Trichostatin A, a Histone Deacetylase Inhibitor, Potentiated Cytotoxic Effect of ionizing Radiation in Human Head and Neck Cancer Cell Lines

히스톤탈아세틸효소 억제제 Trichostatin A에 의한 인간 두경부암 셰포주의 방사선 감수성 증강

  • Kim, Jin Ho (Department of Radiation Oncology, Seoul National University College of Medicine) ;
  • Shin, Jin Hee (Department of Cancer Research Institute, Seoul National University College of Medicine) ;
  • Chie, Eui Kyu (Department of Radiation Oncology, Seoul National University College of Medicine) ;
  • Wu, Hong-Gyun (Department of Radiation Oncology, Seoul National University College of Medicine) ;
  • Kim, Jae Sung (Department of Radiation Oncology, Seoul National University College of Medicine) ;
  • Kim, Il Han (Department of Radiation Oncology, Seoul National University College of Medicine) ;
  • Ha, Sung Whan (Department of Radiation Oncology, Seoul National University College of Medicine) ;
  • Park, Charn Il (Department of Radiation Oncology, Seoul National University College of Medicine) ;
  • Kang, Wee-Saing (Department of Radiation Oncology, Seoul National University College of Medicine)
  • 김진호 (서울대학교 의과대학 방사선종양학교실) ;
  • 신진희 (서울대학교 의과대학 암연구소) ;
  • 지의규 (서울대학교 의과대학 방사선종양학교실) ;
  • 우홍균 (서울대학교 의과대학 방사선종양학교실) ;
  • 김재성 (서울대학교 의과대학 방사선종양학교실) ;
  • 김일한 (서울대학교 의과대학 방사선종양학교실) ;
  • 하성환 (서울대학교 의과대학 방사선종양학교실) ;
  • 박찬일 (서울대학교 의과대학 방사선종양학교실) ;
  • 강위생 (서울대학교 의과대학 방사선종양학교실)
  • Published : 2004.06.01

Abstract

Purpose : We have previously reported that human glioblastoma cells are sensitized to radiation-induced death after their exposure to trichostatin A (TSA), a histone deacetylase inhibitor (HDAC-1), prior to the irradiation. We aimed to measure the magnitude of the radiosensitizing effect of TSA in human head and neck cancer cell lines. Materials and Methods : Human head and neck cancer cell lines, HN-3 and HN-9, were exposed to 0, 50, 100, and 200 nM TSA for 18 hr prior to irradiation. Then, the TSA-treated cells were irradiated with 0, 2, 4, 6, and 8 Gy, and cell survival was measured by clonogenic assay. Results : Pre-irradiation exposure to TSA was found to radiosensitize HN-3 and HN-9 cell lines. In HN-9 cells, the fraction surviving after 2 Gy (SF2) was significantly reduced by treatment of TSA at concentration as low as 50 nM. However, a treatment with 200 nM TSA was required to significantly decrease SF2 in the HN-3 cell line. SER of pre-irradiation treatment with 200 nM TSA was 1.84 in HN-3 and 7.24 in HN-9, respectively. Conclusions : Our results clearly showed that human head and neck cancer cell lines can be sensitized to ionizing radiation by pre-irradiation inhibition of histone deacetylase (HDAC) using TSA, and that this potentiation might well be a general phenomenon.

목적 : 본 연구진이 기왕에 입증한 바 있는 히스톤탈아세틸효소 억제제 trichostatin A (TSA)가 나타내는 방사선 감수성 증강 작용이 두경부암 세포주에서 발생하는 정도를 실험적으로 확인하고자 하였다. 대상 및 방법 : 인간 두경부암 세포주인 HN-3과 HN-9를 0, 50, 100, 200 nM의 TSA에 18시간 동안 전처치시킨 후 각각 0, 2, 4, 6, 8 Gy 방사선을 조사하였다. 세포생존곡선은 clonogenic assay를 이용하여 산출하였고 linear quadratic 모델에 따라 분석하였다 결과 : 방사선 조사 전 TSA처리는 HN-3과 HN-9 세포주의 방사선 감수성을 증강시켰다. 50 nM의 TSA로 처리된 HN-9 세포주에서 2 Gy 조사후 생존분획(SF2)은 유의한 수준으로 감소하였으나, HN-3 세포주는 200 nM의 TSA 처리 후 SF2가 유의하게 감소하였다. HN-3과 HN-9 세포주에서 200 nM TSA의 sensitizer enhancement ratio는 각각 1.84와 7.24였다. 결론 : 방사선 조사 전 히스톤탈아세틸효소 억제는 인간 두경부암 세포주의 방사선 감수성을 증가시켰으며, 이 증강 작용이 암세포주에서의 일반적으로 관찰되는 현상일 가능성이 크다고 제안한다.

Keywords

References

  1. Yoshida M, Furumai R, Nishiyama M, Komatsu Y, Nishino N, Horinouchi S. Histone deacetylase as a new target for cancer chemotherapy. Cancer Chemother Pharmacol 2001;48(Suppl. 1):20-26 https://doi.org/10.1007/s002800100300
  2. Grunstein M. Histone acetylation and chromatin structure and transcription. Nature 1997;389:349-352 https://doi.org/10.1038/38664
  3. Tycko B, Ashkenas J. Epigenetics and its role in disease. J Clin Invest 2000;105:245-246 https://doi.org/10.1172/JCI9319
  4. Verdin E, Dequiedt F, Kasler HG. Class II histone deacetylases: versatile regulators. Trends Genet 2003;19:286-293 https://doi.org/10.1016/S0168-9525(03)00073-8
  5. Juan LJ, Shia WJ, Chen MH, et al. Histone deacetylase specifically down-regulate p53-dependent gene activation. J Biol Chem 2000;275:20436-20433 https://doi.org/10.1074/jbc.M000202200
  6. Luo J, Su F, Chen, D, Shiloh A, Gu W. Deacetylation of p53 modulates its effect on cell growth and apoptosis. Nature 2000;408:377-381 https://doi.org/10.1038/35042612
  7. Li M, Luo J, Brooks CL, Gu W. Acetylation of p53 inhibits its ubiquitination by Mdm2. J Biol Chem 2002;277:50607-50611 https://doi.org/10.1074/jbc.C200578200
  8. Chan HM, Kristic-Demonacos M, Smith L, Demonacos C, La Thangue N. Acetylation control of the retinoblastoma tumor-suppressor protein. Nat Cell Biol 2001;3:667-674 https://doi.org/10.1038/35083062
  9. Martinez-Balbas MA, Bauer UM Nielsen SJ, Brehm A, Kouzarides T. Regulation of E2F1 activity by acetylation. EMBO J 2000;19:662-671 https://doi.org/10.1093/emboj/19.4.662
  10. Marks PA, Richon VM, Rifkind RA. Histone deacetylase inhibitors: inducers of differentiation or apoptosis of transformed cells. J Natl Cancer Inst 2000;92:1210-1216 https://doi.org/10.1093/jnci/92.15.1210
  11. De Ruijter AJM, Van Gennip AH, Caron HN, Kemp S, Van Kuilenburg ABP. Histone deacetylase (HDACs): characterization of the classical HDAC family. Biochem J 2003; 307:737-749
  12. Hoshikawa Y, Kwon HJ, Yoshida M, Horinouchi S, Beppu T. Trichostatin A induces morphological changes and gelsolin expression by inhibiting histone deacetylase in human carcinoma cell lines. Exp Cell Res 1994;214:189-197 https://doi.org/10.1006/excr.1994.1248
  13. Yu X, Guo ZS Marcu MG et al. Modulation of p53, ErbB1, ErbB2, and Raf-1 expression in lung cancer cells by depsipeptide FR901228. J Natl Cancer Inst 2002;94:504-513 https://doi.org/10.1093/jnci/94.7.504
  14. Munster PN, Troso-Sandoval T, Rosen N, Rifkind R, Marks PA, Richon VM. The histone deacetylase inhibitor suberoylanilide hydroxamic acid induces differentiation of human breast cancer cells. Cancer Res 2001;61:8492-8497
  15. Kim YB, Yoshida M, Sueharu H. Selective induction of cyclin-dependent kinase inhibitors and their roles in cell cycle arrest caused by trichostatin A, an inhibitor of histone deacetylase. Ann N Y Acad Sci 1999;886:200-203 https://doi.org/10.1111/j.1749-6632.1999.tb09416.x
  16. Yamashita Y, Shimada M, Harimoto N, et al. Histone deacetylase inhibitor trichostatin A induces cell-cycle arrest/ apoptosis and hepatocyte differentiation in human hepatoma cells. Int J Cancer 2003;103:572-576 https://doi.org/10.1002/ijc.10699
  17. Yoshida M, Beppu T. Reversible arrest of proliferation of Rat 3Y1 fibroblasts in the G1 and G2 phases by Trichostatin A. Exp Cell Res 1988;177:122-131 https://doi.org/10.1016/0014-4827(88)90030-4
  18. Kwon HJ, Kim MS, Kim MJ, Nakajima H, Kim KW. Histone deacetylase inhibitor FK228 inhibits tumor angiogenesis. Int J Cancer 2002;97:290-296 https://doi.org/10.1002/ijc.1602
  19. Herold C, Ganslmayer M, Ocker M, et al. The histonedeacetylase inhibitor trichostatin A block proliferation and triggers apoptotic programs in hepatoma cells. J Hepatol 2002;36:233-240
  20. McBain JA, Eastman A, Nobel CS, Mueller GC. Apoptotic death in adenocarcinoma cell lines induced by butyrate and other histone deacetylase inhibitors. Biochem Pharmacol 1997;53:1357-1368 https://doi.org/10.1016/S0006-2952(96)00904-5
  21. Medina V, Edmonds B, Young GP, James R, Appleton S, Zalewski PD. Induction of caspase-3 protease activity and apoptosis by butyrate and trichostatin A (inhibitors of histone deacetylase): dependence on protein synthesis and synergy with a mitochondrial/cytochrome c-dependent pathway. Cancer Res 1997;57:3697-3707
  22. Engelhard HH, Duncan HA, Kim S, Criswell PS, Van Eldik L. Therapeutic effects of sodium butyrate on glioma cells in vitro and in the rat C6 model. Neurosurgery 2001;48:616-625 https://doi.org/10.1097/00006123-200103000-00035
  23. Fournel M, Trachy-bourget M, Yan PT, et al. Sulfonamide anilides, a novel class of histone deacetylase inhibitors, are antiproliferative against human tumors. Cancer Res 2002;62:4325-4330
  24. Jaboin J, Wild J, Hamidi H, et al. MS-27-275, an inhibitor of histone deacetylase, has marked in vitro and in vivo antitumor activity against pediatric solid tumors. Cancer Res 2002;62:6108-6115
  25. Qui L, Kelso MJ, Hansen C, West ML, Fairlie DP, Parsons PG. Anti-tumor activity in vitro and in vivo of selective differentiating agents containing hydroxamate. Br J Cancer 1999;80:1252-1258 https://doi.org/10.1038/sj.bjc.6690493
  26. Saito A, Yamashita T, Mariko Y, et al. A synthetic inhibitor of histone deacetylase, MS-27-275, with marked in vivo antitumor activity against human tumors. Proc Natl Acad Sci USA. 1999;96:4592-4597
  27. Sasakawa Y, Naoe Y, Inoue T, et al. Effects of FK228, a novel histone deacetylase inhibitor, on human lymphoma U-937 cells in vitro and in vivo. Biochem Pharmacol 2002;64:1079-1090 https://doi.org/10.1016/S0006-2952(02)01261-3
  28. Saunders N, Dicker A, Popa C, Jones S, Dahler A. Histone deacetylase inhibitors as potential anti-skin cancer agents. Cancer Res 1999;59:399-404
  29. Suzuki T, Yokozaki H, Kuniyasu H, et al. Effect of trichostatin A on cell growth and expression of cell cycle- and apoptosis-related molecules in human gastric and oral carcinoma cell lines. Int J Cancer 2000;88:992-997 https://doi.org/10.1002/1097-0215(20001215)88:6<992::AID-IJC24>3.0.CO;2-9
  30. Gilbert J, Baker SD, Bowling MK, et al. A phase I dose escalation and bioavailability study of oral sodium phenylbutyrate in patients with refractory solid tumor malignancies. Clin Cancer Res 2001;7:2292-2300
  31. Marshall JL, Rizvi N, Kauh J, et al. A phase I trial of depsipeptide (FR901228) in patients with advanced cancer (Abstr.) J Exp Ther Oncol 2002;2:325-332
  32. Piekarz RL, Robey R, Sandor V, et al. Inhibitor of histone deacetylase, depsipeptide (FR901228), in the treatment of peripheral and cutaneous T-cell lymphoma: a case report. Blood 2001;98:2865-2868 https://doi.org/10.1182/blood.V98.9.2865
  33. Prakash S, Foster BJ, Meyer M, et al. Chronic oral administration of CI-994: a phase 1 study. Invest New Drugs 2001;19:1-11 https://doi.org/10.1023/A:1006489328324
  34. Sandor V, Bakke S, Robey RW, et al. Phase I trial of the histone deacetylase inhibitor, depsipeptide (FR901228, NSC 630176), in patients with refractory neoplasms. Clin Cancer Res 2002;8:718-728
  35. Sandor V, Bakke S, Robey RW, et al. Phase I trial of the histone deacetylase inhibitor, depsipeptide (FR901228, NSC 630176), in patients with refractory neoplasms. Clin Cancer Res 2002;8:718-728
  36. Biade S, Stobbe, CC, Boyd JT, Chapman JD. Chemical agents that promote chromatin compaction radiosensitize tumour cells. Int J Radiat Biol 2001;77:1033-1042 https://doi.org/10.1080/09553000110066068
  37. Chung YL, Lee YHW, Yen SH, Chi KH. A novel approach for nasopharyngeal carcinoma treatment uses phenyl butyrate as a protein kinase C modulators: implications for radiosensitization and EBV-targeted therapy. Clin Cancer Res 2000;6:1452-1458
  38. Kim JH, Shin JH, Kim IH. Susceptibility and radiosensitization of human glioblastoma cells to trichostatin A, a histone deacetylase inhibitor. Int J Radiat Oncol Biol Phys 2004;59: (in press)
  39. Kim SY, Chu KC, Lee HR, et al. Establishment and characterization of nine new head and neck cancer cell lines. Acta Otolaryngol (Stckh) 1997;117:775-784 https://doi.org/10.3109/00016489709113477
  40. Kim GD, Choi YH, Dimtchev A, Jeong SJ, Dritschilo A, Jung M. Sensing of ionizing radiation-induced DNA damage by ATM through interaction with histone deacetylase. J Biol Chem 1999;274:31127-31130 https://doi.org/10.1074/jbc.274.44.31127
  41. Schmidt DR, Schreiber SL. Molecular association between ATR and tow components of the nucleosome remodeling and deacetylating complex, HDAC2 and CHD4. Biochem 1999;38:14711-14717 https://doi.org/10.1021/bi991614n
  42. Kao GD, McKenna WG, Guenther MG, Muschel RJ, Lazar MA, Yen TJ. Histone deacetylase 4 interacts with 53BP1 to mediate the DNA damage response. J Cell Biol 2003;160:1017-1027 https://doi.org/10.1083/jcb.200209065
  43. Luo J, Nikolaev AY, Imai S, et al. Negative control of p53 by Sir2 (promotes cell survival under stress. Cell 2001;107:137-148 https://doi.org/10.1016/S0092-8674(01)00524-4
  44. Vaziri H, Dessain SK, Eaton EN, et al. hSIR2SIRT1 functions as an NAD-dependent p53 deacetylase. Cell 2001;107:149-159 https://doi.org/10.1016/S0092-8674(01)00527-X