Review article
Hydrogen sulfide in pharmacology and medicine – An update

https://doi.org/10.1016/j.pharep.2015.01.005Get rights and content

Abstract

Hydrogen sulfide (H2S) is the endogenously produced gasotransmitter involved in the regulation of nervous system, cardiovascular functions, inflammatory response, gastrointestinal system and renal function. Together with nitric oxide and carbon monoxide, H2S belongs to a family of gasotransmitters. H2S is synthesized from l-cysteine and/or l-homocysteine by cystathionine β-synthase, cystathionine γ-lyase and cysteine aminotransferase together with 3-mercaptopyruvate sulfurtransferase. Significant progress has been made in recent years in our understanding of H2S biochemistry, signaling mechanisms and physiological role. H2S-mediated signaling may be accounted for not only by the intact compound but also by its oxidized form, polysulfides. The most important signaling mechanisms include reaction with protein thiol groups to form persulfides (protein S-sulfhydration), reaction with nitric oxide and related species such as nitrosothiols to form thionitrous acid (HSNO), nitrosopersulfide (SSNO) and nitroxyl (HNO), as well as reaction with hemoproteins. H2S is enzymatically oxidized in mitochondria to thiosulfate and sulfate by specific enzymes, sulfide:quinone oxidoreductase, persulfide dioxygenase, rhodanese and sulfite oxidase. H2S donors have therapeutic potential for diseases such as arterial and pulmonary hypertension, atherosclerosis, ischemia–reperfusion injury, heart failure, peptic ulcer disease, acute and chronic inflammatory diseases, Parkinson's and Alzheimer's disease and erectile dysfunction. The group of currently available H2S donors includes inorganic sulfide salts, synthetic organic slow-releasing H2S donors, H2S-releasing non-steroidal antiinflammatory drugs, cysteine analogs, nucleoside phosphorothioates and plant-derived polysulfides contained in garlic. H2S is also regulated by many currently used drugs but the mechanism of these effects and their clinical implications are only started to be understood.

Introduction

It was first proposed by Abe and Kimura in 1996 that hydrogen sulfide (H2S) is the endogenously generated neuromodulator [1]. During almost two decades since their seminal paper was published, a large body of data about H2S in biological systems has been accumulated. Currently, there is little doubt that hydrogen sulfide is the third “gasotransmitter” in addition to nitric oxide (NO) and carbon monoxide (CO) [2], [3], [4]. In 2007 we published a review article about H2S in pharmacology in this journal [5]. Herein, I briefly review the progress made in the field since that time. Because H2S literature is now very huge, I will not comprehensively cover its effects in all experimental systems but will focus on general aspects of H2S biochemistry and molecular signaling mechanisms as well as its potential application in pharmacotherapy.

Section snippets

General properties of H2S

H2S is a colorless flammable gas with a strong odor of rotten eggs. It is easily soluble in both water and lipids. At physiological pH (7.4) less than 20% of H2S exists in the solution as the undissociated compound and the rest is dissociated to HS (hydrosulfide anion) and H+. Further dissociation of HS to sulfide anion (S2−) occurs only at high pH and is insignificant at physiological conditions. Since both H2S and HS always coexist in aqueous solution, it is not possible to separate their

Endogenous H2S synthesis

Four enzymatic pathways of H2S production have been described so far. Two of them are catalyzed by cystathionine β-synthase (CBS) and cystathionine γ-lyase (CSE) which are pyridoxal 5′-phosphate (vitamin B6)-dependent cytosolic enzymes of the transsulfuration pathway in which homocysteine is metabolized to cysteine. In the transsulfuration pathway l-homocysteine is condensed by CBS with l-serine to produce l-cystathionine and H2O. However, l-serine may be replaced in this reaction by l-cysteine

H2S concentration in plasma and tissues

Early studies suggested that H2S is present in plasma and tissues at relatively high concentrations; values up to 50–60 μM have been reported in plasma and even >100 μM in the brain. However, it was the artifact resulting from using colorimetric methylene blue method which has multiple disadvantages [6]. This method measures not only free H2S/HS but also other sulfide pools in the sample including sulfane sulfur and acid-labile sulfur; the latter mostly includes iron–sulfur clusters in proteins

H2S metabolism

H2S has been known for decades as one of the most important poisons in human toxicology. The main mechanism of its toxicity is inhibition of mitochondrial respiratory chain by binding to cytochrome c oxidase. Indeed, H2S is the second most potent inhibitor of this enzyme after cyanide [25]. However, one of the most exciting findings of the recent years in the H2S field was that it may also be enzymatically metabolized in mitochondria. H2S is the first and until now the only known inorganic

Mechanisms of H2S signaling in the cells

Biological effects of H2S result from its interactions with three groups of targets: (1) thiols, (2) reactive oxygen and nitrogen species as well as oxidation products of macromolecules, (3) metals and metalloproteins. Below these three targets are briefly characterized.

H2S as a target for pharmacotherapy

Although excess H2S may contribute to the pathogenesis of some diseases such as cancer [88], [89], septic shock [90] or acute pancreatitis [91] and pharmacological or genetic inhibition of H2S production is protective in some models, application of H2S donors and/or augmenting endogenous H2S attracts most attention as the possible therapeutic approach for several reasons. First, protective effects of H2S or its donors were demonstrated more convincingly in many experimental models. Potential

Conclusions and future directions

A significant progress has been made in recent years in the H2S field. The mechanisms of H2S synthesis by CBS and CSE were explained and it was demonstrated that it can be formed not only from cysteine but also from homocysteine. The new 3-MST-dependent pathways of H2S production have been described and its mitochondrial metabolism is now quite well characterized. Several molecular signaling mechanisms have been demonstrated. Many new methods of H2S measurements such as polarographic sensors,

Conflict of interest

None declared.

Acknowledgements

Author's own studies cited in this paper were supported by grant DS 476 from Medical University, Lublin, Poland, as well as by EU Project “The equipment of innovative laboratories doing research on new medicines used in the therapy of civilization and neoplastic diseases” within the Operational Program Development of Eastern Poland 2007–2013, Priority Axis I Modern Economy, Operations I.3 Innovation Promotion.

References (177)

  • N. Helmy et al.

    Oxidation of hydrogen sulfide by human liver mitochondria

    Nitric Oxide

    (2014)
  • M. Ackermann et al.

    The vertebrate homolog of sulfide-quinone reductase is expressed in mitochondria of neuronal tissues

    Neuroscience

    (2011)
  • O. Kabil et al.

    Characterization of patient mutations in human persulfide dioxygenase (ETHE1) involved in H2S catabolism

    J Biol Chem

    (2012)
  • K.R. Olson

    Hydrogen sulfide is an oxygen sensor in the carotid body

    Respir Physiol Neurobiol

    (2011)
  • N. Maenhaut et al.

    Hypoxia enhances the relaxing influence of perivascular adipose tissue in isolated mice aorta

    Eur J Pharmacol

    (2010)
  • K. Módis et al.

    Hydrogen sulfide-mediated stimulation of mitochondrial electron transport involves inhibition of the mitochondrial phosphodiesterase 2A, elevation of cAMP and activation of protein kinase A

    Biochem Pharmacol

    (2013)
  • N. Sen et al.

    Hydrogen sulfide-linked sulfhydration of NF-κB mediates its antiapoptotic actions

    Mol Cell

    (2012)
  • C. Guo et al.

    Hydrogen sulfide protected gastric epithelial cell from ischemia/reperfusion injury by Keap1 S-sulfhydration, MAPK dependent anti-apoptosis and NF-κB dependent anti-inflammation pathway

    Eur J Pharmacol

    (2014)
  • S. Koike et al.

    Polysulfide exerts a protective effect against cytotoxicity caused by t-buthylhydroperoxide through Nrf2 signaling in neuroblastoma cells

    FEBS Lett

    (2013)
  • S. Bruce King

    Potential biological chemistry of hydrogen sulfide (H2S) with the nitrogen oxides

    Free Radic Biol Med

    (2013)
  • M. Whiteman et al.

    Evidence for the formation of a novel nitrosothiol from the gaseous mediators nitric oxide and hydrogen sulfide

    Biochem Biophys Res Commun

    (2006)
  • M.M. Cortese-Krott et al.

    Nitrosopersulfide (SSNO) accounts for sustained NO bioactivity of S-nitrosothiols following reaction with sulfide

    Redox Biol

    (2014)
  • M. Nishida et al.

    Role of 8-nitro-cGMP and its redox regulation in cardiovascular electrophilic signaling

    J Mol Cell Cardiol

    (2014)
  • D.G. Searcy

    HS:O2 oxidoreductase activity of Cu, Zn superoxide dismutase

    Arch Biochem Biophys

    (1996)
  • K. Kashfi et al.

    Biology and therapeutic potential of hydrogen sulfide and hydrogen sulfide-releasing chimeras

    Biochem Pharmacol

    (2013)
  • A. Mijušković et al.

    Comparison of the effects of methanethiol and sodium sulphide on uterine contractile activity

    Pharmacol Rep

    (2014)
  • S. Bekpinar et al.

    Propargylglycine aggravates liver damage in LPS-treated rats: possible relation of nitrosative stress with the inhibition of H2S formation

    Pharmacol Rep

    (2014)
  • M. Sikora et al.

    Exogenous hydrogen sulfide causes different hemodynamic effects in normotensive and hypertensive rats via neurogenic mechanisms

    Pharmacol Rep

    (2014)
  • L. Li et al.

    GYY4137, a novel hydrogen sulfide-releasing molecule, protects against endotoxic shock in the rat

    Free Radic Biol Med

    (2009)
  • K. Abe et al.

    The possible role of hydrogen sulfide as an endogenous neuromodulator

    J Neurosci

    (1996)
  • H. Kimura

    Hydrogen sulfide and polysulfides as biological mediators

    Molecules

    (2014)
  • R. Wang

    Physiological implications of hydrogen sulfide: a whiff exploration that blossomed

    Physiol Rev

    (2012)
  • E. Łowicka et al.

    Hydrogen sulfide H2S – the third gas of interest for pharmacologists

    Pharmacol Rep

    (2007)
  • M. Iciek et al.

    Biosynthesis and biological properties of compounds containing highly reactive: reduced sulfane sulfur

    Pol J Pharmacol

    (2001)
  • J.I. Toohey et al.

    Thiosulfoxide (sulfane) sulfur: new chemistry and new regulatory roles in biology

    Molecules

    (2014)
  • O. Kabil et al.

    Enzymology of H2S biogenesis, decay and signaling

    Antioxid Redox Signal

    (2014)
  • S. Carballal et al.

    Kinetics of reversible reductive carbonylation of heme in human cystathionine β-synthase

    Biochemistry

    (2013)
  • T. Morikawa et al.

    Hypoxic regulation of the cerebral microcirculation is mediated by a carbon monoxide-sensitive hydrogen sulfide pathway

    Proc Natl Acad Sci USA

    (2012)
  • H. Jurkowska et al.

    Cystathionine γ-lyase

    Postepy Hig Med Dosw (Online)

    (2014)
  • T. Ida et al.

    Reactive cysteine persulfides and S-polythiolation regulate oxidative stress and redox signaling

    Proc Natl Acad Sci USA

    (2014)
  • N. Shibuya et al.

    3-Mercaptopyruvate sulfurtransferase produces hydrogen sulfide and bound sulfane sulfur in the brain

    Antioxid Redox Signal

    (2009)
  • N. Shibuya et al.

    Vascular endothelium expresses 3-mercaptopyruvate sulfurtransferase and produces hydrogen sulfide

    J Biochem

    (2009)
  • Y. Mikami et al.

    Thioredoxin and dihydrolipoic acid are required for 3-mercaptopyruvate sulfurtransferase to produce hydrogen sulfide

    Biochem J

    (2011)
  • N. Shibuya et al.

    A novel pathway for the production of hydrogen sulfide from d-cysteine in mammalian cells

    Nat Commun

    (2013)
  • N.L. Whitfield et al.

    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

    (2008)
  • C.E. Cooper et al.

    The inhibition of mitochondrial cytochrome oxidase by the gases carbon monoxide, nitric oxide, hydrogen cyanide and hydrogen sulfide: chemical mechanism and physiological significance

    J Bioenergy Biomembr

    (2008)
  • T.M. Hildebrandt et al.

    Three enzymatic activities catalyze the oxidation of sulfide to thiosulfate in mammalian and invertebrate mitochondria

    FEBS J

    (2008)
  • F. Bouillaud et al.

    Mitochondria and sulfide: a very old story of poisoning, feeding, and signaling?

    Antioxid Redox Signal

    (2011)
  • M.R. Jackson et al.

    Human sulfide:quinone oxidoreductase catalyzes the first step in hydrogen sulfide metabolism and produces a sulfane sulfur metabolite

    Biochemistry

    (2012)
  • K.R. Olson

    Mitochondrial adaptations to utilize hydrogen sulfide for energy and signaling

    J Comp Physiol B

    (2012)
  • Cited by (0)

    View full text