Silibinin Induces Apoptotic Cell Death Via ROS-dependent Mitochondrial Pathway in Human Glioma Cells

  • Shin, Won-Yong (Department of Internal Medicine, College of Korean Medicine, Dongguk University) ;
  • Jeong, Ji-Cheon (Department of Internal Medicine, College of Korean Medicine, Dongguk University)
  • Published : 2009.08.25

Abstract

It has been reported that silibinin, a natural polyphenolic flavonoid, induces cell death in various cancer cell types. However, the underlying mechanisms by which silibinin induces apoptosis in human glioma cells are poorly understood. The present study was therefore undertaken to examine the effect of silibinin on glioma cell apoptosis and to determine its underlying mechanism in human glioma cells. Apoptosis was estimated by FACS analysis. Reactive oxygen species (ROS) generation and mitochondrial membrane potential (${\Psi}m$) were measured using fluorescence dyes DCFH-DA and $DiOC_6$(3), respectively. Cytochrome c release from mitochondria and caspase-3 activation were estimated by Western blot analysis using specific antibodies. Exposure of cells to 30 mM silibinin induced apoptosis starting at 6 h, with increasing effects after 12-48h in a time-dependent manner. Silibinin caused ROS generation and disruption of ym, which were associated with the silibinin-induced apoptosis. The silibinin-induced ROS generation and disruption in ym were prevented by inhibitors of mitochondrial electron transport chain. The hydrogen peroxide scavenger catalase blocked ROS generation and apoptosis induced by silibinin. Silibinin induced cytochrome c release into cytosolic fraction and its effect was prevented by catalase and cyclosporine A. Silibinin treatment caused caspase-3 activation, which was inhibited by DVED-CHO and cyclosporine A. Pretreatment of caspase inhibitors also protected against the silibinin-induced apoptosis. These findings indicate that ROS generation plays a critical role in the initiation of the silibinin-induced apoptotic cascade by mediation of the mitochondrial apoptotic pathway including the disruption of ${\Psi}m$, cytochrome c release, and caspase-3 activation.

Keywords

References

  1. Ohgaki, H., Kleihues, P. Population-based studies on incidence, survival rates, and genetic alterations in astrocytic and oligodendroglial gliomas. J Neuropathol Exp Neurol 64: 479-489, 2005 https://doi.org/10.1093/jnen/64.6.479
  2. DeAngelis, L.M. Brain tumors. N Engl J Med 344: 114-123, 2001 https://doi.org/10.1056/NEJM200101113440207
  3. Sanai, N., Alvarez-Buylla, A., Berger, M.S. Neural stem cells and the origin of gliomas. N Engl J Med 353: 811-822, 2005 https://doi.org/10.1056/NEJMra043666
  4. Fresco, P., Borges, F., Diniz, C., Marques, M.P. New insights on the anticancer properties of dietary polyphenols. Med Res Rev 26: 747-766, 2006 https://doi.org/10.1002/med.20060
  5. Kanadaswami, C., Lee, L.T., Lee, P.P., Hwang, J.J., Ke, F.C., Huang, Y.T., Lee, M.T. The antitumor activities of flavonoids. In Vivo 19: 895-909, 2005
  6. Ren, W., Qiao, Z., Wang, H., Zhu, L., Zhang, L. Flavonoids: promising anticancer agents. Med Res Rev 23: 519-534, 2003 https://doi.org/10.1002/med.10033
  7. Braganhol, E., Zamin, L.L., Canedo, A.D., Horn, F., Tamajusuku, A.S., Wink, M.R., Salbego, C., Battastini, A.M. Antiproliferative effect of quercetin in the human U138MG glioma cell line. Anticancer Drugs 17: 663-671, 2006 https://doi.org/10.1097/01.cad.0000215063.23932.02
  8. Sharma, V., Joseph, C., Ghosh, S., Agarwal, A., Mishra, M.K., Sen, E. Kaempferol induces apoptosis in glioblastoma cells through oxidative stress. Mol Cancer Ther 6: 2544-2553, 2007 https://doi.org/10.1158/1535-7163.MCT-06-0788
  9. Shen, S.C., Lin, C.W., Lee, H.M., Chien, L.L., Chen, Y.C. Lipopolysaccharide plus 12-o-tetradecanoylphorbol 13-acetate induction of migration and invasion of glioma cells in vitro and in vivo: Differential inhibitory effects of flavonoids. Neuroscience 140: 477-489, 2006 https://doi.org/10.1016/j.neuroscience.2006.02.028
  10. Kaur, M., Agarwal, R. Silymarin and epithelial cancer chemoprevention: how close we are to bedside? Toxicol Appl Pharmacol 224: 350-359, 2007 https://doi.org/10.1016/j.taap.2006.11.011
  11. Ramasamy, K., Agarwal, R. Multitargeted therapy of cancer by silymarin. Cancer Lett. 269: 352-362, 2008 https://doi.org/10.1016/j.canlet.2008.03.053
  12. Singh, R.P., Gu, M., Agarwal, R. Silibinin inhibits colorectal cancer growth by inhibiting tumor cell proliferation and angiogenesis. Cancer Res 68: 2043-2050, 2008 https://doi.org/10.1158/0008-5472.CAN-07-6247
  13. Singh, R.P., Mallikarjuna, G.U., Sharma, G., Dhanalakshmi, S., Tyagi, A.K., Chan, D.C., Agarwal, C., Agarwal, R. Oral silibinin inhibits lung tumor growth in athymic nude mice and forms a novel chemocombination with doxorubicin targeting nuclear factor kappaB-mediated inducible chemoresistance. Clin Cancer Res 10: 8641-8647, 2004 https://doi.org/10.1158/1078-0432.CCR-04-1435
  14. Son, Y.G., Kim, E.H., Kim, J.Y., Kim, S.U., Kwon, T.K., Yoon, A.R., Yun, C.O., Choi, K.S. Silibinin sensitizes human glioma cells to TRAIL-mediated apoptosis via DR5 up-regulation and down-regulation of c-FLIP and survivin. Cancer Res 67: 8274-8284, 2007 https://doi.org/10.1158/0008-5472.CAN-07-0407
  15. Pastorino, J.G., Chen, S.T., Tafani, M., Snyder, J.W., Farber, J.L. The overexpression of Bax produces cell death upon induction of the mitochondrial permeability transition. J Biol Chem 273: 7770-7775, 1998 https://doi.org/10.1074/jbc.273.13.7770
  16. Nowak, G. PKC-a and ERK1/2 mediate mitochondrial dysfunction, decreases in active Na+ transport, and cisplatin-induced apoptosis in renal cells. J. Biol. Chem. 277: 43377-43388, 2002 https://doi.org/10.1074/jbc.M206373200
  17. Ramos, S. Effects of dietary flavonoids on apoptotic pathways related to cancer chemoprevention. J Nutr Biochem 18: 427-442, 2007 https://doi.org/10.1016/j.jnutbio.2006.11.004
  18. Watson, W.H., Cai, J., Jones, D.P. Diet and apoptosis. Annu Rev Nutr 20: 485-505, 2000 https://doi.org/10.1146/annurev.nutr.20.1.485
  19. Chen, Y.C., Shen, S.C., Chow, J.M., Ko, C.H., Tseng, S.W. Flavone inhibition of tumor growth via apoptosis in vitro and in vivo. Int J Oncol 25: 661-670, 2004
  20. Kim, E.J., Choi, C.H., Park, J.Y., Kang, S.K., Kim, Y.K. Underlying mechanism of quercetin-induced cell death in human glioma cells. Neurochem Res 33: 971-979, 2008 https://doi.org/10.1007/s11064-007-9416-8
  21. Wang, I.K., Lin-Shiau, S.Y., Lin, J.K. Induction of apoptosis by apigenin and related flavonoids through cytochrome c release and activation of caspase-9 and caspase-3 in leukaemia HL-60 cells. Eur J Cancer 35: 1517-1525, 1999 https://doi.org/10.1016/S0959-8049(99)00168-9
  22. Desagher, S., Martinou, J.C. Mitochondria as the central control point of apoptosis. Trends Cell Biol 10: 369-377, 2000 https://doi.org/10.1016/S0962-8924(00)01803-1
  23. Chandra, D., Liu, J.W., Tang, D.G. Early mitochondrial activation and cytochrome c up-regulation during apoptosis. J Biol Chem 52: 50842-50854, 2002
  24. Green, D.R., Reed, J.C. Mitochondria and apoptosis. Science 281: 1309-1312, 1998 https://doi.org/10.1126/science.281.5381.1309
  25. Zou, H., Henzel, W.J., Liu, X., Lutschg, A., Wang, X. Apaf-1, a human protein homologous to C. elegans CED-4, participates in cytochrome c-dependent activation of caspase-3. Cell 90: 405-413, 1997 https://doi.org/10.1016/S0092-8674(00)80501-2