Skip to main content
Top
Published in: Medical Oncology 2/2012

01-06-2012 | Original Paper

Assessment of the involvement of oxidative stress and Mitogen-Activated Protein Kinase signaling pathways in the cytotoxic effects of arsenic trioxide and its combination with sulindac or its metabolites: sulindac sulfide and sulindac sulfone on human leukemic cell lines

Authors: M. Stępnik, M. Ferlińska, A. Smok-Pieniążek, D. Gradecka-Meesters, J. Arkusz, M. Stańczyk

Published in: Medical Oncology | Issue 2/2012

Login to get access

Abstract

The purpose of the study was to characterize the involvement of reactive oxygen species (ROS) in mediating the cytotoxic effects of arsenic trioxide (ATO) in combination with sulindac or its metabolites: sulfide (SS) and sulfone (SF) on human leukemic cell lines. Jurkat, HL-60, K562, and HPB-ALL cells were exposed to the drugs alone or in combinations. Cell viability was measured using WST-1 or XTT reduction tests and ROS production by dichlorodihydrofluorescein diacetate staining (flow cytometry). Modulation of (a) intracellular glutathione (GSH) level was done by using l-buthionine sulfoximine (BSO) or diethylmaleate (DEM), (b) NADPH oxidase by using diphenyleneiodonium (DPI), and (c) MAP kinases by using SB202190 (p38), SP600125 (JNK), and U0126 (ERK) inhibitors. ATO cytotoxicity (0.5 or 1 μM) was enhanced by sulindacs, with higher activity showed by the metabolites. Strong cytotoxic effects appeared at SS and SF concentrations starting from 50 μM. The induction of ROS production seemed not to be the major mechanism responsible for the cytotoxicity of the combinations. A strong potentiating effect of BSO on ATO cytotoxicity was demonstrated; DEM (10–300 μM) and DPI (0.0025–0.1 μM; 72 h) did not influence the effects of ATO. Some significant decreases in the viability of the cells exposed to ATO in the presence of MAPK inhibitors comparing with the cells exposed to ATO alone were observed; however, the effects likely resulted from a simple additive cytotoxicity of the drugs. The combinations of ATO with sulindacs offer potential therapeutic usefulness.
Literature
1.
go back to reference Giovannucci E, Rimm EB, Stampfer MJ, et al. Aspirin use and the risk for colorectal cancer and adenoma in male health professionals. Ann Int Med. 1994;121:241–6.PubMed Giovannucci E, Rimm EB, Stampfer MJ, et al. Aspirin use and the risk for colorectal cancer and adenoma in male health professionals. Ann Int Med. 1994;121:241–6.PubMed
2.
go back to reference Giardiello FM, Yang VW, Hylind LM, et al. Primary chemoprevention of familial adenomatous polyposis with sulindac. N Engl J Med. 2002;346:1054–9.PubMedCrossRef Giardiello FM, Yang VW, Hylind LM, et al. Primary chemoprevention of familial adenomatous polyposis with sulindac. N Engl J Med. 2002;346:1054–9.PubMedCrossRef
3.
go back to reference Labayle D, Fischer D, Vielh P, et al. Sulindac causes regression of rectal polyps in familial adenomatous polyposis. Gastroenterology. 1991;101:635–9.PubMed Labayle D, Fischer D, Vielh P, et al. Sulindac causes regression of rectal polyps in familial adenomatous polyposis. Gastroenterology. 1991;101:635–9.PubMed
4.
go back to reference Oshima M, Dinchuk JE, Kargman SL, et al. Suppression of intestinal polyposis in Apc delta716 knockout mice by inhibition of cyclooxygenase 2 (COX-2). Cell. 1996;87:803–9.PubMedCrossRef Oshima M, Dinchuk JE, Kargman SL, et al. Suppression of intestinal polyposis in Apc delta716 knockout mice by inhibition of cyclooxygenase 2 (COX-2). Cell. 1996;87:803–9.PubMedCrossRef
5.
6.
go back to reference Yamamoto Y, Yin MJ, Lin KM, Gaynor R. Sulindac inhibits activation of NF-κB pathway. J Biol Chem. 1999;274:27307–14.PubMedCrossRef Yamamoto Y, Yin MJ, Lin KM, Gaynor R. Sulindac inhibits activation of NF-κB pathway. J Biol Chem. 1999;274:27307–14.PubMedCrossRef
7.
go back to reference Shiff SF, Qiao L, Tsai LL, Rigas B. Sulindac sulfide, an aspirin-like compound, inhibits proliferation, causes cell cycle quiescence, and induces apoptosis in HT-29 colon adenocarcinoma cells. J Clin Invest. 1995;96:491–503.PubMedCrossRef Shiff SF, Qiao L, Tsai LL, Rigas B. Sulindac sulfide, an aspirin-like compound, inhibits proliferation, causes cell cycle quiescence, and induces apoptosis in HT-29 colon adenocarcinoma cells. J Clin Invest. 1995;96:491–503.PubMedCrossRef
8.
go back to reference Hanif R, Pittas A, Feng Y, et al. Effects of nonsteroidal anti-inflammatory drugs on proliferation and on induction of apoptosis in colon cancer cells by a prostaglandin-independent pathway. Biochem Pharmacol. 1996;52:237–45.PubMedCrossRef Hanif R, Pittas A, Feng Y, et al. Effects of nonsteroidal anti-inflammatory drugs on proliferation and on induction of apoptosis in colon cancer cells by a prostaglandin-independent pathway. Biochem Pharmacol. 1996;52:237–45.PubMedCrossRef
9.
go back to reference Han EK, Arber N, Yamamoto H, et al. Effects of sulindac and its metabolites on growth and apoptosis in human mammary epithelial and breast carcinoma cell lines. Breast Cancer Res Treat. 1998;48:195–203.PubMedCrossRef Han EK, Arber N, Yamamoto H, et al. Effects of sulindac and its metabolites on growth and apoptosis in human mammary epithelial and breast carcinoma cell lines. Breast Cancer Res Treat. 1998;48:195–203.PubMedCrossRef
10.
go back to reference Rahman MA, Dhar DK, Masunaga R, et al. Sulindac and exisulind exibit a significant antiproliferative effect and induce apoptosis in human hepatocellular carcinoma cell lines. Cancer Res. 2000;60:2085–9.PubMed Rahman MA, Dhar DK, Masunaga R, et al. Sulindac and exisulind exibit a significant antiproliferative effect and induce apoptosis in human hepatocellular carcinoma cell lines. Cancer Res. 2000;60:2085–9.PubMed
11.
go back to reference Lim JTE, Piazza GA, Pamukcu R, et al. Exisulind and related compounds inhibit expression and function of the androgen receptor in human prostate cancer cells. Clin Cancer Res. 2003;9:4972–82.PubMed Lim JTE, Piazza GA, Pamukcu R, et al. Exisulind and related compounds inhibit expression and function of the androgen receptor in human prostate cancer cells. Clin Cancer Res. 2003;9:4972–82.PubMed
12.
go back to reference Piazza GA, Alberts DS, Hixson LJ, et al. Sulindac sulfone inhibits azoxymethane-induced colon carcinogenesis in rats without reducing prostaglandin levels. Cancer Res. 1997;57:2909–15.PubMed Piazza GA, Alberts DS, Hixson LJ, et al. Sulindac sulfone inhibits azoxymethane-induced colon carcinogenesis in rats without reducing prostaglandin levels. Cancer Res. 1997;57:2909–15.PubMed
13.
go back to reference Thompson HJ, Briggs S, Paranka NS, et al. Sulfone metabolite of sulindac inhibits mammary carcinonogenesis. J Nat Cancer Inst. 1995;87:1259–60.PubMed Thompson HJ, Briggs S, Paranka NS, et al. Sulfone metabolite of sulindac inhibits mammary carcinonogenesis. J Nat Cancer Inst. 1995;87:1259–60.PubMed
14.
go back to reference Soriano AF, Helfrich B, Chan DC, et al. Synergistic effects of new chemopreventive agents and conventional cytotoxic agents against human lung cancer cell lines. Cancer Res. 1999;59:6178–84.PubMed Soriano AF, Helfrich B, Chan DC, et al. Synergistic effects of new chemopreventive agents and conventional cytotoxic agents against human lung cancer cell lines. Cancer Res. 1999;59:6178–84.PubMed
15.
go back to reference Chan DC, Earle KA, Zhao TL, et al. Exisulind in combination with docetaxel inhibits growth and metastasis of human lung cancer and prolongs survival in athymic nude rats with orthotopic lung tumors. Clin Cancer Res. 2002;8:904–12.PubMed Chan DC, Earle KA, Zhao TL, et al. Exisulind in combination with docetaxel inhibits growth and metastasis of human lung cancer and prolongs survival in athymic nude rats with orthotopic lung tumors. Clin Cancer Res. 2002;8:904–12.PubMed
16.
go back to reference Whitehead CM, Earle KA, Fetter J, et al. Exisulind-induced apoptosis in a non-small cell lung cancer orthotopic lung tumor model augments docetaxel treatment and contributes to increased survival. Mol Cancer Ther. 2003;2:479–88.PubMed Whitehead CM, Earle KA, Fetter J, et al. Exisulind-induced apoptosis in a non-small cell lung cancer orthotopic lung tumor model augments docetaxel treatment and contributes to increased survival. Mol Cancer Ther. 2003;2:479–88.PubMed
17.
go back to reference Piazza GA, Rahm AK, Finn TS, et al. Apoptosis primarily accounts for the growth-inhibitory properties of sulindac metabolites and involves a mechanism that is independent of cyclooxygenase inhibition, cell cycle arrest, and p53 induction. Cancer Res. 1997;57:2452–9.PubMed Piazza GA, Rahm AK, Finn TS, et al. Apoptosis primarily accounts for the growth-inhibitory properties of sulindac metabolites and involves a mechanism that is independent of cyclooxygenase inhibition, cell cycle arrest, and p53 induction. Cancer Res. 1997;57:2452–9.PubMed
18.
go back to reference Thompson WJ, Piazza GA, Li H, et al. Exisulind induction of apoptosis involves guanosine 3′, 5′-cyclic monophosphate phosphodiesterase inhibition, protein kinase G activation, and attenuated beta-catenin. Cancer Res. 2000;60:3338–42.PubMed Thompson WJ, Piazza GA, Li H, et al. Exisulind induction of apoptosis involves guanosine 3′, 5′-cyclic monophosphate phosphodiesterase inhibition, protein kinase G activation, and attenuated beta-catenin. Cancer Res. 2000;60:3338–42.PubMed
19.
go back to reference Soh JW, Mao Y, Kim MG, et al. Cyclic GMP mediates apoptosis induced by sulindac derivatives via activation of c-Jun NH2-terminal kinase 1. Clin Cancer Res. 2000;6:4136–41.PubMed Soh JW, Mao Y, Kim MG, et al. Cyclic GMP mediates apoptosis induced by sulindac derivatives via activation of c-Jun NH2-terminal kinase 1. Clin Cancer Res. 2000;6:4136–41.PubMed
20.
go back to reference Czibere A, Prall WC, Zerbini LF, et al. Exisulind induces apoptosis in advanced myelodysplastic syndrome (MDS) and acute myeloid leukaemia/MDS. Br J Haematol. 2006;135:355–7.PubMedCrossRef Czibere A, Prall WC, Zerbini LF, et al. Exisulind induces apoptosis in advanced myelodysplastic syndrome (MDS) and acute myeloid leukaemia/MDS. Br J Haematol. 2006;135:355–7.PubMedCrossRef
21.
go back to reference Zhang T, Westervelt P, Hess JL. Pathologic, cytogenetic and molecular assessment of acute promyelocytic leukemia patients treated with arsenic trioxide (As2O3). Mod Pathol. 2000;13:954–61.PubMedCrossRef Zhang T, Westervelt P, Hess JL. Pathologic, cytogenetic and molecular assessment of acute promyelocytic leukemia patients treated with arsenic trioxide (As2O3). Mod Pathol. 2000;13:954–61.PubMedCrossRef
23.
24.
go back to reference Chen D, Chan R, Waxman S, Jing Y. Buthionine sulfoximine enhancement of arsenic trioxide-induced apoptosis in leukemia and lymphoma cells is mediated via activation of c-Jun NH2-terminal kinase and up-regulation of death receptors. Cancer Res. 2006;66:11416–23.PubMedCrossRef Chen D, Chan R, Waxman S, Jing Y. Buthionine sulfoximine enhancement of arsenic trioxide-induced apoptosis in leukemia and lymphoma cells is mediated via activation of c-Jun NH2-terminal kinase and up-regulation of death receptors. Cancer Res. 2006;66:11416–23.PubMedCrossRef
25.
go back to reference Grad JM, Bahlis NJ, Reis I, et al. Ascorbic acid enhances arsenic trioxide-induced cytotoxicity in multiple myeloma cells. Blood. 2001;98:805–13.PubMedCrossRef Grad JM, Bahlis NJ, Reis I, et al. Ascorbic acid enhances arsenic trioxide-induced cytotoxicity in multiple myeloma cells. Blood. 2001;98:805–13.PubMedCrossRef
26.
go back to reference Qazilbash MH, Saliba RM, Nieto Y, et al. Arsenic trioxide with ascorbic acid and high-dose melphalan: results of a phase II randomized trial. Biol Blood Marrow Transplant. 2008;14:1401–7.PubMedCrossRef Qazilbash MH, Saliba RM, Nieto Y, et al. Arsenic trioxide with ascorbic acid and high-dose melphalan: results of a phase II randomized trial. Biol Blood Marrow Transplant. 2008;14:1401–7.PubMedCrossRef
27.
go back to reference Roboz GJ, Ritchie EK, Curcio T, et al. Arsenic trioxide and low-dose cytarabine in older patients with untreated acute myeloid leukemia, excluding acute promyelocytic leukemia. Cancer. 2008;113:2504–11.PubMedCrossRef Roboz GJ, Ritchie EK, Curcio T, et al. Arsenic trioxide and low-dose cytarabine in older patients with untreated acute myeloid leukemia, excluding acute promyelocytic leukemia. Cancer. 2008;113:2504–11.PubMedCrossRef
28.
go back to reference Jin HO, Yoon SI, Seo SK, et al. Synergistic induction of apoptosis by sulindac and arsenic trioxide in human lung cancer A549 cells via reactive oxygen species-dependent down-regulation of survivin. Biochem Pharmacol. 2006;72:1228–36.PubMedCrossRef Jin HO, Yoon SI, Seo SK, et al. Synergistic induction of apoptosis by sulindac and arsenic trioxide in human lung cancer A549 cells via reactive oxygen species-dependent down-regulation of survivin. Biochem Pharmacol. 2006;72:1228–36.PubMedCrossRef
29.
go back to reference Park JH, Kim EJ, Jang HY, et al. Combination treatment with arsenic trioxide and sulindac enhances apoptotic cell death in lung cancer cells via activation of oxidative stress and mitogen-activated protein kinases. Oncol Rep. 2008;20:379–84.PubMed Park JH, Kim EJ, Jang HY, et al. Combination treatment with arsenic trioxide and sulindac enhances apoptotic cell death in lung cancer cells via activation of oxidative stress and mitogen-activated protein kinases. Oncol Rep. 2008;20:379–84.PubMed
30.
go back to reference Jin HO, Seo SK, Woo SH, et al. A combination of sulindac and arsenic trioxide synergistically induces apoptosis in human lung cancer H1299 cells via c-Jun NH2-terminal kinase-dependent Bcl-xL phosphorylation. Lung Cancer. 2008;61:317–27.PubMedCrossRef Jin HO, Seo SK, Woo SH, et al. A combination of sulindac and arsenic trioxide synergistically induces apoptosis in human lung cancer H1299 cells via c-Jun NH2-terminal kinase-dependent Bcl-xL phosphorylation. Lung Cancer. 2008;61:317–27.PubMedCrossRef
31.
go back to reference Lee HR, Cheong HJ, Kim SJ, et al. Sulindac enhances arsenic trioxide-mediated apoptosis by inhibition of NF-kappaB in HCT116 colon cancer cells. Oncol Rep. 2008;20:41–7.PubMed Lee HR, Cheong HJ, Kim SJ, et al. Sulindac enhances arsenic trioxide-mediated apoptosis by inhibition of NF-kappaB in HCT116 colon cancer cells. Oncol Rep. 2008;20:41–7.PubMed
32.
go back to reference Jiang TT, Brown SL, Kim JH. Combined effect of arsenic trioxide and sulindac sulfide in A549 human lung cancer cells in vitro. J Exp Clin Cancer Res. 2004;23:259–62.PubMed Jiang TT, Brown SL, Kim JH. Combined effect of arsenic trioxide and sulindac sulfide in A549 human lung cancer cells in vitro. J Exp Clin Cancer Res. 2004;23:259–62.PubMed
33.
go back to reference Stępnik M, Ferlińska M, Smok-Pieniążek A, Gradecka-Meesters D, Arkusz J, Stańczyk M. Sulindac and its metabolites: sulindac sulfide and sulindac sulfone enhance cytotoxic effects of arsenic trioxide on leukemic cell lines. Toxicol Vitro. doi:10.1016/j.tiv.2011.04.011. Stępnik M, Ferlińska M, Smok-Pieniążek A, Gradecka-Meesters D, Arkusz J, Stańczyk M. Sulindac and its metabolites: sulindac sulfide and sulindac sulfone enhance cytotoxic effects of arsenic trioxide on leukemic cell lines. Toxicol Vitro. doi:10.​1016/​j.​tiv.​2011.​04.​011.
34.
go back to reference IARC Working Group on the Evaluation of Carcinogenic Risks to Humans. Some drinking water disinfectants and contaminants, including arsenic. In: IARC Monographs on the Evaluation of Carcinogenic Risks to Humans, vol 84, France: IARC, Lyon; 2004. p. 228. IARC Working Group on the Evaluation of Carcinogenic Risks to Humans. Some drinking water disinfectants and contaminants, including arsenic. In: IARC Monographs on the Evaluation of Carcinogenic Risks to Humans, vol 84, France: IARC, Lyon; 2004. p. 228.
35.
go back to reference Dilda PJ, Hogg PJ. Arsenical-based cancer drugs. Cancer Treat Rev. 2002;33:542–64.CrossRef Dilda PJ, Hogg PJ. Arsenical-based cancer drugs. Cancer Treat Rev. 2002;33:542–64.CrossRef
36.
go back to reference Au WY, Tam S, Fong BM, Kwong YL. Determinants of cerebrospinal fluid arsenic concentration in patients with acute promyelocytic leukemia on oral arsenic trioxide therapy. Blood. 2008;112:3587–90.PubMedCrossRef Au WY, Tam S, Fong BM, Kwong YL. Determinants of cerebrospinal fluid arsenic concentration in patients with acute promyelocytic leukemia on oral arsenic trioxide therapy. Blood. 2008;112:3587–90.PubMedCrossRef
37.
go back to reference Kumana CR, Au WY, Lee NS, et al. Systemic availability of arsenic from oral arsenic-trioxide used to treat patients with hematological malignancies. Eur J Clin Pharmacol. 2002;58:521–6.PubMedCrossRef Kumana CR, Au WY, Lee NS, et al. Systemic availability of arsenic from oral arsenic-trioxide used to treat patients with hematological malignancies. Eur J Clin Pharmacol. 2002;58:521–6.PubMedCrossRef
38.
go back to reference Reeder MK, Pamakcu R, Weinstein IB, et al. Select cyclic nucleotide phosphodiesterase inhibitors in colon tumor chemoprevention. In: Gary J, Kelloff ET, Hawk C, Sigman C, editors. Cancer chemoprevention: promising cancer chemopreventive agents. Clifton: Humana Press; 2004. p. 401–18. Reeder MK, Pamakcu R, Weinstein IB, et al. Select cyclic nucleotide phosphodiesterase inhibitors in colon tumor chemoprevention. In: Gary J, Kelloff ET, Hawk C, Sigman C, editors. Cancer chemoprevention: promising cancer chemopreventive agents. Clifton: Humana Press; 2004. p. 401–18.
39.
go back to reference Ravis WR, Diskin CJ, Campagna KD, et al. Pharmacokinetics and dialyzability of sulindac and metabolites in patients with end-stage renal failure. J Clin Pharmacol. 1993;33:527–34.PubMed Ravis WR, Diskin CJ, Campagna KD, et al. Pharmacokinetics and dialyzability of sulindac and metabolites in patients with end-stage renal failure. J Clin Pharmacol. 1993;33:527–34.PubMed
40.
go back to reference van Stolk R, Stoner G, Hayton WL, et al. Phase I trial of exisulind (sulindac sulfone, FGN-1) as a chemopreventive agent in patients with familial adenomatous polyposis. Clin Cancer Res. 2000;78:78–89. van Stolk R, Stoner G, Hayton WL, et al. Phase I trial of exisulind (sulindac sulfone, FGN-1) as a chemopreventive agent in patients with familial adenomatous polyposis. Clin Cancer Res. 2000;78:78–89.
41.
go back to reference Chen YC, Lin-Shiau SY, Lin JK. Involvement of reactive oxygen species and caspase activation in arsenite-induced apoptosis. J Cell Physiol. 1998;177:324–33.PubMedCrossRef Chen YC, Lin-Shiau SY, Lin JK. Involvement of reactive oxygen species and caspase activation in arsenite-induced apoptosis. J Cell Physiol. 1998;177:324–33.PubMedCrossRef
42.
go back to reference Coe E and Schimmer AD. Catalase activity and arsenic sensitivity in acute leukemia. Leuk Lymphoma. 2008;49:1976–81.CrossRef Coe E and Schimmer AD. Catalase activity and arsenic sensitivity in acute leukemia. Leuk Lymphoma. 2008;49:1976–81.CrossRef
43.
go back to reference Krance SM, Keng PC, Palis J, Ballatori N. Transient glutathione depletion determines terminal differentiation in HL-60 cells. Oxid Med Cell Longev. 2010;3:53–60.PubMedCrossRef Krance SM, Keng PC, Palis J, Ballatori N. Transient glutathione depletion determines terminal differentiation in HL-60 cells. Oxid Med Cell Longev. 2010;3:53–60.PubMedCrossRef
45.
go back to reference Plummer JL, Smith BR, Sies H, Bend JR. Chemical depletion of glutathione in vivo. Methods Enzymol. 1981;77:50–9.PubMedCrossRef Plummer JL, Smith BR, Sies H, Bend JR. Chemical depletion of glutathione in vivo. Methods Enzymol. 1981;77:50–9.PubMedCrossRef
46.
go back to reference Griffith OW. Depletion of glutathione by inhibition of biosynthesis. Methods Enzymol. 1981;77:59–63.PubMedCrossRef Griffith OW. Depletion of glutathione by inhibition of biosynthesis. Methods Enzymol. 1981;77:59–63.PubMedCrossRef
47.
go back to reference Chou WC, Jie C, Kenedy AA, et al. Role of NADPH oxidase in arsenic-induced reactive oxygen species formation and cytotoxicity in myeloid leukemia cells. Proc Natl Acad Sci USA. 2004;101:4578–83.PubMedCrossRef Chou WC, Jie C, Kenedy AA, et al. Role of NADPH oxidase in arsenic-induced reactive oxygen species formation and cytotoxicity in myeloid leukemia cells. Proc Natl Acad Sci USA. 2004;101:4578–83.PubMedCrossRef
48.
go back to reference Smith KR, Klei LR, Barchowsky A. Arsenite stimulates plasma membrane NADPH oxidase in vascular endothelial cells. Am J Physiol Lung Cell Mol Physiol. 2001;280:L442–9.PubMed Smith KR, Klei LR, Barchowsky A. Arsenite stimulates plasma membrane NADPH oxidase in vascular endothelial cells. Am J Physiol Lung Cell Mol Physiol. 2001;280:L442–9.PubMed
49.
go back to reference Wang W, Adachi M, Kawamura R, et al. Parthenolide-induced apoptosis in multiple myeloma cells involves reactive oxygen species generation and cell sensitivity depends on catalase activity. Apoptosis. 2006;11:2225–35.PubMedCrossRef Wang W, Adachi M, Kawamura R, et al. Parthenolide-induced apoptosis in multiple myeloma cells involves reactive oxygen species generation and cell sensitivity depends on catalase activity. Apoptosis. 2006;11:2225–35.PubMedCrossRef
50.
go back to reference Wang J, Li L, Cang H, Shi G, Yi J. NADPH oxidase-derived reactive oxygen species are responsible for the high susceptibility to arsenic cytotoxicity in acute promyelocytic leukemia cells. Leuk Res. 2007;32:429–36.PubMedCrossRef Wang J, Li L, Cang H, Shi G, Yi J. NADPH oxidase-derived reactive oxygen species are responsible for the high susceptibility to arsenic cytotoxicity in acute promyelocytic leukemia cells. Leuk Res. 2007;32:429–36.PubMedCrossRef
51.
go back to reference Davison K, Mann K, Waxman S, Miller WH Jr. JNK activation is a mediator of arsenic trioxide-induced apoptosis in acute promyelocytic leukemia cells. Blood. 2004;103:3496–502.PubMedCrossRef Davison K, Mann K, Waxman S, Miller WH Jr. JNK activation is a mediator of arsenic trioxide-induced apoptosis in acute promyelocytic leukemia cells. Blood. 2004;103:3496–502.PubMedCrossRef
52.
go back to reference Sánchez Y, Calle C, de Blas E, Aller P. Modulation of arsenic trioxide-induced apoptosis by genistein and functionally related agents in U937 human leukaemia cells. Regulation by ROS and mitogen-activated protein kinases. Chem Biol Interact. 2009;182:37–44.PubMedCrossRef Sánchez Y, Calle C, de Blas E, Aller P. Modulation of arsenic trioxide-induced apoptosis by genistein and functionally related agents in U937 human leukaemia cells. Regulation by ROS and mitogen-activated protein kinases. Chem Biol Interact. 2009;182:37–44.PubMedCrossRef
53.
go back to reference Kajiguchi T, Yamamoto K, Hossain K, et al. Sustained activation of c-jun-terminal kinase (JNK) is closely related to arsenic trioxide-induced apoptosis in an acute myleoid leukemia (M2)-derived cell line, NKM-1. Leukemia. 2003;17:2189–95.PubMedCrossRef Kajiguchi T, Yamamoto K, Hossain K, et al. Sustained activation of c-jun-terminal kinase (JNK) is closely related to arsenic trioxide-induced apoptosis in an acute myleoid leukemia (M2)-derived cell line, NKM-1. Leukemia. 2003;17:2189–95.PubMedCrossRef
54.
go back to reference Kajiguchi T, Yamamoto K, Iida S, et al. Sustained activation of c-jun-N-terminal kinase plays a critical role in arsenic trioxide-induced cell apoptosis in multiple myeloma cell lines. Cancer Sci. 2006;97:540–5.PubMedCrossRef Kajiguchi T, Yamamoto K, Iida S, et al. Sustained activation of c-jun-N-terminal kinase plays a critical role in arsenic trioxide-induced cell apoptosis in multiple myeloma cell lines. Cancer Sci. 2006;97:540–5.PubMedCrossRef
55.
go back to reference Potin S, Bertoglio J, Bréard J. Involvement of a Rho-ROCK-JNK pathway in arsenic trioxide-induced apoptosis in chronic myelogenous leukemia cells. FEBS Lett. 2007;581:118–24.PubMedCrossRef Potin S, Bertoglio J, Bréard J. Involvement of a Rho-ROCK-JNK pathway in arsenic trioxide-induced apoptosis in chronic myelogenous leukemia cells. FEBS Lett. 2007;581:118–24.PubMedCrossRef
56.
go back to reference Wen J, Cheng HY, Feng Y, et al. P38 MAPK inhibition enhancing ATO-induced cytotoxicity against multiple myeloma cells. Br J Haematol. 2008;140:169–80.PubMedCrossRef Wen J, Cheng HY, Feng Y, et al. P38 MAPK inhibition enhancing ATO-induced cytotoxicity against multiple myeloma cells. Br J Haematol. 2008;140:169–80.PubMedCrossRef
57.
go back to reference Ludwig S, Hoffmeyer A, Goebeler M. The stress inducer arsenite activates mitogen-activated protein kinases extracellular signal-regulated kinases 1 and 2 via a MAPK kinase 6/p38-dependent pathway. J Biol Chem. 1998;273:1917–22.PubMedCrossRef Ludwig S, Hoffmeyer A, Goebeler M. The stress inducer arsenite activates mitogen-activated protein kinases extracellular signal-regulated kinases 1 and 2 via a MAPK kinase 6/p38-dependent pathway. J Biol Chem. 1998;273:1917–22.PubMedCrossRef
58.
go back to reference Lunghi P, Giuliani N, Mazzera L, et al. Targeting MEK/MAPK signal transduction module potentiates ATO-induced apoptosis in multiple myeloma cells through multiple signaling pathways. Blood. 2008;112:2450–62.PubMedCrossRef Lunghi P, Giuliani N, Mazzera L, et al. Targeting MEK/MAPK signal transduction module potentiates ATO-induced apoptosis in multiple myeloma cells through multiple signaling pathways. Blood. 2008;112:2450–62.PubMedCrossRef
Metadata
Title
Assessment of the involvement of oxidative stress and Mitogen-Activated Protein Kinase signaling pathways in the cytotoxic effects of arsenic trioxide and its combination with sulindac or its metabolites: sulindac sulfide and sulindac sulfone on human leukemic cell lines
Authors
M. Stępnik
M. Ferlińska
A. Smok-Pieniążek
D. Gradecka-Meesters
J. Arkusz
M. Stańczyk
Publication date
01-06-2012
Publisher
Springer US
Published in
Medical Oncology / Issue 2/2012
Print ISSN: 1357-0560
Electronic ISSN: 1559-131X
DOI
https://doi.org/10.1007/s12032-011-9920-1

Other articles of this Issue 2/2012

Medical Oncology 2/2012 Go to the issue
Live Webinar | 27-06-2024 | 18:00 (CEST)

Keynote webinar | Spotlight on medication adherence

Live: Thursday 27th June 2024, 18:00-19:30 (CEST)

WHO estimates that half of all patients worldwide are non-adherent to their prescribed medication. The consequences of poor adherence can be catastrophic, on both the individual and population level.

Join our expert panel to discover why you need to understand the drivers of non-adherence in your patients, and how you can optimize medication adherence in your clinics to drastically improve patient outcomes.

Prof. Kevin Dolgin
Prof. Florian Limbourg
Prof. Anoop Chauhan
Developed by: Springer Medicine
Obesity Clinical Trial Summary

At a glance: The STEP trials

A round-up of the STEP phase 3 clinical trials evaluating semaglutide for weight loss in people with overweight or obesity.

Developed by: Springer Medicine

Highlights from the ACC 2024 Congress

Year in Review: Pediatric cardiology

Watch Dr. Anne Marie Valente present the last year's highlights in pediatric and congenital heart disease in the official ACC.24 Year in Review session.

Year in Review: Pulmonary vascular disease

The last year's highlights in pulmonary vascular disease are presented by Dr. Jane Leopold in this official video from ACC.24.

Year in Review: Valvular heart disease

Watch Prof. William Zoghbi present the last year's highlights in valvular heart disease from the official ACC.24 Year in Review session.

Year in Review: Heart failure and cardiomyopathies

Watch this official video from ACC.24. Dr. Biykem Bozkurt discusses last year's major advances in heart failure and cardiomyopathies.