Original article
The chemical biology of the persulfide (RSSH)/perthiyl (RSS·) redox couple and possible role in biological redox signaling

https://doi.org/10.1016/j.freeradbiomed.2016.09.020Get rights and content

Highlights

  • Hydropersulfides (RSSH) are readily oxidized to the corresponding perthiyl radical (RSS·).

  • RSS· does not react with O2 or NO.

  • Hydropersulfides are superior reductants compared to thiols (RSH).

  • Perthiyl is less reactive than a thiyl (RS·) species.

  • The RSSH/RSS· redox couple is biologically accessible and possibly relevant.

Abstract

The recent finding that hydropersulfides (RSSH) are biologically prevalent in mammalian systems has prompted further investigation of their chemical properties in order to provide a basis for understanding their potential functions, if any. Hydropersulfides have been touted as hyper-reactive thiol-like species that possess increased nucleophilicity and reducing capabilities compared to their thiol counterparts. Herein, using persulfide generating model systems, the ability of RSSH species to act as one-electron reductants has been examined. Not unexpectedly, RSSH is relatively easily oxidized, compared to thiols, by weak oxidants to generate the perthiyl radical (RSS·). Somewhat surprisingly, however, RSS· was found to be stable in the presence of both O2 and NO and only appears to dimerize. Thus, the RSSH/RSS· redox couple is readily accessible under biological conditions and since dimerization of RSS· may be a rare event due to low concentrations and/or sequestration within a protein, it is speculated that the general lack of reactivity of individual RSS· species may allow this couple to be utilized as a redox component in biological systems.

Introduction

Hydrogen sulfide (H2S) is an endogenously synthesized small molecule bioregulator with numerous physiological activities [1], [2], [3]. For example, H2S has been reported to be involved in the etiology of atherosclerosis [4] and cancer [5], regulate transcription via NRF2/KEAP1 [6] or NF-⎕B [7], control vascular tone [8], inhibit phosphodiesterase activity [9], protect against neurodegenerative diseases [10] and protect from ischemia-reperfusion toxicity [11], just to name a few. Current interest in the chemical biology of H2S has been motivated primarily by the reports of its biological activity and potential therapeutic utility. Despite the increasing recognition that H2S is an important physiological mediator/regulator, the biochemical targets and chemical mechanism(s) associated with these actions are, for the most part, unknown.

In a biological system, H2S can react with disulfides (RSSR), creating an equilibrium involving H2S, RSSR, thiols (RSH) and hydropersulfides (RSSH, note: this abbreviation will be used to denote all generic hydropersulfide species) (Reaction 1) [12], [13].RSSR+H2S⇌RSSH+RSH

The existence of this equilibrium reaction indicates the potential for RSSH formation from H2S when RSSR is present. Moreover, a pathway for H2S biosynthesis, conversion of cystine (Cys-SS-Cys) to H2S and pyruvate, involves the intermediacy of cysteine hydroperpersulfide (Cys-SSH) [14]. Thus, H2S and RSSH are intimately linked chemically and, presumably biologically. In support of this idea, numerous recent studies indicate that RSSH are ubiquitous and highly prevalent in mammals with concentrations of glutathione hydropersulfide (GSSH) as high as 150 micromolar in mouse brain and 50 micromolar in heart and liver [15]. Also, several labs report the prevalence of numerous protein hydropersulfides [15], [16], [17]. Indeed, it has been speculated that RSSH and polysulfides (RSnH or RSnR, n>2) are crucial biological effectors and that H2S can serve as a marker for their presence [18]. Although H2S is likely to have its own functions, it is becoming increasingly evident that RSSH (and derived compounds) can be important biological effectors, regulators and signaling species for example, [19]. Thus, the chemical biology of RSSH becomes a topic of vital importance in the attempt to delineate the mechanisms by which H2S and related species elicit their biological actions.

Recent work from this lab and others has shown that RSSH exhibit enhanced thiol-like chemistry compared to the corresponding thiol [20], [21], [22]. That is, RSSH are more nucleophilic and reducing (both one- and two-electron) compared to thiols and it is speculated that these properties may be important to their physiological utility. Of particular interest to the current study is the ability of RSSH to act as a one-electron reductant. One possible reason for the superior one-electron reducing capability of RSSH species versus the corresponding thiol is the presumed stability of the oxidized product, the perthiyl radical (RSS·), compared to that of a thiyl radical (RS·) (vide infra). Thus, in order to further evaluate and predict the possible physiological utility of hydropersulfides as one-electron donors or redox regulators, the formation, stability and reactivity of RSS· has been examined.

Section snippets

Reagents and instruments

4-Hydroxy-2,2,6,6-tetramethylpiperidine 1-oxyl (TEMPOL), diethylenetriamine pentaacetic acid (DTPA), potassium ferricyanide (K3Fe(CN)3), N-acetyl penicillamine (NAP) and myoglobin (horse heart) were purchased from Sigma-Aldrich (St. Louis, Mo). Sodium hydrogen sulfide (NaSH) was purchased from Strem Chemicals, Inc. (Newburyport, MA). The persulfide donor S-methoxycarbonyl penicillamine disulfide (MCPD) was synthesized as previously described [23]. The N-methoxycarbonyl penicillamine persulfide

Hydropersulfide generation

The in situ generation of hydropersulfides can be accomplished using a penicillamine-based donor species MCPD giving the hydropersulfide MCP-SSH [22], [23], [35] (Fig. 1a).

The reduction of TEMPOL by MCP-SSH generates the reduced TEMPOL-OH species (vide infra) and the corresponding perthiyl radical N-methoxycarbonyl penicillamine perthiyl radical (MCP-SS·) (Fig. 1b). Many of the experiments described herein involve the use of MCPD to make MCP-SSH, which can be oxidized by TEMPOL to give MCP-SS·.

Reduction of TEMPOL by hydropersulfides

Discussion

All of the results presented herein are consistent with the idea that RSSH species are good one-electron reductants. The model alkyl hydropersulfide used in this study, MCP-SSH (generated from decomposition of MCPD) (Fig. 1a), was easily oxidized by the relatively weak one-electron oxidants TEMPOL, TEMPO, K3Fe(CN)6 and MbFeIII (Fig. 2), a reaction that is likely driven by dimerization of the intermediate RSS· species (Fig. 3). EPR results are also consistent with the idea that reaction of RSSH

Acknowledgement

JMF wishes to acknowledge support from the National Institutes of Health (NIH) (HL106598) and the National Science Foundation (NSF) (CHE-1148641). JPT wishes to acknowledge support from the NSF (CHE-1213438 and CHE-1566065) and DJT wishes to acknowledge support from the NSF (CHE-030089) and the American Chemical Society Petroleum Research Fund (52801-ND4). The authors thank Prof. Joseph Lin (Sonoma State University), Prof. Peter Ford and Zhi Li (UCSB) for helpful discussions.

References (55)

  • M.D. Sevilla et al.

    Sulfinyl radical formation from the reaction of cysteine and glutathione thiyl radicals with molecular oxygen

    Biochem. Biophys. Res. Commun.

    (1987)
  • E. Madej et al.

    Thiyl radicals react with nitric oxide to form S-nitrosothiols with rates constants near diffusion-controlled limit

    Free Radic. Biol. Med.

    (2008)
  • M.D. Brookes et al.

    Millimeter-wave spectrum of the NO-dimer

    J. Mol. Spectrosc.

    (1997)
  • M.M. Cortese-Krott et al.

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

    Redox Biol.

    (2014)
  • Y.-M. Go et al.

    The cysteine proteome

    Free Radic. Biol. Med.

    (2015)
  • H. Kimura

    Physiological roles of hydrogen sulfide and polysulfides

    Handb. Exp. Pharm.

    (2015)
  • J.L. Wallace et al.

    Hydrogen sulfide-based therapeutics: exploiting a unique but ubiquitous gasotransmitter

    Nat. Rev.

    (2015)
  • S. Mani et al.

    Decreased endogenous production of hydrogen sulfide accelerates atherosclerosis

    Circulation

    (2013)
  • M.R. Hellmich et al.

    Hydrogen sulfide and cancer

    Handb. Exp. Pharm.

    (2015)
  • G. Yang et al.

    Hydrogen sulfide protects against cellular senescence via S-sulfhydration of KEAP1 and activation of Nrf2

    Antioxid. Redox Signal.

    (2012)
  • N. Sen et al.

    Hydrogen sulfide-linked sulfhydration of NF-⎕B mediates its antiapoptotic actions

    Mol. Cell

    (2013)
  • M. Al-Magableh et al.

    Mechanism of vasorelaxation and role of endogenous hydrogen sulfide production in mouse aorta

    Naunyn-Schmied. Arch. Pharmacol.

    (2011)
  • M. Bucci et al.

    Hydrogen sulfide is an endogenous inhibitor of phosphodiesterase activity

    Arterioscler. Thromb. Vasc. Biol.

    (2010)
  • X. Zhang et al.

    Hydrogen sulfide: a neuromodulator and neuroprotectant in the central nervous system

    ACS Chem. Neurosci.

    (2014)
  • A.L. King et al.

    Cytoprotective actions of hydrogen sulfide in ischemia-reperfusion injury

    Exp. Physiol.

    (2011)
  • D. Cavallini et al.

    Interactions of proteins with sulfide

    Eur. J. Biochem.

    (1970)
  • T. Yamanishi et al.

    The mechanism of the l-cystine cleavage reaction catalyzed by rat liver γ-cystathionase

    J. Biochem.

    (1981)
  • Cited by (87)

    • Versatile roles of cysteine persulfides in tumor biology

      2024, Current Opinion in Chemical Biology
    View all citing articles on Scopus
    View full text