Skip to main content
Proceedings of the National Academy of Sciences of the United States of America logoLink to Proceedings of the National Academy of Sciences of the United States of America
. 2007 Nov 8;104(46):17907–17908. doi: 10.1073/pnas.0709010104

A new gaseous signaling molecule emerges: Cardioprotective role of hydrogen sulfide

David J Lefer 1,*
PMCID: PMC2084269  PMID: 17991773

Cardiovascular (CV) research has largely been focused on two endogenously produced gaseous signaling molecules, namely, nitric oxide (NO) and carbon monoxide (CO). These gaseous messengers are both highly conserved evolutionarily and synthesized by endogenous enzyme systems. Extensive research efforts have uniformly demonstrated that CO- and NO-based therapeutics protect the brain, heart, and circulation against a number of CV disease states (13). However, controversy exists regarding the therapeutic value of both CO and NO, with some studies citing NO cytotoxicity at very high levels and under certain conditions (2). The toxic effects of NO are related to the highly reactive nature of NO and its interaction with superoxide to form the potent oxidant peroxynitrite (ONOO) (2). Very recently, a third endogenously produced gaseous signaling molecule, hydrogen sulfide (H2S), has emerged as a potentially important mediator of CV homeo stasis and cytoprotection (3). In this issue of PNAS, Benavides et al. (4) reveal that the vasculoprotective and antihypertensive effects of dietary garlic (Allium sativum) are mediated in large part via the generation of H2S.

Previous studies indicate that dietary garlic consumption is inversely correlated with the progression and severity of CV disease and that it affects a lower incidence of hypertension (5). However, the mechanism(s) by which consumption of garlic attenuates CV disease has in large part remained a mystery. Benavides et al. (4) elegantly demonstrate that garlic-derived organic polysulfides are converted by red blood cells (RBCs) into hydrogen sulfide gas. Interestingly, the garlic bulbs used for this study were obtained from local supermarkets in the city of Birmingham, AL, and then pressed by the investigators to obtain garlic juice on the day of the experiment. The bioconversion of polysulfides to H2S is enhanced by glucose-maintained cytosolic glutathione levels and depends on reduced thiols in close proximity to the RBC membrane. The authors demonstrate that generation of H2S after bioconversion of garlic-derived polysulfides results in potent vasorelaxation of isolated blood vessels. The authors propose that H2S produced from garlic leads to vasorelaxation via vascular smooth muscle KATP channel-mediated hyperpolarization (3). These novel experimental studies provide important information regarding the mechanisms responsible for garlic-mediated attenuation of hypertension and serve to highlight the potential importance of H2S therapy in CV disease.

H2S is well known as a toxic gas with a repulsive odor often used to describe the smell of rotten eggs (3). Experimental studies reveal that H2S is produced enzymatically at micromolar levels in mammals and exerts a number of physiological actions in the CV system (3, 6, 7). H2S production has been attributed to two key enzymes in the cysteine biosynthesis pathway, cystathionine β-synthase (CBS) and cystathionine γ-lyase (CGL). CBS and CGL are both heme-containing enzymes whose activity depends on the cofactor pyridoxal 5′-phosphate (68). CBS is capable of catalyzing the reaction of cysteine with other free thiols to generate H2S; likewise, thiocysteine generated by CGL can interact with thiols to generate H2S (68). The endogenous production of H2S was initially described in the brain and attributed to CBS (79). In addition, H2S is produced in the vasculature by CGL, where it mediates smooth muscle relaxation and subsequent vasodilation (3). At present, the distribution of CGL and CBS throughout the body are not well characterized. Furthermore, very little is currently known regarding the functions of CBS and CGL under physiological and pathophysiological conditions. As this rapidly emerging field advances, we are likely to learn a great deal regarding the biochemistry and physiology of these H2S-producing enzyme systems.

Since the recent discovery that H2S is a powerful gaseous signaling molecule, studies have begun to characterize the biological profile of this promiscuous gas. Fig. 1 summarizes some of the physiological actions of H2S in the CV system. H2S has been shown to relax vascular smooth muscle, induce vaso dilation of isolated blood vessels, and reduce blood pressure (3, 10). In this regard, H2S might be an important regulator of blood pressure. H2S has also been shown to inhibit leukocyte–endothelial cell interactions in vivo. Zanardo et al. (11) used intravital microscopy to demonstrate that H2S donors inhibit leukocyte adherence in the rat mesenteric microcirculation during vascular inflammation, suggesting that H2S is a potent antiinflammatory molecule. It has also become evident that H2S is as a potent antioxidant and, under chronic conditions, can up-regulate antioxidant defenses (9). In addition, it has been demonstrated that H2S effectively inhibits apoptosis of a number of cell types, and this effect has been shown to promote cytoprotection (9).

Fig. 1.

Fig. 1.

Summary of the physiological actions of hydrogen sulfide (H2S). H2S is produced in micromolar (i.e., 10–100 μM) concentrations in the circulation and exerts a number of critical effects on the CV system. H2S has been shown to induce vasodilation and inhibit leukocyte-endothelial cell interactions in the circulation. H2S is a potent antioxidant and inhibits cellular apoptosis. H2S also has been shown to transiently and reversibly inhibit mitochondrial respiration. Taken together, this physiological profile is ideally suited for protection of the CV system against disease states.

Research interest in H2S has also been focused on its profound metabolic effects, which result in a state of suspended animation with drastic reduction in metabolic demand (12). Initiating suspended animation may prove to be an ideal therapy for various shock or trauma states in which dramatic reductions in metabolic demands may be highly protective. Studies investigating the metabolic effects of H2S have revealed that, similar to NO, H2S exerts potent effects on mitochondrial function and respiration (10, 13). Physiological concentrations have been shown to dose-dependently and transiently inhibit mitochondrial respiration, an effect that could promote myocardial cell survival during myocardial ischemia and reperfusion (I/R) (10). The mitochondrial effects of H2S have been shown to be mediated in part by reversible and potent inhibition of cytochrome c oxidase (complex IV of the mitochondrial electron transport chain) (10, 13). The defined physiological profile of H2S makes this novel gaseous signaling molecule an ideal pharmacological candidate for the treatment of CV disease states.

It appears that H2S possesses all of the positive effects of NO without the capacity to form a toxic metabolite.

A number of recent studies have provided insights into the protective actions of H2S in the setting of myocardial I/R injury (10, 1417). Pretreatment with the H2S donor (NaHS) reduces arrhythmias in isolated hearts subjected to global cardiac I/R and improves myocyte survival (14, 15). Similarly, Johansen et al. (16) reported that NaHS pretreatment significantly attenuates myocardial injury in isolated, perfused rat hearts subjected to cardiac I/R. An article by Sivarajah et al. (17) represents the first report of the cardioprotective actions of H2S in an in vivo model of myocardial I/R injury. The authors reported a 25% reduction in myocardial infarct size after pretreatment with an H2S–donor compound. Further support for the cardioprotective effects of H2S in myocardial I/R injury are provided by Elrod et al. (10), who have reported that administration of H2S at the time of reperfusion dose-dependently attenuates myocardial infarct size and preserves postischemic left ventricular (LV) function in a clinically relevant, in vivo model of acute myocardial infarction. Furthermore, cardiac-specific overexpression of the H2S-generating enzyme CGL significantly reduces myocardial I/R injury and similarly preserves LV function (10). The authors propose that preservation of cardiac mitochondrial structure and function is central in the cardioprotective actions of H2S in I/R injury.

Recent investigations into the vascular effects of H2S have revealed that this physiologically produced gas has profound effects on vascular growth or formation and neointimal hyperplasia. Meng et al. (18) have very recently demonstrated that H2S ameliorates the intimal hyperplasia response in blood vessels after balloon-mediated vascular injury. Furthermore, Cai et al. (19) have reported that physiologically relevant dosages of H2S promote a highly robust angiogenic response that is largely dependent on activation of Akt. Thus, H2S may prove beneficial for the inhibition of coronary artery restenosis and the induction of therapeutic angiogenesis.

Clearly, additional research is warranted to further our understanding of the synthesis, metabolism, and physiological and potentially pathophysiological actions of endogenously produced H2S in mammalian systems. The biological profile of H2S strongly suggests that this gaseous signaling molecule is likely to protect a variety of organs despite its prior reputation as a noxious gas with a repulsive odor. It appears that H2S possesses all of the positive effects of NO without the capacity to form a toxic metabolite such as ONOO. It is clear that H2S will provide many years of exciting research opportunities for scientific investigators in diverse disciplines. The paper by Benavides et al. (4) sets the stage for future research into the physiological and pharmacological actions of H2S in the CV system.

Footnotes

Conflict of interest statement: The author is currently funded by Ikaria Corporation for a research grant and serves as a paid Consultant for Ikaria Corporation. Ikaria is currently developing hydrogen sulfide therapy for a number of disease states.

See the companion article on page 17977.

References

  • 1.Baranano DE, Snyder SH. Proc Natl Acad Sci USA. 2001;98:10996–11002. doi: 10.1073/pnas.191351298. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Elrod JW, Duranski MR, Langston WJ, Greer JJM, Tao L, Dugas TR, Kevil CG, Champion HC, Lefer DJ. Circ Res. 2006;99:78–85. doi: 10.1161/01.RES.0000231306.03510.77. [DOI] [PubMed] [Google Scholar]
  • 3.Pryor WA, Houk KN, Foote CS, Fukuto JM, Ignarro LJ, Squadrito GL, Davies KJ. Am J Physiol. 2006;291:R491–R511. doi: 10.1152/ajpregu.00614.2005. [DOI] [PubMed] [Google Scholar]
  • 4.Benavides GA, Squadrito GL, Mills RW, Patel HD, Isbell TS, Patel RP, Darley-Usmar VM, Doeller JE, Kraus DW. Proc Natl Acad Sci USA. 2007;104:17977–17982. doi: 10.1073/pnas.0705710104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Banerjee SK, Maulik SK. Nutr J. 2002;1:4. doi: 10.1186/1475-2891-1-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Kamoun P. Amino Acids. 2004;26(3):243–254. doi: 10.1007/s00726-004-0072-x. [DOI] [PubMed] [Google Scholar]
  • 7.Boehning D, Snyder SH. Annu Rev Neurosci. 2003;26:105–131. doi: 10.1146/annurev.neuro.26.041002.131047. [DOI] [PubMed] [Google Scholar]
  • 8.Abe K, Kimura H. J Neurosci. 1996;16:1066–1071. doi: 10.1523/JNEUROSCI.16-03-01066.1996. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Kimura Y, Kimura H. FASEB J. 2004;18(10):1165–1170. doi: 10.1096/fj.04-1815fje. [DOI] [PubMed] [Google Scholar]
  • 10.Elrod JW, Calvert JW, Morrison J, Doeller JE, Kraus DW, Tao L, Jiao X, Scalia R, Kiss L, Szabo C, et al. Proc Natl Acad Sci USA. 2007;104:15560–15565. doi: 10.1073/pnas.0705891104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Zanardo RC, Brancaleone V, Distrutti E, Fiorucci S, Cirino G, Wallace JL. FASEB J. 2006;20:2118–21120. doi: 10.1096/fj.06-6270fje. [DOI] [PubMed] [Google Scholar]
  • 12.Blackstone E, Morrison M, Roth MB. Science. 2005;308:518. doi: 10.1126/science.1108581. [DOI] [PubMed] [Google Scholar]
  • 13.Nicholls P. Biochem Soc Trans. 1975;3:316–319. doi: 10.1042/bst0030316. [DOI] [PubMed] [Google Scholar]
  • 14.Bian JS, Yong QC, Pan TT, Feng ZN, Ali MY, Zhou S, Moore PK. J Pharmacol Exp Ther. 2006;316:670–678. doi: 10.1124/jpet.105.092023. [DOI] [PubMed] [Google Scholar]
  • 15.Pan TT, Feng ZN, Lee SW, Moore PK, Bian JS. J Mol Cell Cardiol. 2006;40(1):119–130. doi: 10.1016/j.yjmcc.2005.10.003. [DOI] [PubMed] [Google Scholar]
  • 16.Johansen D, Ytrehus K, Baxter GF. Basic Res Cardiol. 2006;101(1):53–60. doi: 10.1007/s00395-005-0569-9. [DOI] [PubMed] [Google Scholar]
  • 17.Sivarajah A, McDonald MC, Thiemermann C. Shock. 2006;26:154–161. doi: 10.1097/01.shk.0000225722.56681.64. [DOI] [PubMed] [Google Scholar]
  • 18.Meng QH, Yang G, Yang W, Jiang B, Wu L, Wang R. Am J Pathol. 2007;170(4):1406–1414. doi: 10.2353/ajpath.2007.060939. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Cai WJ, Wang MJ, Moore PK, Jin HM, Yao T, Zhu YC. Cardiovasc Res. 2007;76(1):29–40. doi: 10.1016/j.cardiores.2007.05.026. [DOI] [PubMed] [Google Scholar]

Articles from Proceedings of the National Academy of Sciences of the United States of America are provided here courtesy of National Academy of Sciences

RESOURCES