously supplied via osmotic minipump into rats with streptozotocin-induced diabetes did not significantly impact glucose concentration (275). However, in this same study, H2S protected endothelial cells from hyperglycemic-induced ROS formation via preservation of mitochondrial function. Finally, mice that were fed regular chow developed impaired insulin sensitivity upon treatment with a long-term H2S donor, GYY4137, but mice that were on a high-fat diet had increased insulin sensitivity upon GYY4137 treatment, indicating that H2S therapy may potentially only find use in patients with obesity (276).
Despite the multiple studies on the role of H2S in the development of DM that have occurred over the last decade, much of the data are contradictory, and many issues remain unresolved. Nevertheless, H2S-based approaches clearly have promise for the mitigation of DM-induced chronic complications, and H2S study remains a viable candidate in the field of DM treatment.
CONCLUSIONS AND FUTURE DIRECTIONS
This review consolidates recent relevant research on H2S production, metabolism, regulation, and signaling into one article, specifically highlighting the impact of H2S signaling on mitochondria (Fig. 6). Several broad conclusions can be drawn:
Figure 6.
Impacts of H2S signaling on mitochondrial function and disease. H2S can signal through 3 main mechanisms: via metalloprotein interaction and reduction (A), as a potent antioxidant with implications in cellular redox homeostasis (B), and through a post-translational modification of reactive cysteines known as persulfidation (C). Representative mitochondrial targets of metalloprotein interactions are shown in red, redox regulation in blue, and persulfidation in green. This mitochondrion-specific H2S signaling has been reported to be cytoprotective by promoting efficient mitochondrial bioenergetics, resisting apoptosis, and increasing mitochondrial biogenesis. Taken together, H2S cellular protection is implicated in several diseases and may, therefore, be an exciting avenue for developing new targeted therapeutics.
H2S signaling is tightly controlled, yet its apparent enzyme-dependent signaling specificity has not been studied in depth.
Persulfidation mechanisms and signaling by polysulfides require more research; understanding these processes better will advance the H2S field dramatically.
H2S signals in many complex ways, but the mitochondria are an important target of H2S signaling.
At toxic concentrations, H2S impairs mitochondrial respiration and induces cellular apoptosis via the canonical caspase cascade.
Increasing H2S levels enhance mitochondrial metabolism and function, thus conferring cytoprotection.
H2S donors offer exciting therapeutic opportunities in a variety of disease states, including I/R injury, neurodegenerative diseases, and cancer.
Clearly, a more thorough understanding of H2S signaling in mitochondria is critical for developing therapeutics against a whole host of disease states. Physiologic H2S appears to be critical for normal cellular metabolism and redox homeostasis; however, deregulation of H2S-producing or -metabolizing enzymes can throw cell homeostasis off balance and lead to the development of disease, including neurodegenerative and cardiovascular conditions. Moreover, cancer exploits the cytoprotective nature of H2S by up-regulating or enhancing H2S-producing enzyme activity in preclinical models. Therefore, exogenous H2S treatment or specific H2S therapeutic targeting may prove valuable in the clinical setting for the treatment of debilitating and potentially deadly diseases.
Mutations in H2S-producing genes are implicated in cardiovascular and neurovascular diseases, and aberrant H2S expression is implicated in the pathogenesis of cancer. Recent evidence also indicates that H2S plays an important role in mitochondrial biogenesis and morphogenesis. However, regulation of H2S at the molecular, cellular, and organismal levels remains poorly understood. Thus, a critical remaining question is what cues cells rely on to regulate H2S levels in order to trigger either mitochondrial biogenesis or morphogenesis and to meet the cellular metabolic requirement. Furthermore, the solubility of H2S in aqueous systems is relatively high, so it may trigger signaling cascades both locally and at a distance. The role of H2S and H2S-producing enzymes in regulating the cellular microenvironment, and how that relates to both normal and diseased states, is an exciting area for future exploration.
ACKNOWLEDGMENTS
This work is supported by U.S. National Institutes of Health (NIH) National Cancer Institute (NCI) Grants 2R01CA136494, CA213278, and NIH National Heart, Lung, and Blood Institute Grant HL120585. Preparation of this publication was supported, in part, by NCI Cancer Center Support Grant P30CA225520 awarded to The University of Oklahoma Stephenson Cancer Center through the use of services provided by the Office of Cancer Research. The content is solely the responsibility of the authors and does not necessarily represent the official views of the NIH. The authors declare no conflicts of interest.
Glossary
- ΔΨm
mitochondrial membrane potential
- 3-MP
3-mercaptopyruvate
- 3-MST
3-mercaptopyruvate sulfurtransferase
- Aβ
β-amyloid
- AD
Alzheimer disease
- AOAA
aminooxyacetic acid
- APP
amyloid precursor protein
- ATP5A1
ATP synthase F1 subunit α
- BMMSC
bone marrow mesenchymal stem cell
- BSA
bovine serum albumin
- CBS
cystathionine β synthase
- COX
cytochrome c oxidase
- CSE
cystathionine γ lyase
- Cy5
cyanine 5
- DM
diabetes mellitus
- Dnmt3a
DNA methyltransferase 3a
- Drp
dynamin-related protein
- ER
endoplasmic reticulum
- ETC
electron transport chain
- ETHE1
ethylmalonic encephalopathy 1 protein
- Fe2+
ferrous
- Fe3+
ferric
- Fis1
fission 1
- GAPDH
glyceraldehyde 3-phosphate dehydrogenase
- GSH
glutathione
- GSSG
oxidized GSH
- Hb
hemoglobin
- HD
Huntington’s disease
- HEK
human embryonic kidney
- HSNO2
thionitrate
- I/R
ischemia-reperfusion
- IRF-1
IFN regulatory factor 1
- KATP
ATP-sensitive K+
- Keap1
kelch-like ECH-associated protein 1
- LDH
lactate dehydrogenase
- MFN
mitofusin
- mPTP
mitochondrial permeability transition pore
- MS
mass spectrometry
- Nrf2
nuclear factor (erythroid-derived 2)–like 2
- ONO2−
peroxynitrite
- OPA1
optic atrophy 1
- PD
Parkinson’s disease
- PGC-1α
peroxisome proliferator-activated receptor γ 1α
- PLP
pyridoxal 5′-phosphate
- PPRC
peroxisome proliferator–activated receptor-γ coactivator-related protein
- PTP-1b
protein tyrosine phosphatase 1B
- RNS
reactive nitrogen species
- ROS
reactive oxygen species
- SAM
S-adenosylmethionine
- SCA3
spinocerebellar ataxia type 3
- SO42−
sulfate
- SP
specific protein
- SRB
SO42−-reducing bacteria
- SQR
sulfide quinone oxidoreductase
- SUMO
small ubiquitin-like modifier
- TFAM
mitochondrial transcription factor A
- TRP
transient receptor potential
- Trx
thioredoxin
- TXNIP
Trx-interacting protein
- WT
wild type
AUTHOR CONTRIBUTIONS
R. Bhattacharya and P. Mukherjee conceived the idea; B. Murphy, R. Bhattacharya, and P. Mukherjee jointly wrote and edited the manuscript; and B. Murphy illustrated the figures.
REFERENCES
- 1.Giuffrè A., Vicente J. B. (2018) Hydrogen sulfide biochemistry and interplay with other gaseous mediators in mammalian physiology. Oxid. Med. Cell. Longev. 2018, 6290931 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Muyzer G., Stams A. J. (2008) The ecology and biotechnology of sulphate-reducing bacteria. Nat. Rev. Microbiol. 6, 441–454 [DOI] [PubMed] [Google Scholar]
- 3.Vigneron A., Cruaud P., Alsop E., de Rezende J. R., Head I. M., Tsesmetzis N. (2018) Beyond the tip of the iceberg; a new view of the diversity of sulfite- and sulfate-reducing microorganisms. ISME J. 12, 2096–2099 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Roger A. J., Muñoz-Gómez S. A., Kamikawa R. (2017) The origin and diversification of mitochondria. Curr. Biol. 27, R1177–R1192 [DOI] [PubMed] [Google Scholar]
- 5.Araujo-Alvarez J. M., Trujillo-Ferrara J. G. (2002) [“De Morbis Artificum Diatriba.” 1700-2000. ]. Salud Publica Mex. 44, 362–370 [DOI] [PubMed] [Google Scholar]
- 6.Szabo C. (2018) A timeline of hydrogen sulfide (H2S) research: from environmental toxin to biological mediator. Biochem. Pharmacol. 149, 5–19 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Warenycia M. W., Goodwin L. R., Benishin C. G., Reiffenstein R. J., Francom D. M., Taylor J. D., Dieken F. P. (1989) Acute hydrogen sulfide poisoning. Demonstration of selective uptake of sulfide by the brainstem by measurement of brain sulfide levels. Biochem. Pharmacol. 38, 973–981 [DOI] [PubMed] [Google Scholar]
- 8.Goodwin L. R., Francom D., Dieken F. P., Taylor J. D., Warenycia M. W., Reiffenstein R. J., Dowling G. (1989) Determination of sulfide in brain tissue by gas dialysis/ion chromatography: postmortem studies and two case reports. J. Anal. Toxicol. 13, 105–109 [DOI] [PubMed] [Google Scholar]
- 9.Savage J. C., Gould D. H. (1990) Determination of sulfide in brain tissue and rumen fluid by ion-interaction reversed-phase high-performance liquid chromatography. J. Chromatogr. A 526, 540–545 [DOI] [PubMed] [Google Scholar]
- 10.Abe K., Kimura H. (1996) The possible role of hydrogen sulfide as an endogenous neuromodulator. J. Neurosci. 16, 1066–1071 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Zhao W., Wang R. (2002) H(2)S-induced vasorelaxation and underlying cellular and molecular mechanisms. Am. J. Physiol. Heart Circ. Physiol. 283, H474–H480 [DOI] [PubMed] [Google Scholar]
- 12.Chakraborty P. K., Xiong X., Mustafi S. B., Saha S., Dhanasekaran D., Mandal N. A., McMeekin S., Bhattacharya R., Mukherjee P. (2015) Role of cystathionine beta synthase in lipid metabolism in ovarian cancer. Oncotarget 6, 37367–37384 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Kanagy N. L., Szabo C., Papapetropoulos A. (2017) Vascular biology of hydrogen sulfide. Am. J. Physiol. Cell Physiol. 312, C537–C549 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Zhao Z. Z., Wang Z., Li G. H., Wang R., Tan J. M., Cao X., Suo R., Jiang Z. S. (2011) Hydrogen sulfide inhibits macrophage-derived foam cell formation. Exp. Biol. Med. (Maywood) 236, 169–176 [DOI] [PubMed] [Google Scholar]
- 15.Wang C., Du J., Du S., Liu Y., Li D., Zhu X., Ni X. (2018) Endogenous H2S resists mitochondria-mediated apoptosis in the adrenal glands via ATP5A1 S-sulfhydration in male mice. Mol. Cell. Endocrinol. 474, 65–73 [DOI] [PubMed] [Google Scholar]
- 16.Ahmad A., Szabo C. (2016) Both the H2S biosynthesis inhibitor aminooxyacetic acid and the mitochondrially targeted H2S donor AP39 exert protective effects in a mouse model of burn injury. Pharmacol. Res. 113, 348–355 [DOI] [PubMed] [Google Scholar]
- 17.Yamasaki H., Cohen M. F. (2016) Biological consilience of hydrogen sulfide and nitric oxide in plants: gases of primordial earth linking plant, microbial and animal physiologies. Nitric Oxide 55–56, 91–100 [DOI] [PubMed] [Google Scholar]
- 18.Blackstone E., Morrison M., Roth M. B. (2005) H2S induces a suspended animation-like state in mice. Science 308, 518 [DOI] [PubMed] [Google Scholar]
- 19.Shatalin K., Shatalina E., Mironov A., Nudler E. (2011) H2S: a universal defense against antibiotics in bacteria. Science 334, 986–990 [DOI] [PubMed] [Google Scholar]
- 20.Ma L., Yang Y., Hu X. (2015) Roles of H2S in adaptation of alpine plants Lamiophlomis rotata to altitude gradients. Plant Signal. Behav. 10, e1055433 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Yang J., Minkler P., Grove D., Wang R., Willard B., Dweik R., Hine C. (2019) Non-enzymatic hydrogen sulfide production from cysteine in blood is catalyzed by iron and vitamin B6. Commun. Biol. 2, 194. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.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. (2007) Hydrogen sulfide mediates the vasoactivity of garlic. Proc. Natl. Acad. Sci. USA 104, 17977–17982 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.DeLeon E. R., Stoy G. F., Olson K. R. (2012) Passive loss of hydrogen sulfide in biological experiments. Anal. Biochem. 421, 203–207 [DOI] [PubMed] [Google Scholar]
- 24.Powell C. R., Dillon K. M., Matson J. B. (2018) A review of hydrogen sulfide (H2S) donors: chemistry and potential therapeutic applications. Biochem. Pharmacol. 149, 110–123 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Shen X., Carlström M., Borniquel S., Jädert C., Kevil C. G., Lundberg J. O. (2013) Microbial regulation of host hydrogen sulfide bioavailability and metabolism. Free Radic. Biol. Med. 60, 195–200 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Filipovic M. R. (2015) Persulfidation (S-sulfhydration) and H2S. Handb. Exp. Pharmacol. 230, 29–59 [DOI] [PubMed] [Google Scholar]
- 27.Nagahara N., Sawada N. (2006) The mercaptopyruvate pathway in cysteine catabolism: a physiologic role and related disease of the multifunctional 3-mercaptopyruvate sulfurtransferase. Curr. Med. Chem. 13, 1219–1230 [DOI] [PubMed] [Google Scholar]
- 28.Taniguchi S., Kang L., Kimura T., Niki I. (2011) Hydrogen sulphide protects mouse pancreatic β-cells from cell death induced by oxidative stress, but not by endoplasmic reticulum stress. Br. J. Pharmacol. 162, 1171–1178 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Fräsdorf B., Radon C., Leimkühler S. (2014) Characterization and interaction studies of two isoforms of the dual localized 3-mercaptopyruvate sulfurtransferase TUM1 from humans. J. Biol. Chem. 289, 34543–34556 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Wang J., Wang W., Li S., Han Y., Zhang P., Meng G., Xiao Y., Xie L., Wang X., Sha J., Chen Q., Moore P. K., Wang R., Xiang W., Ji Y. (2018) Hydrogen sulfide as a potential target in preventing spermatogenic failure and testicular dysfunction. Antioxid. Redox Signal. 28, 1447–1462 [DOI] [PubMed] [Google Scholar]
- 31.Liang R., Yu W. D., Du J. B., Yang L. J., Shang M., Guo J. Z. (2006) Localization of cystathionine beta synthase in mice ovaries and its expression profile during follicular development. Chin. Med. J. (Engl.) 119, 1877–1883 [PubMed] [Google Scholar]
- 32.Guzmán M. A., Navarro M. A., Carnicer R., Sarría A. J., Acín S., Arnal C., Muniesa P., Surra J. C., Arbonés-Mainar J. M., Maeda N., Osada J. (2006) Cystathionine beta-synthase is essential for female reproductive function. Hum. Mol. Genet. 15, 3168–3176 [DOI] [PubMed] [Google Scholar]
- 33.Miyamoto R., Otsuguro K., Yamaguchi S., Ito S. (2015) Neuronal regulation of expression of hydrogen sulfide-producing enzyme cystathionine β-synthase in rat spinal cord astrocytes. Neurosci. Res. 97, 52–59 [DOI] [PubMed] [Google Scholar]
- 34.Lee M., Schwab C., Yu S., McGeer E., McGeer P. L. (2009) Astrocytes produce the antiinflammatory and neuroprotective agent hydrogen sulfide. Neurobiol. Aging 30, 1523–1534 [DOI] [PubMed] [Google Scholar]
- 35.Mustafa A. K., Gadalla M. M., Snyder S. H. (2009) Signaling by gasotransmitters. Sci. Signal. 2, re2 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Kabil O., Banerjee R. (2014) Enzymology of H2S biogenesis, decay and signaling. Antioxid. Redox Signal. 20, 770–782 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Badiei A., Chambers S. T., Gaddam R. R., Bhatia M. (2016) Cystathionine-γ-lyase gene silencing with siRNA in monocytes/ macrophages attenuates inflammation in cecal ligation and puncture-induced sepsis in the mouse. J. Biosci. 41, 87–95 [DOI] [PubMed] [Google Scholar]
- 38.Lu Y., O’Dowd B. F., Orrego H., Israel Y. (1992) Cloning and nucleotide sequence of human liver cDNA encoding for cystathionine gamma-lyase. Biochem. Biophys. Res. Commun. 189, 749–758 [DOI] [PubMed] [Google Scholar]
- 39.Tripatara P., Patel N. S., Collino M., Gallicchio M., Kieswich J., Castiglia S., Benetti E., Stewart K. N., Brown P. A., Yaqoob M. M., Fantozzi R., Thiemermann C. (2008) Generation of endogenous hydrogen sulfide by cystathionine gamma-lyase limits renal ischemia/reperfusion injury and dysfunction. Lab. Invest. 88, 1038–1048 [DOI] [PubMed] [Google Scholar]
- 40.Dominy J. E., Stipanuk M. H. (2004) New roles for cysteine and transsulfuration enzymes: production of H2S, a neuromodulator and smooth muscle relaxant. Nutr. Rev. 62, 348–353 [DOI] [PubMed] [Google Scholar]
- 41.Kuo M. M., Kim D. H., Jandu S., Bergman Y., Tan S., Wang H., Pandey D. R., Abraham T. P., Shoukas A. A., Berkowitz D. E., Santhanam L. (2016) MPST but not CSE is the primary regulator of hydrogen sulfide production and function in the coronary artery. Am. J. Physiol. Heart Circ. Physiol. 310, H71–H79 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Bearden S. E., Beard R. S., Jr., Pfau J. C. (2010) Extracellular transsulfuration generates hydrogen sulfide from homocysteine and protects endothelium from redox stress. Am. J. Physiol. Heart Circ. Physiol. 299, H1568–H1576 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Vitvitsky V., Yadav P. K., Kurthen A., Banerjee R. (2015) Sulfide oxidation by a noncanonical pathway in red blood cells generates thiosulfate and polysulfides. J. Biol. Chem. 290, 8310–8320 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Donnarumma E., Trivedi R. K., Lefer D. J. (2017) Protective actions of H2S in acute myocardial infarction and heart failure. Compr. Physiol. 7, 583–602 [DOI] [PubMed] [Google Scholar]
- 45.Bhattacharyya S., Saha S., Giri K., Lanza I. R., Nair K. S., Jennings N. B., Rodriguez-Aguayo C., Lopez-Berestein G., Basal E., Weaver A. L., Visscher D. W., Cliby W., Sood A. K., Bhattacharya R., Mukherjee P. (2013) Cystathionine beta-synthase (CBS) contributes to advanced ovarian cancer progression and drug resistance. PLoS One 8, e79167 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Szabo C., Coletta C., Chao C., Módis K., Szczesny B., Papapetropoulos A., Hellmich M. R. (2013) Tumor-derived hydrogen sulfide, produced by cystathionine-β-synthase, stimulates bioenergetics, cell proliferation, and angiogenesis in colon cancer. Proc. Natl. Acad. Sci. USA 110, 12474–12479 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Fu M., Zhang W., Wu L., Yang G., Li H., Wang R. (2012) Hydrogen sulfide (H2S) metabolism in mitochondria and its regulatory role in energy production. Proc. Natl. Acad. Sci. USA 109, 2943–2948 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Kabil O., Zhou Y., Banerjee R. (2006) Human cystathionine beta-synthase is a target for sumoylation. Biochemistry 45, 13528–13536 [DOI] [PubMed] [Google Scholar]
- 49.Agrawal N., Banerjee R. (2008) Human polycomb 2 protein is a SUMO E3 ligase and alleviates substrate-induced inhibition of cystathionine beta-synthase sumoylation. PLoS One 3, e4032 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Filipovic M. R., Zivanovic J., Alvarez B., Banerjee R. (2018) Chemical biology of H2S signaling through persulfidation. Chem. Rev. 118, 1253–1337 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Chen X., Jhee K. H., Kruger W. D. (2004) Production of the neuromodulator H2S by cystathionine beta-synthase via the condensation of cysteine and homocysteine. J. Biol. Chem. 279, 52082–52086 [DOI] [PubMed] [Google Scholar]
- 52.Singh S., Padovani D., Leslie R. A., Chiku T., Banerjee R. (2009) Relative contributions of cystathionine beta-synthase and gamma-cystathionase to H2S biogenesis via alternative trans-sulfuration reactions. J. Biol. Chem. 284, 22457–22466 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Go Y. M., Chandler J. D., Jones D. P. (2015) The cysteine proteome. Free Radic. Biol. Med. 84, 227–245 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Davoli A., Greco V., Spalloni A., Guatteo E., Neri C., Rizzo G. R., Cordella A., Romigi A., Cortese C., Bernardini S., Sarchielli P., Cardaioli G., Calabresi P., Mercuri N. B., Urbani A., Longone P. (2015) Evidence of hydrogen sulfide involvement in amyotrophic lateral sclerosis. Ann. Neurol. 77, 697–709 [DOI] [PubMed] [Google Scholar]
- 55.Sbodio J. I., Snyder S. H., Paul B. D. (2019) Regulators of the transsulfuration pathway. Br. J. Pharmacol. 176, 583–593 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Rose P., Moore P. K., Zhu Y. Z. (2017) H2S biosynthesis and catabolism: new insights from molecular studies. Cell. Mol. Life Sci. 74, 1391–1412 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Meier M., Janosik M., Kery V., Kraus J. P., Burkhard P. (2001) Structure of human cystathionine beta-synthase: a unique pyridoxal 5′-phosphate-dependent heme protein. EMBO J. 20, 3910–3916 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Banerjee R., Zou C. G. (2005) Redox regulation and reaction mechanism of human cystathionine-beta-synthase: a PLP-dependent hemesensor protein. Arch. Biochem. Biophys. 433, 144–156 [DOI] [PubMed] [Google Scholar]
- 59.Vicente J. B., Colaço H. G., Malagrinò F., Santo P. E., Gutierres A., Bandeiras T. M., Leandro P., Brito J. A., Giuffrè A. (2017) A clinically relevant variant of the human hydrogen sulfide-synthesizing enzyme cystathionine β-synthase: increased CO reactivity as a novel molecular mechanism of pathogenicity? Oxid. Med. Cell. Longev. 2017, 8940321 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Kabil O., Weeks C. L., Carballal S., Gherasim C., Alvarez B., Spiro T. G., Banerjee R. (2011) Reversible heme-dependent regulation of human cystathionine β-synthase by a flavoprotein oxidoreductase. Biochemistry 50, 8261–8263 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Taoka S., Banerjee R. (2001) Characterization of NO binding to human cystathionine beta-synthase: possible implications of the effects of CO and NO binding to the human enzyme. J. Inorg. Biochem. 87, 245–251 [DOI] [PubMed] [Google Scholar]
- 62.Taoka S., West M., Banerjee R. (1999) Characterization of the heme and pyridoxal phosphate cofactors of human cystathionine beta-synthase reveals nonequivalent active sites. Biochemistry 38, 7406 [DOI] [PubMed] [Google Scholar]
- 63.Niu W., Wang J., Qian J., Wang M., Wu P., Chen F., Yan S. (2018) Allosteric control of human cystathionine β-synthase activity by a redox active disulfide bond. J. Biol. Chem. 293, 2523–2533 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Finkelstein J. D., Kyle W. E., Martin J. L., Pick A. M. (1975) Activation of cystathionine synthase by adenosylmethionine and adenosylethionine. Biochem. Biophys. Res. Commun. 66, 81–87 [DOI] [PubMed] [Google Scholar]
- 65.Majtan T., Pey A. L., Fernández R., Fernández J. A., Martínez-Cruz L. A., Kraus J. P. (2014) Domain organization, catalysis and regulation of eukaryotic cystathionine beta-synthases. PLoS One 9, e105290 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Niu W. N., Yadav P. K., Adamec J., Banerjee R. (2015) S-glutathionylation enhances human cystathionine β-synthase activity under oxidative stress conditions. Antioxid. Redox Signal. 22, 350–361 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.D’Emmanuele di Villa Bianca R., Mitidieri E., Fusco F., Russo A., Pagliara V., Tramontano T., Donnarumma E., Mirone V., Cirino G., Russo G., Sorrentino R. (2016) Urothelium muscarinic activation phosphorylates CBS(Ser227) via cGMP/PKG pathway causing human bladder relaxation through H2S production. Sci. Rep. 6, 31491 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Sun Q., Collins R., Huang S., Holmberg-Schiavone L., Anand G. S., Tan C. H., van-den-Berg S., Deng L. W., Moore P. K., Karlberg T., Sivaraman J. (2009) Structural basis for the inhibition mechanism of human cystathionine gamma-lyase, an enzyme responsible for the production of H(2)S. J. Biol. Chem. 284, 3076–3085 [DOI] [PubMed] [Google Scholar]
- 69.Renga B., Bucci M., Cipriani S., Carino A., Monti M. C., Zampella A., Gargiulo A., d’Emmanuele di Villa Bianca R., Distrutti E., Fiorucci S. (2015) Cystathionine γ-lyase, a H2S-generating enzyme, is a GPBAR1-regulated gene and contributes to vasodilation caused by secondary bile acids. Am. J. Physiol. Heart Circ. Physiol. 309, H114–H126 [DOI] [PubMed] [Google Scholar]
- 70.Mustafa A. K., Gadalla M. M., Sen N., Kim S., Mu W., Gazi S. K., Barrow R. K., Yang G., Wang R., Snyder S. H. (2009) H2S signals through protein S-sulfhydration. Sci. Signal. 2, ra72 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Yang G., Pei Y., Teng H., Cao Q., Wang R. (2011) Specificity protein-1 as a critical regulator of human cystathionine gamma-lyase in smooth muscle cells. J. Biol. Chem. 286, 26450–26460 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Zhang L., Yang G., Tang G., Wu L., Wang R. (2011) Rat pancreatic level of cystathionine γ-lyase is regulated by glucose level via specificity protein 1 (SP1) phosphorylation. Diabetologia 54, 2615–2625 [DOI] [PubMed] [Google Scholar]
- 73.Renga B., Mencarelli A., Migliorati M., Distrutti E., Fiorucci S. (2009) Bile-acid-activated farnesoid X receptor regulates hydrogen sulfide production and hepatic microcirculation. World J. Gastroenterol. 15, 2097–2108 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Sen N., Paul B. D., Gadalla M. M., Mustafa A. K., Sen T., Xu R., Kim S., Snyder S. H. (2012) Hydrogen sulfide-linked sulfhydration of NF-κB mediates its antiapoptotic actions. Mol. Cell 45, 13–24 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Martín J. A., Pereda J., Martínez-López I., Escrig R., Miralles V., Pallardó F. V., Viña J. R., Vento M., Viña J., Sastre J. (2007) Oxidative stress as a signal to up-regulate gamma-cystathionase in the fetal-to-neonatal transition in rats. Cell. Mol. Biol. 53 (Suppl), OL1010–OL1017 [PubMed] [Google Scholar]
- 76.Hassan M. I., Boosen M., Schaefer L., Kozlowska J., Eisel F., von Knethen A., Beck M., Hemeida R. A., El-Moselhy M. A., Hamada F. M., Beck K. F., Pfeilschifter J. (2012) Platelet-derived growth factor-BB induces cystathionine γ-lyase expression in rat mesangial cells via a redox-dependent mechanism. Br. J. Pharmacol. 166, 2231–2242 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Hourihan J. M., Kenna J. G., Hayes J. D. (2013) The gasotransmitter hydrogen sulfide induces nrf2-target genes by inactivating the keap1 ubiquitin ligase substrate adaptor through formation of a disulfide bond between cys-226 and cys-613. Antioxid. Redox Signal. 19, 465–481 [DOI] [PubMed] [Google Scholar]
- 78.Harding H. P., Zhang Y., Zeng H., Novoa I., Lu P. D., Calfon M., Sadri N., Yun C., Popko B., Paules R., Stojdl D. F., Bell J. C., Hettmann T., Leiden J. M., Ron D. (2003) An integrated stress response regulates amino acid metabolism and resistance to oxidative stress. Mol. Cell 11, 619–633 [DOI] [PubMed] [Google Scholar]
- 79.Hine C., Kim H. J., Zhu Y., Harputlugil E., Longchamp A., Matos M. S., Ramadoss P., Bauerle K., Brace L., Asara J. M., Ozaki C. K., Cheng S. Y., Singha S., Ahn K. H., Kimmelman A., Fisher F. M., Pissios P., Withers D. J., Selman C., Wang R., Yen K., Longo V. D., Cohen P., Bartke A., Kopchick J. J., Miller R., Hollenberg A. N., Mitchell J. R. (2017) Hypothalamic-pituitary axis regulates hydrogen sulfide production. Cell Metab. 25, 1320–1333.e5 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Nandi S. S., Mishra P. K. (2017) H2S and homocysteine control a novel feedback regulation of cystathionine beta synthase and cystathionine gamma lyase in cardiomyocytes. Sci. Rep. 7, 3639 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Gambari L., Lisignoli G., Gabusi E., Manferdini C., Paolella F., Piacentini A., Grassi F. (2017) Distinctive expression pattern of cystathionine-β-synthase and cystathionine-γ-lyase identifies mesenchymal stromal cells transition to mineralizing osteoblasts. J. Cell. Physiol. 232, 3574–3585 [DOI] [PubMed] [Google Scholar]
- 82.Sen S., Kawahara B., Gupta D., Tsai R., Khachatryan M., Roy-Chowdhuri S., Bose S., Yoon A., Faull K., Farias-Eisner R., Chaudhuri G. (2015) Role of cystathionine β-synthase in human breast Cancer. Free Radic. Biol. Med. 86, 228–238 [DOI] [PubMed] [Google Scholar]
- 83.Nagahara N. (2018) Multiple role of 3-mercaptopyruvate sulfurtransferase: antioxidative function, H2 S and polysulfide production and possible SOx production. Br. J. Pharmacol. 175, 577–589 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Nagahara N., Sreeja V. G., Li Q., Shimizu T., Tsuchiya T., Fujii-Kuriyama Y. (2004) A point mutation in a silencer module reduces the promoter activity for the human mercaptopyruvate sulfurtransferase. Biochim. Biophys. Acta 1680, 176–184 [DOI] [PubMed] [Google Scholar]
- 85.Nagahara N., Yoshii T., Abe Y., Matsumura T. (2007) Thioredoxin-dependent enzymatic activation of mercaptopyruvate sulfurtransferase. An intersubunit disulfide bond serves as a redox switch for activation. J. Biol. Chem. 282, 1561–1569 [DOI] [PubMed] [Google Scholar]
- 86.Nagahara N., Katayama A. (2005) Post-translational regulation of mercaptopyruvate sulfurtransferase via a low redox potential cysteine-sulfenate in the maintenance of redox homeostasis. J. Biol. Chem. 280, 34569–34576 [DOI] [PubMed] [Google Scholar]
- 87.Shibuya N., Tanaka M., Yoshida M., Ogasawara Y., Togawa T., Ishii K., Kimura H. (2009) 3-Mercaptopyruvate sulfurtransferase produces hydrogen sulfide and bound sulfane sulfur in the brain. Antioxid. Redox Signal. 11, 703–714 [DOI] [PubMed] [Google Scholar]
- 88.Yadav P. K., Yamada K., Chiku T., Koutmos M., Banerjee R. (2013) Structure and kinetic analysis of H2S production by human mercaptopyruvate sulfurtransferase. J. Biol. Chem. 288, 20002–20013 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Patterson S. E., Moeller B., Nagasawa H. T., Vince R., Crankshaw D. L., Briggs J., Stutelberg M. W., Vinnakota C. V., Logue B. A. (2016) Development of sulfanegen for mass cyanide casualties. Ann. N. Y. Acad. Sci. 1374, 202–209 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Furne J., Saeed A., Levitt M. D. (2008) Whole tissue hydrogen sulfide concentrations are orders of magnitude lower than presently accepted values. Am. J. Physiol. Regul. Integr. Comp. Physiol. 295, R1479–R1485 [DOI] [PubMed] [Google Scholar]
- 91.Vitvitsky V., Kabil O., Banerjee R. (2012) High turnover rates for hydrogen sulfide allow for rapid regulation of its tissue concentrations. Antioxid. Redox Signal. 17, 22–31 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Curtis C. G., Bartholomew T. C., Rose F. A., Dodgson K. S. (1972) Detoxication of sodium 35 S-sulphide in the rat. Biochem. Pharmacol. 21, 2313–2321 [DOI] [PubMed] [Google Scholar]
- 93.Bartholomew T. C., Powell G. M., Dodgson K. S., Curtis C. G. (1980) Oxidation of sodium sulphide by rat liver, lungs and kidney. Biochem. Pharmacol. 29, 2431–2437 [DOI] [PubMed] [Google Scholar]
- 94.Bostelaar T., Vitvitsky V., Kumutima J., Lewis B. E., Yadav P. K., Brunold T. C., Filipovic M., Lehnert N., Stemmler T. L., Banerjee R. (2016) Hydrogen sulfide oxidation by myoglobin. J. Am. Chem. Soc. 138, 8476–8488 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.Ruetz M., Kumutima J., Lewis B. E., Filipovic M. R., Lehnert N., Stemmler T. L., Banerjee R. (2017) A distal ligand mutes the interaction of hydrogen sulfide with human neuroglobin. J. Biol. Chem. 292, 6512–6528 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Cuevasanta E., Denicola A., Alvarez B., Möller M. N. (2012) Solubility and permeation of hydrogen sulfide in lipid membranes. PLoS One 7, e34562 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Wedmann R., Bertlein S., Macinkovic I., Böltz S., Miljkovic J. Lj., Muñoz L. E., Herrmann M., Filipovic M. R. (2014) Working with “H2S”: facts and apparent artifacts. Nitric Oxide 41, 85–96 [DOI] [PubMed] [Google Scholar]
- 98.Filipovic M. R., Miljkovic J., Allgäuer A., Chaurio R., Shubina T., Herrmann M., Ivanovic-Burmazovic I. (2012) Biochemical insight into physiological effects of H2S: reaction with peroxynitrite and formation of a new nitric oxide donor, sulfinyl nitrite. Biochem. J. 441, 609–621 [DOI] [PubMed] [Google Scholar]
- 99.Xie Z. Z., Liu Y., Bian J. S. (2016) Hydrogen sulfide and cellular redox homeostasis. Oxid. Med. Cell. Longev. 2016, 6043038 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.Lu M., Hu L. F., Hu G., Bian J. S. (2008) Hydrogen sulfide protects astrocytes against H(2)O(2)-induced neural injury via enhancing glutamate uptake. Free Radic. Biol. Med. 45, 1705–1713 [DOI] [PubMed] [Google Scholar]
- 101.Kimura Y., Goto Y., Kimura H. (2010) Hydrogen sulfide increases glutathione production and suppresses oxidative stress in mitochondria. Antioxid. Redox Signal. 12, 1–13 [DOI] [PubMed] [Google Scholar]
- 102.Jain S. K., Huning L., Micinski D. (2014) Hydrogen sulfide upregulates glutamate-cysteine ligase catalytic subunit, glutamate-cysteine ligase modifier subunit, and glutathione and inhibits interleukin-1β secretion in monocytes exposed to high glucose levels. Metab. Syndr. Relat. Disord. 12, 299–302 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.Parsanathan R., Jain S. K. (2018) Hydrogen sulfide increases glutathione biosynthesis, and glucose uptake and utilisation in C2C12 mouse myotubes. Free Radic. Res. 52, 288–303 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Matsuzawa A. (2017) Thioredoxin and redox signaling: roles of the thioredoxin system in control of cell fate. Arch. Biochem. Biophys. 617, 101–105 [DOI] [PubMed] [Google Scholar]
- 105.Jha S., Calvert J. W., Duranski M. R., Ramachandran A., Lefer D. J. (2008) Hydrogen sulfide attenuates hepatic ischemia-reperfusion injury: role of antioxidant and antiapoptotic signaling. Am. J. Physiol. Heart Circ. Physiol. 295, H801–H806 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106.Nicholson C. K., Lambert J. P., Molkentin J. D., Sadoshima J., Calvert J. W. (2013) Thioredoxin 1 is essential for sodium sulfide-mediated cardioprotection in the setting of heart failure. Arterioscler. Thromb. Vasc. Biol. 33, 744–751 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Spindel O. N., World C., Berk B. C. (2012) Thioredoxin interacting protein: redox dependent and independent regulatory mechanisms. Antioxid. Redox Signal. 16, 587–596 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108.Tian D., Dong J., Jin S., Teng X., Wu Y. (2017) Endogenous hydrogen sulfide-mediated MAPK inhibition preserves endothelial function through TXNIP signaling. Free Radic. Biol. Med. 110, 291–299 [DOI] [PubMed] [Google Scholar]
- 109.Nagy P., Winterbourn C. C. (2010) Rapid reaction of hydrogen sulfide with the neutrophil oxidant hypochlorous acid to generate polysulfides. Chem. Res. Toxicol. 23, 1541–1543 [DOI] [PubMed] [Google Scholar]
- 110.Kimura H. (2017) Hydrogen sulfide and polysulfide signaling. Antioxid. Redox Signal. 27, 619–621 [DOI] [PubMed] [Google Scholar]
- 111.Kimura H. (2015) Signaling molecules: hydrogen sulfide and polysulfide. Antioxid. Redox Signal. 22, 362–376 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112.Yuan S., Shen X., Kevil C. G. (2017) Beyond a gasotransmitter: hydrogen sulfide and polysulfide in cardiovascular health and immune response. Antioxid. Redox Signal. 27, 634–653 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113.Zhang D., Macinkovic I., Devarie-Baez N. O., Pan J., Park C. M., Carroll K. S., Filipovic M. R., Xian M. (2014) Detection of protein S-sulfhydration by a tag-switch technique. Angew. Chem. Int. Ed. Engl. 53, 575–581 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114.Nagy P. (2015) Mechanistic chemical perspective of hydrogen sulfide signaling. Methods Enzymol. 554, 3–29 [DOI] [PubMed] [Google Scholar]
- 115.Kimura Y., Mikami Y., Osumi K., Tsugane M., Oka J., Kimura H. (2013) Polysulfides are possible H2S-derived signaling molecules in rat brain. FASEB J. 27, 2451–2457 [DOI] [PubMed] [Google Scholar]
- 116.Szabó C., Ischiropoulos H., Radi R. (2007) Peroxynitrite: biochemistry, pathophysiology and development of therapeutics. Nat. Rev. Drug Discov. 6, 662–680 [DOI] [PubMed] [Google Scholar]
- 117.Filipovic M. R., Miljkovic J. Lj., Nauser T., Royzen M., Klos K., Shubina T., Koppenol W. H., Lippard S. J., Ivanović-Burmazović I. (2012) Chemical characterization of the smallest S-nitrosothiol, HSNO; cellular cross-talk of H2S and S-nitrosothiols. J. Am. Chem. Soc. 134, 12016–12027 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118.Eberhardt M., Dux M., Namer B., Miljkovic J., Cordasic N., Will C., Kichko T. I., de la Roche J., Fischer M., Suárez S. A., Bikiel D., Dorsch K., Leffler A., Babes A., Lampert A., Lennerz J. K., Jacobi J., Martí M. A., Doctorovich F., Högestätt E. D., Zygmunt P. M., Ivanovic-Burmazovic I., Messlinger K., Reeh P., Filipovic M. R. (2014) H2S and NO cooperatively regulate vascular tone by activating a neuroendocrine HNO-TRPA1-CGRP signalling pathway. Nat. Commun. 5, 4381 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119.Fukuto J. M., Bianco C. L., Chavez T. A. (2009) Nitroxyl (HNO) signaling. Free Radic. Biol. Med. 47, 1318–1324 [DOI] [PubMed] [Google Scholar]
- 120.Ríos-González B. B., Román-Morales E. M., Pietri R., López-Garriga J. (2014) Hydrogen sulfide activation in hemeproteins: the sulfheme scenario. J. Inorg. Biochem. 133, 78–86 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121.Koenitzer J. R., Isbell T. S., Patel H. D., Benavides G. A., Dickinson D. A., Patel R. P., Darley-Usmar V. M., Lancaster J. R., Jr., Doeller J. E., Kraus D. W. (2007) Hydrogen sulfide mediates vasoactivity in an O2-dependent manner. Am. J. Physiol. Heart Circ. Physiol. 292, H1953–H1960 [DOI] [PubMed] [Google Scholar]
- 122.Nicholls P., Marshall D. C., Cooper C. E., Wilson M. T. (2013) Sulfide inhibition of and metabolism by cytochrome c oxidase. Biochem. Soc. Trans. 41, 1312–1316 [DOI] [PubMed] [Google Scholar]
- 123.Hill B. C., Woon T. C., Nicholls P., Peterson J., Greenwood C., Thomson A. J. (1984) Interactions of sulphide and other ligands with cytochrome c oxidase. An electron-paramagnetic-resonance study. Biochem. J. 224, 591–600 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124.Módis K., Coletta C., Erdélyi K., Papapetropoulos A., Szabo C. (2013) Intramitochondrial hydrogen sulfide production by 3-mercaptopyruvate sulfurtransferase maintains mitochondrial electron flow and supports cellular bioenergetics. FASEB J. 27, 601–611 [DOI] [PubMed] [Google Scholar]
- 125.Pietri R., Román-Morales E., López-Garriga J. (2011) Hydrogen sulfide and hemeproteins: knowledge and mysteries. Antioxid. Redox Signal. 15, 393–404 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126.Collman J. P., Ghosh S., Dey A., Decréau R. A. (2009) Using a functional enzyme model to understand the chemistry behind hydrogen sulfide induced hibernation. Proc. Natl. Acad. Sci. USA 106, 22090–22095 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127.Vitvitsky V., Miljkovic J. L., Bostelaar T., Adhikari B., Yadav P. K., Steiger A. K., Torregrossa R., Pluth M. D., Whiteman M., Banerjee R., Filipovic M. R. (2018) Cytochrome c reduction by H2S potentiates sulfide signaling. ACS Chem. Biol. 13, 2300–2307 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128.Berzofsky J. A., Peisach J., Alben J. O. (1972) Sulfheme proteins. 3. Carboxysulfmyoglobin: the relation between electron withdrawal from iron and ligand binding. J. Biol. Chem. 247, 3774–3782 [PubMed] [Google Scholar]
- 129.Berzofsky J. A., Peisach J., Blumberg W. E. (1971) Sulfheme proteins. II. The reversible oxygenation of ferrous sulfmyoglobin. J. Biol. Chem. 246, 7366–7372 [PubMed] [Google Scholar]
- 130.Carrico R. J., Blumberg W. E., Peisach J. (1978) The reversible binding of oxygen to sulfhemoglobin. J. Biol. Chem. 253, 7212–7215 [PubMed] [Google Scholar]
- 131.Finch C. A. (1948) Methemoglobinemia and sulfhemoglobinemia. N. Engl. J. Med. 239, 470–478 [DOI] [PubMed] [Google Scholar]
- 132.Aravindhan N., Chisholm D. G. (2000) Sulfhemoglobinemia presenting as pulse oximetry desaturation. Anesthesiology 93, 883–884 [DOI] [PubMed] [Google Scholar]
- 133.Potor L., Nagy P., Méhes G., Hendrik Z., Jeney V., Pethő D., Vasas A., Pálinkás Z., Balogh E., Gyetvai Á., Whiteman M., Torregrossa R., Wood M. E., Olvasztó S., Nagy P., Balla G., Balla J. (2018) Hydrogen sulfide abrogates hemoglobin-lipid interaction in atherosclerotic lesion. Oxid. Med. Cell. Longev. 2018, 3812568 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 134.Paul B. D., Snyder S. H. (2015) Protein sulfhydration. Methods Enzymol. 555, 79–90 [DOI] [PubMed] [Google Scholar]
- 135.Pan J., Carroll K. S. (2013) Persulfide reactivity in the detection of protein s-sulfhydration. ACS Chem. Biol. 8, 1110–1116 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 136.Park C. M., Macinkovic I., Filipovic M. R., Xian M. (2015) Use of the “tag-switch” method for the detection of protein S-sulfhydration. Methods Enzymol. 555, 39–56 [DOI] [PubMed] [Google Scholar]
- 137.Wedmann R., Onderka C., Wei S., Szijártó I. A., Miljkovic J. L., Mitrovic A., Lange M., Savitsky S., Yadav P. K., Torregrossa R., Harrer E. G., Harrer T., Ishii I., Gollasch M., Wood M. E., Galardon E., Xian M., Whiteman M., Banerjee R., Filipovic M. R. (2016) Improved tag-switch method reveals that thioredoxin acts as depersulfidase and controls the intracellular levels of protein persulfidation. Chem. Sci. (Camb.) 7, 3414–3426 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 138.Francoleon N. E., Carrington S. J., Fukuto J. M. (2011) The reaction of H(2)S with oxidized thiols: generation of persulfides and implications to H(2)S biology. Arch. Biochem. Biophys. 516, 146–153 [DOI] [PubMed] [Google Scholar]
- 139.Mintel R., Westley J. (1966) The rhodanese reaction. Mechanism of sulfur-sulfur bond cleavage. J. Biol. Chem. 241, 3381–3385 [PubMed] [Google Scholar]
- 140.Saha S., Chakraborty P. K., Xiong X., Dwivedi S. K., Mustafi S. B., Leigh N. R., Ramchandran R., Mukherjee P., Bhattacharya R. (2016) Cystathionine β-synthase regulates endothelial function via protein S-sulfhydration. FASEB J. 30, 441–456 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 141.Yang G., Zhao K., Ju Y., Mani S., Cao Q., Puukila S., Khaper N., Wu L., Wang R. (2013) Hydrogen sulfide protects against cellular senescence via S-sulfhydration of Keap1 and activation of Nrf2. Antioxid. Redox Signal. 18, 1906–1919 [DOI] [PubMed] [Google Scholar]
- 142.Xie L., Gu Y., Wen M., Zhao S., Wang W., Ma Y., Meng G., Han Y., Wang Y., Liu G., Moore P. K., Wang X., Wang H., Zhang Z., Yu Y., Ferro A., Huang Z., Ji Y. (2016) Hydrogen sulfide induces keap1 S-sulfhydration and suppresses diabetes-accelerated atherosclerosis via Nrf2 activation. Diabetes 65, 3171–3184 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 143.Li S., Yang G. (2015) Hydrogen sulfide maintains mitochondrial DNA replication via demethylation of TFAM. Antioxid. Redox Signal. 23, 630–642 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 144.Zhao K., Ju Y., Li S., Altaany Z., Wang R., Yang G. (2014) S-sulfhydration of MEK1 leads to PARP-1 activation and DNA damage repair. EMBO Rep. 15, 792–800 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 145.Krishnan N., Fu C., Pappin D. J., Tonks N. K. (2011) H2S-Induced sulfhydration of the phosphatase PTP1B and its role in the endoplasmic reticulum stress response. Sci. Signal. 4, ra86 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 146.Módis K., Ju Y., Ahmad A., Untereiner A. A., Altaany Z., Wu L., Szabo C., Wang R. (2016) S-Sulfhydration of ATP synthase by hydrogen sulfide stimulates mitochondrial bioenergetics. Pharmacol. Res. 113, 116–124 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 147.Untereiner A. A., Oláh G., Módis K., Hellmich M. R., Szabo C. (2017) H2S-induced S-sulfhydration of lactate dehydrogenase a (LDHA) stimulates cellular bioenergetics in HCT116 colon cancer cells. Biochem. Pharmacol. 136, 86–98 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 148.Liu Y., Yang R., Liu X., Zhou Y., Qu C., Kikuiri T., Wang S., Zandi E., Du J., Ambudkar I. S., Shi S. (2014) Hydrogen sulfide maintains mesenchymal stem cell function and bone homeostasis via regulation of Ca(2+) channel sulfhydration. Cell Stem Cell 15, 66–78 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 149.Naik J. S., Osmond J. M., Walker B. R., Kanagy N. L. (2016) Hydrogen sulfide-induced vasodilation mediated by endothelial TRPV4 channels. Am. J. Physiol. Heart Circ. Physiol. 311, H1437–H1444 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 150.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., Amzel L. M., Berkowitz D. E., Snyder S. H. (2011) Hydrogen sulfide as endothelium-derived hyperpolarizing factor sulfhydrates potassium channels. Circ. Res. 109, 1259–1268 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 151.Yang J., Gupta V., Carroll K. S., Liebler D. C. (2014) Site-specific mapping and quantification of protein S-sulphenylation in cells. Nat. Commun. 5, 4776 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 152.Talipov M. R., Timerghazin Q. K. (2013) Protein control of S-nitrosothiol reactivity: interplay of antagonistic resonance structures. J. Phys. Chem. B 117, 1827–1837 [DOI] [PubMed] [Google Scholar]
- 153.Mueller E. G. (2006) Trafficking in persulfides: delivering sulfur in biosynthetic pathways. Nat. Chem. Biol. 2, 185–194 [DOI] [PubMed] [Google Scholar]
- 154.Ida T., Sawa T., Ihara H., Tsuchiya Y., Watanabe Y., Kumagai Y., Suematsu M., Motohashi H., Fujii S., Matsunaga T., Yamamoto M., Ono K., Devarie-Baez N. O., Xian M., Fukuto J. M., Akaike T. (2014) Reactive cysteine persulfides and S-polythiolation regulate oxidative stress and redox signaling. Proc. Natl. Acad. Sci. USA 111, 7606–7611 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 155.Greiner R., Pálinkás Z., Bäsell K., Becher D., Antelmann H., Nagy P., Dick T. P. (2013) Polysulfides link H2S to protein thiol oxidation. Antioxid. Redox Signal. 19, 1749–1765 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 156.Dóka É., Pader I., Bíró A., Johansson K., Cheng Q., Ballagó K., Prigge J. R., Pastor-Flores D., Dick T. P., Schmidt E. E., Arnér E. S., Nagy P. (2016) A novel persulfide detection method reveals protein persulfide- and polysulfide-reducing functions of thioredoxin and glutathione systems. Sci. Adv. 2, e1500968 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 157.Nicholls P., Kim J. K. (1982) Sulphide as an inhibitor and electron donor for the cytochrome c oxidase system. Can. J. Biochem. 60, 613–623 [DOI] [PubMed] [Google Scholar]
- 158.Völkel S., Grieshaber M. K. (1996) Mitochondrial sulfide oxidation in Arenicola marina. Evidence for alternative electron pathways. Eur. J. Biochem. 235, 231–237 [DOI] [PubMed] [Google Scholar]
- 159.Goubern M., Andriamihaja M., Nübel T., Blachier F., Bouillaud F. (2007) Sulfide, the first inorganic substrate for human cells. FASEB J. 21, 1699–1706 [DOI] [PubMed] [Google Scholar]
- 160.Theissen U., Martin W. (2008) Sulfide: quinone oxidoreductase (SQR) from the lugworm Arenicola marina shows cyanide- and thioredoxin-dependent activity. FEBS J. 275, 1131–1139 [DOI] [PubMed] [Google Scholar]
- 161.Libiad M., Vitvitsky V., Bostelaar T., Bak D. W., Lee H. J., Sakamoto N., Fearon E., Lyssiotis C. A., Weerapana E., Banerjee R. (2019) Hydrogen sulfide perturbs mitochondrial bioenergetics and triggers metabolic reprogramming in colon cells. J. Biol. Chem. 294, 12077–12090 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 162.Szczesny B., Módis K., Yanagi K., Coletta C., Le Trionnaire S., Perry A., Wood M. E., Whiteman M., Szabo C. (2014) 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 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 163.Pun P. B., Lu J., Kan E. M., Moochhala S. (2010) Gases in the mitochondria. Mitochondrion 10, 83–93 [DOI] [PubMed] [Google Scholar]
- 164.Eghbal M. A., Pennefather P. S., O’Brien P. J. (2004) H2S cytotoxicity mechanism involves reactive oxygen species formation and mitochondrial depolarisation. Toxicology 203, 69–76 [DOI] [PubMed] [Google Scholar]
- 165.Leschelle X., Goubern M., Andriamihaja M., Blottière H. M., Couplan E., Gonzalez-Barroso M. D., Petit C., Pagniez A., Chaumontet C., Mignotte B., Bouillaud F., Blachier F. (2005) Adaptative metabolic response of human colonic epithelial cells to the adverse effects of the luminal compound sulfide. Biochim. Biophys. Acta 1725, 201–212 [DOI] [PubMed] [Google Scholar]
- 166.Baskar R., Li L., Moore P. K. (2007) Hydrogen sulfide-induces DNA damage and changes in apoptotic gene expression in human lung fibroblast cells. FASEB J. 21, 247–255 [DOI] [PubMed] [Google Scholar]
- 167.Adhikari S., Bhatia M. (2008) H2S-induced pancreatic acinar cell apoptosis is mediated via JNK and p38 MAP kinase. J. Cell. Mol. Med. 12, 1374–1383 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 168.De Moura Alvorcem L., Britto R., Parmeggiani B., Glanzel N. M., da Rosa-Junior N. T., Cecatto C., Bobermin L. D., Amaral A. U., Wajner M., Leipnitz G. (2019) Evidence that thiol group modification and reactive oxygen species are involved in hydrogen sulfide-induced mitochondrial permeability transition pore opening in rat cerebellum. Mitochondrion 47, 141–150 [DOI] [PubMed] [Google Scholar]
- 169.George L., Ramasamy T., Sirajudeen K., Manickam V. (2019) LPS-induced apoptosis is partially mediated by hydrogen sulphide in RAW 264.7 murine macrophages. Immunol. Invest. 48, 451–465 [DOI] [PubMed] [Google Scholar]
- 170.Hu L. F., Lu M., Wu Z. Y., Wong P. T., Bian J. S. (2009) Hydrogen sulfide inhibits rotenone-induced apoptosis via preservation of mitochondrial function. Mol. Pharmacol. 75, 27–34 [DOI] [PubMed] [Google Scholar]
- 171.Kimura Y., Dargusch R., Schubert D., Kimura H. (2006) Hydrogen sulfide protects HT22 neuronal cells from oxidative stress. Antioxid. Redox Signal. 8, 661–670 [DOI] [PubMed] [Google Scholar]
- 172.Johri A., Chandra A., Flint Beal M. (2013) PGC-1α, mitochondrial dysfunction, and Huntington’s disease. Free Radic. Biol. Med. 62, 37–46 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 173.Untereiner A. A., Wang R., Ju Y., Wu L. (2016) Decreased gluconeogenesis in the absence of cystathionine gamma-lyase and the underlying mechanisms. Antioxid. Redox Signal. 24, 129–140 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 174.Untereiner A. A., Fu M., Módis K., Wang R., Ju Y., Wu L. (2016) Stimulatory effect of CSE-generated H2S on hepatic mitochondrial biogenesis and the underlying mechanisms. Nitric Oxide 58, 67–76 [DOI] [PubMed] [Google Scholar]
- 175.Shimizu Y., Polavarapu R., Eskla K. L., Nicholson C. K., Koczor C. A., Wang R., Lewis W., Shiva S., Lefer D. J., Calvert J. W. (2018) Hydrogen sulfide regulates cardiac mitochondrial biogenesis via the activation of AMPK. J. Mol. Cell. Cardiol. 116, 29–40 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 176.Zhai Y., Behera J., Tyagi S. C., Tyagi N. (2019) Hydrogen sulfide attenuates homocysteine-induced osteoblast dysfunction by inhibiting mitochondrial toxicity. J. Cell. Physiol. 234, 18602–18614 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 177.Kashatus D. F. (2018) The regulation of tumor cell physiology by mitochondrial dynamics. Biochem. Biophys. Res. Commun. 500, 9–16 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 178.Smirnova E., Griparic L., Shurland D. L., van der Bliek A. M. (2001) Dynamin-related protein Drp1 is required for mitochondrial division in mammalian cells. Mol. Biol. Cell 12, 2245–2256 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 179.Santel A., Fuller M. T. (2001) Control of mitochondrial morphology by a human mitofusin. J. Cell Sci. 114, 867–874 [DOI] [PubMed] [Google Scholar]
- 180.Ishihara N., Eura Y., Mihara K. (2004) Mitofusin 1 and 2 play distinct roles in mitochondrial fusion reactions via GTPase activity. J. Cell Sci. 117, 6535–6546 [DOI] [PubMed] [Google Scholar]
- 181.Ishihara N., Fujita Y., Oka T., Mihara K. (2006) Regulation of mitochondrial morphology through proteolytic cleavage of OPA1. EMBO J. 25, 2966–2977 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 182.James D. I., Parone P. A., Mattenberger Y., Martinou J. C. (2003) hFis1, a novel component of the mammalian mitochondrial fission machinery. J. Biol. Chem. 278, 36373–36379 [DOI] [PubMed] [Google Scholar]
- 183.Liu N., Wu J., Zhang L., Gao Z., Sun Y., Yu M., Zhao Y., Dong S., Lu F., Zhang W. (2017) Hydrogen Sulphide modulating mitochondrial morphology to promote mitophagy in endothelial cells under high-glucose and high-palmitate. J. Cell. Mol. Med. 21, 3190–3203 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 184.Qiao P., Zhao F., Liu M., Gao D., Zhang H., Yan Y. (2017) Hydrogen sulfide inhibits mitochondrial fission in neuroblastoma N2a cells through the Drp1/ERK1/2 signaling pathway. Mol. Med. Rep. 16, 971–977 [DOI] [PubMed] [Google Scholar]
- 185.Meng G., Liu J., Liu S., Song Q., Liu L., Xie L., Han Y., Ji Y. (2018) Hydrogen sulfide pretreatment improves mitochondrial function in myocardial hypertrophy via a SIRT3-dependent manner. Br. J. Pharmacol. 175, 1126–1145 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 186.Zhang J., Yu J., Chen Y., Liu L., Xu M., Sun L., Luo H., Wang Y., Meng G. (2018) Exogenous hydrogen sulfide supplement attenuates isoproterenol-induced myocardial hypertrophy in a sirtuin 3-dependent manner. Oxid. Med. Cell. Longev. 2018, 9396089 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 187.Chakraborty P. K., Murphy B., Mustafi S. B., Dey A., Xiong X., Rao G., Naz S., Zhang M., Yang D., Dhanasekaran D. N., Bhattacharya R., Mukherjee P. (2018) Cystathionine β-synthase regulates mitochondrial morphogenesis in ovarian cancer. FASEB J. 32, 4145–4157 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 188.Xu M., Hua Y., Qi Y., Meng G., Yang S. (2019) Exogenous hydrogen sulphide supplement accelerates skin wound healing via oxidative stress inhibition and vascular endothelial growth factor enhancement. Exp. Dermatol. 28, 776–785 [DOI] [PubMed] [Google Scholar]
- 189.Salloum F. N. (2015) Hydrogen sulfide and cardioprotection--mechanistic insights and clinical translatability. Pharmacol. Ther. 152, 11–17 [DOI] [PubMed] [Google Scholar]
- 190.Wu D., Wang J., Li H., Xue M., Ji A., Li Y. (2015) Role of hydrogen sulfide in ischemia-reperfusion injury. Oxid. Med. Cell. Longev. 2015, 186908 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 191.Meng G., Ma Y., Xie L., Ferro A., Ji Y. (2015) Emerging role of hydrogen sulfide in hypertension and related cardiovascular diseases. Br. J. Pharmacol. 172, 5501–5511 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 192.Paul B. D., Snyder S. H. (2018) Gasotransmitter hydrogen sulfide signaling in neuronal health and disease. Biochem. Pharmacol. 149, 101–109 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 193.Kida K., Ichinose F. (2015) Hydrogen sulfide and neuroinflammation. Handb. Exp. Pharmacol. 230, 181–189 [DOI] [PubMed] [Google Scholar]
- 194.Gemici B., Wallace J. L. (2015) Anti-inflammatory and cytoprotective properties of hydrogen sulfide. Methods Enzymol. 555, 169–193 [DOI] [PubMed] [Google Scholar]
- 195.Hellmich M. R., Szabo C. (2015) Hydrogen sulfide and cancer. Handb. Exp. Pharmacol. 230, 233–241 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 196.Hellmich M. R., Coletta C., Chao C., Szabo C. (2015) The therapeutic potential of cystathionine β-synthetase/hydrogen sulfide inhibition in cancer. Antioxid. Redox Signal. 22, 424–448 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 197.Bełtowski J., Jamroz-Wiśniewska A. (2016) Hydrogen sulfide in the adipose tissue-physiology, pathology and a target for pharmacotherapy. Molecules 22, E63 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 198.Bełtowski J., Wójcicka G., Jamroz-Wiśniewska A. (2018) Hydrogen sulfide in the regulation of insulin secretion and insulin sensitivity: implications for the pathogenesis and treatment of diabetes mellitus. Biochem. Pharmacol. 149, 60–76 [DOI] [PubMed] [Google Scholar]
- 199.Kruger W. D. (2017) Cystathionine β-synthase deficiency: of mice and men. Mol. Genet. Metab. 121, 199–205 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 200.Gimbrone M. A., Jr., García-Cardeña G. (2016) Endothelial cell dysfunction and the pathobiology of atherosclerosis. Circ. Res. 118, 620–636 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 201.Konukoglu D., Uzun H. (2017) Endothelial dysfunction and hypertension. Adv. Exp. Med. Biol. 956, 511–540 [DOI] [PubMed] [Google Scholar]
- 202.Gerő D., Torregrossa R., Perry A., Waters A., Le-Trionnaire S., Whatmore J. L., Wood M., Whiteman M. (2016) The novel mitochondria-targeted hydrogen sulfide (H2S) donors AP123 and AP39 protect against hyperglycemic injury in microvascular endothelial cells in vitro. Pharmacol. Res. 113, 186–198 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 203.Zong Y., Huang Y., Chen S., Zhu M., Chen Q., Feng S., Sun Y., Zhang Q., Tang C., Du J., Jin H. (2015) Downregulation of endogenous hydrogen sulfide pathway is involved in mitochondrion-related endothelial cell apoptosis induced by high salt. Oxid. Med. Cell. Longev. 2015, 754670 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 204.Wang Z. J., Wu J., Guo W., Zhu Y. Z. (2017) Atherosclerosis and the hydrogen sulfide signaling pathway - therapeutic approaches to disease prevention. Cell. Physiol. Biochem. 42, 859–875 [DOI] [PubMed] [Google Scholar]
- 205.Jeney V., Komódi E., Nagy E., Zarjou A., Vercellotti G. M., Eaton J. W., Balla G., Balla J. (2009) Supression of hemin-mediated oxidation of low-density lipoprotein and subsequent endothelial reactions by hydrogen sulfide (H(2)S). Free Radic. Biol. Med. 46, 616–623 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 206.Muellner M. K., Schreier S. M., Laggner H., Hermann M., Esterbauer H., Exner M., Gmeiner B. M., Kapiotis S. (2009) Hydrogen sulfide destroys lipid hydroperoxides in oxidized LDL. Biochem. J. 420, 277–281 [DOI] [PubMed] [Google Scholar]
- 207.Madamanchi N. R., Runge M. S. (2007) Mitochondrial dysfunction in atherosclerosis. Circ. Res. 100, 460–473 [DOI] [PubMed] [Google Scholar]
- 208.Stocker R., Keaney J. F., Jr. (2004) Role of oxidative modifications in atherosclerosis. Physiol. Rev. 84, 1381–1478 [DOI] [PubMed] [Google Scholar]
- 209.Wen Y. D., Wang H., Kho S. H., Rinkiko S., Sheng X., Shen H. M., Zhu Y. Z. (2013) Hydrogen sulfide protects HUVECs against hydrogen peroxide induced mitochondrial dysfunction and oxidative stress. PLoS One 8, e53147 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 210.Yu E., Calvert P. A., Mercer J. R., Harrison J., Baker L., Figg N. L., Kumar S., Wang J. C., Hurst L. A., Obaid D. R., Logan A., West N. E., Clarke M. C., Vidal-Puig A., Murphy M. P., Bennett M. R. (2013) Mitochondrial DNA damage can promote atherosclerosis independently of reactive oxygen species through effects on smooth muscle cells and monocytes and correlates with higher-risk plaques in humans. Circulation 128, 702–712 [DOI] [PubMed] [Google Scholar]
- 211.Cheung S. H., Lau J. Y. W. (2018) Hydrogen sulfide mediates athero-protection against oxidative stress via S-sulfhydration. PLoS One 13, e0194176 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 212.Xie Y. H., Zhang N., Li L. F., Zhang Q. Z., Xie L. J., Jiang H., Li L. P., Hao N., Zhang J. X. (2014) Hydrogen sulfide reduces regional myocardial ischemia injury through protection of mitochondrial function. Mol. Med. Rep. 10, 1907–1914 [DOI] [PubMed] [Google Scholar]
- 213.Miao L., Shen X., Whiteman M., Xin H., Shen Y., Xin X., Moore P. K., Zhu Y. Z. (2016) Hydrogen sulfide mitigates myocardial infarction via promotion of mitochondrial biogenesis-dependent M2 polarization of macrophages. Antioxid. Redox Signal. 25, 268–281 [DOI] [PubMed] [Google Scholar]
- 214.Yellon D. M., Hausenloy D. J. (2007) Myocardial reperfusion injury. N. Engl. J. Med. 357, 1121–1135 [DOI] [PubMed] [Google Scholar]
- 215.Halestrap A. P. (2010) A pore way to die: the role of mitochondria in reperfusion injury and cardioprotection. Biochem. Soc. Trans. 38, 841–860 [DOI] [PubMed] [Google Scholar]
- 216.Elrod J. W., Calvert J. W., Morrison J., Doeller J. E., Kraus D. W., Tao L., Jiao X., Scalia R., Kiss L., Szabo C., Kimura H., Chow C. W., Lefer D. J. (2007) Hydrogen sulfide attenuates myocardial ischemia-reperfusion injury by preservation of mitochondrial function. Proc. Natl. Acad. Sci. USA 104, 15560–15565 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 217.Karwi Q. G., Bornbaum J., Boengler K., Torregrossa R., Whiteman M., Wood M. E., Schulz R., Baxter G. F. (2017) 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 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 218.Ikeda K., Marutani E., Hirai S., Wood M. E., Whiteman M., Ichinose F. (2015) Mitochondria-targeted hydrogen sulfide donor AP39 improves neurological outcomes after cardiac arrest in mice. Nitric Oxide 49, 90–96 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 219.Ahmad A., Olah G., Szczesny B., Wood M. E., Whiteman M., Szabo C. (2016) AP39, a mitochondrially targeted hydrogen sulfide donor, exerts protective effects in renal epithelial cells subjected to oxidative stress in vitro and in acute renal injury in vivo. Shock 45, 88–97 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 220.Pan H., Xie X., Chen D., Zhang J., Zhou Y., Yang G. (2014) Protective and biogenesis effects of sodium hydrosulfide on brain mitochondria after cardiac arrest and resuscitation. Eur. J. Pharmacol. 741, 74–82 [DOI] [PubMed] [Google Scholar]
- 221.Roy A., Khan A. H., Islam M. T., Prieto M. C., Majid D. S. (2012) Interdependency of cystathione γ-lyase and cystathione β-synthase in hydrogen sulfide-induced blood pressure regulation in rats. Am. J. Hypertens. 25, 74–81 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 222.Asimakopoulou A., Panopoulos P., Chasapis C. T., Coletta C., Zhou Z., Cirino G., Giannis A., Szabo C., Spyroulias G. A., Papapetropoulos A. (2013) Selectivity of commonly used pharmacological inhibitors for cystathionine β synthase (CBS) and cystathionine γ lyase (CSE). Br. J. Pharmacol. 169, 922–932 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 223.Sun N. L., Xi Y., Yang S. N., Ma Z., Tang C. S. (2007) Plasma hydrogen sulfide and homocysteine levels in hypertensive patients with different blood pressure levels and complications [in Chinese]. Zhonghua Xin Xue Guan Bing Za Zhi 35, 1145–1148 [PubMed] [Google Scholar]
- 224.Ahmad F. U., Sattar M. A., Rathore H. A., Tan Y. C., Akhtar S., Jin O. H., Pei Y. P., Abdullah N. A., Johns E. J. (2014) Hydrogen sulphide and tempol treatments improve the blood pressure and renal excretory responses in spontaneously hypertensive rats. Ren. Fail. 36, 598–605 [DOI] [PubMed] [Google Scholar]
- 225.Sun L., Jin H., Sun L., Chen S., Huang Y., Liu J., Li Z., Zhao M., Sun Y., Tang C., Zhao B., Du J. (2015) Hydrogen sulfide alleviates myocardial collagen remodeling in association with inhibition of TGF-β/Smad signaling pathway in spontaneously hypertensive rats. Mol. Med. 20, 503–515 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 226.Zhao X., Zhang L. K., Zhang C. Y., Zeng X. J., Yan H., Jin H. F., Tang C. S., Du J. B. (2008) Regulatory effect of hydrogen sulfide on vascular collagen content in spontaneously hypertensive rats. Hyperten. Res. 31, 1619–1630 [DOI] [PubMed] [Google Scholar]
- 227.Sikora M., Drapala A., Ufnal M. (2014) Exogenous hydrogen sulfide causes different hemodynamic effects in normotensive and hypertensive rats via neurogenic mechanisms. Pharmacol Rep. 66, 751–758 [DOI] [PubMed] [Google Scholar]
- 228.Huang P., Chen S., Wang Y., Liu J., Yao Q., Huang Y., Li H., Zhu M., Wang S., Li L., Tang C., Tao Y., Yang G., Du J., Jin H. (2015) Down-regulated CBS/H2S pathway is involved in high-salt-induced hypertension in Dahl rats. Nitric Oxide 46, 192–203 [DOI] [PubMed] [Google Scholar]
- 229.Scheltens P., Blennow K., Breteler M. M., de Strooper B., Frisoni G. B., Salloway S., Van der Flier W. M. (2016) Alzheimer’s disease. Lancet 388, 505–517 [DOI] [PubMed] [Google Scholar]
- 230.Sasaguri H., Nilsson P., Hashimoto S., Nagata K., Saito T., De Strooper B., Hardy J., Vassar R., Winblad B., Saido T. C. (2017) APP mouse models for Alzheimer’s disease preclinical studies. EMBO J. 36, 2473–2487 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 231.Hodgson N., Trivedi M., Muratore C., Li S., Deth R. (2013) Soluble oligomers of amyloid-β cause changes in redox state, DNA methylation, and gene transcription by inhibiting EAAT3 mediated cysteine uptake. J. Alzheimers Dis. 36, 197–209 [DOI] [PubMed] [Google Scholar]
- 232.Aoyama K., Suh S. W., Hamby A. M., Liu J., Chan W. Y., Chen Y., Swanson R. A. (2006) Neuronal glutathione deficiency and age-dependent neurodegeneration in the EAAC1 deficient mouse. Nat. Neurosci. 9, 119–126 [DOI] [PubMed] [Google Scholar]
- 233.Liu X. Q., Liu X. Q., Jiang P., Huang H., Yan Y. (2008) Plasma levels of endogenous hydrogen sulfide and homocysteine in patients with Alzheimer’s disease and vascular dementia and the significance thereof [in Chinese]. Zhonghua Yi Xue Za Zhi 88, 2246–2249 [PubMed] [Google Scholar]
- 234.Morrison L. D., Smith D. D., Kish S. J. (1996) Brain S-adenosylmethionine levels are severely decreased in Alzheimer’s disease. J. Neurochem. 67, 1328–1331 [DOI] [PubMed] [Google Scholar]
- 235.Eto K., Asada T., Arima K., Makifuchi T., Kimura H. (2002) Brain hydrogen sulfide is severely decreased in Alzheimer’s disease. Biochem. Biophys. Res. Commun. 293, 1485–1488 [DOI] [PubMed] [Google Scholar]
- 236.Liu Y., Deng Y., Liu H., Yin C., Li X., Gong Q. (2016) Hydrogen sulfide ameliorates learning memory impairment in APP/PS1 transgenic mice: a novel mechanism mediated by the activation of Nrf2. Pharmacol. Biochem. Behav. 150–151, 207–216 [DOI] [PubMed] [Google Scholar]
- 237.He X. L., Yan N., Chen X. S., Qi Y. W., Yan Y., Cai Z. (2016) Hydrogen sulfide down-regulates BACE1 and PS1 via activating PI3K/Akt pathway in the brain of APP/PS1 transgenic mouse. Pharmacol. Rep 68, 975–982 [DOI] [PubMed] [Google Scholar]
- 238.Zhao F. L., Fang F., Qiao P. F., Yan N., Gao D., Yan Y. (2016) 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, 8360738 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 239.Vandiver M. S., Paul B. D., Xu R., Karuppagounder S., Rao F., Snowman A. M., Ko H. S., Lee Y. I., Dawson V. L., Dawson T. M., Sen N., Snyder S. H. (2013) Sulfhydration mediates neuroprotective actions of parkin. Nat. Commun. 4, 1626 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 240.Hu L. F., Lu M., Tiong C. X., Dawe G. S., Hu G., Bian J. S. (2010) Neuroprotective effects of hydrogen sulfide on Parkinson’s disease rat models. Aging Cell 9, 135–146 [DOI] [PubMed] [Google Scholar]
- 241.Kida K., Yamada M., Tokuda K., Marutani E., Kakinohana M., Kaneki M., Ichinose F. (2011) Inhaled hydrogen sulfide prevents neurodegeneration and movement disorder in a mouse model of Parkinson’s disease. Antioxid. Redox Signal. 15, 343–352 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 242.Yin W. L., Yin W. G., Huang B. S., Wu L. X. (2017) Neuroprotective effects of lentivirus-mediated cystathionine-beta-synthase overexpression against 6-OHDA-induced Parkinson’s disease rats. Neurosci. Lett. 657, 45–52 [DOI] [PubMed] [Google Scholar]
- 243.Paul B. D., Sbodio J. I., Xu R., Vandiver M. S., Cha J. Y., Snowman A. M., Snyder S. H. (2014) Cystathionine γ-lyase deficiency mediates neurodegeneration in Huntington’s disease. Nature 509, 96–100 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 244.Sbodio J. I., Snyder S. H., Paul B. D. (2016) Transcriptional control of amino acid homeostasis is disrupted in Huntington’s disease. Proc. Natl. Acad. Sci. USA 113, 8843–8848 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 245.Wright D. J., Gray L. J., Finkelstein D. I., Crouch P. J., Pow D., Pang T. Y., Li S., Smith Z. M., Francis P. S., Renoir T., Hannan A. J. (2016) N-acetylcysteine modulates glutamatergic dysfunction and depressive behavior in Huntington’s disease. Hum. Mol. Genet. 25, 2923–2933 [DOI] [PubMed] [Google Scholar]
- 246.Jiang X., Huang Y., Lin W., Gao D., Fei Z. (2013) Protective effects of hydrogen sulfide in a rat model of traumatic brain injury via activation of mitochondrial adenosine triphosphate-sensitive potassium channels and reduction of oxidative stress. J. Surg. Res. 184, e27–e35 [DOI] [PubMed] [Google Scholar]
- 247.Zou W., Yuan J., Tang Z. J., Wei H. J., Zhu W. W., Zhang P., Gu H. F., Wang C. Y., Tang X. Q. (2017) Hydrogen sulfide ameliorates cognitive dysfunction in streptozotocin-induced diabetic rats: involving suppression in hippocampal endoplasmic reticulum stress. Oncotarget 8, 64203–64216 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 248.Ma S., Zhong D., Ma P., Li G., Hua W., Sun Y., Liu N., Zhang L., Zhang W. (2017) Exogenous hydrogen sulfide ameliorates diabetes-associated cognitive decline by regulating the mitochondria-mediated apoptotic pathway and IL-23/IL-17 expression in db/db mice. Cell. Physiol. Biochem. 41, 1838–1850 [DOI] [PubMed] [Google Scholar]
- 249.Snijder P. M., Baratashvili M., Grzeschik N. A., Leuvenink H. G. D., Kuijpers L., Huitema S., Schaap O., Giepmans B. N. G., Kuipers J., Miljkovic J. L., Mitrovic A., Bos E. M., Szabó C., Kampinga H. H., Dijkers P. F., Bos E. M., Szabó C., Kampinga H. H., Dijkers P. F., Dunnen W. F. A. D., Filipovic M. R., Goor H. V., Sibon O. C. M. (2016) Overexpression of cystathionine γ-lyase suppresses detrimental effects of spinocerebellar ataxia type 3. Mol. Med. 21, 758–768 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 250.Chen H., McCaffery J. M., Chan D. C. (2007) Mitochondrial fusion protects against neurodegeneration in the cerebellum. Cell 130, 548–562 [DOI] [PubMed] [Google Scholar]
- 251.Gao J., Wang L., Liu J., Xie F., Su B., Wang X. (2017) Abnormalities of mitochondrial dynamics in neurodegenerative diseases. Antioxidants 6, E25 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 252.Módis K., Coletta C., Asimakopoulou A., Szczesny B., Chao C., Papapetropoulos A., Hellmich M. R., Szabo C. (2014) Effect of S-adenosyl-L-methionine (SAM), an allosteric activator of cystathionine-β-synthase (CBS) on colorectal cancer cell proliferation and bioenergetics in vitro. Nitric Oxide 41, 146–156 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 253.Senft D., Ronai Z. A. (2016) Regulators of mitochondrial dynamics in cancer. Curr. Opin. Cell Biol. 39, 43–52 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 254.Kong B., Wang Q., Fung E., Xue K., Tsang B. K. (2014) p53 is required for cisplatin-induced processing of the mitochondrial fusion protein L-Opa1 that is mediated by the mitochondrial metallopeptidase Oma1 in gynecologic cancers. J. Biol. Chem. 289, 27134–27145 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 255.Phillips C. M., Zatarain J. R., Nicholls M. E., Porter C., Widen S. G., Thanki K., Johnson P., Jawad M. U., Moyer M. P., Randall J. W., Hellmich J. L., Maskey M., Qiu S., Wood T. G., Druzhyna N., Szczesny B., Módis K., Szabo C., Chao C., Hellmich M. R. (2017) Upregulation of cystathionine-β-synthase in colonic epithelia reprograms metabolism and promotes carcinogenesis. Cancer Res. 77, 5741–5754 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 256.Untereiner A. A., Pavlidou A., Druzhyna N., Papapetropoulos A., Hellmich M. R., Szabo C. (2018) Drug resistance induces the upregulation of H2S-producing enzymes in HCT116 colon cancer cells. Biochem. Pharmacol. 149, 174–185 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 257.Szczesny B., Marcatti M., Zatarain J. R., Druzhyna N., Wiktorowicz J. E., Nagy P., Hellmich M. R., Szabo C. (2016) Inhibition of hydrogen sulfide biosynthesis sensitizes lung adenocarcinoma to chemotherapeutic drugs by inhibiting mitochondrial DNA repair and suppressing cellular bioenergetics. Sci. Rep. 6, 36125 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 258.Lv M., Li Y., Ji M. H., Zhuang M., Tang J. H. (2014) Inhibition of invasion and epithelial-mesenchymal transition of human breast cancer cells by hydrogen sulfide through decreased phospho-p38 expression. Mol. Med. Rep. 10, 341–346 [DOI] [PubMed] [Google Scholar]
- 259.Zhao K., Li S., Wu L., Lai C., Yang G. (2014) Hydrogen sulfide represses androgen receptor transactivation by targeting at the second zinc finger module. J. Biol. Chem. 289, 20824–20835 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 260.Meram A. T., Chen J., Patel S., Kim D. D., Shirley B., Covello P., Coppola D., Wei E. X., Ghali G., Kevil C. G., Shackelford R. E. (2018) Hydrogen sulfide is increased in oral squamous cell carcinoma compared to adjacent benign oral mucosae. Anticancer Res. 38, 3843–3852 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 261.Turbat-Herrera E. A., Kilpatrick M. J., Chen J., Meram A. T., Cotelingam J., Ghali G., Kevil C. G., Coppola D., Shackelford R. E. (2018) Cystathione β-synthase is increased in thyroid malignancies. Anticancer Res. 38, 6085–6090 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 262.Kaneko Y., Kimura Y., Kimura H., Niki I. (2006) L-cysteine inhibits insulin release from the pancreatic beta-cell: possible involvement of metabolic production of hydrogen sulfide, a novel gasotransmitter. Diabetes 55, 1391–1397 [DOI] [PubMed] [Google Scholar]
- 263.Wu L., Yang W., Jia X., Yang G., Duridanova D., Cao K., Wang R. (2009) Pancreatic islet overproduction of H2S and suppressed insulin release in Zucker diabetic rats. Lab. Invest. 89, 59–67 [DOI] [PubMed] [Google Scholar]
- 264.Kaneko Y., Kimura T., Taniguchi S., Souma M., Kojima Y., Kimura Y., Kimura H., Niki I. (2009) Glucose-induced production of hydrogen sulfide may protect the pancreatic beta-cells from apoptotic cell death by high glucose. FEBS Lett. 583, 377–382 [DOI] [PubMed] [Google Scholar]
- 265.Yang G., Yang W., Wu L., Wang R. (2007) H2S, endoplasmic reticulum stress, and apoptosis of insulin-secreting beta cells. J. Biol. Chem. 282, 16567–16576 [DOI] [PubMed] [Google Scholar]
- 266.Ali M. Y., Whiteman M., Low C. M., Moore P. K. (2007) Hydrogen sulphide reduces insulin secretion from HIT-T15 cells by a KATP channel-dependent pathway. J. Endocrinol. 195, 105–112 [DOI] [PubMed] [Google Scholar]
- 267.Tang G., Zhang L., Yang G., Wu L., Wang R. (2013) Hydrogen sulfide-induced inhibition of L-type Ca2+ channels and insulin secretion in mouse pancreatic beta cells. Diabetologia 56, 533–541 [DOI] [PubMed] [Google Scholar]
- 268.Yang W., Yang G., Jia X., Wu L., Wang R. (2005) Activation of KATP channels by H2S in rat insulin-secreting cells and the underlying mechanisms. J. Physiol. 569, 519–531 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 269.Takahashi N., Wei F. Y., Watanabe S., Hirayama M., Ohuchi Y., Fujimura A., Kaitsuka T., Ishii I., Sawa T., Nakayama H., Akaike T., Tomizawa K. (2017) Reactive sulfur species regulate tRNA methylthiolation and contribute to insulin secretion. Nucleic Acids Res. 45, 435–445 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 270.Zhang L., Yang G., Untereiner A., Ju Y., Wu L., Wang R. (2013) Hydrogen sulfide impairs glucose utilization and increases gluconeogenesis in hepatocytes. Endocrinology 154, 114–126 [DOI] [PubMed] [Google Scholar]
- 271.Gonzalez-Franquesa A., Patti M. E. (2017) Insulin Resistance and Mitochondrial Dysfunction. Adv. Exp. Med. Biol. 982, 465–520 [DOI] [PubMed] [Google Scholar]
- 272.Wang B., Zeng J., Gu Q. (2017) Exercise restores bioavailability of hydrogen sulfide and promotes autophagy influx in livers of mice fed with high-fat diet. Can. J. Physiol. Pharmacol. 95, 667–674 [DOI] [PubMed] [Google Scholar]
- 273.Patel M., Shah G. (2010) Possible role of hydrogen sulfide in insulin secretion and in development of insulin resistance. J. Young Pharm. 2, 148–151 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 274.Vellecco V., Mancini A., Ianaro A., Calderone V., Attanasio C., Cantalupo A., Andria B., Savoia G., Panza E., Di Martino A., Cirino G., Bucci M. (2016) Cystathionine β-synthase-derived hydrogen sulfide is involved in human malignant hyperthermia. Clin. Sci. (Lond.) 130, 35–44 [DOI] [PubMed] [Google Scholar]
- 275.Suzuki K., Olah G., Modis K., Coletta C., Kulp G., Gerö D., Szoleczky P., Chang T., Zhou Z., Wu L., Wang R., Papapetropoulos A., Szabo C. (2011) Hydrogen sulfide replacement therapy protects the vascular endothelium in hyperglycemia by preserving mitochondrial function. Proc. Natl. Acad. Sci. USA 108, 13829–13834 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 276.Geng B., Cai B., Liao F., Zheng Y., Zeng Q., Fan X., Gong Y., Yang J., Cui Q. H., Tang C., Xu G. H. (2013) Increase or decrease hydrogen sulfide exert opposite lipolysis, but reduce global insulin resistance in high fatty diet induced obese mice. PLoS One 8, e73892 [DOI] [PMC free article] [PubMed] [Google Scholar]