Browse > Article
http://dx.doi.org/10.5352/JLS.2016.26.4.431

Effects of Sodium Butyrate, a Histone Deacetylase Inhibitor, on TRAIL-mediated Apoptosis in Human Bladder Cancer Cells  

Han, Min-Ho (Natural products Research Team, National Marine Biodiversity Institute of Korea)
Choi, Yung Hyun (Department of Biochemistry, Dongeui University College of Korean Medicine)
Publication Information
Journal of Life Science / v.26, no.4, 2016 , pp. 431-438 More about this Journal
Abstract
The tumour necrosis factor (TNF)-related apoptosis-inducing ligand (TRAIL) is considered a promising anticancer agent due to its unique ability to induce cancer cell death having only negligible effects on normal cells. However, many cancer cells tend to be resistant to TRAIL. In this study, we investigated the effects and molecular mechanisms of sodium butyrate (SB), a histone deacetylase inhibitor, in sensitizing TRAIL-induced apoptosis in 5637 human bladder cancer cells. Our results indicated that co-treatment with SB and TRAIL significantly increased the apoptosis induction, compared with treatment with either agent alone. Co-treatment with SB and TRAIL effectively increased the cell-surface expression of death receptor (DR) 5, but not DR4, which was associated with the inhibition of cellular Fas-associated death domain (FADD)-like interleukin-1β-converting enzyme (FLICE) inhibitory protein (c-FLIP). Furthermore, the activation of caspases (caspase-3, -8 and -9) and degradation of poly(ADP-ribose) were markedly increased in 5637 cells co-treated with SB and TRAIL; however, the synergistic effect was perfectly attenuated by caspase inhibitors. We also found that combined treatment with SB and TRAIL effectively induced the expression of pro-apoptotic Bax, cytosolic cytochrome c and cleave Bid to truncated Bid (tBid), along with down-regulation of anti-apoptotic Bcl-xL expression. These results collectively suggest that a combined regimen of SB plus TRAIL may offer an effective therapeutic strategy for safely and selectively treating TRAIL-resistant bladder cancer cells.
Keywords
Apoptosis; Caspase; DR5; Sodium butyrate; TRAIL ((TNF)-related apoptosis-inducing ligand);
Citations & Related Records
Times Cited By KSCI : 3  (Citation Analysis)
연도 인용수 순위
1 Abdulghani, J. and El-Deiry, W. S. 2010. TRAIL receptor signaling and therapeutics. Expert. Opin. Ther. Targets 14, 1091-1108.   DOI
2 Barneda-Zahonero, B. and Parra, M. 2012. Histone deacetylases and cancer. Mol. Oncol. 6, 579-589.   DOI
3 Benayoun, B., Baghdiguian, S., Lajmanovich, A., Bartoli, M., Daniele, N., Gicquel, E., Bourg, N., Raynaud, F., Pasquier, M. A., Suel, L., Lochmuller, H., Lefranc, G. and Richard, I. 2008. NF-kappaB-dependent expression of the antiapoptotic factor c-FLIP is regulated by calpain 3, the protein involved in limb-girdle muscular dystrophy type 2A. FASEB J. 22, 1521-1529.   DOI
4 Billen, L. P., Shamas-Din, A. and Andrews, D. W. 2008. Bid: a Bax-like BH3 protein. Oncogene 27, S93-104.   DOI
5 Carew, J. S., Giles, F. J. and Nawrocki, S. T. 2008. Histone deacetylase inhibitors: mechanisms of cell death and promise in combination cancer therapy. Cancer Lett. 269, 7-17.   DOI
6 Crowder, R. N. and El-Deiry, W. S. 2012. Caspase-8 regulation of TRAIL-mediated cell death. Exp. Oncol. 34, 160-164.
7 Donovan, M. and Cotter, T. G. 2004. Control of mitochondrial integrity by Bcl-2 family members and caspase-independent cell death. Biochim. Biophys. Acta 1644, 133-147.   DOI
8 Earel, J. K. Jr., VanOosten, R. L. and Griffith, T. S. 2006. Histone deacetylase inhibitors modulate the sensitivity of tumor necrosis factor-related apoptosis-inducing ligand-resistant bladder tumor cells. Cancer Res. 66, 499-507.   DOI
9 Fulda, S. 2012. Histone deacetylase (HDAC) inhibitors and regulation of TRAIL-induced apoptosis. Exp. Cell Res. 318, 1208-1212.   DOI
10 Ghobrial, I. M., Witzig, T. E. and Adjei, A. A. 2005. Targeting apoptosis pathways in cancer therapy. CA Cancer J. Clin. 55, 178-194.   DOI
11 Giménez-Bonafé, P., Tortosa, A. and Pérez-Tomás, R. 2009. Overcoming drug resistance by enhancing apoptosis of tumor cells. Curr. Cancer Drug Targets 9, 320-340.   DOI
12 Hussein, M. R., Haemel, A. K. and Wood, G. S. 2003. Apoptosis and melanoma: molecular mechanisms. J. Pathol. 199, 275-288.   DOI
13 Han, M. H., Park, C., Kwon, T. K., Kim, G. Y., Kim, W. J., Hong, S. H., Yoo, Y. H. and Choi, Y. H. 2015. The Histone Deacetylase Inhibitor Trichostatin A Sensitizes Human Renal Carcinoma Cells to TRAIL-Induced Apoptosis through Down-Regulation of c-FLIPL. Biomol. Ther. (Seoul) 23, 31-38.   DOI
14 Han, S. I., Kim, Y. S. and Kim, T. H. 2008. Role of apoptotic and necrotic cell death under physiologic conditions. BMB Rep. 41, 1-10.   DOI
15 Holcik, M., Gibson, H. and Korneluk, R. G. 2001. XIAP: apoptotic brake and promising therapeutic targe. Apoptosis 6, 253-261.   DOI
16 Inoue, H., Shiraki, K., Ohmori, S., Sakai, T., Deguchi, M., Yamanaka, T., Okano, H. and Nakano, T. 2002. Histone deacetylase inhibitors sensitize human colonic adenocarcinoma cell lines to TNF-related apoptosis inducing ligand-mediated apoptosis. Int. J. Mol. Med. 9, 521-525.
17 Jin, C. Y., Park, C., Cheong, J., Choi, B. T., Lee, T. H., Lee, J. D., Lee, W. H., Kim, G. Y., Ryu, C. H. and Choi, Y. H. 2007. Genistein sensitizes TRAIL-resistant human gastric adenocarcinoma AGS cells through activation of caspase-3. Cancer Lett. 257, 56-64.   DOI
18 Jin, C. Y., Park, C., Hong, S. H., Han, M. H., Jeong, J. W., Xu, H., Liu, H., Kim, G. Y., Kim, W. J., Yoo, Y. H. and Choi, Y. H. 2013. Synergistic induction of TRAIL-mediated apoptosis by anisomycin in human hepatoma cells via the BH3-only protein Bid and c-Jun/AP-1 signaling pathway. Biomed. Pharmacother. 67, 321-328.   DOI
19 Jin, C. Y., Park, C., Hwang, H. J., Kim, G. Y., Choi, B. T., Kim, W. J. and Choi, Y. H. 2011. Naringenin up-regulates the expression of death receptor 5 and enhances TRAIL-induced apoptosis in human lung cancer A549 cells. Mol. Nutr. Food Res. 55, 300-309.   DOI
20 Jin, Z. and El-Deiry, W. S. 2005. Overview of cell death signaling pathways. Cancer Biol. Ther. 4, 139-163.
21 Kantari, C. and Walczak, H. 2011. Caspase-8 and bid: caught in the act between death receptors and mitochondria. Biochim. Biophys. Acta 1813, 558-563.   DOI
22 Kelly, M. M., Hoel, B. D. and Voelkel-Johnson, C. 2002. Doxorubicin pretreatment sensitizes prostate cancer cell lines to TRAIL induced apoptosis which correlates with the loss of c-FLIP expression. Cancer Biol. Ther. 1, 520-527.   DOI
23 Kim, E. Y., Ryu, J. H. and Kim, A. K. 2013. CAPE promotes TRAIL-induced apoptosis through the upregulation of TRAIL receptors via activation of p38 and suppression of JNK in SK-Hep1 hepatocellular carcinoma cells. Int. J. Oncol. 43, 1291-1300.   DOI
24 Kim, Y. H., Park, J. W., Lee, J. Y. and Kwon, T. K. 2004. Sodium butyrate sensitizes TRAIL-mediated apoptosis by induction of transcription from the DR5 gene promoter through Sp1 sites in colon cancer cells. Carcinogenesis 25, 1813-1820.   DOI
25 Kouraklis, G. and Theocharis, S. 2002. Histone deacetylase inhibitors and anticancer therapy. Curr. Med. Chem. Anticancer Agents 2, 477-484.   DOI
26 Lazebnik, Y. A., Kaufmann, S. H., Desnoyers, S., Poirier, G. G. and Earnshaw, W. C. 1994. Cleavage of poly(ADP-ribose) polymerase by a proteinase with properties like ICE. Nature 371, 346-347.   DOI
27 Lee, H. H., Jeong, J. W., Lee, J. H., Kim, G. Y., Cheong, J., Jeong, Y. K., Yoo, Y. H. and Choi, Y. H. 2013. Cordycepin increases sensitivity of Hep3B human hepatocellular carcinoma cells to TRAIL-mediated apoptosis by inactivating the JNK signaling pathway. Oncol. Rep. 30, 1257-1264.   DOI
28 Mühlethaler-Mottet, A., Flahaut, M., Bourloud, K. B., Auderset, K., Meier, R., Joseph, J. M. and Gross, N. 2006. Histone deacetylase inhibitors strongly sensitise neuro-blastoma cells to TRAIL-induced apoptosis by a caspases-dependent increase of the pro- to anti-apoptotic proteins ratio. BMC Cancer 6, 214.   DOI
29 Mérino, D., Lalaoui, N., Morizot, A., Solary, E. and Micheau, O. 2007. TRAIL in cancer therapy: present and future challenges. Expert. Opin. Ther. Targets 11, 1299-1314.   DOI
30 Monneret, C. 2005. Histone deacetylase inhibitors. Eur. J. Med. Chem. 40, 1-13.   DOI
31 Park, C., Jin, C. Y., Hwang, H. J., Kim, G. Y., Jung, J. H., Kim, W. J., Yoo, Y. H. and Choi, Y. H. 2012. J7, a methyl jasmonate derivative, enhances TRAIL-mediated apoptosis through up-regulation of reactive oxygen species generation in human hepatoma HepG2 cells. Toxicol. In Vitro 26, 86-93.   DOI
32 Rae, C., Langa, S., Tucker, S. J. and MacEwan, D. J. 2007. Elevated NF-kappaB responses and FLIP levels in leukemic but not normal lymphocytes: reduction by salicylate allows TNF-induced apoptosis. Proc. Natl. Acad. Sci. USA 104, 12790-12795.   DOI
33 Rosato, R. R., Almenara, J. A., Dai, Y. and Grant, S. 2003. Simultaneous activation of the intrinsic and extrinsic pathways by histone deacetylase (HDAC) inhibitors and tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) synergistically induces mitochondrial damage and apoptosis in human leukemia cells. Mol. Cancer Ther. 2, 1273-1284.
34 Safa, A. R. and Pollok, K. E. 2011. Targeting the Anti-Apoptotic Protein c-FLIP for Cancer Therapy. Cancers (Basel) 3, 1639-1671.   DOI
35 Teraishi, F., Kagawa, S., Watanabe, T., Tango, Y., Kawashima, T., Umeoka, T., Nisizaki, M., Tanaka, N. and Fujiwara, T. 2005. ZD1839 (Gefitinib, 'Iressa'), an epidermal growth factor receptor-tyrosine kinase inhibitor, enhances the anti-cancer effects of TRAIL in human esophageal squamous cell carcinoma. FEBS Lett. 579, 4069-4075.   DOI
36 Seo, O. W., Kim, J. H., Lee, K. S., Lee, K. S., Kim, J. H., Won, M. H., Ha, K. S., Kwon, Y. G. and Kim, Y. M. 2012. Kurarinone promotes TRAIL-induced apoptosis by inhibiting NF-κB-dependent cFLIP expression in HeLa cells. Exp. Mol. Med. 44, 653-664.   DOI
37 Sonnemann, J., Gänge, J., Kumar, K. S., Müller, C., Bader, P. and Beck, J. F. 2005. Histone deacetylase inhibitors interact synergistically with tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) to induce apoptosis in carcinoma cell lines. Invest. New Drugs 23, 99-109.   DOI
38 Srivastava, R. K. 2001. TRAIL/Apo-2L: mechanisms and clinical applications in cancer. Neoplasia 3, 535-546.   DOI