2,3,7,8-TCDD의 세포형질전환 및 내성획득에 관여하는 세포내 인자에 관한 연구

Studies on Cellular Factors Responsible for 2,3,7,8-TCDD Resistency and Cellular Transformation

  • 발행 : 2006.03.01

초록

To enhance our understanding of toxicity mediated through the pathway by which TCDD stimulates gene expression, we have investigated genes whose expressions are changed after treatment with TCDD and/or MNNG in human Chang liver cell. First, we treated with MNNG and TCDD for two weeks to transform human Chang liver cell. We obtained cell looks like to be transformed and compared the differential gene expression by using cDNA chip (Macrogen) which carrys genes related with signal transduction pathways, oncogenes and tumor suppressor genes, etc. We found that TCDD up- or down-regulated 203 and 111 genes including oncogenes and tumor suppressor genes in human Chang liver cell two fold or more, respectively. Second, we compared the differential gene expression after treatment with TCDD only by using cDNA chip (Superarray) which carrys genes related with cell cycle regulations, and found that TCDD up regulated genes related with cell proliferation as well as cell growth inhibition in human Chang liver cell two fold or more, respectively. These results suggest that toxicity induced by TCDD may reflect sustained alterations in the expression of many genes and that the changes reflect both direct and indirect effects of TCDD.

키워드

참고문헌

  1. Bowes, R.C., Parrish, A.R., Steinberg, M.A., Willett, K.L., Zhao, W., Savas, U., Jefcoate, C.R., Safe, S.H., and Ramos, K.S. (1996) Atypical cytochrome P45 0 induction profiles in glomerular mesangial cells at the mRNA and enzyme level. Evidence for CYP1A1 and CYP1B1 expression and their involvement in benzo[a]pyrene metabolism. Biochem. Pharmacol. 52, 587-595 https://doi.org/10.1016/0006-2952(96)00310-3
  2. Davis, J.W. 2nd, Melendez, K., Salas, V.M., Lauer, F.T., Burchiel, S.W. (2000) 2,3,7,8-Tetrachlorodibenzo-p-dioxin (TCDD) inhibits growth factor withdrawal-induced apoptosis in the human mammary epithelial cell line, MCF-10A. Carcinogenesis. 21, 881-6 https://doi.org/10.1093/carcin/21.5.881
  3. Ge, N-L. and Elferink, J.A. (1998) direct interaction between the aryl hydrocarbon receptor and retinoblastoma protein. Linking dioxin signaling to the cell cycle. J. Biol. Chem. 273, 22708-22713 https://doi.org/10.1074/jbc.273.35.22708
  4. Gottlicher, M. and Cikryt, P. (1987) Induction of the aromatic hydrocarbon receptor by trans-4-acetylaminostilbene in rat liver. Comparison with other aromatic amines. Carcinogenesis 8, 1021-1023 https://doi.org/10.1093/carcin/8.7.1021
  5. Heimler, I, Rawlins, R.G., Owen, H., Hutz, R.J. (1998) Dioxin perturbs, in a dose- and time-dependent fashion, steroid secretion, and induces apoptosis of human luteinized granulosa cells. Endocrinology. 139, 4373-9 https://doi.org/10.1210/en.139.10.4373
  6. Herschman, H.R. (1991) Primary response genes induced by growth factors and tumor promotors. Annu. Rev. Biochem. 60, 281-319 https://doi.org/10.1146/annurev.bi.60.070191.001433
  7. Hoffman, R.E., Stehr-Green, P.A., Webb, K.B., Evans, R.G., Knutsen, A.P., Schramm, W.R.F., Staake, J.L., Gibson, B.B., and Steinberg, K.K. (1986) Health effects of long-term exposure to 2,3,7,8-tetrachlorodibenzo-p-dioxin. JAMA, 255, 2031- 2038 https://doi.org/10.1001/jama.255.15.2031
  8. Kharat, I. and Saatcioglu, F. (1996) Antiestrogenic effects of 2,3,7,8-tetrachlorodibenzo -p-dioxin are mediated by direct transcriptional interference with the liganded estrogen receptor. Cross-talk between aryl hydrocarbon- and estrogen-mediated signaling. J. Biol. Chem. 271, 10533-7 https://doi.org/10.1074/jbc.271.18.10533
  9. Kolluri, S.K., Weiss, C., Koff, A. and Gottlicher M, (1999) p27 kip1 induction and inhibition of proliferation by the intracellular Ah receptor in developing thymus and hepatoma cells. Genes. Dev. 13, 1742-1753 https://doi.org/10.1101/gad.13.13.1742
  10. Ma, X. and Babish, J.G. (1993) Acute 2,3,7,8-tetrachlorodibenzo-p-dioxin exposure results in enhanced tyrosylphosphorylation and expression of murine hepatic cyclin dependent kinases. Biochem. Biophys. Res. Commun. 197, 1070-1077 https://doi.org/10.1006/bbrc.1993.2587
  11. Olsen, A., Teglund, S., Nelson, D., Gordon, L., Copeland, A., Georgescu, A., Carrano, A., Hammarstrom, S. (1994) Gene organization of the pregnancy-specific glycoprotein region on human chromosome assembly and analysis of a 700-kb cosmidcontig spanning the region. Genomics 23, 659-668 https://doi.org/10.1006/geno.1994.1555
  12. Reyes, H., Reisz-Porszasz, S., and Hankinson, O. (1992) Identification of the Ah receptor nuclear translocator protein (Arnt) as a component of the DNA binding form of the Ah receptor. Science, 256, 1193-1195 https://doi.org/10.1126/science.256.5060.1193
  13. Puga, A., Barnes, S.J., Dalton, T.P., Chang, C.y., Knudsen, E.S., Maier, M.A. (2000) Aromatic hydrocarbon receptor interaction with the retinoblastoma protein potentiates repression of E2F-dependent transcription and cell cycle arrest. J. Biol Chem. 275, 2943-50 https://doi.org/10.1074/jbc.275.4.2943
  14. Rininger, J., Ma, X. and Babish, J.G., (1995) Increased hepatic expression and phosphorylation of the murine p53 tumor suppressor protein and expression of $p21^{WAF1}$ CDK-inhibitory protein following acute dosing of TCDD. Proc 15th Int. Symp. Chlor. Dioxins 25, 367-372
  15. Safe, S., Wang, F., Porter, W., Duan, R. and McDougal, A. (1998) Ah receptor agonists as endocrine disruptors, Antiestrogenic activity and mechanisms. Toxicol. Lett. 102, 343-347 https://doi.org/10.1016/S0378-4274(98)00331-2
  16. Sidransky, D., Von Eschenbach, A., Tsai, Y.C., Jones, P., Summerhayes, I., Marshall, F., Paul, M., Green, P., Hamilton, S.R., Frost, P. and Vogelstein, B. (1991) Identification of p53 gene mutations in bladder cancers and urine samples. Science. 252, 706-709 https://doi.org/10.1126/science.2024123
  17. Weiss, C., Kolluri, S.K., Kiefer, F. and Gottlicher, M. (1996) Complementation of Ah receptor deficiency in hepatoma cells, Negative feedback regulation and cell cycle control by the Ahreceptor. Exp. Cell. Res. 226, 154-163 https://doi.org/10.1006/excr.1996.0214
  18. Whitelock, J.P., (1999) Induction of cytochrome P4501A1. Annu. Rev. Pharmacol .Toxicol. 39, 103-125 https://doi.org/10.1146/annurev.pharmtox.39.1.103