Trichostatin A induces apoptosis in lung cancer cells viasimultaneous activation of the death receptor-mediated andmitochondrial pathway

Kim, Hak-Ryeol;Kim, Eun-Jeong;Yang, Se-Hun;Jeong, Eun-Taek;Park, Chae-Ni;Lee, Jae-Hyeong;Yun, Myeong-Ja;So, Hong-Seop;Park, Rae-Gil
김학렬;김은정;양세훈;정은택;박채니;이재형;윤명자;소홍섭;박래길

  • Published : 20060000

Abstract

Trichostatin A (TSA), originally developed as an antifungal agent, is one of potent histone deace-tylase (HDAC) inhibitors, which are known to cause growth arrest and apoptosis induction of trans-formed cells, including urinary bladder, breast, prostate, ovary, and colon cancers. However, the effect of HDAC inhibitors on human non-small cell lung cancer cells is not clearly known yet. Herein, we demonstrated that treatment of TSA resulted in a significant decrease of the viability of H157 cells in a dose-dependent manner, which was revealed as apoptosis accompanying with nuclear fragmen-tation and an increase in sub-G0/G1 fraction. In addition, it induced the expression of Fas/FasL, which further triggered the activation of caspase-8. Catalytic activation of caspase-9 and decreased expression of anti-aptototic Bcl-2 and Bcl-XL pro-teins were observed in TSA-treated cells. Catalytic activation of caspase-3 by TSA was further con-firmed by cleavage of pro-caspase-3 and intracellular substrates, including poly (ADP-ribose) polymerase (PARP) and inhibitor of caspase-activated deoxy-ribonuclease (ICAD). In addition, a characteristic phenomenon of mitochondrial dysfunction, inc-luding mitochondrial membrane potential transition and release of mitochondrial cytochrome c into the cytosol was apparent in TSA-treated cells. Taken together, our data indicate that inhibition of HDAC by TSA induces the apoptosis of H157 cells through signaling cascade of Fas/FasL-mediated extrinsic and mitocondria-mediated intrinsic caspases path-way.

Keywords

References

  1. Choi YH. Induction of apoptosis by trichostatin A, a histone deacetylase inhibitor, is associated with inhibition of cyclooxygenase- 2 activity in human non-small cell lung cancer cells. Int J Oncol 2005;27:473-9
  2. Cousens LS, Gallwitz D, Alberts BM. Different accessibilities in chromatin to histone acetylase. J Biol Chem 1979;254: 1716-23
  3. Crissman HA, Steinkamp JA. Cell cycle-related changes in chromatin structure detected by flow cytometry using multiple DNA fluorochromes. Eur J Histochem 1993;37:129-38
  4. Fokkema E, Groen HJ, Meijer C, Timens W, de Jong S, de Vries EG. The role of apoptosis-related genes in non-small-cell lung cancer. Clin Lung Cancer 2002;4:174-82 https://doi.org/10.3816/CLC.2002.n.025
  5. Glick RD, Swendeman SL, Coffey DC, Rifkind RA, Marks PA, Richon VM, La Quaglia MP. Hybrid polar histone deacetylase inhibitor induces apoptosis and CD95/CD95 ligand expression in human neuroblastoma. Cancer Res 1999;59: 4392-9
  6. Green DR, Reed JC. Mitochondria and apoptosis. Science 1998;281:1309-12 https://doi.org/10.1126/science.281.5381.1309
  7. Grunstein M. Histone acetylation in chromatin structure and transcription. Nature 1997;389:349-52 https://doi.org/10.1038/38664
  8. Han JW, Ahn SH, Park SH, Wang SY, Bae GU, Seo DW, Kwon HK, Hong S, Lee HY, Lee YW, Lee HW. Apicidin, a histone deacetylase inhibitor, inhibits proliferation of tumor cells via induction of p21WAF1/Cip1 and gelsolin. Cancer Res 2000; 60:6068-74
  9. 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;214:189-97 https://doi.org/10.1006/excr.1994.1248
  10. Jemal A, Tiwari RC, Murray T, Ghafoor A, Samuels A, Ward E, Feuer EJ, Thun MJ. Cancer statistics. CA Cancer J Clin 2004;54:8-29 https://doi.org/10.3322/canjclin.54.1.8
  11. Kijima M, Yoshida M, Sugita K, Horinouchi S, Beppu T. Trapoxin, an antitumor cyclic tetrapeptide, is an irreversible inhibitor of mammalian hisone deacetylase. J Biol Chem 1993;268:22429-35
  12. Kim DH, Shim JS, Kwon HJ. Coordinated transcriptional regulation of calmegin, a testis-specific molecular chaperon, by histone deacetylase and CpG methyltransferase. Exp Mol Med 2005;37:492-6 https://doi.org/10.1038/emm.2005.61
  13. Kim MS, So HS, Lee KM, Park JS, Lee JH, Moon SK, Ryu DG, Chung SY, Jung BH, Kim YK, Moon G, Park R. Activation of caspase cascades in Korean mistletoe (Viscum album var. coloratum) lectin-II-induced apoptosis of human myeloleukemic U937 cells. Gen Pharmacol 2000;34:349-55 https://doi.org/10.1016/S0306-3623(01)00072-6
  14. Kim YB, Lee KH, Sugita K, Yoshida M, Horinouchi S. Oxamflatin is a novel antitumor compound that inhibits mammalian histone deacetylase. Oncogene 1999;18:2461-70 https://doi.org/10.1038/sj.onc.1202564
  15. Kwon HJ, Owa T, Hassig CA, Shimada J, Schreiber SL. Depudecin induces morphological reversion of transformed fibroblasts via the inhibition of histone deacetylase. Proc Natl Acad Sci USA 1998;95:3356-61 https://doi.org/10.1073/pnas.95.7.3356
  16. Kwon SH, Ahn SH, Kim YK, Bae GU, Yoon JW, Hong S, Lee HY, Lee YW, Lee HW, Han JW. Apicidin, a histone deacetylase inhibitor, induces apoptosis and Fas/Fas ligand expression in human acute promyelocytic leukemia cells. J Biol Chem 2002;277:2073-80 https://doi.org/10.1074/jbc.M106699200
  17. Li H, Zhu H, Xu CJ, Yuan J. Cleavage of BID by caspase 8 mediates the mitochondrial damage in the Fas pathway of apoptosis. Cell 1998;94:491-501 https://doi.org/10.1016/S0092-8674(00)81590-1
  18. Luger K, Mader AW, Richmond RK, Sargent DF, Richmond TJ. Crystal structure of the nucleosome core particle at 2.8 A resolution. Nature 1997;389:251-60 https://doi.org/10.1038/38444
  19. Mahlknecht U, Hoelzer D. Histone acetylation modifiers in the pathogenesis of malignant disease. Mol Med 2000;6:623-44
  20. Mandal M, Adam L, Kumar R. Redistribution of activated caspase- 3 to the nucleus during butyric acid-induced apoptosis. Biochem Biophys Res Commun 1999;260:775-80 https://doi.org/10.1006/bbrc.1999.0966
  21. Marks PA, Rifkind RA. Erythroleukemic differentiation. Annu Rev Biochem 1978;47:419-48 https://doi.org/10.1146/annurev.bi.47.070178.002223
  22. Marks PA, Richon VM, Rifkind RA. Histone deacetylase inhibitors: inducers of differentiation or apoptosis of transformed cells. J Natl Cancer Inst 2000;92:1210-6 https://doi.org/10.1093/jnci/92.15.1210
  23. Marks PA, Richon VM, Breslow R, Rifkind RA. Histone deacetylase inhibitors as new cancer drugs. Curr Opin Oncol 2001;13:477-83 https://doi.org/10.1097/00001622-200111000-00010
  24. Mayo MW, Denlinger CE, Broad RM, Yeung F, Reilly ET, Shi Y, Jones DR. Ineffectiveness of histone deacetylase inhibitors to induce apoptosis involves the transcriptional activation of NF-kappa B through the Akt pathway. J Biol Chem 2003; 278:18980-9 https://doi.org/10.1074/jbc.M211695200
  25. 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;57:3697-707
  26. Mei S, Ho AD, Mahlknecht U. Role of histone deacetylase inhibitors in the treatment of cancer. Int J Oncol 2004;25:1509-19
  27. Monneret C. Histone deacetylase inhibitors. Eur J Med Chem 2005;40:1-13 https://doi.org/10.1016/j.ejmech.2004.10.001
  28. Nakajima H, Kim YB, Terano H, Yoshida M, Horinouchi S. FR901228, a potent antitumor antibiotic, is a novel histone deacetylase inhibitor. Exp Cell Res 1998;241:126-33 https://doi.org/10.1006/excr.1998.4027
  29. Park HW, Song JY, Kim KS, Han Y, Kim CW, Yi SY, Yun YS. Enhancement of radiosensitivity by combined ceramide and dimethylsphingosine treatment in lung cancer cells. Exp Mol Med 2004;36:411-9 https://doi.org/10.1038/emm.2004.53
  30. Peterson CL, Laniel MA. Histones and histone modifications. Curr Biol 2004;14:R546-51 https://doi.org/10.1016/j.cub.2004.07.007
  31. Roh MS, Kim CW, Park BS, Kim GC, Jeong JH, Kwon HC, Suh DJ, Cho KH, Yee SB, Yoo YH. Mechanism of histone deacetylase inhibitor Trichostatin A induced apoptosis in human osteosarcoma cells. Apoptosis 2004;9:583-9 https://doi.org/10.1023/B:APPT.0000038037.68908.6e
  32. Rosato RR, Grant S. Histone deacetylase inhibitors in cancer therapy. Cancer Biol Ther 2003;2:30-7
  33. Ruefli AA, Ausserlechner MJ, Bernhard D, Sutton VR, Tainton KM, Kofler R, Smyth MJ, Johnstone RW. The histone deacetylase inhibitor and chemotherapeutic agent suberoylanilide hydroxamic acid (SAHA) induces a cell-death pathway characterized by cleavage of Bid and production of reactive oxygen species. Proc Natl Acad Sci USA 2001;98:10833-8 https://doi.org/10.1073/pnas.191208598
  34. Saito A, Yamashita T, Mariko Y, Nosaka Y, Tsuchiya K, Ando T, Suzuki T, Tsuruo T, Nakanishi O. A synthetic inhibitor of histone deactylase, MS-27-275, with marked in vivo antitumor activity against human tumors. Proc Natl Acad Sci USA 1999; 96:4592-7
  35. Schiller JH, Harrington D, Belani CP, Langer C, Sandler A, Krook J, Zhu J, Johnson DH. Comparison of four chemotherapy regimens for advanced non-small-cell lung cancer. N Engl J Med 2002;346:92-8 https://doi.org/10.1056/NEJMoa011954
  36. Timmermann S, Lehrmann H, Polesskaya A, Harel-Bellan A. Histone acetylation and disease. Cell Mol Life Sci 2001;58: 728-36 https://doi.org/10.1007/PL00000896
  37. Tsuji N, Kobayashi M, Nagashima K, Wakisaka Y, Koizumi K. A new antifungal antibiotic, trichostatin. J Antibiot 1979;29:1-6
  38. Vander Heiden MG, Chandel NS, Williamson EK, Schumacker PT, Thomson CB. Bcl-xL regulates the membrane potential and volume homeostasis of mitochondria. Cell 1997;91: 627-37 https://doi.org/10.1016/S0092-8674(00)80450-X
  39. Wolf CM, Eastman A. The temporal relationship between protein phosphatase, mitochondrial cytochrome c release, and caspase activation in apoptosis. Exp Cell Res 1999;247: 505-13 https://doi.org/10.1006/excr.1998.4380
  40. Yee SB, Kim MS, Baek SJ, Kim GC, Yoo KS, Yoo YH, Park BS. Trichostatin A induces apoptosis of p815 mastocytoma cells in histone acetylation-and mitochondria-dependent fashion. Int J Oncol 2004;25:1431-6