Abstract
Significance: Hydrogen sulfide (H2S) plays critical roles in redox biology, and its regulatory effects are tightly controlled by its cellular location and concentration. The imbalance of H2S is believed to contribute to some pathological processes.
Recent Advances: Downregulation of H2S requires chemical tools such as inhibitors of H2S-producing enzymes and H2S scavengers. Recent efforts have discovered some promising inhibitors and scavengers. These advances pave the road toward better understanding of the functions of H2S.
Critical Issues: Precise H2S downregulation is challenging. The potency and specificity of current inhibitors are still far from ideal. H2S-producing enzymes are involved in complex sulfur metabolic pathways and ubiquitously present in biological matrices. The inhibition of these enzymes can cause unwanted side effects. H2S scavengers allow targeted H2S clearance, but their options are still limited. In addition, the scavenging process often results in biologically active by-products.
Future Directions: Further development of potent and specific inhibitors for H2S-producing enzymes is needed. Scavengers that can rapidly and selectively remove H2S while generating biocompatible by-products are needed. Potential therapeutic applications of scavengers and inhibitors are worth exploring. Antioxid. Redox Signal. 36, 294–308.
Keywords: hydrogen sulfide, inhibitor, scavenger, cystathionine-β-synthase, cystathionine-γ-lyase, 3-mercaptopyruvate sulfur transferase
Introduction
Hydrogen sulfide (H2S) is the newest member of the gasotransmitter family, which also includes nitric oxide (NO) and carbon monoxide (CO). Studies have shown that H2S regulates various physiological and pathological responses (22, 44, 62, 74). The first notable function of H2S in mammalian systems was discovered by Abe and Kimura (1), which demonstrated H2S as a neuromodulator in the brain. Following this seminal work, many other functions of H2S (such as antioxidation, anti- or proinflammation, and vasodilation) were reported. H2S is a reactive and diffusible molecule, and it can be further converted into other reactive sulfur species in biological systems. The functions of H2S are believed to be the result of its concentration, location, and biochemical reactions (7, 19, 33, 35). Dysregulated H2S exerts disparate impacts in pathological processes, such as cancer, inflammation, and cardiovascular diseases (13, 70). In this regard, the development of H2S regulating methods or tools has become an important research field, and a large number of such tools have been reported. So far, some excellent review articles have been published, which cover the design, mode of action, properties, and applications of H2S-releasing or supplying agents (i.e., prodrugs or donors) (34, 53, 81, 83). On the contrary, reviews on H2S-suppressing agents are relatively rare. Herein, we summarize reports on H2S-downregulating agents, mainly focusing on inhibitors of H2S-producing enzymes and H2S scavengers. Their design principle, activity, applications, and potential limitations are discussed.
Biosynthesis of H2S
Up until now, at least four enzymes (cystathionine-β-synthase [CBS], cystathionine-γ-lyase [CSE], 3-mercaptopyruvate sulfur transferase [3-MST], and cysteinyl-tRNA synthetases [CARS]) are believed to contribute to the production of H2S in mammalian systems (Fig. 1). The expressions of H2S-producing enzymes are tissue specific. For example, CBS is the predominate source of H2S in liver, brain, and nervous tissues (20, 54, 55). CBS is a pyridoxal-5′-phosphate (PLP)-dependent enzyme, which catalyzes the conversion of L-homocysteine or L-cysteine to L-cystathionine, lanthionine, L-serine, and H2S in cytoplasm. CSE is critical for H2S production in vasculature. It catalyzes PLP-dependent reactions converting L-homocysteine, L-cystathionine, and L-cysteine to α-ketobutyrate or pyruvate and releases H2S. It should be noted that CBS and CSE are important enzymes in metabolism, and they are involved in other critical transformations. For example, a primary role of CBS is to convert L-serine and L-homocysteine to form L-cystathionine. Therefore, the significance of CBS/CSE should not just be attributed to H2S formation. CBS and CSE are predominantly located in the cytoplasm. However, under some conditions of stress, they can translate to nuclei or mitochondria (19, 21, 65). 3-MST is expressed in both cytoplasm and mitochondria. In cooperating with cysteine aminotransferase (CAT), 3-MST catalyzes H2S formation by employing L-cysteine as the substrate. In this process, CAT first converts L-cysteine to 3-mercaptopyruvate (3-MP) with α-ketoglutarate as the cosubstrate (Fig. 1c). Then, 3-MP transfers its sulfur to the cysteine residue of 3-MST to form 3-MST persulfide, which finally reacts with thioredoxin or thiols to produce H2S. D-Cysteine can also be catalyzed by 3-MST to release H2S with the help of D-amino acid oxidase (DAO). Again, 3-MP is the key intermediate. However, this is not a physiological production of H2S as D-cysteine needs to be exogenously administered. CARS is the most recently discovered H2S-releasing enzyme (4). The primary products of CARS are cysteine persulfide and polysulfides, which are unstable species and can easily be converted to H2S, especially in the presence of cellular thiols (Fig. 1d). As such, H2S can be considered as a downstream product from CARS reactions, and CARS is at least partially responsible for H2S biosynthesis. The two different isoforms of CARS, CARS1 and CARS2, are found in mitochondria and cytoplasm, respectively.
FIG. 1.
Enzyme-mediated H2S generation. H2S, hydrogen sulfide.
In addition to these enzyme-mediated generations, H2S can be produced in nonenzymatic pathways (Fig. 2). For example, highly reactive sulfane sulfur species such as allyl trisulfide can readily react with glutathione (GSH) in red blood cells to form H2S (6). Less reactive sulfane sulfur species, such as elemental sulfur (S8), are not very reactive toward GSH. However, in human erythrocytes S8 can be effectively converted to H2S by cellular reducing equivalents obtained from glucose oxidation (e.g., glucose glycolysis) (59). These reducing equivalents are found to be nicotinamide adenine dinucleotide phosphate (NADPH)/nicotinamide adenine dinucleotide (NADH), in conjugation with GSH. While the detailed reaction mechanism is still unclear, it was found that combinations of GSH and NAD(P)H were synergistic. It should be noted that allyl trisulfide and elemental sulfur are normally given exogenously (such as from food). Therefore, these pathways are not considered as physiological ways of H2S production. Very recently, it was discovered that cysteine (both D- and L-) could be converted to H2S via coordinated catalysis by vitamin B6, pyridoxal(phosphate), and iron under physiological conditions (80). Initially, cysteine-aldimine is formed between cysteine and the pyridoxal (phosphate) aldehyde. Then, iron derives the formation of cysteine-quinonoid, -SH elimination, and hydrolysis of desulfurated aldimine. Eventually this reaction produces pyruvate, ammonia (NH3), and H2S.
FIG. 2.
Nonenzyme-mediated H2S generation.
H2S Catabolism
The accumulation of H2S could cause severe organ toxicity. In mammalian systems, currently known H2S catabolic pathways include oxidation, methylation, and scavenging by metallo- or sulfur-containing macromolecules. In mitochondria, a series of enzymes including sulfide quinone oxidoreductase, sulfurdioxygenase, and rhodanese catalyze H2S oxidation to form thiosulfate, sulfite, and sulfate sequentially (36). As the end-product, sulfate can be disposed by urinary excretion. In addition to these enzymes, ferric hemoglobin and myoglobin can promote H2S oxidation to form thiosulfate and iron-bound persulfide species (9). H2S methylation mainly occurs in the cytosol. Catalyzed by thiol S-methyltransferase, H2S is first converted to methanethiol and then to nontoxic dimethyl sulfide. Compared with oxidation, this process is much slower (37). Therefore, it is considered a minor H2S catabolism pathway. Finally, H2S can also react with macromolecules such as hemoglobin to form H2S bound species such as sulfhemoglobin (14). This may serve as an internal storage of H2S. It should be noted that these H2S catabolism pathways, especially its oxidation in mitochondrial, do not simply occur as a way for H2S detoxification. These are physiological pathways normally happening in cells. For example, the mitochondrial oxidation is critical in cellular bioenergetics as H2S serves as an electron donor for electron transport chain.
H2S in Pathological Processes
Understanding the steady state of H2S in circulation is still a challenge. This is mainly due to the high reactivity toward molecular oxygen and the high volatility of H2S. In complex biological environments, such as live cells and tissues, the presence of other reactive sulfur species—especially biothiols and persulfides—often results in spuriously high H2S concentrations. In addition, the methods used in H2S measurement can make a difference as they tend to have varied sensitivity and selectivity (46, 76). Because of these constraints, the steady level of endogenous H2S and its ideal therapeutic concentration are controversial. It is now accepted that H2S acts as a double-edged sword in pathological processes. For example, cytoprotective effects of H2S were found in cardiac diseases, asthma, and reperfusion injury. In a myocardial ischemia–reperfusion injury murine model, the generation of H2S was deficient. H2S-releasing compounds such as JK-1 could be used to restore cellular H2S levels and exhibit promising cardioprotective effects (32). On the contrary, overproduction of H2S can induce adverse impacts in cancer, inflammation, and even hypertension. Compared with noncancerous cells, the rate of H2S generation in colon cancer-derived cells was much higher (26). By silencing CBS, the growth of colon cancer and blood flow around tumors was found to be suppressed in nude mice (63). In a burn-induced inflammation model, H2S generation was upregulated in plasma and liver. Different from the control (saline injected) and propargylglycine (PAG, CSE inhibitor)-injected mice, burned mice with injected NaHS solution exhibited significantly aggravated systemic inflammation (82). Furthermore, H2S is known as a cyanide-like highly toxic substance. It inhibits cytochrome C oxidase in electron transfer chain and affects adenosine triphosphate (ATP) synthesis (49). Short-time exposure to high levels of H2S can lead to irreversible neurologic, respiratory, and cardiovascular deficits (56). While H2S poisoning has been a concern for many years, specific H2S antidotes are still lacking. On the contrary, H2S at physiological concentration was found to directly reduce cytochrome C and produce a HS•/S•− radical. This process could stimulate protein persulfidation, and thus plays an important role in H2S signaling (69).
Inhibitors of H2S-Producing Enzymes
CBS inhibitors
Aminooxy (R-ONH2)-based CBS inhibitors
Aminooxyacetic acid (AOAA) is one of the most well-known inhibitors of CBS. The understanding of AOAA's responding mechanism started with the discovery of the reaction between AOAA and vitamin B6. In 1961, McCormick and Snell found that AOAA was able to react with the aldehyde group of vitamin B6 (pyridoxal form) and generate a stable oxime 1 in aqueous solutions (Fig. 3) (37). PLP is the metabolically active form of vitamin B6, which participates in a series of enzymatic reactions in biological systems. PLP is the cofactor of CBS. Through embedding PLP into the active center, the Schiff base 2 is generated. After a series of additions and eliminations, H2S is formed from cysteine or homocysteine. While in the presence of AOAA, oxime 3 is generated irreversibly and therefore, inhibits CBS-catalyzed H2S generation. In sodium phosphate buffer, half-maximal inhibitory concentration (IC50) of AOAA on CBS is 8.52 ± 0.71 μM (5). AOAA has been wildly used in the past years. For example, in the construction of H2S detection platform HSN2-BG, Montoya and Pluth employed AOAA to down regulate the production of endogenous H2S in cytoplasm of Chinese hamster ovary (CHO) cells (40). Zhao and colleagues treated HCT116 cells with AOAA to establish the negative control for examining the detection ability of a cancerous cell-targeted H2S probe (75). Previous research indicated that the expression of CBS in Down syndrome cells (DSCs) was higher than that in normal cells. Szabo and colleagues used AOAA to silence CBS in DSCs. They discovered that by normalizing H2S levels, mitochondrial electron transport and proliferation ability of DSCs were restored (47). Even with these applications, it is worthy to note that AOAA is not a specific inhibitor for CBS. About 40 PLP-dependent enzymes, such as decarboxylase, aminotransferase, and racemase, have been found to be affected by AOAA in either in vitro or in vivo tests (24, 28, 78). Interestingly, Asimakopoulou et al. indicated that even though AOAA was always considered as an inhibitor for CBS, the inhibitory efficiency of AOAA on CSE (IC50 = 1.09 ± 0.12 μM) was tested to be stronger than that of CBS (5). Another drawback of AOAA is the low lipophilicity (water/octanol coefficient: 0.0019). While in aqueous solutions the IC50 of AOAA for CBS is at low micromole level, its efficiency significantly drops in cells due to poor lipophilicity. In HCT116 colon cancer cells, up to 100 μM AOAA was needed to induce noticable inhibition of H2S generation, presumably due to low cellular uptake (43).
FIG. 3.
Inhibitory mechanism of AOAA and the structure of YD0171. AOAA, aminooxyacetic acid.
To improve its lipophilicity, a prodrug of AOAA, YD0171, was developed by Szabo and colleagues (15). Compared with AOAA, YD0171 showed weaker inhibition activity toward CBS (IC50 = 300 μM). However, YD0171's lipophilicity was enhanced to 0.121, 63.8-fold higher than AOAA. After penetrating cell membrane, the ester linkage on YD0171 can be easily hydrolyzed by cellular esterase to form AOAA. As such, the efficiency of YD0171 was improved. CBS inhibition caused H2S downregulation upon treating HCT116 cells with 30 μM YD0171, while 100 μM AOAA was needed for similar effects. In animal models, injecting 1 mg/kg/day of YD0171 into mice for 3 weeks led to the volume suppression of xenograft colon tumor by 90% (similar results were achieved by injecting 9 mg/kg/day of AOAA during the same period). Furthermore, YD0171 was shown to be safer than AOAA. The mortality in mice injected with YD0171 (1 mg/kg/day) was 14% after 21 days, while the mortality of AOAA (9 mg/kg/day)-injected mice was up to 53%. The inhibitory effects of YD0171 were found to be selective for cancer cells. The expression of CBS was not limited to colon tumor. Compared with the inhibitory effects on colon cancer proliferation, the effects of YD0171 on hepatocytes were negligible, which was due to either fast metabolism in hepatocytes or different turnover rates of CBS in tumor cells versus hepatocytes. Like AOAA, YD0171 is a nonspecific inhibitor for PLP-dependent enzymes. Its inhibition on other PLP-dependent enzymes, such as glutamic oxaloacetic transaminase 1 (GOT1), may account for the suppression of cancer growth as well.
In addition to AOAA and YD0171, the simplest compound bearing the aminooxy moiety, hydroxylamine (NH2OH), has been considered as a CBS inhibitor. The original use of hydroxylamine was to separate ketones and aldehydes from biological matrix (12). By undergoing imine formation, hydroxylamine can convert water-soluble ketones and aldehydes to insoluble oximes, which then precipitate out from cell lysates. Based on this reactivity, hydroxylamine was used in deactivating aldehyde-contained PLP-dependent enzymes (5). Although it showed some inhibitory activity on CBS (IC50 = 278.0 ± 22.0 μM), the application of hydroxylamine as a CBS inhibitor was still limited. This was because (i) hydroxylamine showed higher inhibition activity toward CSE (IC50 = 4.83 ± 0.31 μM); (ii) hydroxylamine was unstable in aqueous solution. At room temperature (25°C), hydroxylamine can decompose to form a series of reactive nitrogen species, such as nitrous oxide (N2O), NH3, and tiny amount of NO (73). Almost all these nitrogen species have their own unique biological functions. Therefore, the use of hydroxylamine as a CBS inhibitor can be compromised.
Hydrazine (H2NNH2)-based CBS inhibitors
Some hydrazine derivatives, such as isoniazid, are known as antibiotic drugs for treating tuberculosis. However, high-dose isoniazid can cause a series of vitamin B6 antagonism-related side effects, including pellagra, convulsion, and peripheral neuropathy (51). These findings inspired the study of inhibitory effects of hydrazine derivatives on PLP-dependent enzymes. A hydrazine derivative 2,3,4-trihydroxylbenzylhydrazine (THBH) was found to be a CBS inhibitor (IC50 = 30 μM) (18). In HCT116 cells, obvious inhibition in proliferation was noted when treating with 30–100 μM THBH for 48 h. The inhibition mechanism of THBH was studied by structural modeling. The formation of a PLP-THBH hydrazone derivative 4 (Fig. 4) was found to be favored. In the same way, 3-hydroxylbenzylhydrazine was also used as a CBS inhibitor.
FIG. 4.
Hydrazine-based CBS inhibitors and inhibitory mechanism of THBH. CBS, cystathionine-β-synthase; THBH, 2,3,4-trihydroxylbenzylhydrazine.
Benserazide, a THBH derivative, was found to be another CBS inhibitor (66). Compared with the control (containing CBS, L-cysteine, homocysteine, and PLP), H2S generation in the benserazide-treated group was significantly decreased. Under the same conditions, H2S formation from a donor GYY4137 was not affected by benserazide, indicating that benserazide downregulated H2S concentration by inhibiting CBS, not by scavenging H2S. In biological systems, benserazide could degrade (by hydrolysis or deacylation) to form THBH and thus achieve inhibition. However, benserazide also contains a free amine group, which could conjugate with the PLP aldehyde to form a Schiff base and cause inhibition. Docking studies revealed that the latter was favored. Benserazide is not a potent inhibitor as its IC50 toward CBS (in buffers) is only 30 μM, much weaker than AOAA. However, in cell-based studies its IC50 was shown as 20 μM, which could be attributed to good cellular uptake (18). Under standard conditions (2 mM L-cysteine and homocysteine for CBS; 10 mM L-cysteine for CSE; 10 mM 3-MP for 3-MST), benserazide exerted some selectivity toward CBS. Up to 66% CBS activity was inhibited after treating 100 μM benserazide for 2 h. Under the same conditions, the inhibitory effects on CSE and 3-MST were only 16% and 35%, respectively. It should be noted that the inhibitory effect of benserazide was related to the concentration of substrates. Using CBS as an example, when the concentrations of L-cysteine and homocysteine (substrates of CBS) were increased to 10 mM, the inhibitory effect of benserazide (100 μM) was decreased to 10% ± 2%. Therefore, one should not draw the conclusion that benserazide showed selectivity toward CBS by simply comparing inhibitory effects when concentrations of substrates are different.
In 2016, Berkowitz and colleagues reported a “zipped synthesis” approach for the development of CBS inhibitors (39). The primary function of CBS is to covert homocysteine and serine to form cystathionine, which is a pseudo-C2 symmetric molecule. It is known that cystathionine possesses much higher binding recognition toward CBS than other substrates, including serine, cysteine, and homocysteine (Michaelis constant [Km] ∼83 μM for cystathionine vs. mM values for other substrates) (3). Based on the structural and charge identity of cystathionine, a series of cystathionine analogs with vicinal heteroatomic groups (-NHNH2, -ONH2, -NHOH) replacing the two α-NH2 groups were prepared and tested. Among these analogs, the hydrazine compound 6S was found to be most promising. Ultraviolet/visible spectroscopy demonstrated that 6S was able to engage the PLP cofactor in a stable hydrazone adduct. In both in vitro kinetic assays and brain whole-cell lysates 6S showed good activity in attenuating H2S production (inhibition constant [Ki] ∼50 μM). In an in vivo rat stroke model (transient middle cerebral artery occlusion), 6S showed significant activity in decreasing infarction volume. The administration of 6S (1.6 μmol/kg, intracerebroventricular injection) led to 83% or 66% reduction in infraction volume.
High-throughput screening identified CBS inhibitors
High-throughput screening has been used in finding CBS inhibitors. Barrios and colleagues used a H2S fluorescent probe 7-azido-4-methylcoumarin (AzMC) for this purpose (66). This probe could sense the production of H2S and provided H2S-concentration-dependent fluorescence readouts. It was used to evaluate the activity of CBS (or CSE), as well as the inhibitory efficacy of inhibitors. Using this method, 12 of 1900 compounds were initially found to be potent (with significant inhibition at 150 μM) for CBS. However, some of them were later found to be false positives due to two possible reasons: (i) some compounds reacted with H2S directly and led to low fluorescence responses; (ii) some compounds quenched the fluorophore (coumarin) and weakened fluorescent signals. In addition, the candidates' selectivity for CSE was tested. Eventually, tangeritin and 1,4-naphthoquinone (Fig. 5) were found to be relatively selective and potent for CBS.
FIG. 5.
High-throughput screening identified CBS inhibitors.
In 2013, Wu and coworkers reported a tandem-microwell-based assay for measuring CBS/CSE-mediated H2S production (84). This method was used to screen 21,599 compounds, and several potential CBS inhibitors with IC50 values in low two-digit μM range were identified. For example, NSC111041 and NSC67078 were found to be most potent for CBS (IC50 = 4 and 12 μM, respectively). However, their selectivity was less satisfactory (IC50 for CSE were 2.5 and 30 μM, respectively). Quinaldine blue and MBS08407 were less potent for CBS (IC50 = 20 and 25 μM, respectively) but more selective as they hardly affected CSE. A follow-up study by the same group identified a more potent and selective CBS inhibitor—CH004 (71). The IC50 of CH004 for CBS was ∼1 μM, which was ∼30-fold stronger than that for CSE. As for the mode of action, it was found that CH004 binds reversibly to CBS. In kinetic studies, CH004 showed noncompetitive inhibition toward PLP, and mixed-type inhibition toward Cys and Hcys. Surface plasmon resonance studies showed that CH004 binds to CBS with a KD value of 0.6 μM. CH004 was used to understand the links between liver cancer and CBS. It was found that CH004-mediated CBS inhibition could lead to ferroptotic cancer death in cell models and effective reduction of tumor growth in a liver tumor xenograft mice model.
Another known reaction of CBS is that it can catalyze the transformation of methylcysteine to produce serine and methanethiol (42). The production of methanethiol can be continuously monitored by 7-diethylamino-3-(4-maleimidophenyl)-4-methylcoumarin (CPM), a commercial fluorescent thiol probe. This was used as a high-throughput method to identify CBS inhibitors by Niu et al. (41). A natural product library with 6491 compounds was screened, and 11 compounds with IC50 <20 μM were selected. Among these hits, hypericin was most potent with IC50 of 3 μM. Hypericin also showed >10-fold selectivity for CBS over CSE. Another hit, sikokianin C, showed higher IC50 for CBS (9 μM) but better selectivity (IC50 >400 μM for CSE) than hypericin.
CSE inhibitors
In 1973, Abeles and Walsh reported PAG as an irreversible CSE inhibitor (2). Different from most PLP-dependent enzyme inhibitors, the mechanism of PAG inhibition is controlled by several key amino acid residues of CSE, including Arg62, Lys212, and Tyr114 (61). As shown in Figure 6, the α-amino group of PAG is first deprotonated by Arg62 to create the activated species, which then undergoes transaldimination to form intermediate 5. Lys212 next abstracts a proton from the alkyne to form an activated allene 6. Finally, the hydroxyl group of Tyr114 reacts with allene to produce vinylether 7. The internal aldimine can be regenerated by subsequent transaldimination with Lys212. This process irreversibly modifies Tyr114 in the active center of CSE, and therefore, deactivates the enzyme function. Although PAG is a commonly used CSE inhibitor, it has two limitations: (i) PAG is not a very potent inhibitor with IC50 of 40 μM (5). In living cells, much higher PAG concentrations (1–10 mM) are needed to block H2S generation. (ii) In addition to CSE, PAG acts on other enzymes. For example, Morino and colleagues demonstrated that PAG decreased the activity of alanine aminotransferase, because increased alanine level was found in urine of PAG-treated mice (60). Compared with PAG, β-cyano-L-alanine (BCA) and L-aminoethoxyvinylglycine (AVG) are more potent CSE inhibitors with IC50 of 14 and 1 μM, respectively (5, 50). In cell- and tissue-based studies, the treatment with BCA inhibits H2S generation in a concentration-dependent manner, while data illustrating biological function of AVG at cellular or tissue level are still lacking (64). Both BCA and AVG suffer selectivity issues. For example, BCA is potentially neurotoxic, and AVG inhibits other PLP-dependent enzymes, such as cystathionine-β-lyase (CBL) (16, 57).
FIG. 6.
Inhibitory mechanism of PAG and PAG-derived CSE inhibitors. CSE, cystathionine-γ-lyase; PAG, propargylglycine.
Inspired by the inhibitory mechanism of PAG, a series of compounds, which fused the propargyl group with modified cysteine structures, were prepared and tested by Caliendo and colleagues (17). Among these compounds, oxothiazolidine derivative 8 showed the maximum inhibitory effect on purified CSE without affecting CBS. Compared with PAG, the IC50 of 8 was improved by 100-fold at tissue level. In addition, the inhibitory effect of 8 on CSE is more comprehensive. The reactions catalyzed by CSE are not only limited to converting L-cysteine to pyruvate, ammonia, and H2S, but also include converting L-cysteine to lanthionine. 8 induced the inhibition of both functions, whereas PAG failed to block CSE-promoted lanthionine generation. The inhibition of CSE by 8 involves a competitive mechanism, while PAG is an irreversible inhibitor.
D-Penicillamine was used as a drug for ameliorating rheumatic disease. Taking advantage of its structural similarity to cysteine (a CSE substrate), Cirino and colleagues applied D-penicillamine as a CSE inhibiter in 2016 (10). After treating CSE with D-penicillamine, reduced H2S generation was observed in cell-free assay. D-Penicillamine was found to be relatively selective toward CSE. The activity of D-penicillamine toward CSE (IC50 = 0.27 mM) was ∼31-fold stronger than that for CBS (IC50 = 8.5 mM). PLP was believed to be the reacting site of D-penicillamine as the addition of extra PLP reversed D-penicillamine-induced inhibition. The activity of D-penicillamine was also confirmed by in vitro experiments. For example, the treatment of D-penicillamine decreased H2S generation and exacerbated vascular inflammation in a mouse model.
High-throughput screening was also used in the discovery of CSE inhibitors. Wardrop and colleagues screened >100,000 compounds against recombinant human CSE and identified Schiff base 9 (Fig. 7) as a hit (8). They further prepared 32 analogs and studied the structure–activity relationships (SARs). It was found that the 2-pyridyl group (R1) and the hydrogen atom (R2) on 9 were essential for maintaining high inhibitory efficacy on CSE, while the thioester substituent could be modified. This work led to the discovery of compound 10 as the most potent and selective CSE inhibitor. Its IC50 for CSE was 1.2 μM, ∼400-fold lower than that for CBS. Nevertheless, the application of 10 in cell- or animal-based studies is still lacking.
FIG. 7.
High-throughput screening identified CSE inhibitors.
Wu and coworkers developed a tandem-microwell-based assay that can constantly measure enzyme-catalyzed H2S generation (84). This method was first used in the identification of CBS inhibitors. Very recently, it was also used in CSE inhibitor discovery (29). Eleven thousand nine hundred fifty-four compounds were screened against hCSE, and one potent hit-NSC4056 was identified. In buffers, dose-dependent CSE inhibition was induced by NSC4056, with an IC50 of 0.6 μM. This compound also showed high specificity for CSE. The IC50 for two other PLP-dependent enzymes, CBS and DOPA decarboxylase (DDC), were 137- and 143-fold higher than those for CSE. Counterscreen assay demonstrated that the effect of NSC4056 on H2S reduction was not due to direct absorption. The study of SAR revealed that the carboxyl groups and tripod-like moiety of NSC4056 were essential. In Raw264.7 cells NSC4056 was found to reduce endogenous H2S levels in a dose-dependent manner with an IC50 of 43.2 μM. It also effectively relieved hypotension in hemorrhagic shock rats.
Wang et al. used a virtual screening technique to discover CSE inhibitors (72). A SPECS compound library (∼200,000 compounds) was created and used to study their binding affinity toward CSE. High-throughput rigid docking followed by flexible docking with force-field refinement was used to rank the compounds. I157172 was identified as the top hit with the highest binding affinity. Methylene blue assay showed that I157172 significantly decreased H2S generation in MCF7 breast cancer cells. Interestingly, Western blot analysis also revealed that I157172 inhibited CSE expression in MCF7 cells. Eventually I157172 was found to effectively inhibit the proliferation, migration, and invasion of MCF7 cells. Mechanistic studies suggested that the effects of I157172 were linked to the sirtuin 1 (SIRT1)/acetyl-STAT3 (signal transducer and activator of transcription 3) signaling pathway. In human breast cancer cells and tissues, the expressions of CSE and SIRT1 were negatively associated. I157172-triggered CSE inhibition would promote SIRT1-mediated deacetylation of STAT3 in breast cancer cells and consequently inhibit the growth of breast cancer cells.
3-MST inhibitors
In biological systems, 3-MST not only catalyzes H2S generation but also promotes the conversion of cyanide to thiocyanate. Wing and Baskin tested a series of compounds as potential inhibitors by measuring their effects on the rate of thiocyanate formation (77). Phenylpyruvate showed the highest activity. Under the treatment of 30 mM phenylpyruvate, the enzyme's activity dropped by 99.8% in bovine kidney extract or by 89% with purified 3-MST. 3-MP is known as the substrate of 3-MST. Two 3-MP derivatives, 3-mercaptopropionic acid (3-MPA) and 2-mercaptopropionic acid (2-MPA), were also tested (52). Both were found to be weak inhibitors (while IC50 data were not provided) but with different binding mechanisms. 3-MPA can bind to both free and substrate-complexed enzymes as a noncompetitive inhibitor. 2-MPA can only bind to the latter, therefore being an uncompetitive inhibitor. These studies provided a good starting point for characterizing 3-MST functions. However, their application as 3-MST inhibitors is limited due to their low potency and unclear specificity for 3-MST.
To identify more effective and selective 3-MST inhibitors, Hanaoka et al. screened 174,118 compounds using HSip-1 (a H2S fluorescent probe) assay and identified a lead compound 11 (Fig. 8) (25). 11 inhibited 3-MST-catalyzed H2S generation with an IC50 of 2.7 μM. 11 was also found to be selective for 3-MST as it was almost inactive for CBS, CSE, and rhodanese. When 1 μM 11 was applied to 3-MST-overexpressed COS7 cells, the activity of 3-MST was completely suppressed, demonstrating 11 as a cell-permeable inhibitor. Theoretical calculations were used to understand the interaction between 11 and the enzyme. The target of 11 was believed to be the persulfidated cysteine residue in the active site of 3-MST. A long-range electrostatic interaction between positively charged carbonyl carbon of pyrimidone moiety in 11 and persulfidated cysteine anion in 3-MST was identified as the main factor. 11should be a useful tool in 3-MST research. For example, Panagaki et al. used 11 to reveal biological roles of 3-MST and H2S in endoplasmic reticulum (ER) stress (48). NaHS treatment mitigated aspects of ER-stress, while the inhibition or silencing of 3-MST exacerbated ER-stress-related readouts.
FIG. 8.
3-MST inhibitors. 3-MST, 3-mercaptopyruvate sulfur transferase.
H2S scavengers
As H2S-producing enzymes are ubiquitously present and involved in complex sulfur metabolisms, interfering with the activity of these enzymes may lead to unwanted side effects, and this is a general concern of the inhibitors. An alternative approach is to develop small-molecule H2S scavengers. Ideally the scavengers should specifically and rapidly remove H2S, therefore, only eliminating the functions of H2S from certain biological systems. By doing so, interference with other necessary enzyme activities unrelated to H2S could be avoided. Another advantage of using scavengers is that they can be delivered to specific cellular locations when conjugated with certain targeting moieties. As such, this would allow targeted H2S clearance. So far, reports on H2S scavengers have been very limited and are summarized below.
Hydroxocobalamin
H2S is also known as a highly toxic species due to its environmental and industrial exposure. Its toxic mechanism is believed to inhibit mitochondrial respiration by complexing cytochrome c oxidase copper and/or heme iron site, as well as by inducing hepatocyte reactive oxygen species (ROS) formation. Hydroxocobalamin (HC; e.g., vitamin B12a) has been investigated as a potential antidote for H2S poisoning. As such, HC may be considered as a potential H2S scavenger. In the report by Truong et al., HC was found to be effective at preventing NaSH-induced mice death at all concentrations tested (67). In a cell-free system, HC increased the rate of oxygen consumption induced by NaSH, which was prevented by catalase. Catalytic concentrations of HC (25–150 μM) depleted ∼95% NaSH from the original concentration (500 μM) in 1 h. A more recent work showed that within 10 min of mixing an equimolar concentration of HC and NaHS (100 μM), the sulfide concentration dropped by 95% (68). HC has been suggested as a useful H2S-scavenging tool for studying H2S functions in cells. In a report by Obeso and colleagues, HC was used to study the involvement of endogenous H2S in carotid body (CB) oxygen sensing (23). It was found that a H2S donor GYY4137 elicited catecholamine release from the whole carotid bodies with HC preventing this response. HC also abolished the rise of [Ca2+] evoked by NaHS in enzymatically dispersed CB glomus cells. Salnikov et al. carried out a detailed kinetic study of the reaction between HC and H2S in water (pH 1–10) (58). Their suggested reaction mechanism is shown in Figure 9. H2S or HS− first forms a complex with aquacobalamin (the conjugate acid of hydroxocobalamin). This then promotes inner-sphere electron transfer to oxidize H2S to S−• and reduce the cobalt(III) ion (Co3+) in the HC core to cobalt(II) ion (Co2+). Finally, the addition of another molecule of HS− to the bound S−• produces SSH2−•, which is released as anion radical SSH2−• in acidic medium. This unstable species eventually should degrade to form elemental sulfur. The problem of HC as a H2S scavenger is its poor specificity. In addition to H2S, HC scavenges cyanide, NO, CO, and ROS (11, 31, 67). This nonspecificity may be fine for HC as an antidote for H2S poisoning but should limit its use as a selective H2S scavenger. Nevertheless, NO, CO, or even some ROS are known as signaling molecules. Removing all of these could cause unexpected consequences.
FIG. 9.
The reaction between HC and H2S. HC, hydroxocobalamin.
Sulfonyl azides
Our laboratory explored small-molecule compounds as H2S scavengers (79). We envisioned the following criteria for “ideal” scavengers: (i) the scavengers should possess high reactivity to H2S. The reaction should be completed within minutes, not in hours. (ii) The scavengers should be highly selective to H2S. They should only react with H2S, not with other cellular species. (iii) The scavengers and their H2S reaction products should possess minimum biological activity. We recognized that these criteria are very much the same as the criteria for H2S sensors. H2S sensors have been extensively studied, and hundreds of such sensors have been reported. Therefore, we built up a comprehensive H2S sensor database and analyzed their reactions with H2S. We focused on the time needed for the sensors to complete the reaction with H2S and the specific H2S-reactive functional groups. This data-driven approach allowed us to identify possible chemical entities that could react with H2S rapidly and specifically. We then prepared a number of such compounds, and tested their H2S-scavenging ability and specificity. A Unisense H2S microsensor (Tueager 1, Denmark) was used in these studies. Eventually, a series of sulfonyl azides (Fig. 10) were found to be potent scavengers. These compounds could remove H2S from buffers in a few minutes, and the presence of biothiols (Cys or GSH) did not seem to interfere with their scavenging ability. One scavenger SS19 showed dose-dependent H2S removal in an enzymatic sulfide generation system (using EcCARS). Another scavenger SS17 was tested in cell models to mitigate the effects of H2S. For example, the pretreatment with SS17 for 30 min before H2S exposure markedly attenuated the effects of H2S on cell growth in SNU398 cells. However, it should be noted that the pretreatment with scavengers in cell models is not an ideal way for the evaluation of scavengers. To better mimic the actual scenarios, scavengers should be applied after H2S overproduction is induced. In a mouse model of H2S intoxication, SS20 showed excellent activity as the antidote at 250 μmol/kg via intraperitoneal injection. SS20 was also used by Olson et al. in removing endogenously formed H2S, which served as part of the evidence to show lipoic acid could concentration dependently induce H2S production (45). The reaction between H2S and sulfonyl azides is shown in Figure 10. It is a redox reaction, similar to the reaction between H2S and aryl-azides (27). Sulfonyl azides are converted to sulfonamides, and H2S is converted to S8. In this process, hydrogen polysulfides (H2Sn) may be formed as the intermediates. However, H2Sn should also react rapidly with sulfonyl azides to form S8.
FIG. 10.
Sulfonyl azide-based H2S scavengers.
7-Nitro-1,2,3-benzoxadiazole amines
Ismail et al. reported a series of 7-nitro-1,2,3-benzoxadiazole (NBD) amines as potential H2S scavengers (30). These compounds are based on NBD-type H2S fluorescent sensors. It was found that these compounds could specifically react with H2S via thiolysis to form NBD-SH and the corresponding amines (Fig. 11). The reaction kinetics are dependent on the structures of amines. One compound, NBD-S8, showed the highest reactivity toward H2S (k2 = 116.1 M−1·s−1). It also showed good stability in buffers and high specificity for H2S over biothiols. H2S-scavenging ability of NBD-S8 was further evaluated in 10% fetal bovine serum, cells (HeLa and FHC), and mice, by monitoring H2S concentration changes in these systems with fluorescence measurements. Overall, NBD-S8 showed satisfactory results, demonstrating that it is a promising scavenger.
FIG. 11.
NBD amine-based H2S scavenger. NBD, 7-nitro-1,2,3-benzoxadiazole.
Conclusions
H2S has been classified as an important signaling molecule, and dysregulation of H2S is linked to a number of pathological processes, including cancer, inflammation, and neurodegenerative diseases. Regulation of H2S levels is believed to have therapeutic potential, and this requires chemical compounds that can precisely regulate local H2S concentrations. This is still a challenging topic as the therapeutic window of H2S is very narrow. In the past decade, many inhibitors of H2S-producing enzymes have been reported, and recently H2S scavengers started to attract some attention. Nevertheless, there are still limitations with these chemicals. The general concerns regarding inhibitors are their specificity and possible side effects. H2S-producing enzymes are present ubiquitously in biological matrices and involved in complex sulfur metabolic pathways. The blockage of the metabolic pathways catalyzed by these enzymes thereby may affect the downstream cysteine production rather than H2S per se. Any pharmacological consequences obtained by the inhibitors can not to be solely attributed to the reduction of H2S biosynthesis. This mechanism-based problem seems to be unavoidable even for the most selective inhibitors. Scavengers have the potential to remove H2S in certain specific tissue or cellular locations. However, up to now, the options of specific H2S scavengers have been very limited. In addition, the use of scavengers would generate H2S-derived reaction products, which could cause unexpected side effects. Some precautions should be kept in mind when using scavengers in research. For example, the sulfonyl azide-based scavengers would produce sulfonamides and S8 when reacting with H2S. Control experiments with the corresponding sulfonamide and S8 should be performed to rule out the effects from these by-products. Another problem is that current H2S scavengers are based on stoichiometric reactions with H2S. Therefore, high doses of scavengers are needed if H2S is continuously generated. This problem may be solved if catalytic H2S scavengers are developed, but this has yet to be explored. We expect to see more research on H2S-suppressing methods in the coming years.
Abbreviations Used
- 2-MPA
2-mercaptopropionic acid
- 3-MP
3-mercaptopyruvate
- 3-MPA
3-mercaptopropionic acid
- 3-MST
3-mercaptopyruvate sulfur transferase
- AOAA
aminooxyacetic acid
- Arg
arginine
- AVG
L-aminoethoxyvinylglycine
- BCA
β-cyano-L-alanine
- Ca2+
calcium ion
- CARS
cysteinyl-tRNA synthetases
- CAT
cysteine aminotransferase
- CB
carotid body
- CBS
cystathionine-β-synthase
- CO
carbon monoxide
- CSE
cystathionine-γ-lyase
- DSCs
Down syndrome cells
- ER
endoplasmic reticulum
- GSH
glutathione
- H2S
hydrogen sulfide
- H2Sn
hydrogen polysulfide
- HC
hydroxocobalamin
- IC50
half-maximal inhibitory concentration
- Lys
lysine
- NADPH
nicotinamide adenine dinucleotide phosphate
- NBD
7-nitro-1,2,3-benzoxadiazole
- NH3
ammonia
- NO
nitric oxide
- PAG
propargylglycine
- PLP
pyridoxal-5′-phosphate
- ROS
reactive oxygen species
- S8
elemental sulfur
- SARs
structure–activity relationships
- SIRT1
sirtuin 1
- STAT3
signal transducer and activator of transcription 3
- THBH
2,3,4-trihydroxylbenzylhydrazine
- Tyr
tyrosine
Authors' Contributions
Y.W. and M.X. conceived the presented review. Y.W., X.N., R.C., C.M., Y.L., B.B., G.R., and M.X. contributed to the writing of the article.
Author Disclosure Statement
There are no conflicts to disclose.
Funding Information
This work was supported by the National Institute of Health (R01HL151398) and NSF (CHE2100870).
References
- 1. Abe K and Kimura H. The possible role of hydrogen sulfide as an endogenous neuromodulator. J Neurosci 16: 1066–1071, 1996. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2. Abeles RH and Walsh CT. Acetylenic enzyme inactivators. Inactivation of .gamma.-cystathionase, in vitro and in vivo by propargylglycine. J Am Chem Soc 95: 6124–6125, 1973. [DOI] [PubMed] [Google Scholar]
- 3. Aitken SM and Kirsch JF. Kinetics of the yeast cystathionine β-synthase forward and reverse reactions: continuous assays and the equilibrium constant for the reaction. Biochemistry 42: 571–578, 2003. [DOI] [PubMed] [Google Scholar]
- 4. Akaike T, Ida T, Wei F-Y, Nishida M, Kumagai Y, Alam MM, Ihara H, Sawa T, Matsunaga T, Kasamatsu S, Nishimura A, Morita M, Tomizawa K, Nishimura A, Watanabe S, Inaba K, Shima H, Tanuma N, Jung M, Fujii S, Watanabe Y, Ohmuraya M, Nagy P, Feelisch M, Fukuto JM, and Motohashi H. Cysteinyl-tRNA synthetase governs cysteine polysulfidation and mitochondrial bioenergetics. Nat Commun 8: 1177, 2017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5. Asimakopoulou A, Panopoulos P, Chasapis CT, Coletta C, Zhou Z, Cirino G, Giannis A, Szabo C, Spyroulias GA, and Papapetropoulos A. Selectivity of commonly used pharmacological inhibitors for cystathionine β synthase (CBS) and cystathionine γ lyase (CSE). Br J Pharmacol 169: 922–932, 2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6. Benavides GA, Squadrito GL, Mills RW, Patel HD, Isbell TS, Patel RP, Darley-Usmar VM, Doeller JE, and Kraus DW. Hydrogen sulfide mediates the vasoactivity of garlic. Proc Natl Acad Sci U S A 104: 17977–17982, 2007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7. Benchoam D, Cuevasanta E, Möller MN, and Alvarez B. Hydrogen sulfide and persulfides oxidation by biologically relevant oxidizing species. Antioxidants (Basel) 8: 48, 2019. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8. Bhattacharjee A, Sinha A, Ratia K, Yin L, Delgado-Rivera L, Petukhov PA, Thatcher GRJ, and Wardrop DJ. 2-Arylidene hydrazinecarbodithioates as potent, selective inhibitors of cystathionine γ-lyase (CSE). ACS Med Chem Lett 8: 1241–1245, 2017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9. Bostelaar T, Vitvitsky V, Kumutima J, Lewis BE, Yadav PK, Brunold TC, Filipovic M, Lehnert N, Stemmler TL, and Banerjee R. Hydrogen sulfide oxidation by myoglobin. J Am Chem Soc 138: 8476–8488, 2016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10. Brancaleone V, Esposito I, Gargiulo A, Vellecco V, Asimakopoulou A, Citi V, Calderone V, Gobbetti T, Perretti M, Papapetropoulos A, Bucci M, and Cirino G. D-Penicillamine modulates hydrogen sulfide (H2S) pathway through selective inhibition of cystathionine-γ-lyase. Br J Pharmacol 173: 1556–1565, 2016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11. Brenner M, Benavides S, Mahon SB, Lee J, Yoon D, Mukai D, Viseroi M, Chan A, Jiang J, Narula N, Azer SM, Alexander C, and Boss GR. The vitamin B12 analog cobinamide is an effective hydrogen sulfide antidote in a lethal rabbit model. Clin Toxicol (Phila) 52: 490–497, 2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12. Bryant WMD and Smith DM. Improved hydroxylamine method for the determination of aldehydes and ketones. Displacement of oxime equilibria by means of pyridine. J Am Chem Soc 57: 57–61, 1935. [Google Scholar]
- 13. Cao X, Ding L, Xie Z-Z, Yang Y, Whiteman M, Moore PK, and Bian J-S. A review of hydrogen sulfide synthesis, metabolism, and measurement: is modulation of hydrogen sulfide a novel therapeutic for cancer? Antioxid Redox Signal 31: 1–38, 2019. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14. Cerda-Colón JF, Silfa E, and López-Garriga J. Unusual rocking freedom of the heme in the hydrogen sulfide-binding hemoglobin from lucina pectinata. J Am Chem Soc 120: 9312–9317, 1998. [DOI] [PubMed] [Google Scholar]
- 15. Chao C, Zatarain JR, Ding Y, Coletta C, Mrazek AA, Druzhyna N, Johnson P, Chen H, Hellmich JL, Asimakopoulou A, Yanagi K, Olah G, Szoleczky P, Törö G, Bohanon FJ, Cheema M, Lewis R, Eckelbarger D, Ahmad A, Módis K, Untereiner A, Szczesny B, Papapetropoulos A, Zhou J, Hellmich MR, and Szabo C. Cystathionine-beta-synthase inhibition for colon cancer: enhancement of the efficacy of aminooxyacetic acid via the prodrug approach. Mol Med 22: 361–379, 2016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16. Clausen T, Huber R, Messerschmidt A, Pohlenz H-D, and Laber B. Slow-binding inhibition of Escherichia coli cystathionine β-lyase by l-aminoethoxyvinylglycine: a kinetic and X-ray study. Biochemistry 36: 12633–12643, 1997. [DOI] [PubMed] [Google Scholar]
- 17. Corvino A, Severino B, Fiorino F, Frecentese F, Magli E, Perissutti E, Santagada V, Bucci M, Cirino G, Kelly G, Servillo L, Popowicz G, Pastore A, and Caliendo G. Fragment-based de novo design of a cystathionine γ-lyase selective inhibitor blocking hydrogen sulfide production. Sci Rep 6: 34398, 2016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18. Druzhyna N, Szczesny B, Olah G, Módis K, Asimakopoulou A, Pavlidou A, Szoleczky P, Gerö D, Yanagi K, Törö G, López-García I, Myrianthopoulos V, Mikros E, Zatarain JR, Chao C, Papapetropoulos A, Hellmich MR, and Szabo C. Screening of a composite library of clinically used drugs and well-characterized pharmacological compounds for cystathionine β-synthase inhibition identifies benserazide as a drug potentially suitable for repurposing for the experimental therapy of colon cancer. Pharmacol Res 113: 18–37, 2016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19. Filipovic MR, Zivanovic J, Alvarez B, and Banerjee R. Chemical biology of H2S signaling through persulfidation. Chem Rev 118: 1253–1337, 2018. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20. Fiorucci S, Distrutti E, Cirino G, and Wallace JL. The emerging roles of hydrogen sulfide in the gastrointestinal tract and liver. Gastroenterology 131: 259–271, 2006. [DOI] [PubMed] [Google Scholar]
- 21. Fu M, Zhang W, Wu L, Yang G, Li H, and Wang R. Hydrogen sulfide (H2S) metabolism in mitochondria and its regulatory role in energy production. Proc Natl Acad Sci U S A 109: 2943–2948, 2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22. Fukuto JM, Carrington SJ, Tantillo DJ, Harrison JG, Ignarro LJ, Freeman BA, Chen A, and Wink DA. Small molecule signaling agents: the integrated chemistry and biochemistry of nitrogen oxides, oxides of carbon, dioxygen, hydrogen sulfide, and their derived species. Chem Res Toxicol 25: 769–793, 2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23. Gallego-Martin T, Prieto-Lloret J, Aaronson PI, Rocher A, and Obeso A. Hydroxycobalamin reveals the involvement of hydrogen sulfide in the hypoxic responses of rat carotid body chemoreceptor cells. Antioxidants 8: 62, 2019. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24. Grossfeld RM, Yancey SW, and Baxter CF. Inhibitors of crayfish glutamic acid decarboxylase. Neurochem Res 9: 947–963, 1984. [DOI] [PubMed] [Google Scholar]
- 25. Hanaoka K, Sasakura K, Suwanai Y, Toma-Fukai S, Shimamoto K, Takano Y, Shibuya N, Terai T, Komatsu T, Ueno T, Ogasawara Y, Tsuchiya Y, Watanabe Y, Kimura H, Wang C, Uchiyama M, Kojima H, Okabe T, Urano Y, Shimizu T, and Nagano T. Discovery and mechanistic characterization of selective inhibitors of H2S-producing enzyme: 3-mercaptopyruvate sulfurtransferase (3MST) targeting active-site cysteine persulfide. Sci Rep 7: 40227, 2017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26. Hellmich MR, Coletta C, Chao C, and Szabo C. The therapeutic potential of cystathionine β-synthetase/hydrogen sulfide inhibition in cancer. Antioxid Redox Signal 22: 424–448, 2015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27. Henthorn HA and Pluth MD. Mechanistic insights into the H2S-mediated reduction of aryl azides commonly used in H2S detection. J Am Chem Soc 137: 15330–15336, 2015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28. Hotta SS. Oxidative metabolism of isolated brain mitochondria: changes caused by aminooxyacetate. Arch Biochem Biophys 127: 132–139, 1968. [DOI] [PubMed] [Google Scholar]
- 29. Hu Y, Wang L, Han X, Zhou Y, Zhang T, Wang L, Hong T, Zhang W, Guo X-X, Sun J, Qi Y, Yu J, Liu H, and Wu F. Discovery of a bioactive inhibitor with a new scaffold for cystathionine γ-Lyase. J Med Chem 62: 1677–1683, 2019. [DOI] [PubMed] [Google Scholar]
- 30. Ismail I, Chen Z, Sun L, Ji X, Ye H, Kang X, Huang H, Song H, Bolton SG, Xi Z, Pluth MD, and Yi L. Highly efficient H2S scavengers via thiolysis of positively-charged NBD amines. Chem Sci 11: 7823–7828, 2020. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31. Jiang J, Chan A, Ali S, Saha A, Haushalter KJ, Lam W-LM, Glasheen M, Parker J, Brenner M, Mahon SB, Patel HH, Ambasudhan R, Lipton SA, Pilz RB, and Boss GR. Hydrogen sulfide—mechanisms of toxicity and development of an antidote. Sci Rep 6: 20831, 2016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32. Kang J, Li Z, Organ CL, Park C-M, Yang C-t, Pacheco A, Wang D, Lefer DJ, and Xian M. pH-controlled hydrogen sulfide release for myocardial ischemia-reperfusion injury. J Am Chem Soc 138: 6336–6339, 2016. [DOI] [PubMed] [Google Scholar]
- 33. Kimura H. Production and physiological effects of hydrogen sulfide. Antioxid Redox Signal 20: 783–793, 2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34. Levinn CM, Cerda MM, and Pluth MD. Activatable small-molecule hydrogen sulfide donors. Antioxid Redox Signal 32: 96–109, 2019. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35. Li L, Rose P, and Moore PK. Hydrogen sulfide and cell signaling. Annu Rev Pharmacol Toxicol 51: 169–187, 2011. [DOI] [PubMed] [Google Scholar]
- 36. Libiad M, Yadav PK, Vitvitsky V, Martinov M, and Banerjee R. Organization of the human mitochondrial hydrogen sulfide oxidation pathway. J Biol Chem 289: 30901–30910, 2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37. McCormick DB and Snell EE. Pyridoxal phosphokinases. II. Effects of inhibitors. J Biol Chem 236: 2085–2088, 1961. [PubMed] [Google Scholar]
- 38. This reference has been deleted.
- 39. McCune CD, Chan SJ, Beio ML, Shen W, Chung WJ, Szczesniak LM, Chai C, Koh SQ, Wong PTH, and Berkowitz DB. “Zipped synthesis” by cross-metathesis provides a cystathionine β-synthase inhibitor that attenuates cellular H2S levels and reduces neuronal infarction in a rat ischemic stroke model. ACS Cent Sci 2: 242–252, 2016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40. Montoya LA and Pluth MD. Organelle-targeted H2S probes enable visualization of the subcellular distribution of H2S donors. Anal Chem 88: 5769–5774, 2016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41. Niu W, Wu P, Chen F, Wang J, Shang X, and Xu C. Discovery of selective cystathionine β-synthase inhibitors by high-throughput screening with a fluorescent thiol probe. Med Chem Comm 8: 198–201, 2017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42. Niu W-N, Yadav PK, Adamec J, and Banerjee R. S-Glutathionylation enhances human cystathionine β-synthase activity under oxidative stress conditions. Antioxid Redox Signal 22: 350–361, 2015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43. Oláh G, Módis K, Törö G, Hellmich MR, Szczesny B, and Szabo C. Role of endogenous and exogenous nitric oxide, carbon monoxide and hydrogen sulfide in HCT116 colon cancer cell proliferation. Biochem Pharmacol 149: 186–204, 2018. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44. Olson KR. A practical look at the chemistry and biology of hydrogen sulfide. Antioxid Redox Signal 17: 32–44, 2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45. Olson KR, Briggs A, Devireddy M, Xian M, and Gao Y. Are the beneficial effects of ‘antioxidant’ lipoic acid mediated through metabolism of reactive sulfur species? Free Radic Biol Med 146: 139–149, 2020. [DOI] [PubMed] [Google Scholar]
- 46. Olson KR, DeLeon ER, and Liu F. Controversies and conundrums in hydrogen sulfide biology. Nitric Oxide 41: 11–26, 2014. [DOI] [PubMed] [Google Scholar]
- 47. Panagaki T, Randi EB, Augsburger F, and Szabo C. Overproduction of H2S, generated by CBS, inhibits mitochondrial complex IV and suppresses oxidative phosphorylation in Down syndrome. Proc Natl Acad Sci U S A 116: 18769–18771, 2019. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48. Panagaki T, Randi EB, and Szabo C. Role of hydrogen sulfide and 3-mercaptopyruvate sulfurtransferase in the regulation of the endoplasmic reticulum stress response in hepatocytes. Biomolecules 10: 1692, 2020. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49. Petersen LC. The effect of inhibitors on the oxygen kinetics of cytochrome c oxidase. Biochim Biophys Acta 460: 299–307, 1977. [DOI] [PubMed] [Google Scholar]
- 50. Pfeffer M and Ressler C. β-Cyanoalanine, an inhibitor of rat liver cystathionase. Biochem Pharmacol 16: 2299–2308, 1967. [DOI] [PubMed] [Google Scholar]
- 51. Pleasure H. Physchiatric and neurological side-effects of isoniazid and iproniazid. AMA Arch Neur Psych 72: 313–320, 1954. [DOI] [PubMed] [Google Scholar]
- 52. Porter DW and Baskin SI. Specificity studies of 3-mercaptopyruvate sulfurtransferase. J Biochem Toxicol 10: 287–292, 1995. [DOI] [PubMed] [Google Scholar]
- 53. Powell CR, Dillon KM, and Matson JB. A review of hydrogen sulfide (H2S) donors: chemistry and potential therapeutic applications. Biochem Pharmacol 149: 110–123, 2018. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54. Qu K, Lee SW, Bian JS, Low CM, and Wong PTH. Hydrogen sulfide: neurochemistry and neurobiology. Neurochem Int 52: 155–165, 2008. [DOI] [PubMed] [Google Scholar]
- 55. Régnier V, Billard J-M, Gupta S, Potier B, Woerner S, Paly E, Ledru A, David S, Luilier S, Bizot J-C, Vacano G, Kraus JP, Patterson D, Kruger WD, Delabar JM, and London J. Brain phenotype of transgenic mice overexpressing cystathionine β-synthase. PLoS One 7: e29056, 2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56. Reiffenstein RJ, Hulberta WC, and Roth SH. Toxicology of hydrogen sulfide. Annu Rev Pharmacol Toxicol 32: 109–134, 1992. [DOI] [PubMed] [Google Scholar]
- 57. Ressler C, Nigam SN, and Giza YH. Toxic principle in vetch. Isolation and identification of .alpha.-L-glutamyl-L-.beta.-cyanoalanine from common vetch seeds. Distribution in some legumes. J Am Chem Soc 91: 2758–2765, 1969. [DOI] [PubMed] [Google Scholar]
- 58. Salnikov DS, Kucherenko PN, Dereven'kov IA, Makarov SV, and van Eldik R. Kinetics and mechanism of the reaction of hydrogen sulfide with cobalamin in aqueous solution. Eur J Inorg Chem 2014: 852–862, 2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59. Searcy DG and Lee SH. Sulfur reduction by human erythrocytes. J Exp Zool 282: 310–322, 1998. [DOI] [PubMed] [Google Scholar]
- 60. Shinozijka S, Tanase S, and Morino Y. Metabolic consequences of affinity labeling of cystathionase and alanine aminotransferase by l-propargylglycine in vivo. Eur J Biochem 124: 377–382, 1982. [DOI] [PubMed] [Google Scholar]
- 61. Sun Q, Collins R, Huang S, Holmberg-Schiavone L, Anand GS, Tan C-H, van-den-Berg S, Deng L-W, Moore PK, Karlberg T, and Sivaraman J. Structural basis for the inhibition mechanism of human cystathionine γ-lyase, an enzyme responsible for the production of H2S. J Biol Chem 284: 3076–3085, 2009. [DOI] [PubMed] [Google Scholar]
- 62. Szabó C. Hydrogen sulphide and its therapeutic potential. Nat Rev Drug Discov 6: 917–935, 2007. [DOI] [PubMed] [Google Scholar]
- 63. Szabo C, Coletta C, Chao C, Módis K, Szczesny B, Papapetropoulos A, and Hellmich MR. Tumor-derived hydrogen sulfide, produced by cystathionine-β-synthase, stimulates bioenergetics, cell proliferation, and angiogenesis in colon cancer. Proc Natl Acad Sci U S A 110: 12474–12479, 2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64. Szabo C and Papapetropoulos A. International union of basic and clinical pharmacology. CII: pharmacological modulation of H2S levels: H2S donors and H2S biosynthesis inhibitors. Pharmacol Rev 69: 497–564, 2017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65. Teng H, Wu B, Zhao K, Yang G, Wu L, and Wang R. Oxygen-sensitive mitochondrial accumulation of cystathionine β-synthase mediated by Lon protease. Proc Natl Acad Sci U S A 110: 12679–12684, 2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66. Thorson MK, Majtan T, Kraus JP, and Barrios AM. Identification of cystathionine β-synthase inhibitors using a hydrogen sulfide selective probe. Angew Chem Int Ed Engl 52: 4641–4644, 2013. [DOI] [PubMed] [Google Scholar]
- 67. Truong DH, Mihajlovic A, Gunness P, Hindmarsh W, and O'Brien PJ. Prevention of hydrogen sulfide (H2S)-induced mouse lethality and cytotoxicity by hydroxocobalamin (vitamin B12a). Toxicology 242: 16–22, 2007. [DOI] [PubMed] [Google Scholar]
- 68. Van de Louw A and Haouzi P. Ferric iron and cobalt (III) compounds to safely decrease hydrogen sulfide in the body? Antioxid Redox Signal 19: 510–516, 2013. [DOI] [PubMed] [Google Scholar]
- 69. Vitvitsky V, Miljkovic JL, Bostelaar T, Adhikari B, Yadav PK, Steiger AK, Torregrossa R, Pluth MD, Whiteman M, Banerjee R, and Filipovic MR. Cytochrome c reduction by H2S potentiates sulfide signaling. ACS Chem Biol 13: 2300–2307, 2018. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70. Wallace JL and Wang R. Hydrogen sulfide-based therapeutics: exploiting a unique but ubiquitous gasotransmitter. Nat Rev Drug Discov 14: 329–345, 2015. [DOI] [PubMed] [Google Scholar]
- 71. Wang L, Cai H, Hu Y, Liu F, Huang S, Zhou Y, Yu J, Xu J, and Wu F. A pharmacological probe identifies cystathionine β-synthase as a new negative regulator for ferroptosis. Cell Death Dis 9: 1005, 2018. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72. Wang L, Shi H, Zhang X, Zhang X, Liu Y, Kang W, Shi X, and Wang T. I157172, a novel inhibitor of cystathionine γ-lyase, inhibits growth and migration of breast cancer cells via SIRT1-mediated deacetylation of STAT3. Oncol Rep 41: 427–436, 2019. [DOI] [PubMed] [Google Scholar]
- 73. Wang Q, Wei C, Pérez LM, Rogers WJ, Hall MB, and Mannan MS. Thermal decomposition pathways of hydroxylamine: theoretical investigation on the initial steps. J Phys Chem A 114: 9262–9269, 2010. [DOI] [PubMed] [Google Scholar]
- 74. Wang R. Physiological implications of hydrogen sulfide: a whiff exploration that blossomed. Physiol Rev 92: 791–896, 2012. [DOI] [PubMed] [Google Scholar]
- 75. Wang R, Gu X, Li Q, Gao J, Shi B, Xu G, Zhu T, Tian H, and Zhao C. Aggregation enhanced responsiveness of rationally designed probes to hydrogen sulfide for targeted cancer imaging. J Am Chem Soc 142: 15084–15090, 2020. [DOI] [PubMed] [Google Scholar]
- 76. Whitfield NL, Kreimier EL, Verdial FC, Skovgaard N, and Olson KR. Reappraisal of H2S/sulfide concentration in vertebrate blood and its potential significance in ischemic preconditioning and vascular signaling. Am J Physiol Regul Integr Comp Physiol 294: R1930–R1937, 2008. [DOI] [PubMed] [Google Scholar]
- 77. Wing DA and Baskin SI. Modifiers of mercaptopyruvate sulfurtransferase catalyzed conversion of cyanide to thiocyanate in vitro. J Biochem Toxicol 7: 65–72, 1992. [DOI] [PubMed] [Google Scholar]
- 78. Wolosker H, Blackshaw S, and Snyder SH. Serine racemase: a glial enzyme synthesizing D-serine to regulate glutamate-N-methyl-D-aspartate neurotransmission. Proc Natl Acad Sci U S A 96: 13409–13414, 1999. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79. Yang C-T, Wang Y, Marutani E, Ida T, Ni X, Xu S, Chen W, Zhang H, Akaike T, Ichinose F, and Xian M. Data-driven identification of hydrogen sulfide scavengers. Angew Chem Int Ed Engl 58: 10898–10902, 2019. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80. Yang J, Minkler P, Grove D, Wang R, Willard B, Dweik R, and Hine C. Non-enzymatic hydrogen sulfide production from cysteine in blood is catalyzed by iron and vitamin B6. Commun Biol 2: 194, 2019. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81. Zaorska E, Tomasova L, Koszelewski D, Ostaszewski R, and Ufnal M. Hydrogen sulfide in pharmacotherapy, beyond the hydrogen sulfide-donors. Biomolecules 10: 323, 2020. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82. Zhang J, Sio SWS, Moochhala S, and Bhatia M. Role of hydrogen sulfide in severe burn injury-induced inflammation in mice. Mol Med 16: 417–424, 2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83. Zhao Y, Biggs TD, and Xian M. Hydrogen sulfide (H2S) releasing agents: chemistry and biological applications. Chem Commun 50: 11788–11805, 2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84. Zhou Y, Yu J, Lei X, Wu J, Niu Q, Zhang Y, Liu H, Christen P, Gehring H, and Wu F. High-throughput tandem-microwell assay identifies inhibitors of the hydrogen sulfide signaling pathway. Chem Commun 49: 11782–11784, 2013. [DOI] [PubMed] [Google Scholar]











