Skip to main content
Top
Published in: Annals of Surgical Oncology 3/2013

01-12-2013 | Translational Research and Biomarkers

Valproic Acid Sensitizes TRAIL-Resistant Anaplastic Thyroid Carcinoma Cells to Apoptotic Cell Death

Authors: Hyun-Young Cha, Bok-Soon Lee, PhD, Sam Kang, Yoo Seob Shin, MD, Jae Won Chang, MD, Eun-Sil Sung, PhD, Yong-Sung Kim, PhD, Jae Won Choi, Jang Hee Kim, MD, Chul-Ho Kim, MD, PhD

Published in: Annals of Surgical Oncology | Special Issue 3/2013

Login to get access

Abstract

Background

Anaplastic thyroid carcinoma (ATC) is an aggressive human tumor associated with a median survival of 2–6 months. TRAIL, as a ligand of death receptors, is known to induce apoptotic cell death in several cancer cells. However, TRAIL treatment alone is not effective against TRAIL-resistant cancer cells. This study was designed to investigate whether valproic acid (VPA) enhances apoptotic cell death of TRAIL-resistant ATC cells and to identify the mechanism of cell death of ATC cells by combination treatment with VPA and TRAIL.

Methods

To evaluate the cytotoxic effect of TRAIL and/or VPA on ATC cells, we used the MTT assay. The effects of VPA and TRAIL on apoptosis were assessed using FACS analysis (Annexin-V/PI stain) and Western blotting.

Results

The combination of VPA with TRAIL significantly induced apoptotic cell death compared with 8505C and ARO cells treated with TRAIL alone. The protein levels of cleaved caspase-8, -3, and PARP were increased in VPA and TRAIL co-treated ARO cells. The combination induced the activation of JNK and the phosphorylation of FADD and c-Jun but not p38. However, pretreatment with caspase inhibitors reduced the expression of cleaved caspase-8, -3, and PARP in co-treated ARO cells. SP600125 remarkably reduced the expression of cleaved caspase-8, -3, and PARP and the phosphorylation of FADD and c-Jun, as well as apoptotic cell death.

Conclusions

VPA sensitized TRAIL-resistant ATC cells to apoptotic cell death through involvement of the JNK pathway. Thus, the combination of VPA and TRAIL may be a promising therapy for ATC.
Literature
2.
go back to reference Tan RK, Finley RK 3rd, Driscoll D, Bakamjian V, Hicks WL Jr, Shedd DP. Anaplastic carcinoma of the thyroid: a 24-year experience. Head Neck. 1995;17(1):41–7; discussion 47–8. Tan RK, Finley RK 3rd, Driscoll D, Bakamjian V, Hicks WL Jr, Shedd DP. Anaplastic carcinoma of the thyroid: a 24-year experience. Head Neck. 1995;17(1):41–7; discussion 47–8.
4.
go back to reference Conticello C, Adamo L, Giuffrida R, et al. Proteasome inhibitors synergize with tumor necrosis factor-related apoptosis-induced ligand to induce anaplastic thyroid carcinoma cell death. J Clin Endocrinol Metab. 2007;92(5):1938–42.PubMedCrossRef Conticello C, Adamo L, Giuffrida R, et al. Proteasome inhibitors synergize with tumor necrosis factor-related apoptosis-induced ligand to induce anaplastic thyroid carcinoma cell death. J Clin Endocrinol Metab. 2007;92(5):1938–42.PubMedCrossRef
5.
go back to reference Arlot-Bonnemains Y, Baldini E, Martin B, et al. Effects of the Aurora kinase inhibitor VX-680 on anaplastic thyroid cancer-derived cell lines. Endocr Relat Cancer. 2008;15(2):559–68.PubMedCrossRef Arlot-Bonnemains Y, Baldini E, Martin B, et al. Effects of the Aurora kinase inhibitor VX-680 on anaplastic thyroid cancer-derived cell lines. Endocr Relat Cancer. 2008;15(2):559–68.PubMedCrossRef
6.
go back to reference Libertini S, Abagnale A, Passaro C, et al. AZD1152 negatively affects the growth of anaplastic thyroid carcinoma cells and enhances the effects of oncolytic virus dl922-947. Endocr Relat Cancer. 2011;18(1):129–41.PubMedCrossRef Libertini S, Abagnale A, Passaro C, et al. AZD1152 negatively affects the growth of anaplastic thyroid carcinoma cells and enhances the effects of oncolytic virus dl922-947. Endocr Relat Cancer. 2011;18(1):129–41.PubMedCrossRef
7.
go back to reference Wunderlich A, Roth S, Ramaswamy A, et al. Combined inhibition of cellular pathways as a future therapeutic option in fatal anaplastic thyroid cancer. Endocrine. 2012;42(3):637–46.PubMedCrossRef Wunderlich A, Roth S, Ramaswamy A, et al. Combined inhibition of cellular pathways as a future therapeutic option in fatal anaplastic thyroid cancer. Endocrine. 2012;42(3):637–46.PubMedCrossRef
8.
go back to reference Ahmad M, Shi Y. TRAIL-induced apoptosis of thyroid cancer cells: potential for therapeutic intervention. Oncogene. 2000;19(30):3363–71.PubMedCrossRef Ahmad M, Shi Y. TRAIL-induced apoptosis of thyroid cancer cells: potential for therapeutic intervention. Oncogene. 2000;19(30):3363–71.PubMedCrossRef
9.
go back to reference LeBlanc HN, Ashkenazi A. Apo2L/TRAIL and its death and decoy receptors. Cell Death Differ. 2003;10(1):66–75.PubMedCrossRef LeBlanc HN, Ashkenazi A. Apo2L/TRAIL and its death and decoy receptors. Cell Death Differ. 2003;10(1):66–75.PubMedCrossRef
10.
go back to reference Ozoren N, El-Deiry WS. Cell surface death receptor signaling in normal and cancer cells. Semin Cancer Biol. 2003;13(2):135–47.PubMedCrossRef Ozoren N, El-Deiry WS. Cell surface death receptor signaling in normal and cancer cells. Semin Cancer Biol. 2003;13(2):135–47.PubMedCrossRef
11.
go back to reference Sung ES, Park KJ, Choi HJ, Kim CH, Kim YS. The proteasome inhibitor MG132 potentiates TRAIL receptor agonist-induced apoptosis by stabilizing tBid and Bik in human head and neck squamous cell carcinoma cells. Exp Cell Res. 2012;318(13):1564–76.PubMedCrossRef Sung ES, Park KJ, Choi HJ, Kim CH, Kim YS. The proteasome inhibitor MG132 potentiates TRAIL receptor agonist-induced apoptosis by stabilizing tBid and Bik in human head and neck squamous cell carcinoma cells. Exp Cell Res. 2012;318(13):1564–76.PubMedCrossRef
12.
go back to reference Kahana S, Finniss S, Cazacu S, et al. Proteasome inhibitors sensitize glioma cells and glioma stem cells to TRAIL-induced apoptosis by PKCepsilon-dependent downregulation of AKT and XIAP expressions. Cell Signal. 2011;23(8):1348–57.PubMedCrossRef Kahana S, Finniss S, Cazacu S, et al. Proteasome inhibitors sensitize glioma cells and glioma stem cells to TRAIL-induced apoptosis by PKCepsilon-dependent downregulation of AKT and XIAP expressions. Cell Signal. 2011;23(8):1348–57.PubMedCrossRef
13.
go back to reference Sung ES, Kim A, Park JS, Chung J, Kwon MH, Kim YS. Histone deacetylase inhibitors synergistically potentiate death receptor 4-mediated apoptotic cell death of human T-cell acute lymphoblastic leukemia cells. Apoptosis. 2010;15(10):1256–69.PubMedCrossRef Sung ES, Kim A, Park JS, Chung J, Kwon MH, Kim YS. Histone deacetylase inhibitors synergistically potentiate death receptor 4-mediated apoptotic cell death of human T-cell acute lymphoblastic leukemia cells. Apoptosis. 2010;15(10):1256–69.PubMedCrossRef
14.
go back to reference Srivastava RK, Kurzrock R, Shankar S. MS-275 sensitizes TRAIL-resistant breast cancer cells, inhibits angiogenesis and metastasis, and reverses epithelial-mesenchymal transition in vivo. Mol Cancer Ther. 2010;9(12):3254–66.PubMedCrossRef Srivastava RK, Kurzrock R, Shankar S. MS-275 sensitizes TRAIL-resistant breast cancer cells, inhibits angiogenesis and metastasis, and reverses epithelial-mesenchymal transition in vivo. Mol Cancer Ther. 2010;9(12):3254–66.PubMedCrossRef
15.
go back to reference Iacomino G, Medici MC, Russo GL. Valproic acid sensitizes K562 erythroleukemia cells to TRAIL/Apo2L-induced apoptosis. Anticancer Res. 2008;28(2A):855–64.PubMed Iacomino G, Medici MC, Russo GL. Valproic acid sensitizes K562 erythroleukemia cells to TRAIL/Apo2L-induced apoptosis. Anticancer Res. 2008;28(2A):855–64.PubMed
16.
go back to reference Brautigam K, Biernath-Wupping J, Bauerschlag DO, et al. Combined treatment with TRAIL and PPARγ ligands overcomes chemoresistance of ovarian cancer cell lines. J Cancer Res Clin Oncol. 2011;137(5):875–86.PubMedCrossRef Brautigam K, Biernath-Wupping J, Bauerschlag DO, et al. Combined treatment with TRAIL and PPARγ ligands overcomes chemoresistance of ovarian cancer cell lines. J Cancer Res Clin Oncol. 2011;137(5):875–86.PubMedCrossRef
17.
go back to reference Munshi A, McDonnell TJ, Meyn RE. Chemotherapeutic agents enhance TRAIL-induced apoptosis in prostate cancer cells. Cancer Chemother Pharmacol. 2002;50(1):46-52.PubMedCrossRef Munshi A, McDonnell TJ, Meyn RE. Chemotherapeutic agents enhance TRAIL-induced apoptosis in prostate cancer cells. Cancer Chemother Pharmacol. 2002;50(1):46-52.PubMedCrossRef
18.
go back to reference Nakata S, Yoshida T, Horinaka M, Shiraishi T, Wakada M, Sakai T. Histone deacetylase inhibitors upregulate death receptor 5/TRAIL-R2 and sensitize apoptosis induced by TRAIL/APO2-L in human malignant tumor cells. Oncogene. 2004;23(37):6261–71.PubMedCrossRef Nakata S, Yoshida T, Horinaka M, Shiraishi T, Wakada M, Sakai T. Histone deacetylase inhibitors upregulate death receptor 5/TRAIL-R2 and sensitize apoptosis induced by TRAIL/APO2-L in human malignant tumor cells. Oncogene. 2004;23(37):6261–71.PubMedCrossRef
19.
go back to reference Nightingale KP, O’Neill LP, Turner BM. Histone modifications: signalling receptors and potential elements of a heritable epigenetic code. Curr Opin Gen Dev. 2006;16(2):125–36.PubMedCrossRef Nightingale KP, O’Neill LP, Turner BM. Histone modifications: signalling receptors and potential elements of a heritable epigenetic code. Curr Opin Gen Dev. 2006;16(2):125–36.PubMedCrossRef
20.
go back to reference Kouzarides T. Histone acetylases and deacetylases in cell proliferation. Curr Opin Gen Dev. 1999;9(1):40–8.CrossRef Kouzarides T. Histone acetylases and deacetylases in cell proliferation. Curr Opin Gen Dev. 1999;9(1):40–8.CrossRef
21.
go back to reference Csordas A. On the biological role of histone acetylation. Biochem J. 1990;265(1):23–38.PubMed Csordas A. On the biological role of histone acetylation. Biochem J. 1990;265(1):23–38.PubMed
22.
go back to reference Fulda S. Histone deacetylase (HDAC) inhibitors and regulation of TRAIL-induced apoptosis. Exp Cell Res. 2012;318(11):1208–12.PubMedCrossRef Fulda S. Histone deacetylase (HDAC) inhibitors and regulation of TRAIL-induced apoptosis. Exp Cell Res. 2012;318(11):1208–12.PubMedCrossRef
23.
go back to reference Perego P, Zuco V, Gatti L, Zunino F. Sensitization of tumor cells by targeting histone deacetylases. Biochem Pharmacol. 2012;83(8):987–94.PubMedCrossRef Perego P, Zuco V, Gatti L, Zunino F. Sensitization of tumor cells by targeting histone deacetylases. Biochem Pharmacol. 2012;83(8):987–94.PubMedCrossRef
24.
go back to reference Frew AJ, Lindemann RK, Martin BP, et al. Combination therapy of established cancer using a histone deacetylase inhibitor and a TRAIL receptor agonist. Proc Natl Acad Sci USA. 2008;105(32):11317–22.PubMedCrossRef Frew AJ, Lindemann RK, Martin BP, et al. Combination therapy of established cancer using a histone deacetylase inhibitor and a TRAIL receptor agonist. Proc Natl Acad Sci USA. 2008;105(32):11317–22.PubMedCrossRef
25.
go back to reference Carew JS, Giles FJ, Nawrocki ST. Histone deacetylase inhibitors: mechanisms of cell death and promise in combination cancer therapy. Cancer Lett. 2008;269(1):7–17.PubMedCrossRef Carew JS, Giles FJ, Nawrocki ST. Histone deacetylase inhibitors: mechanisms of cell death and promise in combination cancer therapy. Cancer Lett. 2008;269(1):7–17.PubMedCrossRef
26.
go back to reference Lee BS, Cha HY, Shin YS, Kim YS, Kim CH. AY4, an agonistic anti-death receptor 4 monoclonal antibody, induces apoptotic cell death in anaplastic thyroid cancer cells via down-regulation of Bcl-xL with reactive oxygen species generation. Endocr Relat Cancer. 2013;20:283–91.PubMedCrossRef Lee BS, Cha HY, Shin YS, Kim YS, Kim CH. AY4, an agonistic anti-death receptor 4 monoclonal antibody, induces apoptotic cell death in anaplastic thyroid cancer cells via down-regulation of Bcl-xL with reactive oxygen species generation. Endocr Relat Cancer. 2013;20:283–91.PubMedCrossRef
27.
go back to reference Lee BS, Kang SU, Hwang HS, et al. An agonistic antibody to human death receptor 4 induces apoptotic cell death in head and neck cancer cells through mitochondrial ROS generation. Cancer Lett. 2012;322(1):45–57.PubMedCrossRef Lee BS, Kang SU, Hwang HS, et al. An agonistic antibody to human death receptor 4 induces apoptotic cell death in head and neck cancer cells through mitochondrial ROS generation. Cancer Lett. 2012;322(1):45–57.PubMedCrossRef
28.
go back to reference Lim YC, Park HY, Hwang HS, et al. (-)-Epigallocatechin-3-gallate (EGCG) inhibits HGF-induced invasion and metastasis in hypopharyngeal carcinoma cells. Cancer Lett. 2008;271(1):140–52.PubMedCrossRef Lim YC, Park HY, Hwang HS, et al. (-)-Epigallocatechin-3-gallate (EGCG) inhibits HGF-induced invasion and metastasis in hypopharyngeal carcinoma cells. Cancer Lett. 2008;271(1):140–52.PubMedCrossRef
29.
go back to reference Kim CH, Koh YW, Han JH, et al. c-Met expression as an indicator of survival outcome in patients with oral tongue carcinoma. Head Neck. 2010;32(12):1655–64.PubMed Kim CH, Koh YW, Han JH, et al. c-Met expression as an indicator of survival outcome in patients with oral tongue carcinoma. Head Neck. 2010;32(12):1655–64.PubMed
30.
go back to reference Tang F, Tang G, Xiang J, Dai Q, Rosner MR, Lin A. The absence of NF-kappaB-mediated inhibition of c-Jun N-terminal kinase activation contributes to tumor necrosis factor alpha-induced apoptosis. Mol Cell Biol. 2002;22(24):8571–9.PubMedCrossRef Tang F, Tang G, Xiang J, Dai Q, Rosner MR, Lin A. The absence of NF-kappaB-mediated inhibition of c-Jun N-terminal kinase activation contributes to tumor necrosis factor alpha-induced apoptosis. Mol Cell Biol. 2002;22(24):8571–9.PubMedCrossRef
31.
go back to reference Muscarella DE, Bloom SE. The contribution of c-Jun N-terminal kinase activation and subsequent Bcl-2 phosphorylation to apoptosis induction in human B-cells is dependent on the mode of action of specific stresses. Toxicol Appl Pharmacol. 2008;228(1):93–104.PubMedCrossRef Muscarella DE, Bloom SE. The contribution of c-Jun N-terminal kinase activation and subsequent Bcl-2 phosphorylation to apoptosis induction in human B-cells is dependent on the mode of action of specific stresses. Toxicol Appl Pharmacol. 2008;228(1):93–104.PubMedCrossRef
32.
33.
go back to reference Luong QT, O’Kelly J, Braunstein GD, Hershman JM, Koeffler HP. Antitumor activity of suberoylanilide hydroxamic acid against thyroid cancer cell lines in vitro and in vivo. Clin Cancer Res. 2006;12(18):5570–7.PubMedCrossRef Luong QT, O’Kelly J, Braunstein GD, Hershman JM, Koeffler HP. Antitumor activity of suberoylanilide hydroxamic acid against thyroid cancer cell lines in vitro and in vivo. Clin Cancer Res. 2006;12(18):5570–7.PubMedCrossRef
34.
go back to reference Sonnemann J, Gange J, Kumar KS, Muller C, Bader P, Beck JF. Histone deacetylase inhibitors interact synergistically with tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) to induce apoptosis in carcinoma cell lines. Investig New Drugs. 2005;23(2):99–109.CrossRef Sonnemann J, Gange J, Kumar KS, Muller C, Bader P, Beck JF. Histone deacetylase inhibitors interact synergistically with tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) to induce apoptosis in carcinoma cell lines. Investig New Drugs. 2005;23(2):99–109.CrossRef
35.
go back to reference Chopin V, Slomianny C, Hondermarck H, Le Bourhis X. Synergistic induction of apoptosis in breast cancer cells by cotreatment with butyrate and TNF-alpha, TRAIL, or anti-Fas agonist antibody involves enhancement of death receptors’ signaling and requires P21(waf1). Exp Cell Res. 2004;298(2):560–73.PubMedCrossRef Chopin V, Slomianny C, Hondermarck H, Le Bourhis X. Synergistic induction of apoptosis in breast cancer cells by cotreatment with butyrate and TNF-alpha, TRAIL, or anti-Fas agonist antibody involves enhancement of death receptors’ signaling and requires P21(waf1). Exp Cell Res. 2004;298(2):560–73.PubMedCrossRef
36.
go back to reference Fan M, Goodwin ME, Birrer MJ, Chambers TC. The c-Jun NH(2)-terminal protein kinase/AP-1 pathway is required for efficient apoptosis induced by vinblastine. Cancer Res. 2001;61(11):4450–8.PubMed Fan M, Goodwin ME, Birrer MJ, Chambers TC. The c-Jun NH(2)-terminal protein kinase/AP-1 pathway is required for efficient apoptosis induced by vinblastine. Cancer Res. 2001;61(11):4450–8.PubMed
37.
go back to reference Sharma V, Koul N, Joseph C, Dixit D, Ghosh S, Sen E. HDAC inhibitor, scriptaid, induces glioma cell apoptosis through JNK activation and inhibits telomerase activity. J Cell Mol Med. 2010;14(8):2151–61.PubMedCrossRef Sharma V, Koul N, Joseph C, Dixit D, Ghosh S, Sen E. HDAC inhibitor, scriptaid, induces glioma cell apoptosis through JNK activation and inhibits telomerase activity. J Cell Mol Med. 2010;14(8):2151–61.PubMedCrossRef
38.
go back to reference Xiao D, Pinto JT, Soh JW, et al. Induction of apoptosis by the garlic-derived compound S-allylmercaptocysteine (SAMC) is associated with microtubule depolymerization and c-Jun NH(2)-terminal kinase 1 activation. Cancer Res. 2003;63(20):6825–37.PubMed Xiao D, Pinto JT, Soh JW, et al. Induction of apoptosis by the garlic-derived compound S-allylmercaptocysteine (SAMC) is associated with microtubule depolymerization and c-Jun NH(2)-terminal kinase 1 activation. Cancer Res. 2003;63(20):6825–37.PubMed
39.
go back to reference Benoit M, Dormond-Meuwly A, Demartines N, Dormond O. Targeting the JNK signaling pathway potentiates the antiproliferative efficacy of rapamycin in LS174T colon cancer cells. J Surg Res. 2011;167(2):e193–8.PubMedCrossRef Benoit M, Dormond-Meuwly A, Demartines N, Dormond O. Targeting the JNK signaling pathway potentiates the antiproliferative efficacy of rapamycin in LS174T colon cancer cells. J Surg Res. 2011;167(2):e193–8.PubMedCrossRef
40.
go back to reference Higuchi H, Grambihler A, Canbay A, Bronk SF, Gores GJ. Bile acids up-regulate death receptor 5/TRAIL-receptor 2 expression via a c-Jun N-terminal kinase-dependent pathway involving Sp1. J Biol Chem. 2004;279(1):51–60.PubMedCrossRef Higuchi H, Grambihler A, Canbay A, Bronk SF, Gores GJ. Bile acids up-regulate death receptor 5/TRAIL-receptor 2 expression via a c-Jun N-terminal kinase-dependent pathway involving Sp1. J Biol Chem. 2004;279(1):51–60.PubMedCrossRef
41.
go back to reference Hwang MK, Ryu BJ, Kim SH. AW00179 potentiates TRAIL-mediated death of human lung cancer H1299 cells through ROS-JNK-c-Jun-mediated up-regulation of DR5 and down-regulation of anti-apoptotic molecules. Amino Acids. 2012;43(4):1679–87.PubMedCrossRef Hwang MK, Ryu BJ, Kim SH. AW00179 potentiates TRAIL-mediated death of human lung cancer H1299 cells through ROS-JNK-c-Jun-mediated up-regulation of DR5 and down-regulation of anti-apoptotic molecules. Amino Acids. 2012;43(4):1679–87.PubMedCrossRef
Metadata
Title
Valproic Acid Sensitizes TRAIL-Resistant Anaplastic Thyroid Carcinoma Cells to Apoptotic Cell Death
Authors
Hyun-Young Cha
Bok-Soon Lee, PhD
Sam Kang
Yoo Seob Shin, MD
Jae Won Chang, MD
Eun-Sil Sung, PhD
Yong-Sung Kim, PhD
Jae Won Choi
Jang Hee Kim, MD
Chul-Ho Kim, MD, PhD
Publication date
01-12-2013
Publisher
Springer US
Published in
Annals of Surgical Oncology / Issue Special Issue 3/2013
Print ISSN: 1068-9265
Electronic ISSN: 1534-4681
DOI
https://doi.org/10.1245/s10434-013-3232-y

Other articles of this Special Issue 3/2013

Annals of Surgical Oncology 3/2013 Go to the issue