Cancer Res Treat.  2005 Apr;37(2):122-128.

A Histone Deacetylase Inhibitor, Trichostatin A, Enhances Radiosensitivity by Abrogating G2/M Arrest in Human Carcinoma Cells

Affiliations
  • 1Department of Radiation Oncology, Seoul National University College of Medicine, Seoul, Korea. ihkim@snu.ac.kr
  • 2Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea.
  • 3Institute of Radiation Medicine, Seoul National University College of Medicine, Seoul, Korea.
  • 4Department of Radiation Oncology, Seoul National University Bundang Hospital, Seongnam, Korea.
  • 5Department of Radiation Oncology, Soonchunhyang University College of Medicine, Bucheon, Korea.
  • 6Department of Radiation Oncology, Dankook University College of Medicine, Cheonan, Korea.
  • 7Department of Radiation Oncology, Konkuk University College of Medicine, Seoul, Korea.
  • 8Department of Radiation Oncology, Chung-Ang University College of Medicine, Seoul, Korea.

Abstract

PURPOSE
Histone deacetylase inhibitors (HDIs) are emerging as potentially useful components in anticancer therapy. In this study, we tried to confirm the radiosensitizing effect of trichostatin A (TSA) on a panel of human carcinoma cell lines and elucidate its mechanism of interaction. MATERIALS AND METHODS: A549, HeLa and Caski cells were exposed to TSA for 18 hr prior to irradiation, and the cell survival then measured using a clonogenic assay. Western blot and flow cytometric analyses, for histone acetylation, and cell cycle and apoptosis, respectively, were also performed. RESULTS: TSA increased the acetylation of histone H3. The pretreatment of TSA consistently radiosensitized all three cell lines. The SF2 (surviving fraction at 2 Gy) of TSA-treated cells was significantly lower than that of mock treated cells. The SER (sensitizer enhancement ratio) increased in all 3 cell lines, in concentration dependent manners. The TSA treated cells showed abrogation of radiation-induced G2/M arrest, in a concentration dependent manner. CONCLUSION: The pretreatment of TSA enhanced the radiosensitivity of a panel of human carcinoma cells, which was attributed, in part, to the abrogation of radiationinduced G2/M arrest.

Keyword

Trichostatin A; Histone deacetylase inhibitor; Radiosensitization; G2/M arrest

MeSH Terms

Acetylation
Apoptosis
Blotting, Western
Cell Cycle
Cell Line
Cell Survival
Histone Deacetylase Inhibitors*
Histone Deacetylases*
Histones*
Humans*
Radiation Tolerance*
Radiation-Sensitizing Agents
Histone Deacetylase Inhibitors
Histone Deacetylases
Histones
Radiation-Sensitizing Agents

Figure

  • Fig. 1 TSA increased hyperacetylation of histone H3. Cells were treated 200 nM TSA for 18 hr, the proteins harvested, separated on SDS-PAGE and electroblotted. The membrane was probed with anti-histone H3 antibody. α-tubulin was used as a loading control. The same amount of drug carrier (DMSO) treated cells were used as a mock control. (A) A549; (B) HeLa.

  • Fig. 2 Survival curves for cells treated with trichostatin A (TSA) prior to irradiation. Cells were pretreated with 0, 50 or 200 nM TSA for 18 hr, and then irradiated with 4 MV X-rays after the removal of the TSA from the culture media. The survival was measured using a clonogenic assay. Each point on the survival curves represents the mean surviving fraction from at least three dishes. Experiments were repeated a minimum of three times. The error bars represent the standard errors. (A) A549; (B) HeLa; (C) Caski.

  • Fig. 3 Surviving fraction of cells pretreated with trichostatin A (TSA) prior to 2 Gy (SF2) of irradiation. Cells were pretreated with 0, 50, 100 or 200 nM TSA for 18 hr, and then irradiated with 4 MV X-rays after the removal of the TSA from the culture media. The survival was measured using a clonogenic assay. The SF2s of the TSA-treated cells were compared with those of mock treated cells using the t test. The error bars represent the standard errors.

  • Fig. 4 Sensitizer enhancement ratio (SER) of trichostatin A (TSA). The SER was defined as the ratio of the radiation dose required to obtain a surviving fraction (SF) of 0.5, without TSA pretreatment, to that required to obtain the same SF after TSA pretreatment. Cells were pretreated with 0, 50, 100 or 200 nM TSA for 18 hr, and then irradiated with 4 MV X-rays after the removal of the TSA from the culture media. The survival was measured using a clonogenic assay.

  • Fig. 5 Cell cycle analysis for A549 cells treated with trichostatin A (TSA). Cells were treated with 0~1,000 nM TSA for 18 hr, and collected, fixed and stained with propidium iodide, and then analyzed using flow cytometry.

  • Fig. 6 Sub-G0/G1 proportion in A549 cells pretreated with trichostatin A (TSA). Cells were treated with 0~1,000 nM TSA for 6~48 hr, and the cell cycle distribution then determined using flow cytometry. The proportions of cells with DNA contents less than that in the G0/G1 phase cells were defined as the sub-G0/G1 proportions.

  • Fig. 7 The change in the G2/M proportions of cells following the combination of TSA and irradiation. A549 cells were pretreated with 0 nM (-○-), 200 nM (-□-) and 400 nM(-△-) TSA for 18 hr. The TSA was then removed from the media, and the cells irradiated with 4 MV X-rays at 0 Gy (A), 2 Gy (B) or 8 Gy (C) at 0 hr. After irradiation, the cells were collected, fixed and stained with propidium iodide, and then analyzed using flow cytometry at 2, 4, 6, 8, 12, 16, 20 and 24 hr.


Reference

1. Yoshida M, Furumai R, Nishiyama M, Komatsu Y, Nishino N, Horinouchi S, et al. Histone deacetylase as a new target for cancer chemotherapy. Cancer Chemother Pharmacol. 2001; 48(Suppl. 1):S20–S26. PMID: 11587361.
Article
2. Grunstein M. Histone acetylation and chromatin structure and transcription. Nature. 1997; 389:349–352. PMID: 9311776.
3. 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. PMID: 10922406.
Article
4. Juan LJ, Shia WJ, Chen MH, Yang WM, Seto E, Lin YS, et al. Histone deacetylase specifically down-regulate p53-dependent gene activation. J Biol Chem. 2000; 275:20436–20443. PMID: 10777477.
5. 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. PMID: 11099047.
Article
6. Verdin E, Dequiedt F, Kasler HG. Class II histone deacetylases: versatile regulators. Trends Genet. 2003; 19:286–293. PMID: 12711221.
Article
7. de Ruijter AJ, van Gennip AH, Caron HN, Kemp S, van Kuilenburg AB. Histone deacetylase (HDACs): characterization of the classical HDAC family. Biochem J. 2003; 370:737–749. PMID: 12429021.
8. Kim TY, Bang YJ. Development of new anti-cancer agents targeting histone deacetylase. Cancer Res Treat. 2002; 34(Suppl. 2):9S–16S.
9. Gilbert J, Baker SD, Bowling MK, Grochow L, Figg WD, Zabelina Y, 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. PMID: 11489804.
10. Marshall JL, Rizvi N, Kauh J, Dahut W, Figuera M, Kang MH, et al. A phase I trial of depsipeptide (FR901228) in patients with advanced cancer. J Exp Ther Oncol. 2002; 2:325–332. PMID: 12440223.
Article
11. Sandor V, Bakke S, Robey RW, Kang MH, Blagosklonny MV, Bender J, 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. PMID: 11895901.
12. Arundel C, Glicksman A, Leith J. Enhancement of radiation injury in human colon tumor cells by the maturational agent sodium butyrate (NaB). Radiat Res. 1985; 104:443–448. PMID: 4080986.
Article
13. Chung YL, Lee YH, Yen SH, Chi KH. A novel approach for nasopharyngeal carcinoma treatment uses phenylbutyrate as a protein kinase C modulators: implications for radiosensitization and EBV-targeted therapy. Clin Cancer Res. 2000; 6:1452–1458. PMID: 10778977.
14. 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. PMID: 11682008.
Article
15. Camphausen K, Burgan W, Cerra M, Oswald KA, Trepel JB, Lee MJ, et al. Enhanced radiation-induced cell killing and prolongation of γH2AX foci expression by the histone deacetylase inhibitor MS-275. Cancer Res. 2004; 64:316–321. PMID: 14729640.
Article
16. Zhang Y, Adachi M, Zhao X, Kawamura R, Imai K. Histone deacetylase inhibitors FK228, N-(2-aminophenyl)-4-[N-(pyridin-3-yl-methoxycarbonyl)amino-methyl]benzamide and m-carboxycinnamic acid bis-hydroxamide augment radiation-induced cell death in gastrointestinal adenocarcinoma cells. Int J Cancer. 2004; 110:301–308. PMID: 15069698.
17. 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; 59:1174–1180. PMID: 15234053.
Article
18. Furumai R, Komatsu Y, Nishino N, Khochbin S, Yoshida , Horinouchi S. Potent histone deacetylase inhibitors built from trichostatin A and cyclic tetrapeptide antibiotics including trapoxin. Proc Natl Acad Sci USA. 2001; 98:87–92. PMID: 11134513.
Article
19. 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. PMID: 9214697.
Article
20. Iliakis G, Wang Y, Guan J, Wang H. DNA damage checkpoint control in cells exposed to ionizing radiation. Oncogene. 2003; 22:5834–5847. PMID: 12947390.
Article
21. Su LN, Little JB. Prolonged cell cycle delay in radioresistant human cell lines transfected with activated ras oncogene and/or simian virus 40T-antigen. Radiat Res. 1993; 133:73–79. PMID: 8434116.
22. 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. PMID: 10531300.
Article
23. 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. PMID: 12668657.
Article
24. Van Lint C, Emiliani S, Verdn E. The expression of small fraction of cellular genes is changed in response to histone hyperacetylation. Gene Expr. 1996; 5:245–253. PMID: 8723390.
25. Butler LM, Webb Y, Agus D, Higgins B, Tolentino TR, Kutko MC, et al. Inhibition of transformed cell growth and induction of cellular differentiation by pyroxamide: an inhibitor of histone deacetylase. Clin Cancer Res. 2001; 7:962–970. PMID: 11309347.
Full Text Links
  • CRT
Actions
Cited
CITED
export Copy
Close
Share
  • Twitter
  • Facebook
Similar articles
Copyright © 2024 by Korean Association of Medical Journal Editors. All rights reserved.     E-mail: koreamed@kamje.or.kr