Elsevier

Nitric Oxide

Volume 50, 15 November 2015, Pages 38-45
Nitric Oxide

Review
Hydrogen sulfide in cancer: Friend or foe?

https://doi.org/10.1016/j.niox.2015.08.004Get rights and content

Highlights

  • Hydrogen sulfide (H2S) has opposite effects on cancer: pro-cancer and anti-cancer.

  • Endogenous H2S promotes cancer development and progression.

  • Inhibition of endogenous H2S production may be a new strategy for cancer treatment.

  • Relatively high concentration of exogenous H2S suppresses growth of cancer cells.

  • H2S donors and H2S-releasing hybrids could be developed as novel anti-cancer drugs.

Abstract

Hydrogen sulfide (H2S) is the third gaseous signaling molecule that plays important roles in cancer biological processes. Recent studies indicate that H2S has both pro-cancer and anti-cancer effects. Endogenous H2S can exert pro-cancer functions through induction of angiogenesis regulation of mitochondrial bioenergetics, acceleration of cell cycle progression, and anti-apoptosis mechanisms. Thus, the inhibition of the production of H2S in cancer cells may be a new cancer treatment strategy. In contrast to the pro-cancer effect of H2S, relatively high concentrations of exogenous H2S could suppress the growth of cancer cells by inducing uncontrolled intracellular acidification, inducing cell cycle arrest, and promoting apoptosis. Therefore, H2S donors and H2S-releasing hybrids could be designed and developed as novel anti-cancer drugs. In this review, the production and metabolism of H2S in cancer cells are summarized and the role and mechanism of H2S in cancer development and progression are further discussed.

Introduction

Hydrogen sulfide (H2S) was known only as an environmental pollutant for several centuries but is now widely considered an important biological and pharmacological mediator. In mammals, endogenous H2S mainly comes from the metabolism of l-cysteine and homocysteine by the catalysis of two pyridoxal-5′-phosphate (PLP)-dependent enzymes, termed cystathionine γ-lyase (CSE) and cystathionine β-synthase (CBS). Both CSE and CBS are cytosolic enzymes [1], [2], [3]. 3-Mercaptopyruvate sulfurtransferase (3-MST), a PLP-independent enzyme, acts in combination with cysteine aminotransferase (CAT) to produce H2S from l-cysteine in the presence of α-ketoglutarate [4]. CAT and 3-MST are localized in both the cytosol and mitochondria [1], [5]. Once formed, H2S can be immediately released or stored as bound and acid-labile sulfur in the cells [6].

H2S has been recognized as the third endogenous gasotransmitter in mammals, along with nitric oxide and carbon monoxide [7], [8], [9]. A number of studies have shown that H2S is involved in many physiological and pathophysiological functions [1], [2], [5], [8], [9], [10], [11], [12]. Nevertheless, there has been some controversy on the role of H2S in cancer development and progression. In multicellular organisms, normal cellular homeostasis is maintained through a balance between the processes of cell proliferation and cell death. Aberrations in cell survival could disrupt the normal cell cycle regulation and initiate tumor formation and metastasis [13], [14], [15]. In recent years, an increasing amount of evidence suggests that exogenously administered and/or endogenously produced H2S could exhibit two obviously opposite functions on the growth of cancer cells [16], [17], [18], [19]. Therefore, it is urgent and essential to illuminate the effect and mechanism of H2S on the growth and proliferation of cancer cells.

In this review, we highlight recent studies that provide new insight into the production and metabolism of H2S in cancer cells, as well as further discuss the role and mechanism of H2S in cancer development and progression.

Section snippets

CSE

Accumulating evidence indicates that CSE plays important roles in many different types of cancer cells. For example, knockdown of CSE by shRNA or its inhibition by dl-propargylglycine inhibits the proliferation and migration of SW480 human colon cancer cells [20]. Similarly, CSE expression can be detected at both the mRNA and protein levels in human colon cancer HCT116 cells [8], [21]. Marked CSE expression is also observed in WiRd, another colon cancer cell line [22]. These results suggest

The catabolism of H2S in cancer

H2S can be metabolized through several enzymatic and nonenzymatic processes in normal mammalian cells [50], [51]. The main pathway of H2S catabolism occurs in mitochondria [52]. H2S could be converted into thiosulfate through several enzymes including quinone oxidoreductase, S-dioxygenase, and S-transferase. Thiosulfate is further metabolized to sulfate via the actions of thiosulfate reductase and sulfite oxidase [50], [51]. H2S can also be methylated by thiol S-methyltransferase to form

The pro-cancer effect of H2S

Recent findings indicate that there are many mechanisms contributing to the pro-cancer effect of H2S, including induction of angiogenesis, regulation of mitochondrial bioenergetics, acceleration of cell cycle progression, and anti-apoptosis function (Fig. 1).

The anti-cancer effect of H2S

In addition to its pro-cancer effect, H2S could also inhibit the proliferation of cancer cells mainly through induction of uncontrolled intracellular acidification, induction of cell cycle arrest, and promotion of apoptosis (Fig. 2).

Conclusions and future directions

In mammals, CSE, CBS and 3-MST are three key H2S-producing enzymes. These enzymes could be detected in many different tumor types, and their expression levels are significantly different and have been shown to have cancer-specific characteristics. Therefore, CSE, CBS and 3-MST could be potential biomarkers and novel molecular targets for cancer diagnostics and treatment. Although the major breakdown product of H2S could be detected in samples of exhaled air and flatus from cancer patients,

Competing interest

The authors declare that they have no conflicts of interest with regard to this work.

Acknowledgments

This study was supported by grants from the National Natural Science Foundation of China (No. 81000045), the Natural Science Foundation of Education Department of Henan Province, China (No. 15A310017), and the Foundation of Science & Technology Department of Henan Province, China (No. 132300410012).

References (91)

  • Y. Pei et al.

    Hydrogen sulfide mediates the anti-survival effect of sulforaphane on human prostate cancer cells

    Toxicol. Appl. Pharmacol.

    (2011)
  • K. Módis et al.

    Effect of S-adenosyl-L-methionine (SAM), an allosteric activator of cystathionine-β-synthase (CBS) on colorectal cancer cell proliferation and bioenergetics in vitro

    Nitric Oxide

    (2014)
  • S. Sen et al.

    Role of cystathionine β-synthase in human breast cancer

    Free Radic. Biol. Med.

    (2015)
  • D. Sumi et al.

    Sp1 transcription factor expression is regulated by estrogen-related receptor alpha1

    Biochem. Biophys. Res. Commun.

    (2005)
  • K.N. Maclean et al.

    The dominant role of Sp1 in regulating the cystathionine beta-synthase -1a and -1b promoters facilitates potential tissue-specific regulation by Kruppel-like factors

    J. Biol. Chem.

    (2004)
  • N. Wu et al.

    Ischemia/reperfusion reduces transcription factor Sp1-mediated cystathionine beta-synthase expression in the kidney

    J. Biol. Chem.

    (2010)
  • K. Módis et al.

    Oxidative stress suppresses the cellular bioenergetic effect of the 3-mercaptopyruvate sulfurtransferase/hydrogen sulfide pathway

    Biochem. Biophys. Res. Commun.

    (2013)
  • O. Kabil et al.

    Redox biochemistry of hydrogen sulfide

    J. Biol. Chem.

    (2010)
  • K. Kashfi et al.

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

    Biochem. Pharmacol.

    (2013)
  • D. Wu et al.

    Anti-tumor effects of a novel chimeric peptide on S180 and H22 xenografts bearing nude mice

    Peptides

    (2010)
  • B. Li et al.

    Ginsenoside Rh2 induces apoptosis and paraptosis-like cell death in colorectal cancer cells through activation of p53

    Cancer Lett.

    (2011)
  • S. Fulda

    Targeting apoptosis for anticancer therapy

    Semin. Cancer Biol.

    (2015)
  • K. Fernald et al.

    Evading apoptosis in cancer

    Trends Cell Biol.

    (2013)
  • D. Wu et al.

    Peptide-based cancer therapy: opportunity and challenge

    Cancer Lett.

    (2014)
  • T. Murata et al.

    Differential susceptibility to hydrogen sulfide-induced apoptosis between PHLDA1-overexpressing oral cancer cell lines and oral keratinocytes: role of PHLDA1 as an apoptosis suppressor

    Exp. Cell Res.

    (2014)
  • R. Wang

    Physiological implications of hydrogen sulfide: a whiff exploration that blossomed

    Physiol. Rev.

    (2012)
  • B.D. Paul et al.

    H2S signalling through protein sulfhydration and beyond

    Nat. Rev. Mol. Cell Biol.

    (2012)
  • K.R. Olson

    Hydrogen sulfide as an oxygen sensor

    Antioxid. Redox. Signal.

    (2015)
  • H. Kimura

    Hydrogen sulfide: its production, release and functions

    Amino Acids

    (2011)
  • C. Szabó

    Hydrogen sulphide and its therapeutic potential

    Nat. Rev. Drug Discov.

    (2007)
  • C. Szabo et al.

    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.

    (2013)
  • G. Yang et al.

    H2S as a physiologic vasorelaxant: hypertension in mice with deletion of cystathionine gamma-lyase

    Science

    (2008)
  • C. Szabo

    Roles of hydrogen sulfide in the pathogenesis of diabetes mellitus and its complications

    Antioxid. Redox. Signal.

    (2012)
  • J.W. Calvert et al.

    Genetic and pharmacologic hydrogen sulfide therapy attenuates ischemia-induced heart failure in mice

    Circulation

    (2010)
  • R. Clarke et al.

    Endoplasmic reticulum stress, the unfolded protein response, autophagy, and the integrated regulation of breast cancer cell fate

    Cancer Res.

    (2012)
  • Z.W. Lee et al.

    Utilizing hydrogen sulfide as a novel anti-cancer agent by targeting cancer glycolysis and pH imbalance

    Br. J. Pharmacol.

    (2014)
  • C. Szabo et al.

    Endogenously produced hydrogen sulfide supports tumor cell growth and proliferation

    Cell Cycle

    (2013)
  • B. Renga

    Hydrogen sulfide generation in mammals: the molecular biology of cystathionine-β-synthase (CBS) and cystathionine-γ-lyase (CSE)

    Inflamm. Allergy Drug Targets

    (2011)
  • W.J. Cai et al.

    Hydrogen sulfide induces human colon cancer cell proliferation: role of Akt, ERK and p21

    Cell Biol. Int.

    (2010)
  • Q. Cao et al.

    Butyrate-stimulated H2S production in colon cancer cells

    Antioxid. Redox. Signal.

    (2010)
  • K. Ma et al.

    H2S donor, S-propargyl-cysteine, increases CSE in SGC-7901 and cancer-induced mice: evidence for a novel anti-cancer effect of endogenous H2S?

    PLoS One

    (2011)
  • H. Guo et al.

    Characterization of hydrogen sulfide and its synthases, cystathionine β-synthase and cystathionine γ-lyase, in human prostatic tissue and cells

    Urology

    (2012)
  • S. Bhattacharyya et al.

    Cystathionine beta-synthase (CBS) contributes to advanced ovarian cancer progression and drug resistance

    PLoS One

    (2013)
  • S. Maor et al.

    Estrogen receptor regulates insulin-like growth factor-I receptor gene expression in breast tumor cells: involvement of transcription factor Sp1

    J. Endocrinol.

    (2006)
  • Y. Ge et al.

    Transcriptional regulation of the human cystathionine beta-synthase -1b basal promoter: synergistic transactivation by transcription factors NF-Y and Sp1/Sp3

    Biochem. J.

    (2001)
  • Cited by (166)

    View all citing articles on Scopus
    View full text