DOI QR코드

DOI QR Code

Modulacon of Cell Cycle Control by Histone Deacetylase Inhibitor Trichostatin A in A549 Human Non-small Cell Lung Cancer Cells

인체폐암세포 A549의 세포주기 조절인자에 미치는 histone deacetylase inhibitor trichostatin A의 영향

  • 황지원 (동의대학교 한의과대학 생화학교실) ;
  • 김영민 (한국과학영재학교) ;
  • 홍수현 (동의대학교 한의과대학 생화학교실) ;
  • 최병태 (동의대학교 해부학교실 및 대학원 바이오물질제어학과) ;
  • 이원호 (부산대학교 생물학과) ;
  • 최영현 (동의대학교 한의과대학 생화학교실)
  • Published : 2005.10.01

Abstract

Histone deacetylase (HDAC) inhibitors target key steps of tumor development. They inhibit proliferation, induce differentiation and/or apoptotic cell death, and exhibit potent antimetastatic and antiangiogenic properties in cancer cells in vitro and in vivo. Although they are emerging as a promising new treatment strategy in malignancy, how they exert their effect on human non-small cell lung cancer cells is as yet unclear. The present study was undertaken to investiate the underlying mechanism of a HDAC inhibitor trichostatin A (TSA)-induced growth arrest and its effect on the cell cycle control gene products in a human lung carcinoma cell line A549. TSA treaoent induced the growth inhibition and morphological changes in a concentration-dependent manner. Treatment of A549 cells with TSA resulted in a concentration-dependent increased G1 (under 100 ng/ml) and/or G2/M (200 ng/ml) cell population of the cell cycle as determined by flow cytometry Moreover, 200 ng/ml TSA treatment significantly induced the population of sub-G1 cells (23.0 fold of control). This anti-proliferative effect of TSA was accompanied by a marked inhibition of cyclins, positive regulators of cell cycle progression, and cyclin-dependent kinases (Cdks) expression and concomitant induction of tumor suppressor p53 and Cdk inhibitors such as p21 and p27 Although further studies are needed, these findings provide important insights into the possible molecular mechanisms of the anti-cancer activity of TSA in human lung carcinoma cells.

Histone deacetylase (HDAC) 억제제가 새로운 항암치료제 후보물질로서 유용성이 높은 것으로 평가되지만, 아직까지 인체폐암세포에 관한 연구는 상대적으로 미미한 실정이다. 따라서 본 연구에서는 폐암세포에 미치는 HDAC 억제제의 항암작용 기전을 조사하기 위하여 A549 인체폐암세포주를 대상으로 암세포의 증식에 미치는 대표적인 HDAC 억제제인 tichostatin A (TSA)에 의한 영향을 세포주기 조절관련인자 중심으로 조사하였다. TSA의 처리에 의하여 A549 폐암세포의 증식은 처리 농도 의존적으로 억제되었으며, 심한 형태적 변형을 동반하였다. 저농도 처리군에서는 TSA 농도가 증가할수록 세포주기 G1기의 빈도가 증가하였으나, 고농도 처리군에서는 G2/M기에 속하는 세포의 빈도가 증가되었다. 또한 apoptosis 유발의 간접적인 지표가 되는 sub-G1기에 속하는 세포의 빈도 역시 TSA 처리 농도 의존적으로 매우 증가되었다. 이러한 TSA의 A549 폐암세포 증식억제 효과는 cyclins 및 CdkS의 발현 억제, 종양억제유전자인 p53 및 Cdks 억제제인 p21과 p27의 발현 증가와도 연관성이 있었다. TSA의 항암 기전을 규명하기 위해서는 더 많은 연구가 부가적으로 필요하겠지만, 본 연구의 결과들에 의하면 TSA는 강력한 인체폐암세포의 증식 억제 및 항암작용이 있음을 시사하여 준다고 할 수 있다.

Keywords

References

  1. Aksan, J. 2002. Chromatin goes global. Trends Biochem. Sci. 27, 7-8
  2. Bordonaro, M., J. M. Mariadason, F. Aslam, B. G. Heerdt and J. H. Augenlicht. 1999. Butyrate-induced apoptotic cascade in colonic carcinoma cells: modulation of the ${\beta}$ -catenin-Tcf pathway and concordance with effects of sulindac and trichostatin A but not curcumin. Cell Growth Differ. 10, 713-720
  3. Bouchain, G. and D. Delorme. 2003. Novel hydroxamate and anilide derivatives as potent histone deacetylase inhibitors: synthesis and antiproliferative evaluation. Curro Med. Chem. 10, 2359-2372 https://doi.org/10.2174/0929867033456585
  4. Chen, J. S. and D. V. Faller. 2005. Histone deacetylase inhibition-mediated post-translational elevation of p27KIPl protein levels is required for G1 arrest in fibroblasts. J. Cell. Physiol. 202, 87-99 https://doi.org/10.1002/jcp.20094
  5. Chen, Y. X., J. Y. Fang, J. Lu and D. K. Qiu. 2004. Regulation of histone acetylation on the expression of cell cycle-associated genes in human colon cancer cell lines, Zhonghua Yi Xue Za Zhi 84, 312-317
  6. Elledge, S. J. and J. W. Harper. 1994. Cdk inhibitors: on the threshold of checkpoints and development. Curro Opin. Cell Bioi. 6, 847-852 https://doi.org/10.1016/0955-0674(94)90055-8
  7. Fukuoka, K, J. Adachi, K. Nishio, H. Arioka, H. Kurokawa, H. Fukumoto, T. Ishida, T. Nomoto, H. Yokote, A. Tomonari, N. Narita, J. Yokota and N. Saijo. 1977. p16lNK4 expression is associated with the increased sensitivity of human nonsmall cell lung cancer cells to DNA topoisomerase I inhibitors. Jpn. J. Cancer Res. 88, 1009-1016
  8. Gerber, M and A. Shilatifard. 2003. Transcriptional elongation by RNA polymerase II and histone methylation. J. Biol. Chem. 278, 26303-26306 https://doi.org/10.1074/jbc.R300014200
  9. Harper, J. W. 1997. Cyclin dependent kinase inhibitors. Cencer Surv 29, 91-107
  10. Herold, C., M. Ganslmayer, M. Ocker, M. Hermann, A. Geerts, E. G. Hahn and D. Schupp an. 2002. The histonedeacetylase inhibitor Trichostatin A blocks proliferation and triggers apoptotic programs in hepatoma cells. J. Hepatol. 36, 233-240
  11. Hirsch, C. J. and K. Bonham. 2004. Histone deacetylase inhibitors regulate p21WAF1 gene expression at the posttranscriptional level in HepG2 cells. FEBS Lett. 570, 37-40 https://doi.org/10.1016/j.febslet.2004.06.018
  12. Hitomi, T., Y. Matsuzaki, T. Yokota, Y. Takaoka and T. Sakai. 2003. 5 (INK4b) in HDAC inhibitor-induced growth arrest. FEBS Left. 554, 347-350 https://doi.org/10.1016/S0014-5793(03)01186-4
  13. Joung, K. E., D. K. Kim and Y. Y. Sheen. 2004. Antiproliferative effect of trichostatin A and HC-toxin in T47D human breast cancer cells. Arch. Pharm. Res. 27, 640-645 https://doi.org/10.1007/BF02980164
  14. Kim, Y. B., S. W. Ki, M. Yoshida and S. Horinouchi. 2000. Mechanism of cell cycle arrest caused by histone deacetylase inhibitors in human carcinoma cells. J. Antibiot. (Tokyo) 53, 1191-200 https://doi.org/10.7164/antibiotics.53.1191
  15. Kuo, M. H. and C. D. Allis. 1998. Roles of histone acetyltransferases and deacetylases in gene regulation. Bioessays 20, 615-626 https://doi.org/10.1002/(SICI)1521-1878(199808)20:8<615::AID-BIES4>3.0.CO;2-H
  16. Lagger, G., A. Doetzlhofer, B. Schuettengruber, E. Haidweger, E. Simboeck, J. Tischler, S. Chiocca, G. Suske, H. Rotheneder, E. Wintersberger and C. Seiser. 2003. The tumor suppressor p53 and histone deacetylase 1 are antagonistic regulators of the eyclin-dependent kinase inhibitor p21/W AF1/CIP1 gene. Mol. Cell. BioI. 23, 2669-2679 https://doi.org/10.1128/MCB.23.8.2669-2679.2003
  17. Li, H. and X. Wu. 2004. Histone deacetylase inhibitor, Trichostatin A, activates p21 W AFl/CIPl expression through downregulation of c-myc and release of the repression of c-myc from the promoter in human cervical cancer cells. Biochem. Biophys. Res. Commun. 324, 860-867 https://doi.org/10.1016/j.bbrc.2004.09.130
  18. Li, Y., C. W. Jenkins, M. A. Nichols and Y. Xiong. 1994. Cell cycle expression and p53 regulation of the cyclin-dependent kinase inhibitor p21. Oncogene 9, 2261-2268
  19. Lopatina, N. G., J. C. Poole, S. N. Saldanha, N. J. Hansen, J. S. Key, M. A. Pita, J. G. Andrews and T. O. Tollefsbol. 2003. Control mechanisms in the regulation of telomerase reverse transcriptase expression in differentiating human teratocarcinoma cells. Biochem. Biophys. Res. Commun. 306, 650-659 https://doi.org/10.1016/S0006-291X(03)01033-7
  20. Margueron, R., V. Duong, A Castet and V. Cavailles. 2004. Histone deacetylase inhibition and estrogen signalling in human breast cancer cells. Biochem. Pharmacal. 68, 1239-1246 https://doi.org/10.1016/j.bcp.2004.04.031
  21. Marks, P. A, V. M. Richon, T. Miller and W. K. Kelly. 2004. Histone deacefylllse inhibitors. Adv. Cancer Res. 91, 137-168 https://doi.org/10.1016/S0065-230X(04)91004-4
  22. Moore, P. S., S. Barbi, M. Donadelli, C. Costanzo, C. Bassi, M. Palmieri and A Scarpa. 2004. Gene expression profiling after treatment with the histone deacetylase inhibitor trichostatin A reveals altered expression of both pro- and antiapoptotic genes in pancreatic adenocarcinoma cells. Biochim. Biophys. Acta 1693, 167-176 https://doi.org/10.1016/j.bbamcr.2004.07.001
  23. Noh, E. J. and J. S. Lee. 2003. Functional interplay between modulation of histone deacetylase activity and its regulatory role in G2-M transition. Biochem. Biophys. Res. Commun. 310, 267-273 https://doi.org/10.1016/j.bbrc.2003.09.013
  24. Papeleu, P. T. Vanhaecke, G. Elaut, M. Vinken, T. Henkens, S. Snykers and V. Rogiers. 2005. Differential effects of histone deacetylase inhibitors in tumor and normal cells-what is the toxicological relevance? Crit. Rev. Toxical. 35, 363-378 https://doi.org/10.1080/10408440590935639
  25. Roh, M.S., C. W. Kim, B. S. Park, G. C. Kim, J. H. Jeong, H. C. Kwon, D. J. Suh, K. H. Cho, S. B. Yee and Y. H. Yoo. 2004. Mechanism of histone deacetylase inhibitor Trichostatin A induced apoptosis in human osteosarcoma cells. Apoptosis 9, 583-589 https://doi.org/10.1023/B:APPT.0000038037.68908.6e
  26. Salminen, A, T. Tapiola, P. Korhonen and T. Suuronen. 1998. Neuronal apoptosis induced by histone deacetylase inhibitors. Brain Res. Mol. Brain Res. 61, 203-206 https://doi.org/10.1016/S0169-328X(98)00210-1
  27. Sherr, C. J. 2000. The Pezcoller lecture: cancer cell cycles revisited. Cancer Res. 60, 3689-3695
  28. Sowa, Y., T. Orita, S. Minamikawa, K. Nakano, T. Mizuno, H. Nomura and T. Sakai. 1997. Histone deacetylase inhibitor activates the WAFI/Cipl gene promoter through the Spl sites. Biochem. Biophys. Res. Commun. 241, 142-150 https://doi.org/10.1006/bbrc.1997.7786
  29. Suzuki, T., H. Yokozaki, H. Kuniyasu, K. Hayashi, K. Naka, S. Ono, T. Ishikawa, E. Tahara and W. Yasui. 2000. Effect of trichostatin A on cell growth and expression of cell cyeleand apoptosis-related molecules in human gastric and oral carcinoma cell lines. Int. J. Cancer 88, 992-997, 2000 https://doi.org/10.1002/1097-0215(20001215)88:6<992::AID-IJC24>3.0.CO;2-9
  30. Vanhaecke, T., P. Papeleu, G. Elaut and V. Rogiers. 2004. Trichostatin A-like hydroxamate histone deacetylase inhibitors as therapeutic agents: toxicological point of view. Curr. Med. Chem. 11, 1629-1643 https://doi.org/10.2174/0929867043365099
  31. Weinberg, R. A 1995. The retinoblastoma protein and cell cyele control. Cell 81, 323-330 https://doi.org/10.1016/0092-8674(95)90385-2
  32. Wharton, W., J. Savell, W. D. Cress, E. Seto and W. J. Pledger. 2000. Inhibition of mitogenesis in Balb/c-3T3 cells by Trichostatin A Multiple alterations in the induction and activation of cyelin-cyelin-dependent kinase complexes. J. Biol. Chem. 275, 33981-33987 https://doi.org/10.1074/jbc.M005600200
  33. Xiao, H., T. Hasegawa and K. Isobe. 1999. Both Sp1 and Sp3 are responsible for p2lwafl promoter activity induced by histone deacetylase inhibitor in NIH3T3 cells. J. Cell. Biochem. 73, 291-302 https://doi.org/10.1002/(SICI)1097-4644(19990601)73:3<291::AID-JCB1>3.0.CO;2-5
  34. Xiong, Y., G. J. Hannon, H. Zhang, D. Casso, R. Kobayashi and D. Beach. 1993. p21 is a universal inhibitor of cyclin kinases. Nature 366, 701-704 https://doi.org/10.1038/366701a0
  35. Yamashita, Y., M. Shimada, N. Harimoto, T. Rikimaru, K. Shirabe, S. Tanaka and K. Sugimachi. 2003. Histone deacetylase inhibitor trichostatin A induces cell-cycle arrest/ apoptosis and hepatocyte differentiation in human hepatoma cells. Int. J. Cancer 103, 572-576 https://doi.org/10.1002/ijc.10699
  36. Yoshida, M., R. Furumai, M. Nishiyama, Y. Komatsu, N. Nishino and S. Horinouchi. 2001. Histone deacetylase as a new target for cancer chemotherapy. Cancer Chemother. Pharmacal. 48 (Suppl 1), 520-26
  37. Yoshida, M., M. Kijima, M. Akita and T. Beppu. 1990. Potent and specific inhibition of mammalian histone deacetylase both in vivo and in vitro by trichostatin A. J. Biol. Chem. 265, 17174-17179
  38. Yoshida, M., M. Matsuyama, Y. Komatsu and N. Nishino. 2003. From discovery to the coming generation of histone deacetylase inhibitors. Curro Med. Chem. 10, 2351-2358 https://doi.org/10.2174/0929867033456602

Cited by

  1. Effects of Bcl-2 Overexpressing on the Apoptotic Cell Death Induced by HDAC Inhibitors in Human Leukemic U937 Cells vol.17, pp.4, 2007, https://doi.org/10.5352/JLS.2007.17.4.552