Next Article in Journal
The Role of Branch Cell Symmetry and Other Critical Nanoscale Design Parameters in the Determination of Dendrimer Encapsulation Properties
Next Article in Special Issue
Role of 3-Mercaptopyruvate Sulfurtransferase in the Regulation of Proliferation and Cellular Bioenergetics in Human Down Syndrome Fibroblasts
Previous Article in Journal
Meridianins and Lignarenone B as Potential GSK3β Inhibitors and Inductors of Structural Neuronal Plasticity
Previous Article in Special Issue
Multidirectional Changes in Parameters Related to Sulfur Metabolism in Frog Tissues Exposed to Heavy Metal-Related Stress
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Editorial

H2S, Polysulfides, and Enzymes: Physiological and Pathological Aspects

by
Noriyuki Nagahara
1,*,† and
Maria Wróbel
2,*,†
1
Nippon Medical School, Isotope Research Institute, 1-1-5 Sendagi, Bunkyo-ku, Tokyo 113-8602, Japan
2
Faculty of Medicine, Jagiellonian University Medical College, Kopernika 7 Cracow, 31-034 Krakow, Poland
*
Authors to whom correspondence should be addressed.
These authors contributed equally to this paper.
Biomolecules 2020, 10(4), 640; https://doi.org/10.3390/biom10040640
Submission received: 10 April 2020 / Accepted: 17 April 2020 / Published: 21 April 2020

Abstract

:
We have been studying the general aspects of the functions of H2S and polysulfides, and the enzymes involved in their biosynthesis, for more than 20 years. Our aim has been to elucidate novel physiological and pathological functions of H2S and polysulfides, and unravel the regulation of the enzymes involved in their biosynthesis, including cystathionine β-synthase (EC 4.2.1.22), cystathionine γ-lyase (EC 4.4.1.1), thiosulfate sulfurtransferase (rhodanese, EC 2.8.1.1), and 3-mercaptopyruvate sulfurtransferase (EC 2.8.1.2). Physiological and pathological functions, alternative biosynthetic processes, and additional functions of H2S and polysulfides have been reported. Further, the structure and reaction mechanisms of related enzymes have also been reported. We expect this issue to advance scientific knowledge regarding the detailed functions of H2S and polysulfides as well as the general properties and regulation of the enzymes involved in their metabolism. We would like to cover four topics: the physiological and pathological functions of H2S and polysulfides, the mechanisms of the biosynthesis of H2S and polysulfides, the properties of the biosynthetic enzymes, and the regulation of enzymatic activity. The knockout mouse technique is a useful tool to determine new physiological functions, especially those of H2S and polysulfides. In the future, we shall take a closer look at symptoms in the human congenital deficiency of each enzyme. Further studies on the regulation of enzymatic activity by in vivo substances may be the key to finding new functions of H2S and polysulfides.

1. Enzyme Production of H2S and Polysulfides

Cystathionine β-synthase (CBS) was first reported to produce H2S and polysulfides by Abe and Kimura in 1996 [1]. Further, cystathionine γ-lyase (CGL) was reported by Hosoki et al. [2], and 3-mercaptopyruvate sulfurtransferase (MST) was reported by Shibuya et al. [3,4,5], Mikami et al. [6,7], Modin et al. [8], Yadav et al. [9], Kimura et al. [10], and Nagahara et al. [11]. Thiosulfate sulfurtransferase (TST) was reported by Mikami et al. [7] and Kimura et al. [10]. These enzymes catalyze a transsulfuration reaction from a sulfur-donor substrate to a sulfur acceptor substrate. Then, the persulfurated or polysulfurated substrate is reduced to produce H2S and polysulfides during this reaction.
On the other hand, Nagahara et al. [11] recently demonstrated in vitro that MST transfers a sulfur atom from 3-mercptopyruvate (MP) to the catalytic site cysteine to form stable persulfide (polysulfide) as a reaction intermediate. It is interesting that as an alternative production process, thiol-containing compounds attack the persulfide (polysulfide) formed at the catalytic site and a new persulfide (polysulfide) molecule is formed at the thiol-containing compound. Then, dithiol is reduced by thioredoxin (Trx) or dihydrolipoic acid to release H2S or polysufides. This process may be autoreduction. Yadav et al. [10] performed enzyme kinetics analysis for human MST in the production process of hydrogen disulfide.

2. Physiological Functions of H2S and Polysulfides

Kimura reviewed the physiological activities of H2S and polysulfides in 2016 [12]. The functions of H2S and polysulfides are summarized in Table 1 and Table 2, respectively, [1,2,13,14,15,16,17,18,19,20,21,22,23,24,25,26,27,28,29,30,31,32].
Sulfane-sulfur binding proteins (SSBPs), ubiquitous in cells and tissues, can be regarded as potentially H2S-releasing molecules when under proper redox conditions and affected by specific stimuli that induce their release. Three sulfurtransferases, i.e., CGL, MST, and TST, carry sulfane sulfur, which can be released as H2S/HS [33]. The released H2S can be enzymatically oxidized to sulfane sulfur by sulfide quinone oxidoreductase with an acceptor of sulfane sulfur, such as GSH [34].
To understand the physiological function of sulfane sulfur, its levels in biological samples (tissues, cell cultures) were determined using the reaction with cyanide, and subsequently investigating the thiocyanate yield of the complex with Fe3+, which is detectable by spectrophotometry [35]. Although the method is not very sensitive, it showed that sulfane sulfur levels were quite similar in various animal tissues [36,37] and in murine macrophages despite stimulation with lipopolysaccharide and interferon-γ [38]. Thus, an argument may be put forward about homeostasis of sulfane sulfur levels in biological systems. Moreover, a negative feedback regulation between CBS and CTH was suggested by Nandi and Mishra [39] and confirmed by Bronowicka et al. [38]. The adaptive cellular response to stimulation with both IFNγ and LPS caused a decreased level of H2S-associated low CBS expression and increased CTH expression.
The adaptive cellular response to electrophiles (electron-deficient species) represented by heavy metal ions [40] involves sulfane sulfur atoms of numerous SSBPs. Protection against electrophilic stresses involves persulfides rather than thiol groups because of their higher nucleophilicity [41]. Electrophiles are captured by reactive persulfide/polysulfide species, resulting in formation of their sulfur adducts [42]. In bovine aortic endothelial cells, CSE knockdown potentiated Cd-induced cytotoxicity but CSE overexpression provided protection [43]. In vivo experiments showed that CSE-knockout mice were sensitive to Cd-induced hepatotoxicity [44]. Adaptive changes in the activity and expression of CGL, MST, and TST in various frog tissues in response to exposure to lead, mercury, and cadmium confirmed the protective function of these enzymatic proteins against electrophilic stress [45,46].

3. Possible Production of Other Sulfur-Containing Substances, Sulfur Oxides

Physiological roles of sulfur dioxide were reported by Liu et al. [47] in 2016 and include vasorelaxation [48,49,50] as well as myocardial injury [48]. MALDI-TOF-MS analysis provided supporting evidence for sulfur oxide production (SO, SO2, and SO3) in the redox cycle of sulfane sulfur as a reaction intermediate of MST by Nagahara et al. [51] in 2012. The persulfurated catalytic site cysteine was oxidized to form Cys-thiosulfenate (Cys-Sγ-SO), Cys-thiosulfinate (Cys-Sγ-SO2), and Cys-thiosulfonate (Cys-Sγ-SO3). Reducing agents such as DTT and Trx produce sulfur oxides [51].

4. Knockout of H2S and Polysulfides-Producing Enzymes

Knockout (KO) technique is a good tool to clarify the physiological functions of proteins. Congenial deficiency of CBS causes hyperhomocysteinemia or homocystinuria in humans. Watanabe et al. produced CBS-KO mice [52], and the mice were afflicted with chronic renal dysfunction. The mice showed growth retardation and died within 5 weeks.
Congenial deficiency of CGL causes cystathioninuria in humans. CGL-KO mice were produced by Yang et al. [53], and the mice displayed low levels of H2S associated with hypertension.
Congenial deficiency of MST causes mercaptolactate-cysteine disulfidria in humans. MST-KO mice were produced by Nagahara et al. [54]; however, mercaptolactate-cysteine disulfiduria has not been examined. Peleli et al. [55] recently reported that MST-KO mice were protected against ischemic reperfusion of the heart. Nasi et al. [56] reported that mice showed accelerated joint calcification and osteoarthritis due to an increase in chondrocyte mineralization. Interestingly, these two findings indicate that MST demonstrates both good and bad effects on living organisms.
TST-KO mice have not been obtained; however, a double KO mouse for both TST and MST has been produced (unpublished data).

5. Regulation of Enzymatic Activity by In Vivo Substances

These four enzymes have been reported to regulate enzymatic activities in vivo. CBS activity is inhibited by CO [57], NO [58], L-cystathione [59], and L-homocysteine [60]. On the other hand, CBS activation by in vivo substances has not been reported. CGL activity is inhibited by acetoacetate [61], alanine [62], cysteine [63], glycine [62], serine [62], Cd2+ [64], Cu2+ [64], H2O2 [61], and O2 [65]. It is interesting that oxidative stress inhibits CBS activity. However, CGL is activated by L-cysteine [66] and 2-mercaptoethanol (probably other reducing agents also reactivate) [62]. Thus, reducing conditions reactivate CBS. TST activity is inhibited by Ca2+ [67], Zn2+ [67], Cu2+ [68], oxaloacetate [69], pyruvate [69], H2S [70], SO32− [67], SO42− [69], sulfide [71], sulfite [72], and H2O2 [73]. Oxidative stress also inhibits TST activity. On the other hand, TST is activated by L-cysteine [74], glutathione [75], and reduced glutathione [75,76]. Thus, reducing conditions also reactivate TST. MST activity is inhibited by alpha-ketobutyrate [77], alpha-ketoglutarate [77], pyruvate [78], cysteine [78], sulfite [6], glutathione [78], and H2O2 [79]. Oxidative stress also inhibits MST activity, while MST is activated by thioredoxin [79,80]. Thus, reducing conditions also reactivate MST. In the three enzymes, these enzymatic activities are regulated by the redox state.

6. Conclusions

Four cysteine-containing enzymes (CBS, CGL, MST, and TST) produce H2S and polysulfides via the reduction of persulfurated or polysulfurated substrates. MST is also produced via the reduction of stable persulfide (polysulfide) formed at a catalytic-site cysteine as a reaction intermediate. Sulfur oxide can also be produced from a catalytic site cysteine of MST. These products play an important role in living organisms. Furthermore, studies using KO mice of each enzyme have clarified the physiological function of these enzymes, which cannot be assessed in wild-type animals. Regulation of enzymatic activity by in vivo substances may be related to the functions of H2S and polysulfides.

Author Contributions

These authors contributed equally to this work. All authors have read and agreed to the published version of the manuscript.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Abe, K.; Kimura, H. The possible role of hydrogen sulfide as an endogenous neuromodulator. J. Neurosci. 1996, 16, 1066–1071. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Hosoki, R.; Matsuki, N.; Kimura, H. The possible role of hydrogen sulfide as an endogenous smooth muscle relaxant in synergy with nitric oxide. Biochem. Biophys. Res. Commun. 1997, 237, 527–531. [Google Scholar] [CrossRef] [PubMed]
  3. Shibuya, N.; Tanaka, M.; Yoshida, M.; Ogasawara, Y.; Togawa, T.; Ishii, K.; Kimura, H. 3-Mercaptopyruvate sulfurtransferase produces hydrogen sulfide and bound sulfane sulfur in the brain. Antioxid. Redox Signal. 2009, 11, 703–714. [Google Scholar] [CrossRef] [PubMed]
  4. Shibuya, N.; Mikami, Y.; Kimura, Y.; Nagahara, N.; Kimura, H. Vascular endothelium expresses 3-mercaptopyruvate sulfurtransferase and produces hydrogensulfide. J. Biochem. 2009, 146, 623–626. [Google Scholar] [CrossRef]
  5. Shibuya, N.; Koike, S.; Tanaka, M.; Ishigami-Yuasa, M.; Kimura, Y.; Ogasawara, Y.; Fukui, K.; Nagahara, N.; Kimura, H. A novel pathway for the production of hydrogen sulfide from D-cysteine in mammalian cells. Nat. Commun. 2013, 4, 1366. [Google Scholar] [CrossRef] [Green Version]
  6. Mikami, Y.; Shibuya, N.; Kimura, Y.; Nagahara, N.; Ogasawara, Y.; Kimura, H. Thioredoxin and dihydrolipoic acid are required for 3-mercaptopyruvate sulfurtransferase to produce hydrogen sulfide. Biochem. J. 2011, 439, 479–485. [Google Scholar] [CrossRef] [Green Version]
  7. Mikami, Y.; Shibuya, N.; Kimura, Y.; Nagahara, N.; Yamada, M.; Kimura, H. Hydrogen sulfide protects the retina from light-induced degeneration by the modulation of Ca21 influx. J. Biol. Chem. 2011, 286, 39379–39386. [Google Scholar] [CrossRef] [Green Version]
  8. Modis, K.; Asimakopoulou, A.; Colettal, C.; Papapetropoulos, A.; Szabo, C. Oxidative stress suppresses the cellular bioenergetic effect of the 3-mercaptopyruvate sulfurtransferase/hydrogen sulfide pathway. Biochem. Biophys. Res. Commun. 2013, 433, 401–407. [Google Scholar] [CrossRef]
  9. Yadav, P.K.; Yamada, K.; Chiku, T.; Koutmos, M.; Banerjee, R. Structure and kinetic analysis of H2S production by human mercaptopyruvate sulfurtransferase. J. Biol. Chem. 2013, 288, 20002–20013. [Google Scholar] [CrossRef] [Green Version]
  10. Kimura, Y.; Toyofuku, Y.; Koike, S.; Shibuya, N.; Nagahara, N.; Lefer, D.; Ogasawara, Y.; Kimura, H. Identification of H2S3 and H2S produced by 3-mercaptopyruvate sulfurtransferase in the brain. Sci. Rep. 2015, 5, 14774. [Google Scholar] [CrossRef] [Green Version]
  11. Nagahara, N.; Koike, S.; Nirasawa, T.; Kimura, H.; Ogasawara, Y. Alternative pathway of H2S and polysulfides production from sulfurated catalytic-cysteine of reaction intermediates of 3-mercaptopyruvate sulfurtransferase. Biochem. Biophys. Res. Commun. 2018, 496, 648–653. [Google Scholar] [CrossRef] [PubMed]
  12. Kimura, H. Physiological roles of hydrogen sulfide and polysulfides. Folia Pharm. Jpn. 2016, 147, 23–29. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Kimura, Y.; Kimura, H. Hydrogen sulfide protects neurons from oxidative stress. FASEB J. 2004, 8, 1165–1167. [Google Scholar] [CrossRef] [PubMed]
  14. Yang, W.; Yang, G.; Jia, X.; Wu, L.; Wang, R. Activation of KATP channels by H2S in rat insulin-secreting cells and the underlying mechanisms. J. Physiol. 2005, 569, 519–531. [Google Scholar] [CrossRef] [PubMed]
  15. Kaneko, Y.; Kimura, Y.; Kimura, H.; Niki, I. L-Cysteine inhibits insulin release from the pancreatic β-cell. Diabetes 2006, 55, 1391–1397. [Google Scholar] [CrossRef] [Green Version]
  16. Olson, K.R.; Dombkowski, R.A.; Russell, M.J.; Doellman, M.M.; Head, S.K.; Whitfield, N.L.; Madden, J.A. Hydrogen sulfide as an oxygen sensor/transducer in vertebrate hypoxic vasoconstriction and hypoxic vasodilation. J. Exp. Biol. 2006, 209, 4011–4023. [Google Scholar] [CrossRef] [Green Version]
  17. Peng, Y.J.; Nanduri, J.; Raghuraman, G.; Souvannakitti, D.; Gadalla, M.M.; Kumar, G.K.; Snyder, S.H.; Prabhakar, N.R. H2S mediates O2 sensing in the carotid body. Proc. Natl. Acad. Sci. USA 2010, 107, 10719–10724. [Google Scholar] [CrossRef] [Green Version]
  18. Zanardo, R.C.O.; Brancaleone, V.; Distrutti, E.; Fiorucci, S.; Cirino, G.; Wallace, J.L. Hydrogen sulfide is an endogenous modulator of leukocyte-mediated inflammation. FASEB J. 2006, 20, 2118–2120. [Google Scholar] [CrossRef]
  19. Elrod, J.W.; Calvert, J.W.; Morrison, J.; Doeller, J.E.; Kraus, D.W.; Tao, L.; Jiao, X.; Scalia, R.; Kiss, L.; Szabo, C.; et al. Hydrogen sulfide attenuates myocardial ischemia-reperfusion injury by preservation of mitochondrial function. Proc. Natl. Acad. Sci. USA 2007, 104, 15560–15565. [Google Scholar] [CrossRef] [Green Version]
  20. Tripatara, P.; Patel, N.; Collino, M.; Gallicchio, M.; Kieswich, J.; Castiglia, S.; Benetti, E.; Stewart, K.N.; Brown, P.A.; Yaqoob, M.M.; et al. Generation of endogenous hydrogen sulfide by cystathionine γ-lyase limits renal ischemia/reperfusion injury and dysfunction. Lab. Invest. 2008, 88, 1038–1048. [Google Scholar] [CrossRef] [Green Version]
  21. Cai, W.J.; Wang, M.J.; Moore, P.K.; Jin, H.M.; Yao, T.; Zhu, Y.C. The novel proangiogenic effect of hydrogen sulfide is dependent on Akt phosphorylation. Cardiovasc Res. 2007, 76, 29–40. [Google Scholar] [CrossRef]
  22. Papapetropoulos, A.; Pyriochou, A.; Altaany, Z.; Yang, G.; Marazioti, A.; Zhou, Z.; Jeschke, M.G.; Branski, L.K.; Herndon, D.N.; Wang, R.; et al. Hydrogen sulfide is an endogenous stimulator of angiogenesis. Proc. Natl. Acad. Sci. USA 2009, 106, 21972–21977. [Google Scholar] [CrossRef] [Green Version]
  23. Krishnan, N.; Fu, C.; Pappin, D.J.; Tonks, N.K. H2S-induced sulfhydration of the phosphatase PTP1B and its role in the endoplasmic reticulum stress response. Sci. Signal. 2011, 4, ra86. [Google Scholar] [CrossRef] [Green Version]
  24. Shatalin, K.; Shatalina, E.; Mironov, A.; Nudler, E. H2S: A universal defense against antibiotics in bacteria. Science 2011, 334, 986–990. [Google Scholar] [CrossRef]
  25. Kimura, H. Physiological role of hydrogen sulfide and polysulfide in the central nervous system. Neurochem. Int. 2013, 63, 492–497. [Google Scholar] [CrossRef]
  26. Kimura, H. Hydrogen sulfide and polysulfides as signaling molecules. Proc. Jpn. Acad. Ser. B 2015, 91, 131–159. [Google Scholar] [CrossRef] [Green Version]
  27. Nagai, Y.; Tsugane, M.; Oka, J.I.; Kimura, H. Hydrogen sulfide induces calcium waves in astrocytes. FASEB J. 2004, 18, 557–559. [Google Scholar] [CrossRef] [PubMed]
  28. Kimura, Y.; Mikami, Y.; Osumi, K.; Tsugane, M.; Oka, J.; Kimura, H. Polysulfides are possible H2S-derived signaling molecules in rat brain. FASEB J. 2013, 27, 2451–2457. [Google Scholar] [CrossRef] [PubMed]
  29. Greiner, R.; Pálinkás, Z.; Bäsell, K.; Becher, D.; Antelmann, H.; Nagy, P.; Dick, T.P. Polysulfides link H2S to protein thiol oxidation. Antioxid Redox Signal. 2013, 19, 1749–1765. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  30. Koike, S.; Ogasawara, Y.; Shibuya, N.; Kimura, H.; Ishii, K. Polysulfide exerts a protective effect against cytotoxicity caused by t-buthylhydroperoxide through Nrf2 signaling in neuroblastoma cells. FEBS Lett. 2013, 587, 3548–3555. [Google Scholar] [CrossRef] [Green Version]
  31. Koike, S.; Shibuya, N.; Kimura, H.; Ishii, K.; Ogasawara, Y. Polysulfide promotes neuroblastoma cell differentiation by accelerating calcium influx. Biochem. Biophys.Res. Commun. 2015, 459, 488–492. [Google Scholar] [CrossRef] [PubMed]
  32. Koike, S.; Kayama, T.; Yamamoto, S.; Komine, D.; Tanaka, R.; Nishimoto, S.; Suzuki, T.; Kishida, A.; Ogasawara, Y. Polysulfides protect SH-SY5Y cells from methylglyoxal-induced toxicity by suppressing protein carbonylation: A possible physiological scavenger for carbonyl stress in the brain. NeuroToxicol 2016, 55, 13–19. [Google Scholar] [CrossRef] [PubMed]
  33. Toohey, J.I. Sulphane sulphur in biological systems: A possible regulatory role. Biochem. J. 1989, 264, 625–632. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Libiad, M.; Yadav, P.K.; Vitvitsky, V.; Martinov, M.; Banerjee, R. Organization of the human mitochondrial hydrogen sulfide oxidation pathway. J. Biol. Chem. 2014, 289, 30901–30910. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  35. Wood, J.L. Sulfane sulfur. Methods Enzymol. 1987, 143, 25–29. [Google Scholar] [PubMed]
  36. Wróbel, M.; P Sura, Z.; Srebro, Z. Sulfurtransferases and the content of cysteine, glutathione and sulfane sulfur in tissues of the frog Rana temporaria. Comp. Biochem. Physiol. Part B 2000, 125, 211–217. [Google Scholar] [CrossRef]
  37. Wróbel, M.; Czubak, J.; Bronowicka-Adamska, P.; Jurkowska, H.; Adamek, D.; Papla, B. Is development of high-grade gliomas sulfur-dependent. Molecules 2014, 19, 21350–21362. [Google Scholar] [CrossRef] [Green Version]
  38. Bronowicka-Adamska, P.; Jurkowska, H.; Gawda, A.; Skalska, P.; Nazimek, K.; Marcinkiewicz, J.; Wróbel, M. Expression and activity of hydrogen sulfide generating enzymes in murine macrophages stimulated with lipopolysaccharide and interferon-γ. Mol. Biol. Rep. 2019, 46, 2791–2798. [Google Scholar] [CrossRef] [Green Version]
  39. Nandi, S.S.; Mishra, P.K. H2S and homocysteine control a novel feedback regulation of cystathionine beta synthase and cystathionine gamma lyase in cardiomyocytes. Sci. Rep. 2017, 7, 3639. [Google Scholar] [CrossRef]
  40. Nishida, M.; Kumagai, Y.; Ihara, H.; Fujii, S.; Motohashi, H.; Akaike, T. Redox signaling regulated by electrophiles and reactive sulfur species. J. Clin. Biochem. Nutr. 2016, 58, 91–98. [Google Scholar] [CrossRef] [Green Version]
  41. Yasuhiro, S.; Yoshito, K. Sulfane Sulfur in Toxicology: A Novel Defense System Against Electrophilic Stress. Toxicol. Sci. 2019, 170, 3–9. [Google Scholar]
  42. Nishida, M.; Sawa, T.; Kitajima, N.; Ono, K.; Inoue, H.; Ihara, H.; Motohashi, H.; Yamamoto, M.; Suematsu, M.; Kurose, H.; et al. Hydrogen sulfide anion regulates redox signaling via electrophile sulfhydration. Nat. Chem. Biol. 2012, 8, 714–724. [Google Scholar] [CrossRef] [PubMed]
  43. Shinkai, Y.; Masuda, A.; Akiyama, M.; Xian, M.; Kumagai, Y. Cadmium-mediated activation of the HSP90/HSF1 pathway regulated by reactive persulfides/polysulfides. Toxicol. Sci. 2017, 156, 412–421. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Akiyama, M.; Shinkai, Y.; Unoki, T.; Shim, I.; Ishii, I.; Kumagai, Y. The capture of cadmium by reactive polysulfides attenuates cadmium-induced adaptive responses and hepatotoxicity. Chem. Res. Toxicol. 2017, 30, 2209–2217. [Google Scholar] [CrossRef]
  45. Kaczor, M.; Sura, P.; Bronowicka-Adamska, P.; Wróbel, M. Exposure to lead in water and cysteine non-oxidative metabolism in Pelophylax ridibundus tissues. Aquat. Toxicol. 2013, 127, 72–77. [Google Scholar] [CrossRef]
  46. Kaczor, M.; Sura, P.; Wróbel, M. Changes in activity of three sulfurtransferases in response to exposure to cadmium, lead and mercury ions. J. Environ. Prot. 2013, 4, 19–28. [Google Scholar] [CrossRef] [Green Version]
  47. Liu, J.; Huang, Y.; Chen, S.; Tang, C.; Jin, H.; Du, J. Role of endogenous sulfur dioxide in regulating vascular structural remodeling in hypertension. Oxid. Med. Cell Longev. 2016, 2016, 4529060. [Google Scholar] [CrossRef] [Green Version]
  48. Du, S.X.; Jin, H.F.; Bu, D.F.; Zhao, X.; Geng, B.; Tang, C.S.; Du, J.B. Endogenously generated sulfur dioxide and its vasorelaxant effect in rats. Acta. Pharm. Sin. 2008, 29, 923–930. [Google Scholar] [CrossRef] [Green Version]
  49. Meng, Z.; Zhang, H. The vasodilator effect and its mechanism of sulfur dioxide-derivatives on isolated aortic rings of rats. Inhal. Toxicol. 2007, 19, 979–986. [Google Scholar] [CrossRef]
  50. Wang, Y.K.; Ren, A.J.; Yang, X.Q.; Wang, L.G.; Rong, W.F.; Tang, C.S.; Yuan, W.J.; Lin, L. Sulfur dioxide relaxes rat aorta by endothelium-dependent and -independent mechanisms. Physiol. Res. 2009, 58, 521–527. [Google Scholar]
  51. Nagahara, N.; Nirasawa, T.; Yoshii, T.; Niimura, Y. Is novel signal transducer sulfur oxide involved in the redox cycle of persulfide at the catalytic site cysteine in a stable reaction intermediate of mercaptopyruvate sulfurtransferase? Antioxid Redox Signal. 2012, 16, 747–753. [Google Scholar] [CrossRef] [PubMed]
  52. Watanabe, M.; Osada, J.; Aratani, Y.; Kluckman, K.; Reddick, R.; Malinow, M.R.; Maeda, N. Mice deficient in cystathionine γ-synthase: Animal models for mild and severe homocyst(e)inemia. Proc. Natl. Acad. Sci. USA 1995, 92, 1585–1589. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  53. Yang, G.; Wu, L.; Jiang, B.; Yang, W.; Qi, J.; Cao, K.; Meng, Q.; Mustafa, A.K.; Mu, W.; Zhang, S.; et al. H2S as a physiologic vasorelaxant: Hypertension in mice with deletion of cystathionine gamma-lyase. Science 2008, 322, 587–590. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Nagahara, N.; Nagano, M.; Ito, T.; Shimamura, K.; Akimoto, T.; Suzuki, H. Antioxidant enzyme, 3-mercaptopyruvate sulfurtransferase-knockout mice exhibit increased anxiety-like behaviors: A model for human mercaptolactatecysteine disulfiduria. Sci. Rep. 2013, 3, 1986. [Google Scholar] [CrossRef] [Green Version]
  55. Peleli, M.; Bibli, S.I.; Li, Z.; Chatzianastasiou, A.; Varela, A.; Katsouda, A.; Zukunft, S.; Bucci, M.; Vellecco, V.; Davos, C.H.; et al. Cardiovascular phenotype of mice lacking 3-mercaptopyruvate sulfurtransferase. Biochem. Pharm. 2020. in print. [Google Scholar] [CrossRef]
  56. Nasi, S.; Ehirchiou, D.; Chatzianastasiou, A.; Nagahara, N.; Papapetropoulos, A.; Bertrand, J.; Cirino, G.; So, A.; Busso, B. The protective role of the 3-mercaptopyruvate sulfurtransferase (3-MST)-hydrogen sulfide (H2S) pathway against experimental osteoarthritis. Arthritis Res. 2020. in print. [Google Scholar] [CrossRef] [Green Version]
  57. Taoka, S.; West, M.; Banerjee, R. Characterization of the heme and pyridoxal phosphate cofactors of human cystathionine beta-synthase reveals nonequivalent active sites. Biochemistry 1999, 38, 2738–2744. [Google Scholar] [CrossRef]
  58. Taoka, S.; Banerjee, R. Characterization of NO binding to human cystathionine beta-synthase: Possible implications of the effects of CO and NO binding to the human enzyme. J. Inorg. Biochem. 2001, 87, 245–251. [Google Scholar] [CrossRef]
  59. Aitken, S.M.; Kirsch, J.F. Kinetics of the yeast cystathionine beta-synthase forward and reverse reactions: Continuous assays and the equilibrium constant for the reaction. Biochemistry 2003, 42, 571–578. [Google Scholar] [CrossRef]
  60. Belew, M.S.; Quazi, F.I.; Willmore, W.G.; Aitken, S.M. Kinetic characterization of recombinant human cystathionine beta-synthase purified from E. coli. Protein Expr. Purif. 2009, 64, 139–145. [Google Scholar] [CrossRef]
  61. Manna, P.; Gungor, N.; McVie, R.; Jain, S.K. Decreased cystathionine-γ-lyase (CSE) activity in livers of type 1 diabetic rats and peripheral blood mononuclear cells (PBMC) of type 1 diabetic patients. J. Biol. Chem. 2014, 289, 11767–11778. [Google Scholar] [CrossRef] [Green Version]
  62. Chu, L.; Ebersole, J.L.; Kurzban, G.P.; Holt, S.C. Cystalysin, a 46-kDa L-cysteine desulfhydrase from Treponema denticola: Biochemical and biophysical characterization. Clin. Infect. Dis. 1999, 28, 442–450. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Braunstein, A.E.; Goryachenkova, E.V. The beta-replacement-specific pyridoxal-P-dependent lyases. Adv. Enzym. Relat. Areas Mol. Biol. 1984, 56, 1–89. [Google Scholar]
  64. Matsuo, Y.; Greenberg, D.M. A crystalline enzyme that cleaves homoserine and cystathionine. III. Coenzyme resolution, activators, and inhibitors. J. Biol. Chem. 1959, 234, 507–515. [Google Scholar] [PubMed]
  65. Olson, K.R.; Healy, M.J.; Qin, Z.; Skovgaard, N.; Vulesevic, B.; Duff, D.W.; Whitfield, N.L.; Yang, G.; Wang, R.; Perry, S.F. Hydrogen sulfide as an oxygen sensor in trout gill chemoreceptors. Am. J. Physiol. Regul. Integr. Comp. Physiol. 2008, 29, R669–R680. [Google Scholar] [CrossRef] [Green Version]
  66. Wróbel, M.; Lewandowska, I.; Bronowicka-Adamska, P.; Paszewski, A. The level of sulfane sulfur in the fungus Aspergillus nidulans wild type and mutant strains. Amino Acids 2009, 37, 565–571. [Google Scholar] [CrossRef] [PubMed]
  67. Westley, J. Rhodanese. Adv. Enzym. Relat. Areas Mol. Biol. 1973, 39, 327–368. [Google Scholar]
  68. Lee, C.Y.; Hwang, J.H.; Lee, Y.S.; Cho, K.S. Purification and characterization of mouse liver rhodanese. J. Biochem. Mol. Biol. 1995, 28, 170–176. [Google Scholar]
  69. Oi, S. Inhibition of rat liver rhodanese by di-, tricarboxylic, and alpha-keto acids. J. Biochem. 1975, 78, 825–834. [Google Scholar] [CrossRef]
  70. Picton, R.; Eggo, M.C.; Merrill, G.A.; Langman, M.J.; Singh, S. Mucosal protection against sulphide: Importance of the enzyme rhodanese. Gut 2002, 50, 201–205. [Google Scholar] [CrossRef] [Green Version]
  71. Ramasamy, S.; Singh, S.; Taniere, P.; Langman, M.J.; Eggo, M.C. Sulfide-detoxifying enzymes in the human colon are decreased in cancer and upregulated in differentiation. Am. J. Physiol. Gastrointest Liver Physiol. 2006, 291, G288–G296. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  72. Vazquez, E.; Gazzaniga, S.; Polo, C.; Batlle, A. Mitochondrial and cytosolic rhodanese from liver of DAB-treated mice. III. Inhibition kinetic studies. Cancer Biochem. Biophys. 1997, 15, 285–293. [Google Scholar] [PubMed]
  73. Nandi, D.L.; Horowitz, P.M.; Westley, J. Rhodanese as a thioredoxin oxidase. Int J. Biochem. Cell Biol. 2000, 32, 465–473. [Google Scholar] [CrossRef]
  74. Anosike, E.O.; Jack, A.S. A comparison of some biochemical properties of liver thiosulphate sulphurtransferase from guinea pig (Lepus caniculus) & albino rat. Indian J. Biochem. Biophys. 1982, 19, 13–16. [Google Scholar]
  75. Turkowsky, A.; Blotevogel, K.H.; Fischer, U. Properties of a soluble thiosulfate sulfur transferase (rhodanese) of the marine methanogen Methanosarcina frisia. FEMS Microbiol. Lett. 1991, 81, 251–256. [Google Scholar] [CrossRef]
  76. Vandenbergh, P.A.; Berk, R.S. Purification and characterization of rhodanese from Acinetobacter calcoaceticus. Can. J. Microbiol. 1980, 26, 281–286. [Google Scholar] [CrossRef]
  77. Porter, D.W.; Baskin, S.I. The effect of three alpha-keto acids on 3-mercaptopyruvate sulfurtransferase activity. J. Biochem. Toxicol. 1996, 11, 45–50. [Google Scholar] [CrossRef]
  78. Vachek, H.; Wood, J.L. Purification and properties of mercaptopyruvate sulfur transferase of Escherichia coli. Biochim. Biophys. Acta 1972, 258, 133–146. [Google Scholar] [CrossRef]
  79. Nagahara, N.; Katayama, A. Post-translational regulation of mercaptopyruvate sulfurtransferase via a low redox potential cysteine-sulfenate in the maintenance of redox homeostasis. J. Biol. Chem. 2005, 280, 34569–34576. [Google Scholar] [CrossRef] [Green Version]
  80. Nagahara, N.; Yoshii, T.; Abe, Y.; Matsumura, T. Thioredoxin-dependent enzymatic activation of mercaptopyruvate sulfurtransferase. An intersubunit disulfide bond serves as a redox switch for activation. J. Biol. Chem. 2007, 282, 1561–1569. [Google Scholar] [CrossRef] [Green Version]
Table 1. Physiological function of H2S.
Table 1. Physiological function of H2S.
FunctionReference
Induction of long-term potentiation in the hippocampus as a synaptic model of memoryAbe and Kimura, 1996 [1]
Effect on smooth muscle relaxant activityHosoki et al., 1997 [2]
Protective action of nerve cells from oxidative stressKimura and Kimura, 2004 [13]
Regulation of insulin secretionYang et al., 2005 [14]; Kaneko et al., 2006 [15]
Oxygen sensorOlson et al., 2006 [16]; Peng et al., 2010 [17]
AntiinfectionZanardo et al., 2006 [18]
Protective action of myocardium and kidney from ischemia reperfusion injuryElrod et al., 2007 [19]; Tripatara et al., 2008 [20]
Angiogenic effectCai et al., 2007 [21]; Papapetropoulos et al., 2009 [22]
Protection of retinal neurons from light-induced damage and apoptosisMikami et al., 2011b [7]
Regulation of endoplasmic reticulum stressKrishnan et al., 2011 [23]
Bacterial resistance against antibioticsShatalin et al., 2011 [24]
Reduction of disulfide bonds in a ligand-binding domain of N-methyl-D-aspartic acid receptorsKimura, 2013 [25]; 2015 [26]
Amplification of the activity of N-methyl-D-aspartic acid receptor upon activation by neurotransmittersKimura, 2015 [26]
Activation of H+-ATPase resulting in decrease of calcium influx into photoreceptor cells of the retinaKimura, 2016 [12]
Table 2. Physiological function of polysulfides.
Table 2. Physiological function of polysulfides.
FunctionReference
Induction of calcium influx by activating a cation channel, subfamily A, and member 1 in astrocytesNagai et al., 2004 [27]; Kimura et al., 2013 [28]
Inhibition of tumor suppressor lipid phosphatase and tensin homolog by changing the protein to its oxidized formGreiner et al., 2013 [29]
Upregulation of antioxidant genes, such as heme oxygenase 1 and glutamate cysteine ligaseKoike et al., 2013 [30]
Induction of long-term potentiation in the hippocampus due to activation of N-methyl-D-aspartic acid receptors Kimura, 2015 [26]
Upregulation of antioxidant genes such as heme oxygenase 1 and glutamate cysteine ligaseKimura, 2015 [26]
Decrease in toxic carbonyl stress in neuroblastoma cellsKoike et al., 2015 [31]
Induction of neural outgrowth and cell differentiation of neuroblastoma cellsKoike et al., 2016 [32]

Share and Cite

MDPI and ACS Style

Nagahara, N.; Wróbel, M. H2S, Polysulfides, and Enzymes: Physiological and Pathological Aspects. Biomolecules 2020, 10, 640. https://doi.org/10.3390/biom10040640

AMA Style

Nagahara N, Wróbel M. H2S, Polysulfides, and Enzymes: Physiological and Pathological Aspects. Biomolecules. 2020; 10(4):640. https://doi.org/10.3390/biom10040640

Chicago/Turabian Style

Nagahara, Noriyuki, and Maria Wróbel. 2020. "H2S, Polysulfides, and Enzymes: Physiological and Pathological Aspects" Biomolecules 10, no. 4: 640. https://doi.org/10.3390/biom10040640

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop