DOI QR코드

DOI QR Code

Effect of methylsulfonylmethane on oxidative stress and CYP3A93 expression in fetal horse liver cells

  • Kim, Kyoung Hwan (Department of Animal Science, College of Natural Resources and Life Sciences, Pusan National University) ;
  • Park, Jeong-Woong (Department of Animal Science, College of Natural Resources and Life Sciences, Pusan National University) ;
  • Yang, Young Mok (Department of Pathology, School of Medicine, Institute of Biomedical Science and Technology, Konkuk University) ;
  • Song, Ki-Duk (Department of Agriculture Convergence Technology, Jeonbuk National University) ;
  • Cho, Byung-Wook (Department of Animal Science, College of Natural Resources and Life Sciences, Pusan National University)
  • Received : 2020.02.05
  • Accepted : 2020.07.28
  • Published : 2021.02.01

Abstract

Objective: Stress-induced cytotoxicity caused by xenobiotics and endogenous metabolites induces the production of reactive oxygen species and often results in damage to cellular components such as DNA, proteins, and lipids. The cytochrome P450 (CYP) family of enzymes are most abundant in hepatocytes, where they play key roles in regulating cellular stress responses. We aimed to determine the effects of the antioxidant compound, methylsulfonylmethane (MSM), on oxidative stress response, and study the cytochrome P450 family 3 subfamily A (CYP3A) gene expression in fetal horse hepatocytes. Methods: The expression of hepatocyte markers and CYP3A family genes (CYP3A89, CYP3A93, CYP3A94, CYP3A95, CYP3A96, and CYP3A97) were assessed in different organ tissues of the horse and fetal horse liver-derived cells (FHLCs) using quantitative reverse transcription polymerase chain reaction. To elucidate the antioxidant effects of MSM on FHLCs, cell viability, levels of oxidative markers, and gene expression of CYP3A were investigated in H2O2-induced oxidative stress in the presence and absence of MSM. Results: FHLCs exhibited features of liver cells and simultaneously maintained the typical genetic characteristics of normal liver tissue; however, the expression profiles of some liver markers and CYP3A genes, except that of CYP3A93, were different. The expression of CYP3A93 specifically increased after the addition of H2O2 to the culture medium. MSM treatment reduced oxidative stress as well as the expression of CYP3A93 and heme oxygenase 1, an oxidative marker in FHLCs. Conclusion: MSM could reduce oxidative stress and hepatotoxicity in FHLCs by altering CYP3A93 expression and related signaling pathways.

Keywords

References

  1. Guengerich FP. Cytochrome P450s and other enzymes in drug metabolism and toxicity. AAPS J 2006;8:E101-11. https://doi.org/10.1208/aapsj080112
  2. Nelson DR. Cytochrome P450 nomenclature, 2004. Methods Mol Biol 2006;320:1-10. https://doi.org/10.1385/1-59259-998-2:1
  3. Zanger UM, Schwab M. Cytochrome P450 enzymes in drug metabolism: regulation of gene expression, enzyme activities, and impact of genetic variation. Pharmacol Ther 2013;138:103-41. https://doi.org/10.1016/j.pharmthera.2012.12.007
  4. Ding X, Kaminsky LS. Human extrahepatic cytochromes P450: function in xenobiotic metabolism and tissue-selective chemical toxicity in the respiratory and gastrointestinal tracts. Annu Rev Pharmacol Toxicol 2003;43:149-73. https://doi.org/10.1146/annurev.pharmtox.43.100901.140251
  5. Schmitz A, Zielinski J, Dick B, Mevissen M. In vitro metabolism of testosterone in the horse liver and involvement of equine CYPs 3A89, 3A94 and 3A95. J Vet Pharmacol Ther 2014;37:338-47. https://doi.org/10.1111/jvp.12106
  6. Nakayama SMM, Ikenaka Y, Hayami A, et al. Characterization of equine cytochrome P450: role of CYP3A in the metabolism of diazepam. J Vet Pharmacol Ther 2016;39:478-87. https://doi.org/10.1111/jvp.12303
  7. Liu YT, Hao HP, Liu CX, Wang GJ, Xie HG. Drugs as CYP3A probes, inducers, and inhibitors. Drug Metab Rev 2007;39:699-721. https://doi.org/10.1080/03602530701690374
  8. Schmitz A, Demmel S, Peters LM, Leeb T, Mevissen M, Haase B. Comparative human-horse sequence analysis of the CYP3A subfamily gene cluster. Anim Genet 2010;41(Suppl 2):72-9. https://doi.org/10.1111/j.1365-2052.2010.02111.x
  9. Deavall DG, Martin EA, Horner JM, Roberts R. Drug-induced oxidative stress and toxicity. J Toxicol 2012;2012:645460. https://doi.org/10.1155/2012/645460
  10. Mishin V, Heck DE, Laskin DL, Laskin JD. Human recombinant cytochrome P450 enzymes display distinct hydrogen peroxide generating activities during substrate independent NADPH oxidase reactions. Toxicol Sci 2014;141:344-52. https://doi.org/10.1093/toxsci/kfu133
  11. Zangar RC, Davydov DR, Verma S. Mechanisms that regulate production of reactive oxygen species by cytochrome P450. Toxicol Appl Pharmacol 2004;199:316-31. https://doi.org/10.1016/j.taap.2004.01.018
  12. Kensler TW, Wakabayashi N. Nrf2: friend or foe for chemoprevention? Carcinogenesis 2010;31:90-9. https://doi.org/10.1093/carcin/bgp231
  13. Loboda A, Damulewicz M, Pyza E, Jozkowicz A, Dulak J. Role of Nrf2/HO-1 system in development, oxidative stress response and diseases: an evolutionarily conserved mechanism. Cell Mol Life Sci 2016;73:3221-47. https://doi.org/10.1007/s00018-016-2223-0
  14. Butawan M, Benjamin RL, Bloomer RJ. Methylsulfonylmethane: applications and safety of a novel dietary supplement. Nutrients 2017;9:290. https://doi.org/10.3390/nu9030290
  15. Mohammadi S, Najafi M, Hamzeiy H, et al. Protective effects of methylsulfonylmethane on hemodynamics and oxidative stress in monocrotaline-induced pulmonary hypertensive rats. Adv Pharmacol Pharm Sci 2012;2012:507278. https://doi.org/10.1155/2012/507278
  16. Tyden E, Lofgren M, Hakhverdyan M, Tjalve H, Larsson P. The genes of all seven CYP3A isoenzymes identified in the equine genome are expressed in the airways of horses. J Vet Pharmacol Ther 2013;36:370-5. https://doi.org/10.1111/jvp.12012
  17. DiMaio Knych HK, DeStefano Shields C, Buckpitt AR, Stanley SD. Equine cytochrome P450 2C92: cDNA cloning, expression and initial characterization. Arch Biochem Biophys 2009;485:49-55. https://doi.org/10.1016/j.abb.2009.02.009
  18. DiMaio Knych HK, McKemie DS, Stanley SD. Molecular cloning, expression, and initial characterization of members of the CYP3A family in horses. Drug Metab Dispos 2010;38:1820-7. https://doi.org/10.1124/dmd.110.032953
  19. Zabulica M, Srinivasan RC, Vosough M, et al. Guide to the assessment of mature liver gene expression in stem cell-derived hepatocytes. Stem Cells Dev 2019;28:907-19. https://doi.org/10.1089/scd.2019.0064
  20. Lee SJ, Hedstrom OR, Fischer K, et al. Immunohistochemical localization and differential expression of cytochrome P450 3A27 in the gastrointestinal tract of rainbow trout. Toxicol Appl Pharmacol 2001;177:94-102. https://doi.org/10.1006/taap.2001.9289
  21. Cotreau MM, von Moltke LL, Beinfeld MC, Greenblatt DJ. Methodologies to study the induction of rat hepatic and intestinal cytochrome P450 3A at the mRNA, protein, and catalytic activity level. J Pharmacol Toxicol Methods 2000;43:41-54. https://doi.org/10.1016/S1056-8719(00)00086-1
  22. Zhao H, Kalivendi S, Zhang H, et al. Superoxide reacts with hydroethidine but forms a fluorescent product that is distinctly different from ethidium: potential implications in intracellular fluorescence detection of superoxide. Free Radic Biol Med 2003;34:1359-68. https://doi.org/10.1016/S0891-5849(03)00142-4
  23. Tyden E, Lofgren M, Pegolo S, Capolongo F, Tjalve H, Larsson P. Differential gene expression of CYP3A isoforms in equine liver and intestines. J Vet Pharmacol Ther 2012;35:588-95. https://doi.org/10.1111/j.1365-2885.2012.01379.x
  24. Sp N, Kang DY, Kim DH, et al. Methylsulfonylmethane inhibits cortisol-induced stress through p53-mediated SDHA/HPRT1 expression in racehorse skeletal muscle cells: a primary step against exercise stress. Exp Ther Med 2020;19:214-22. https://doi.org/10.3892/etm.2019.8196
  25. Withee ED, Tippens KM, Dehen R, Tibbitts D, Hanes D, Zwickey H. Effects of methylsulfonylmethane (MSM) on exercise-induced oxidative stress, muscle damage, and pain following a half-marathon: a double-blind, randomized, placebo-controlled trial. J Int Soc Sports Nutr 2017;14:24. https://doi.org/10.1186/s12970-017-0181-z

Cited by

  1. Conjugation with Methylsulfonylmethane Improves Hyaluronic Acid Anti-Inflammatory Activity in a Hydrogen Peroxide-Exposed Tenocyte Culture In Vitro Model vol.21, pp.21, 2020, https://doi.org/10.3390/ijms21217956