Trichostatin A, a Histone Deacetylase Inhibitor Stimulate CYP3A4 Proximal Promoter Activity in Hepa-I Cells

  • Published : 2004.04.01

Abstract

Cytochrome P450 3A4 (CYP3A4) is the most abundant CYPs in human liver, comprising approximately $30\%$ of the total liver CYPs contents and is involved in the metabolism of more than $60\%$ of currently used therapeutic drugs. However, the molecular mechanisms underly-ing regulation of CYP3A4 gene expression have not been understood. Thus, this study has been carried out to gain the insight of the molecular mechanism of CYP3A4 gene expression, investigating if the histone deacetylation is involved in the regulation of CYP3A4 gene expression by proximal promoter. Also SXR was investigated to see if they were involved in the regulation of CYP3A4 proximal promoter activity. Hepa-1 cells were transfected with a plasmid containing ${\~}1kb$ of the human CYP3A4 proximal promoter region (863 to +64 bp) cloned in front of a reporter gene, luciferase, in the presence or absence of SXR. Transfected cells were treated with CYP3A4 inducers such as rifampicin, PCN and RU 486, in order to examine the regulation of CYP3A4 gene expression in the presence or absence of trichostatin A (TSA). In Hepa-1 cells, CYP3A4 inducers increased modestly the luciferase activity when TSA was co-treated, but this increment was not enhanced by SXR cotransfection. Taken together, these results indicated that the inhibition of histone deacetylation was required to SXR-mediated increase in CYP3A4 proximal promoter region when rifampicin, or PCN was treated. Further a trans-activation by SXR may demand other species-specific transcription factors.

Keywords

References

  1. Butler, L. M., Zhou, X., Xu, W. S., Scher, H. I., Rifkind, R. A., Marks, P. A., and Richon, V. M., The histone deacetylase inhibitor SAHA arrests cancer cell growth, up-regulates thioredoxin-binding protein-2, and down-regulates thioredoxin. Proc. Natl. Acad. Sci. USA, 99,11700-11705 (2002) https://doi.org/10.1073/pnas.182372299
  2. Chen, J. D. and Evans, R. M., A transcriptional co-repressor that interacts with nuclear hormone receptors. Nature, 377, 454-457 (1995) https://doi.org/10.1038/377454a0
  3. Drocourt, L., Pascussi, J. M., Assenat, E., Fabre, J. M., Maurel, P., and Vilarem, M. J., Calcium channel modulators of the dihydropyridine family are human pregnane X receptor activators and inducers of CYP3A, CYP2B, andCYP2C in human HepaItocytes. DrugMetab. Dispos., 29,1325-1331 (2001)
  4. Freiman, R. N. and Tjian, R., Regulating the regulators: lysine modifications maketheirmark. Cell, 112, 11-17 (2003) https://doi.org/10.1016/S0092-8674(02)01278-3
  5. Gibson, G. G., Regulation of the CYP3A4 gene by hydrocortisone andxenobiotics: Role oftheglucocorticoid andpregnane X receptors. DrugMetab. Dispos., 28,493-496 (2000)
  6. Goodwin, B., Hodgson, E., and Liddle, C., The orphan human pregnane X receptor mediates the transcriptional activation of CYP3A4 by rifampicin through a distal enhancer module. Mol. Pharmacal., 56,1329-1339 (1999) https://doi.org/10.1124/mol.56.6.1329
  7. Guengerich, F. P., Cytochrome p-450 3A4: Regulation and role in drug metabolism, Annu. Rev. Pharmacol. Toxicol., 39, 1-17 (1999) https://doi.org/10.1146/annurev.pharmtox.39.1.1
  8. Hashimoto, H., Toide, K., Kitamura, R., Fujita, M., Tagawa, S., Itoh, S., and Kamataki, T., Gene structure of CYP3A4, an adult-specific form of cytochrome P450 in human livers, and its transcriptional control. Eur J. Biochem., 218, 585-595 (1993) https://doi.org/10.1111/j.1432-1033.1993.tb18412.x
  9. Horlein, A. J., Naar, A. M., Heinzel, T., Torchia, J., Gloss, B., Kurokawa, R., Ryan, A., Kamei, Y., Soderstrom, M., Glass, C. K., and Rosenfeld, M. G., Ligand-independent repression by the thyroid hormone receptor mediated by a nuclear receptor co-repressor. Nature, 377,397-404 (1995) https://doi.org/10.1038/377397a0
  10. Jones, S. A, Moore, L. B., Shenk, J. L., Wisely, G. B., Hamilton, G. A, McKee, D. D., Tomkinson, N. C., LeCluyse, E. L., Lambert, M. H., Willson, T. M., Kliewer, S. A, and Moore, J. T, The pregnane X receptor: a promiscuous xenobiotic receptor that has diverged during evolution. Mol. Endocrinol., 14,27-39 (2000) https://doi.org/10.1210/me.14.1.27
  11. Khochbin, S., Verdel, A, Lemercier, C., and Seigneurin-Berny, D., Functional significance of histone deacetylase diversity. Curr Opin. Genet. Dev., 11, 162-166 (2001) https://doi.org/10.1016/S0959-437X(00)00174-X
  12. Kim, J. Y., Ahn, M. R., and Sheen, Y. Y., HDAC inhibitor stimulate CYP3A4 proximal promoter activity in HepG2 cells. Arch. Pharm. Res., 27(4) In press (2004)
  13. Kliewer, S. A., Moore, J. T., Wade, L., Staudinger, J. L., Watson, M. A., Jones, S. A., Mckee, D. D., Oliver, B. B., Willson, T. M., Zetterstrom, R. H., Perlmann, T., and Lehmann, J. M., An orphan nuclear receptor activated by pregnanes defines a novel steroid signaling pathway. Cell, 92, 73-82 (1998) https://doi.org/10.1016/S0092-8674(00)80900-9
  14. Kouzarides, T., Acetylation: a regulatory modification to rival phosphorylation? EMBO J., 19, 1176-1179 (2000) https://doi.org/10.1093/emboj/19.6.1176
  15. Lehmann, J. M., McKee, D. D., Watson, M. A., Willson, T. M., Moore, J. T., and Kliewer, S. A., The human orphan nuclear receptor PXR is activated by compounds that regulate CYP3A4 gene expression and cause drug interactions. J. Clin.lnvest., 102, 1016-1023 (1998) https://doi.org/10.1172/JCI3703
  16. Luo, G., Cunningham, M., Kim S., Burn, T., Lin, J., Sinz, M., Hamilton, G., Rizzo, C., Jolley, S., Gilbert, D., Downey, A., MUdra, D., Graham, R., Carroll, K., Xie, J., Madan, A., Parkinson, A., Christ, D., Selling, B., Lecluyse, E., and Gan, L., CYP3A4 induction by drugs: Correlation between a pregnane X receptor reporter gene assay and CYP3A4 expression in human Hepa-Itocytes. Drug Metab. Dispos., 30, 795-804 (2002) https://doi.org/10.1124/dmd.30.7.795
  17. Marks, P. A., Miller, T., and Richon, V. M., Histone deacetylases. Curr. Opin. Pharmacol., 3, 344-351 (2003) https://doi.org/10.1016/S1471-4892(03)00084-5
  18. Mathur, M., Tucker P. W., and Samuels, H. H., PSF is a novel corepressor that mediates its effect through Sin3A and the DNA binding domain of nuclear hormone receptors. Mol. Cell Biol., 21, 2298-2311 (2001) https://doi.org/10.1128/MCB.21.7.2298-2311.2001
  19. McKinsey, T. A., Zhang, C. L., and Olson, E. N., Control of muscle development by dueling HATs and HDACs. Curr. Opin. Genet. Dev., 11,497-504 (2001) https://doi.org/10.1016/S0959-437X(00)00224-0
  20. Ogg, M. S., Williams, J. M., Tarbit, M., Goldfarb, P. S., Grays, T. J. B., and Gibson, G. G., A reporter gene assay to assess the molecular mechanisms of xenobiotic-dependent induction of the human CYP3A4 gene in vitro. Xenobiotica, 29, 269-279 (1999) https://doi.org/10.1080/004982599238669
  21. Pascussi, J. M., Drocourt, L., Gerbal-Chaloin, S., Fabre, J. M., Maurel, P., and Vilarem, M. J., Dual effect of dexamethasone on CYP3A4 gene expression in human Hepa-Itocytes. Eur J. Biochem., 268, 6346-6357 (2001) https://doi.org/10.1046/j.0014-2956.2001.02540.x
  22. Pascussi, J. M., Gerbal-Chaloin, S., Drocourt, L., Maurel, P., and Vilarem, M. J., The expression of CYP2B6, CYP2C9 and CYP3A4 genes, a tangle of networks of nuclear and steroid receptors. Biochim. Biophysic. Act., 1619,243-253 (2003) https://doi.org/10.1016/S0304-4165(02)00483-X
  23. Polevoda, B., and Sherman, F., The diversity of acetylated proteins. Genome Biol., 3, 1-6 (2002)
  24. Staudinger, J. L., Goodwin, B., Jones, S. A., Hawkins-Brown, D., MacKenzie, K. I., LaTour, A, Liu, Y., Klaassen, C. D., Brown, K. K., Reinhard, J., Willson, T. M., Koller, B. H., and Kliewer, S. A., The nuclear receptor PXR is a lithocholic acid sensor that protects against liver toxicity. Proc. Natl. Acad. Sci. USA, 98, 3369-3374 (2001) https://doi.org/10.1073/pnas.051551698
  25. Suzuki, H., Gabrielson, E., Chen, W., Anbazhagan, R., van Engeland, M., Weijenberg, M. P., Herman, J. G., and Baylin, S. B., A genomic screen for genes upregulated by demethylation and histone deacetylase inhibition in human colorectal cancer. Nat. Genet., 31,141-149 (2002) https://doi.org/10.1038/ng892
  26. Synold, T. W., Dussault, I., and Forman, B. M., The orphan nuclear receptor SXR coordinately regulates drug metabolism and efflux. Nat. Med., 7, 584-590 (2001) https://doi.org/10.1038/87912
  27. Takeshita, A, Taguchi, M., Koibuchi, N., and Ozawa, Y., Putative role of the orphan nuclear receptor SXR in the mechanism of CYP3A4 inhibition by Xenobiotics. J. Biol. Chem., 277, 32453-32458 (2002) https://doi.org/10.1074/jbc.M111245200
  28. Van Lint, C., Emiliani, S., and Verdin, E., The expression of a small fraction of cellular genes is changed in response to histone hyperacetylation. Gene Expr, 5,245-253 (1996)
  29. Watkins, P., The clinical significance of CYP3A enzymes. Pharmacogenetics, 4,171-184 (1994) https://doi.org/10.1097/00008571-199408000-00001
  30. Xie, W., Radominska-Pandya, A., Shi, Y., Simon, C. M., Nelson, M. C., Ong, E. S., Waxman, D. J., and Evans, R. M., An essential role for nuclear receptors SXR/PXR in detoxification of cholestatic bile acids. Proc. Natl. Acad. Sci. U.S.A., 98, 3375-3380 (2001) https://doi.org/10.1073/pnas.051014398
  31. Yoshida, M., Kijima, M., Akita, M., and Beppu, T., Potent and specific inhibition of mammalian histone deacetylase both in vivo and in vitro by trichostatin A. J. Biol. Chem., 265, 17174-17179 (1990)
  32. Yu, X., Guo, Z. S., Mareu, M. G., Neckers,L., Nguyen, D. M., Chen, G. A., and Schrump, D. S., Modulation of p53, ErbB1, ErbB2, and Raf-1 expression in lung cancer cells by depsipeptide FR901228.J. Natl. Cancer Inst., 94, 504-513 (2002) https://doi.org/10.1093/jnci/94.7.504