DOI QR코드

DOI QR Code

Apoptotic Effect of Co-Treatment with Valproic Acid and 17AAG on Human Osteosarcoma Cells

Valproic acid와 17AAG의 병용처리가 사람골육종세포에 미치는 세포자멸사 효과에 대한 연구

  • Park, Jun-Young (Department of Oral Anatomy, School of Dentistry, Pusan National University) ;
  • Park, Se-Jin (Department of Oral Anatomy, School of Dentistry, Pusan National University) ;
  • Kim, In-Ryoung (Department of Oral Anatomy, School of Dentistry, Pusan National University) ;
  • Park, Bong-Soo (Department of Oral Anatomy, School of Dentistry, Pusan National University) ;
  • Jeong, Sung-Hee (Department of Oral Medicine, School of Dentistry, Pusan National University) ;
  • Ko, Myung-Yun (Department of Oral Medicine, School of Dentistry, Pusan National University) ;
  • Ahn, Yong-Woo (Department of Oral Medicine, School of Dentistry, Pusan National University)
  • 박준영 (부산대학교 치의학전문대학원 구강해부학교실) ;
  • 박세진 (부산대학교 치의학전문대학원 구강해부학교실) ;
  • 김인령 (부산대학교 치의학전문대학원 구강해부학교실) ;
  • 박봉수 (부산대학교 치의학전문대학원 구강해부학교실) ;
  • 정성희 (부산대학교 치의학전문대학원 구강내과학교실) ;
  • 고명연 (부산대학교 치의학전문대학원 구강내과학교실) ;
  • 안용우 (부산대학교 치의학전문대학원 구강내과학교실)
  • Received : 2010.12.04
  • Accepted : 2011.01.30
  • Published : 2011.03.30

Abstract

Valproic acid (VPA) is a well-known anticonvulsive agent and has been used in the treatment of epilepsy for almost 30 years. VPA emerged in 1997 as an antineoplastic agent. And it is known that antitmor activity of VPA is associated with its targeted at histone deacetylases. 17AAG, Inhibition of HSP90 leads to the proteasome degradation of the HSP90 client proteins, such as Akt, Raf/Ras, Erk, VEGF, cyclin D and p53, and causes potent antitumor activity. It is reported that 17AAG-induced HSP90 inhibition results in prevention of cell proliferation and induction of apoptosis in several types of cancer. This study was undertaken to investigate the synergistic apoptotic effect of co-treatment with the histone deacetylases inhibitor, VPA and the HSP90 inhibitor, 17AAG on human osteosarcoma (HOS) cells. Cell viability was evaluated by trypan-blue exclusion. Induction and augmentation of apoptosis were confirmed by Hoechst staining, flow cytometry (DNA hypoploidy and MMP change), Westen blot analysis and immunofluorescent staining. In this study, HOS cells co-treated with VPA and 17AAG showed several lines of apoptotic manifestation such as nuclear condensations, the reduction of MMP, the decrease of DNA content, the release of cytochrome c into cytosol, the translocation of AIF onto nuclei, and activation of caspase-3, caspase-7 and PARP whereas each single treated HOS cells did not. Although the single treatment of 1 mM VPA or 0.5 ${\mu}M$ 17AAG for 48 h did not induce apoptosis, the co-treatment with them induced prominently apoptosis. Therefore our data in this study provide the possibility that combination therapy with VPA and 17AAG could be considered as a novel therapeutic strategy for human osteosarcoma.

Valproic acid(VPA)는 아주 잘 알려진 항경련제로서, 30년 동안 간질치료제로서 사용되어져 왔다. VPA는 1997년에 최초로 항암제의 효능이 밝혀졌으며, VPA의 항암효과는 히스톤탈아세틸화효소 억제제의 기전에 기인한다고 규명되었다. 17AAG(17-Allyamnio-17-demethoxygeldanamycin)는 HSP90의 억제제이며, HSP90은 세포증식과 세포생존에 관여하며, 최근 17AAG가 세포자멸사를 유도한다는 연구들이 보고되어지고 있다. 본 연구는 히스톤탈아세틸화효소억제제인 VPA와 HSP90 억제제인 17AAG의 병용처리가 사람골육종세포에 상승 세포자멸사 효과가 있는지를 알기 위해서 수행되었다. VPA과 17AAG의 병용처리가 단독처리에 비해서 효과적인 세포생존율 감소가 있는지 확인하기 위해서 trypan-blue법을 시행하였고, 세포자멸사의 유도와 증가를 확인하기 위해서 Hoechst 염색법, flow cytometry(DNA hypoploidy와 MMP 측정), Western bot 분석법 그리고 면역형광염색법을 수행하였다. 병용처리 된 사람골육종세포는 단독처리 된 사람골육종세포에서 거의 관찰할 수 없었던 핵 응축과 조각남, 사립체막 전위와 DNA 양의 감소, cytochrome c의 세포질로의 유리, AIF의 핵으로의 이동, caspase-3과 caspase-7의 파괴 및 PARP의 분절화와 같은 세포자멸사 증거를 보였다. 48시간 동안 1 mM의 VPA와 0.5 ${\mu}M$ 17AAG을 각기 단독처리 한 결과에서는 세포자멸사를 유도 못했으나, 병용처리한 결과에는 아주 탁월한 세포자멸사의 유도를 보였다. 이러한 병용처리 결과는 사람골육종의 새로운 치료적 전략으로 응용될 수 있다고 생각한다.

Keywords

References

  1. Marks PA, Rifkind RA. Erythroleukemic differentiation. Annu Rev Biochem 1978;47:419-448. https://doi.org/10.1146/annurev.bi.47.070178.002223
  2. Kim MS, Kwon HJ, Lee YM, Baek JH, Jang JE, Lee SW, Moon EJ, Kim HS, Lee SK, Chung HY, Kim CW, Kim KW. Histone deacetylases induce angiogenesis by negative regulation of tumor suppressor genes. Nat Med 2001;7:437-443. https://doi.org/10.1038/86507
  3. 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
  4. Deroanne CF, Bonjean K, Servotte S, Devy L, Colige A, Clausse N, Blacher S, Verdin E, Foidart JM, Nusgens BV and Castronovo V. Histone deacetylases inhibitors as anti-angiogenic agents altering vascular endothelial growth factor signaling. Oncogene 2002;21: 427-436. https://doi.org/10.1038/sj.onc.1205108
  5. Marks PA, Richon VM, Rifkind RA. Histone deacetylase inhibitors: induces of differentiation or apoptosis of transformed cells. J Natl Cancer Inst 2000;92:1210-1216. https://doi.org/10.1093/jnci/92.15.1210
  6. De Ruijter AJ, van Gennip AH, Caron HN, Kemp S, van Kuilenburg AB. Histone deacetylases (HDACs): characterization of the classical HDAC family. Biochem J 2003;370:737-749. https://doi.org/10.1042/BJ20021321
  7. Cinatl J Jr, Cinatl J, Driever PH, Kotchetkov R, Pouckova P, Kornhuber B, Schwabe D. Sodium valproate inhibits in vivo growth of human neuroblastoma cells. Anticancer Drugs 1997;8:958-963. https://doi.org/10.1097/00001813-199711000-00007
  8. Gurvich N, Tsygankova OM, Meinkoth JL, Klein PS. Histone deacetylase is a target of valproic acidmediated cellular differentiation. Cancer Res 2004;64: 1079-1086. https://doi.org/10.1158/0008-5472.CAN-03-0799
  9. Kramer OH, Zhu P, Ostendorff HP, Golebiewski M, Tiefenbach J, Peters MA, Brill B, Groner B, Bach I, Heinzel T, Gottlicher M. The histone deacetylase inhibitor valproic acid selectively induces proteasomal degradation of HDAC2. EMBO J 2003;22:3411-3420. https://doi.org/10.1093/emboj/cdg315
  10. Blaheta RA, Cinatl J Jr. Anti-tumor mechanisms of valproate: a novel role for an old drug. Med Res Rev 2002;22:492-511. https://doi.org/10.1002/med.10017
  11. Cinatl J Jr, Kotchetkov R, Blaheta R, Driever PH, Vogel JU, Cinatl J. Induction of differentiation and suppression of malignant phenotype of human neuroblastoma BE(2)-C cells by valproic acid: enhancement by combination with interferon-alpha. Int J Oncol 2002;20:97-106.
  12. Villa R, Folini M, Porta CD, Valentini A, Pennati M, Daidone MG, Zaffaroni N. Inhibition of telomerase activity by geldanamycin and 17-allylamino, 17- demethoxygeldanamycin in human melanoma cells. Carcinogenesis 2003;24:851-859. https://doi.org/10.1093/carcin/bgg028
  13. Babchia N, Calipel A, Mouriaux F, Faussat AM, Mascarelli F. 17-AAG and 17-DMAG-induced inhibition of cell proliferation through B-Raf downregulation in WT B-Raf-expressing uveal melanoma cell lines. Invest Ophthalmol Vis Sci 2008;49: 2348-2356. https://doi.org/10.1167/iovs.07-1305
  14. Gazitt Y, Kolaparthi V, Moncada K, Thomas C, Freeman J. Targeted therapy of human osteosarcoma with 17AAG or rapamycin: characterization of induced apoptosis and inhibition of mTOR and Akt/MAPK/ Wnt pathways. Int J Oncol 2009;34:551-561.
  15. Williams GT. Programmed cell death: Apoptosis and oncogenesis. Cell 1991;65:1097-1098. https://doi.org/10.1016/0092-8674(91)90002-G
  16. Yuan J. Evolutionary conservation of a genetic pathway of programmed cell death. J Cell Biochem 1996;60:4-11. https://doi.org/10.1002/(SICI)1097-4644(19960101)60:1<4::AID-JCB2>3.0.CO;2-1
  17. Susin SA, Lorenzo HK, Zamzami N, Marzo I, Snow BE, Brothers GM, Mangion J, Jacotot E, Costantini P, Loeffler M, Larochette N, Goodlett DR, Aebersold R, Siderovski DP, Penninger JM, Kroemer G. Molecular characterization of mitochondrial apoptosis-inducing factor. Nature 1999;397:441-446. https://doi.org/10.1038/17135
  18. Orlowski RZ. The role of the ubiquitin-proteasome pathway in apoptosis. Cell Death Differ 1999;6: 303-313. https://doi.org/10.1038/sj.cdd.4400505
  19. Lu Y, Yagi T. Apoptosis of human tumor cells by chemotherapeutic anthracyclines is enhanced by Bax overexpression. J Radiat Res(Tokyo) 1999;40:263-272. https://doi.org/10.1269/jrr.40.263
  20. Fellenberg J, Mau H, Nedel S, Ewerbeck V, Debatin KM. Drug-induced apoptosis in osteosarcoma cell lines is mediated by caspase activation independent of CD95-receptor/ligand interaction. J Orthop Res 2000;18:10-17. https://doi.org/10.1002/jor.1100180103
  21. Seki K, Yoshikawa H, Shiiki K, Hamada Y, Akamatsu N, Tasaka K. Cisplatin (CDDP) specifically induces apoptosis via sequential activation of caspase-8, 3 and -6 in osteosarcoma. Cancer Chemother Pharmacol 2000;45:199-206. https://doi.org/10.1007/s002800050030
  22. Kieslich M, Schwabe D, Cinatl J Jr, Driever PH. Increase of fetal hemoglobin synthesis indicating differentiation induction in children receiving valproic acid. Pediatr Hematol Oncol 2003;20:15-22. https://doi.org/10.1080/08880010390158496
  23. Li XN, Shu Q, Su JM, Perlaky L, Blaney SM, Lau CC. Valproic acid induces growth arrest, apoptosis, and senescence in medulloblastomas by increasing histone hyperacetylation and regulating expression of p21Cip1, CDK4, and CMYC. Mol Cancer Ther 2005;4:1912-1922. https://doi.org/10.1158/1535-7163.MCT-05-0184
  24. Takai N, Desmond JC, Kumagai T, Gui D, Said JW, Whittaker S, Miyakawa I, Koeffler HP. Histone deacetylase inhibitors have a profound antigrowth activity in endometrial cancer cells. Clin Cancer Res 2004;10:1141-1149. https://doi.org/10.1158/1078-0432.CCR-03-0100
  25. Witt O, Schweigerer L, Driever PH, Wolff J, Pekrun A. Valproic acid treatment of glioblastoma multiforme in a child. Pediatr Blood Cancer 2004;43:181. https://doi.org/10.1002/pbc.20083
  26. Warrell RP Jr, He LZ, Richon V, Calleja E, Pandolfi PP. Therapeutic targeting of transcription in acute promyelocytic leukemia by use of an inhibitor of histone deacetylase. J Natl Cancer Inst 1998;90: 1621-1625. https://doi.org/10.1093/jnci/90.21.1621
  27. Graziani G, Tentori L, Portarena I, Vergati M, Navarra P. Valproic acid increases the stimulatory effect of estrogens on proliferation of human endometrial adenocarcinoma cells. Endocrinology 2003;144:2822-2828. https://doi.org/10.1210/en.2002-0180
  28. Mongan NP, Gudas LJ. Valproic acid, in combination with all-trans retinoic acid and 5-aza-2'- deoxycytidine, restores expression of silenced RARbeta2 in breast cancer cells. Mol Cancer Ther 2005;4:477-486.
  29. Karkoulis PK, Stravopodis DJ, Margaritis LH, Voutsinas GE. 17-Allylamino-17-demethoxygeldanamycin induces downregulation of critical Hsp90 protein clients and results in cell cycle arrest and apoptosis of human urinary bladder cancer cells. BMC cancer 2010;10:481-495. https://doi.org/10.1186/1471-2407-10-481
  30. Susin SA, Lorenzo HK, Zamzami N, Marzo I, Snow BE, Brothers GM, Mangion J, Jacotot E, Costantini P, Loeffler M, Larochette N, Goodlett DR, Aebersold R, Siderovski DP, Penninger JM, Kroemer G. Molecular characterization of mitochondrial apoptosis-inducing factor. Nature 1999;397:441-446. https://doi.org/10.1038/17135
  31. Green DR, Reed JC. Mitochondria and apoptosis. Science 1998;281:1309-1312. https://doi.org/10.1126/science.281.5381.1309
  32. Golab J, Stoklosa T, Czajka A, Dabrowska A, Jakobisiak M, Zagozdzon R, Wojcik C, Marczak M, Wilk S. Synergistic antitumor effects of a selective proteasome inhibitor and TNF in mice. Anticancer Res 2000;20:1717-1721.
  33. Golab J, Stoklosa T, Czajka A, Dabrowska A, Jakobisiak M, Zagozdzon R, Wojcik C, Marczak M, Wilk S. Synergistic antitumor effects of a selective proteasome inhibitor and TNF in mice. Anticancer Res 2000;20:1717-1721.
  34. Wagenknecht B, Hermisson M, Groscurth P, Liston P, Krammer PH, Weller M. Proteasome inhibitorinduced apoptosis of glioma cells involves the processing of multiple caspases and cytochrome c release. J Neurochem 2000;75:2288-2297.
  35. Marshansky V, Wang X, Bertrand R, Luo H, Duguid W, Chinnadurai G, Kanaan N, Vu MD, Wu J. Proteasomes modulate balance among proapoptotic and antiapoptotic Bcl-2 family members and compromise functioning of the electron transport chain in leukemic cells. J Immunol 2001;166:3130-3142. https://doi.org/10.4049/jimmunol.166.5.3130
  36. Dauglas E, Susin SA, Zamzami N, Ferri KF, Irinopoulou T, Larochette N, Prevost MC, Leber B, Andrews D, Penninger J, Kroemer G. Mitochondrionuclear translocation of AIF in apoptosis and necrosis. FASEB J 2000;14:729-739. https://doi.org/10.1096/fasebj.14.5.729
  37. Acehan D, Jiang X, Morgan DG, Heuser JE, Wang X, Akey CW. Three-dimensional structure of the apoptosome: Implications for assembly, procaspase-9 binding, and activation. Mol Cell 2002;9:423-432. https://doi.org/10.1016/S1097-2765(02)00442-2