Skip to main content
Top
Published in: Medical Oncology 3/2010

01-09-2010 | Original Paper

Arsenic trioxide induces apoptosis in NB-4, an acute promyelocytic leukemia cell line, through up-regulation of p73 via suppression of nuclear factor kappa B-mediated inhibition of p73 transcription and prevention of NF-κB-mediated induction of XIAP, cIAP2, BCL-XL and survivin

Authors: Majid Momeny, Majid Zakidizaji, Reza Ghasemi, Ahmad R. Dehpour, Maryam Rahimi_Balaei, Yassan Abdolazimi, Ardeshir Ghavamzadeh, Kamran Alimoghaddam, Seyed H. Ghaffari

Published in: Medical Oncology | Issue 3/2010

Login to get access

Abstract

The purpose of the present study is to evaluate the effects of arsenic trioxide (ATO) on human acute promyelocytic leukemia NB-4 cells. Microculture tetrazolium test, bromodeoxyuridine (BrdU) cell proliferation assay, caspase 3 activity assay, cell-based nuclear factor kappa B (NF-κB) phosphorylation measurement by ELISA and real-time RT-PCR were employed to appraise the effects of ATO on metabolic activity, DNA synthesis, induction of programmed cell death and NF-κB activation. The suppressive effects of ATO on metabolic potential, cell proliferation and NF-κB activation were associated with induction of apoptosis in NB-4 cells. In addition, an expressive enhancement in mRNA levels of p73, cyclin-dependent kinase inhibitor 1A (p21), tumor protein 53-induced nuclear protein 1 (TP53INP1), WNK lysine deficient protein kinase 2 (WNK2) and lipocalin 2 coupled with a significant reduction in transcriptional levels of NF-κB inhibitor beta (IKK2), Nemo, BCL2-like 1 (BCL-XL), inhibitor of apoptosis protein 1 (cIAP2), X-linked inhibitor of apoptosis protein (XIAP), survivin, Bcl-2, TIP60, ataxia telangiectasia (ATM), SHP-2 and sirtuin (SIRT1) were observed. Altogether, these issues show for the first time that ATO treatment could trammel cell growth and proliferation as well as induces apoptosis in NB-4 cells through induction of transcriptional levels of p73, TP53INP1, WNK2, lipocalin 2 as well as suppression of NF-κB-mediated induction of BCL-XL, cIAP2, XIAP and survivin. Furthermore, the inductionary effects of ATO on transcriptional stimulation of p73 might be through cramping the NF-κB module (through suppression of p65 phosphorylation as well as transcriptional hindering of IKK2, ATM and Nemo) along with diminishing the mRNA expression of TIP60, SHP-2 and SIRT1.
Literature
1.
go back to reference Lo Coco F, Diverio D, Falini B, Biondi A, Nervi C, et al. Genetic diagnosis and molecular monitoring in the management of acute promyelocytic leukemia. Blood. 1999;94:12–22.PubMed Lo Coco F, Diverio D, Falini B, Biondi A, Nervi C, et al. Genetic diagnosis and molecular monitoring in the management of acute promyelocytic leukemia. Blood. 1999;94:12–22.PubMed
2.
go back to reference Di Croce L, Raker VA, Corsaro M, Fazi F, Fanelli M, et al. Methyltransferase recruitment and DNA hypermethylation of target promoters by an oncogenic transcription factor. Science. 2002;295:1079–82.CrossRefPubMed Di Croce L, Raker VA, Corsaro M, Fazi F, Fanelli M, et al. Methyltransferase recruitment and DNA hypermethylation of target promoters by an oncogenic transcription factor. Science. 2002;295:1079–82.CrossRefPubMed
3.
go back to reference Pettersson HM, Pietras A, Munksgaard Persson M, Karlsson J, Johansson L, et al. Arsenic trioxide is highly cytotoxic to small cell lung carcinoma cells. Mol Cancer Ther. 2009;8:160–70.CrossRefPubMed Pettersson HM, Pietras A, Munksgaard Persson M, Karlsson J, Johansson L, et al. Arsenic trioxide is highly cytotoxic to small cell lung carcinoma cells. Mol Cancer Ther. 2009;8:160–70.CrossRefPubMed
4.
go back to reference Yu J, Qian H, Li Y, Wang Y, Zhang X, et al. Arsenic trioxide (As2O3) reduces the invasive and metastatic properties of cervical cancer cells in vitro and in vivo. Gynecol Oncol. 2007;106:400–6.CrossRefPubMed Yu J, Qian H, Li Y, Wang Y, Zhang X, et al. Arsenic trioxide (As2O3) reduces the invasive and metastatic properties of cervical cancer cells in vitro and in vivo. Gynecol Oncol. 2007;106:400–6.CrossRefPubMed
5.
go back to reference Park M, Lee J, Kwak H, Park C, Lee H, et al. Arsenic trioxide (As2O3) inhibits invasion of HT1080 human fibrosarcoma cells: role of nuclear factor-kB and reactive oxygen species. J Cell Biochem. 2005;95:955–69.CrossRefPubMed Park M, Lee J, Kwak H, Park C, Lee H, et al. Arsenic trioxide (As2O3) inhibits invasion of HT1080 human fibrosarcoma cells: role of nuclear factor-kB and reactive oxygen species. J Cell Biochem. 2005;95:955–69.CrossRefPubMed
6.
go back to reference Vuky J, Yu R, Schwartz L, Motzer RJ. Phase II trial of arsenic trioxide in patients with metastatic renal cell carcinoma. Investig New Drugs. 2002;20:327–30.CrossRef Vuky J, Yu R, Schwartz L, Motzer RJ. Phase II trial of arsenic trioxide in patients with metastatic renal cell carcinoma. Investig New Drugs. 2002;20:327–30.CrossRef
7.
go back to reference Morales AA, Gutman D, Lee KP, Boise LH. BH3-only proteins Noxa, Bmf, and Bim are necessary for arsenic trioxide-induced cell death in myeloma. Blood. 2008;111:5152–62.CrossRefPubMed Morales AA, Gutman D, Lee KP, Boise LH. BH3-only proteins Noxa, Bmf, and Bim are necessary for arsenic trioxide-induced cell death in myeloma. Blood. 2008;111:5152–62.CrossRefPubMed
8.
go back to reference Kanzawa T, Kondo Y, Ito H, Kondo S, Germano I. Induction of autophagic cell death in malignant glioma cells by arsenic trioxide. Cancer Res. 2003;63:2103–8.PubMed Kanzawa T, Kondo Y, Ito H, Kondo S, Germano I. Induction of autophagic cell death in malignant glioma cells by arsenic trioxide. Cancer Res. 2003;63:2103–8.PubMed
9.
go back to reference Li H, He J, Ju P, Zhong X, Liu J. Studies on the mechanism of arsenic trioxide-induced apoptosis in HepG2 human hepatocellular carcinoma cells. Chin J Clin Oncol. 2008;5:22–5.CrossRef Li H, He J, Ju P, Zhong X, Liu J. Studies on the mechanism of arsenic trioxide-induced apoptosis in HepG2 human hepatocellular carcinoma cells. Chin J Clin Oncol. 2008;5:22–5.CrossRef
10.
go back to reference Zhou L, Chan JY, Wang J, Fung K. Anti-proliferation of MDA-MB-231 human breast tumor cells by arsenic trioxide via induction of apoptosis. Res J Med Sci. 2008;2:236–43. Zhou L, Chan JY, Wang J, Fung K. Anti-proliferation of MDA-MB-231 human breast tumor cells by arsenic trioxide via induction of apoptosis. Res J Med Sci. 2008;2:236–43.
11.
go back to reference Cheung WM, Chu PW, Kwong YL. Effects of arsenic trioxide on the cellular proliferation, apoptosis and differentiation of human neuroblastoma cells. Cancer Lett. 2007;246:122–8.CrossRefPubMed Cheung WM, Chu PW, Kwong YL. Effects of arsenic trioxide on the cellular proliferation, apoptosis and differentiation of human neuroblastoma cells. Cancer Lett. 2007;246:122–8.CrossRefPubMed
12.
go back to reference Venepalli NK, Altman JK, Tallman MS. Evolving role of arsenic trioxide in acute promyelocytic leukemia. Clin Leuk. 2006;1:36–40.CrossRef Venepalli NK, Altman JK, Tallman MS. Evolving role of arsenic trioxide in acute promyelocytic leukemia. Clin Leuk. 2006;1:36–40.CrossRef
13.
go back to reference Miller WH Jr, Schipper HM, Lee JS, Singer J, Waxman S. Mechanisms of action of arsenic trioxide. Cancer Res. 2002;62:3893–903.PubMed Miller WH Jr, Schipper HM, Lee JS, Singer J, Waxman S. Mechanisms of action of arsenic trioxide. Cancer Res. 2002;62:3893–903.PubMed
14.
go back to reference Wei L, Lai K, Chen C, Cheng C, Huang Y, et al. Arsenic trioxide prevents radiation-enhanced tumor invasiveness and inhibits matrix metalloproteinase-9 through downregulation of nuclear factor κB. Oncogene. 2005;24:390–8.CrossRefPubMed Wei L, Lai K, Chen C, Cheng C, Huang Y, et al. Arsenic trioxide prevents radiation-enhanced tumor invasiveness and inhibits matrix metalloproteinase-9 through downregulation of nuclear factor κB. Oncogene. 2005;24:390–8.CrossRefPubMed
15.
go back to reference Jiang XH, Wong BC, Yuen ST, Jiang SH, Cho CH, et al. Arsenic trioxide induces apoptosis in human gastric cancer cells through up-regulation of p53 and activation of caspase-3. Int J Cancer. 2001;91:173–9.CrossRefPubMed Jiang XH, Wong BC, Yuen ST, Jiang SH, Cho CH, et al. Arsenic trioxide induces apoptosis in human gastric cancer cells through up-regulation of p53 and activation of caspase-3. Int J Cancer. 2001;91:173–9.CrossRefPubMed
16.
go back to reference Akao Y, Mizoguchi H, Kojima S, Naoe T, Ohishi N, et al. Arsenic induces apoptosis in B-cell leukaemic cell lines in vitro: activation of caspases and down-regulation of Bcl-2 protein. Br J Haematol. 1998;102:1055–60.CrossRefPubMed Akao Y, Mizoguchi H, Kojima S, Naoe T, Ohishi N, et al. Arsenic induces apoptosis in B-cell leukaemic cell lines in vitro: activation of caspases and down-regulation of Bcl-2 protein. Br J Haematol. 1998;102:1055–60.CrossRefPubMed
17.
go back to reference Chou WC, Hawkins AL, Barrett JF, Griffin CA, Dang CV. Arsenic inhibition of telomerase transcription leads to genetic instability. J Clin Investig. 2001;108:1541–7.PubMed Chou WC, Hawkins AL, Barrett JF, Griffin CA, Dang CV. Arsenic inhibition of telomerase transcription leads to genetic instability. J Clin Investig. 2001;108:1541–7.PubMed
18.
go back to reference Yih LH, Lee T. Arsenite induces p53 accumulation through an ATM dependent pathway in human fibroblasts. Cancer Res. 2000;60:6346–52.PubMed Yih LH, Lee T. Arsenite induces p53 accumulation through an ATM dependent pathway in human fibroblasts. Cancer Res. 2000;60:6346–52.PubMed
19.
go back to reference Kroemer G, de The′ H. Arsenic trioxide, a novel mitochondriotoxic anticancer agent? J Natl Cancer Inst. 1999;91:743–5.CrossRefPubMed Kroemer G, de The′ H. Arsenic trioxide, a novel mitochondriotoxic anticancer agent? J Natl Cancer Inst. 1999;91:743–5.CrossRefPubMed
20.
go back to reference Shen Z, Shen J, Cai W, Hong C, Zheng MH. The alteration of mitochondria is an early event of arsenic trioxide induced apoptosis in esophageal carcinoma cells. Int J Mol Med. 2000;5:155–8.PubMed Shen Z, Shen J, Cai W, Hong C, Zheng MH. The alteration of mitochondria is an early event of arsenic trioxide induced apoptosis in esophageal carcinoma cells. Int J Mol Med. 2000;5:155–8.PubMed
22.
go back to reference Kaghad M, Bonnet H, Yang A, Creancier L, Biscan JC, et al. Monoallelically expressed gene related to p53 at 1p36, a region frequently deleted in neuroblastoma and other human cancers. Cell. 1997;90:809–19.CrossRefPubMed Kaghad M, Bonnet H, Yang A, Creancier L, Biscan JC, et al. Monoallelically expressed gene related to p53 at 1p36, a region frequently deleted in neuroblastoma and other human cancers. Cell. 1997;90:809–19.CrossRefPubMed
23.
go back to reference Melino G, De Laurenzi V, Vousden KH. p73: friend or foe in tumorigenesis. Nat Rev Cancer. 2002;2:605–15.CrossRefPubMed Melino G, De Laurenzi V, Vousden KH. p73: friend or foe in tumorigenesis. Nat Rev Cancer. 2002;2:605–15.CrossRefPubMed
24.
go back to reference Melino G, Bernassola F, Ranalli M, Yee K, Xing Zong W, et al. p73 induces apoptosis via puma transactivation and bax mitochondrial translocation. J Biol Chem. 2004;279:8076–83.CrossRefPubMed Melino G, Bernassola F, Ranalli M, Yee K, Xing Zong W, et al. p73 induces apoptosis via puma transactivation and bax mitochondrial translocation. J Biol Chem. 2004;279:8076–83.CrossRefPubMed
25.
go back to reference Yang A, McKeon F. P63 and P73:P53 mimics, menaces and more. Nat Rev Mol Cell Biol. 1997;1:199–207.CrossRef Yang A, McKeon F. P63 and P73:P53 mimics, menaces and more. Nat Rev Mol Cell Biol. 1997;1:199–207.CrossRef
26.
go back to reference Tomasini R, Seux M, Nowak J, Bontemps C, Carrier A, et al. TP53INP1 is a novel p73 target gene that induces cell cycle arrest and cell death by modulating p73 transcriptional activity. Oncogene. 2005;24:8093–104.PubMed Tomasini R, Seux M, Nowak J, Bontemps C, Carrier A, et al. TP53INP1 is a novel p73 target gene that induces cell cycle arrest and cell death by modulating p73 transcriptional activity. Oncogene. 2005;24:8093–104.PubMed
27.
go back to reference Stiewe T, Putzer BM. Role of p73 in malignancy: tumor suppressor or oncogene? Cell Death Differ. 2002;9:237–45.CrossRefPubMed Stiewe T, Putzer BM. Role of p73 in malignancy: tumor suppressor or oncogene? Cell Death Differ. 2002;9:237–45.CrossRefPubMed
28.
go back to reference Dai JM, Wang ZY, Sun DC, Lin RX, Wang SQ. SIRT1 interacts with p73 and suppresses p73-dependent transcriptional activity. J Cell Physiol. 2007;210:161–6.CrossRefPubMed Dai JM, Wang ZY, Sun DC, Lin RX, Wang SQ. SIRT1 interacts with p73 and suppresses p73-dependent transcriptional activity. J Cell Physiol. 2007;210:161–6.CrossRefPubMed
29.
go back to reference Ruhul Amin A, Thakur VS, Paul RK, Feng GS, Qu C, et al. SHP-2 tyrosine phosphatase inhibits p73-dependent apoptosis and expression of a subset of p53 target genes induced by EGCG. PNAS. 2007;104:5419–24.CrossRef Ruhul Amin A, Thakur VS, Paul RK, Feng GS, Qu C, et al. SHP-2 tyrosine phosphatase inhibits p73-dependent apoptosis and expression of a subset of p53 target genes induced by EGCG. PNAS. 2007;104:5419–24.CrossRef
30.
go back to reference Kim JW, Song PI, Jeong MH, An JH, Lee SY, et al. TIP60 represses transcriptional activity of p73beta via an MDM2-bridged ternary complex. J Biol Chem. 2008;283:20077–86.CrossRefPubMed Kim JW, Song PI, Jeong MH, An JH, Lee SY, et al. TIP60 represses transcriptional activity of p73beta via an MDM2-bridged ternary complex. J Biol Chem. 2008;283:20077–86.CrossRefPubMed
31.
go back to reference Baud V, Karin M. Is NFκB a good target for cancer therapy? Hopes and pitfalls. Nat Rev Drug Discov. 2009;8:33–40.CrossRefPubMed Baud V, Karin M. Is NFκB a good target for cancer therapy? Hopes and pitfalls. Nat Rev Drug Discov. 2009;8:33–40.CrossRefPubMed
32.
33.
go back to reference Karin M, Cao Y, Greten F, Li Z. NF-κB in cancer: from innocent bystander to major culprit. Nat Rev Cancer. 2002;2:301–10.CrossRefPubMed Karin M, Cao Y, Greten F, Li Z. NF-κB in cancer: from innocent bystander to major culprit. Nat Rev Cancer. 2002;2:301–10.CrossRefPubMed
34.
go back to reference Bonizzi G, Karin M. The two NF-kappa activation pathways and their role in innate and adaptive immunity. Trends Immunol. 2004;25:280–8.CrossRefPubMed Bonizzi G, Karin M. The two NF-kappa activation pathways and their role in innate and adaptive immunity. Trends Immunol. 2004;25:280–8.CrossRefPubMed
35.
go back to reference Kikuchi H, Ozaki T, Furuya K, Hanamoto T, Nakanishi M, et al. NF-κB regulates the stability and activity of p73 by inducing its proteolytic degradation through a ubiquitin-dependent proteasome pathway. Oncogene. 2006;25:7608–17.CrossRefPubMed Kikuchi H, Ozaki T, Furuya K, Hanamoto T, Nakanishi M, et al. NF-κB regulates the stability and activity of p73 by inducing its proteolytic degradation through a ubiquitin-dependent proteasome pathway. Oncogene. 2006;25:7608–17.CrossRefPubMed
36.
go back to reference Moniz S, Veríssimo F, Matos P, Brazão R, Silva E, et al. Protein kinase WNK2 inhibits cell proliferation by negatively modulating the activation of MEK1/ERK1/2. Oncogene. 2007;26:6071–81.CrossRefPubMed Moniz S, Veríssimo F, Matos P, Brazão R, Silva E, et al. Protein kinase WNK2 inhibits cell proliferation by negatively modulating the activation of MEK1/ERK1/2. Oncogene. 2007;26:6071–81.CrossRefPubMed
37.
go back to reference Hanai J, Mammoto T, Seth P, Mori K, Karumanchi SA, et al. Lipocalin 2 diminishes invasiveness and metastasis of Ras-transformed cells. J Biol Chem. 2006;280:13641–7.CrossRef Hanai J, Mammoto T, Seth P, Mori K, Karumanchi SA, et al. Lipocalin 2 diminishes invasiveness and metastasis of Ras-transformed cells. J Biol Chem. 2006;280:13641–7.CrossRef
38.
go back to reference Grosjean-Raillard J, Tailler M, Ades L, Perfettini JL, Fabre C, et al. ATM mediates constitutive NF-κB activation in high-risk myelodysplastic syndrome and acute myeloid leukemia. Oncogene. 2009;28:1099–109.CrossRefPubMed Grosjean-Raillard J, Tailler M, Ades L, Perfettini JL, Fabre C, et al. ATM mediates constitutive NF-κB activation in high-risk myelodysplastic syndrome and acute myeloid leukemia. Oncogene. 2009;28:1099–109.CrossRefPubMed
39.
40.
go back to reference Shen ZX, Chen GQ, Ni JH, Li XS, Xiong SM, et al. Use of arsenic trioxide (As2O3) in the treatment of acute promyelocytic leukemia (APL): II. Clinical efficacy and pharmacokinetics in relapsed patients. Blood. 1997;89:3354–60.PubMed Shen ZX, Chen GQ, Ni JH, Li XS, Xiong SM, et al. Use of arsenic trioxide (As2O3) in the treatment of acute promyelocytic leukemia (APL): II. Clinical efficacy and pharmacokinetics in relapsed patients. Blood. 1997;89:3354–60.PubMed
41.
go back to reference Schmittgen TD, Livak KJ. Analyzing real-time PCR data by the comparative CT method. Nat Protoc. 2008;3:1101–8.CrossRefPubMed Schmittgen TD, Livak KJ. Analyzing real-time PCR data by the comparative CT method. Nat Protoc. 2008;3:1101–8.CrossRefPubMed
42.
go back to reference Ghavamzadeh A, Alimoghaddam K, Ghaffari SH, Rostami S, Jahani M, et al. Treatment of acute promyelocytic leukemia with arsenic trioxide without ATRA and/or chemotherapy. Ann Oncol. 2005;17:131–4.CrossRefPubMed Ghavamzadeh A, Alimoghaddam K, Ghaffari SH, Rostami S, Jahani M, et al. Treatment of acute promyelocytic leukemia with arsenic trioxide without ATRA and/or chemotherapy. Ann Oncol. 2005;17:131–4.CrossRefPubMed
43.
go back to reference Alimoghaddam K, Shariftabrizi A, Tavangar M, Sanaat Z, Rostami S, et al. Anti-leukemic and anti-angiogenesis efficacy of arsenic trioxide in new cases of acute promyelocytic leukemia. Leuk Lymphoma. 2006;47:81–8.CrossRefPubMed Alimoghaddam K, Shariftabrizi A, Tavangar M, Sanaat Z, Rostami S, et al. Anti-leukemic and anti-angiogenesis efficacy of arsenic trioxide in new cases of acute promyelocytic leukemia. Leuk Lymphoma. 2006;47:81–8.CrossRefPubMed
44.
go back to reference Ghaffari SH, Rostami S, Bashash D, Alimoghaddam K, Ghavamzadeh A. Real-time PCR analysis of PML-RAR in newly diagnosed acute promyelocytic leukaemia patients treated with arsenic trioxide as a front-line therapy. Ann Oncol. 2006;17:1553–9.CrossRefPubMed Ghaffari SH, Rostami S, Bashash D, Alimoghaddam K, Ghavamzadeh A. Real-time PCR analysis of PML-RAR in newly diagnosed acute promyelocytic leukaemia patients treated with arsenic trioxide as a front-line therapy. Ann Oncol. 2006;17:1553–9.CrossRefPubMed
45.
go back to reference Mathews V, George B, Lakshmi KM, Viswabandya A, Bajel A, et al. Single-agent arsenic trioxide in the treatment of newly diagnosed acute promyelocytic leukemia: durable remissions with minimal toxicity. Blood. 2006;107:2627–32.CrossRefPubMed Mathews V, George B, Lakshmi KM, Viswabandya A, Bajel A, et al. Single-agent arsenic trioxide in the treatment of newly diagnosed acute promyelocytic leukemia: durable remissions with minimal toxicity. Blood. 2006;107:2627–32.CrossRefPubMed
46.
go back to reference Chen GQ, Shi XG, Tang W, Xiong SM, Zhu J, et al. Use of arsenic trioxide (As2O3) in the treatment of acute promyelocytic leukemia (APL): I. As2O3 exerts dose-dependent dual effects on APL cells. Blood. 1997;89:3345–53.PubMed Chen GQ, Shi XG, Tang W, Xiong SM, Zhu J, et al. Use of arsenic trioxide (As2O3) in the treatment of acute promyelocytic leukemia (APL): I. As2O3 exerts dose-dependent dual effects on APL cells. Blood. 1997;89:3345–53.PubMed
47.
go back to reference Sherr CJ, Roberts JM. Inhibitors of mammalian G1 cyclin-dependent kinases. Genes Dev. 1995;9:1149–63.CrossRefPubMed Sherr CJ, Roberts JM. Inhibitors of mammalian G1 cyclin-dependent kinases. Genes Dev. 1995;9:1149–63.CrossRefPubMed
48.
go back to reference Kim H, Rafiuddin-Shah M, Tu H, Jeffers JR, Zambetti GP, et al. Hierarchical regulation of mitochondrion-dependent apoptosis by BCL-2 subfamilies. Nat Cell Biol. 2006;8:1348–58.CrossRefPubMed Kim H, Rafiuddin-Shah M, Tu H, Jeffers JR, Zambetti GP, et al. Hierarchical regulation of mitochondrion-dependent apoptosis by BCL-2 subfamilies. Nat Cell Biol. 2006;8:1348–58.CrossRefPubMed
49.
go back to reference Irwin MS, Kondo K, Marin MC, Cheng LS, Hahn WC, et al. Chemosensitivity linked to p73 function. Cancer Cell. 2003;3:403–10.CrossRefPubMed Irwin MS, Kondo K, Marin MC, Cheng LS, Hahn WC, et al. Chemosensitivity linked to p73 function. Cancer Cell. 2003;3:403–10.CrossRefPubMed
50.
go back to reference Muscolini M, Cianfrocca R, Sajeva A, Mozzetti S, Ferrandina G, et al. Trichostatin A up-regulates p73 and induces bax-dependent apoptosis in cisplatin-resistant ovarian cancer cells. Mol Cancer Ther. 2008;7:1410–9.CrossRefPubMed Muscolini M, Cianfrocca R, Sajeva A, Mozzetti S, Ferrandina G, et al. Trichostatin A up-regulates p73 and induces bax-dependent apoptosis in cisplatin-resistant ovarian cancer cells. Mol Cancer Ther. 2008;7:1410–9.CrossRefPubMed
51.
go back to reference Song X, Sheppard HM, Norman AW, Liu X. Mitogen-activated protein kinase is involved in the degradation of p53 protein in the bryostatin-1-induced differentiation of the acute promyelocytic leukemia NB4 cell line. J Biol Chem. 1999;274:1677–82.CrossRefPubMed Song X, Sheppard HM, Norman AW, Liu X. Mitogen-activated protein kinase is involved in the degradation of p53 protein in the bryostatin-1-induced differentiation of the acute promyelocytic leukemia NB4 cell line. J Biol Chem. 1999;274:1677–82.CrossRefPubMed
52.
go back to reference Tomasini R, Azizi Samir A, Carrier A, Isnardon D, Cecchinelli B, et al. TP53INP1s and homeodomain-interacting protein kinase-2 (HIPK2) are partners in regulating p53 activity. J Biol Chem. 2003;278:37722–9.CrossRefPubMed Tomasini R, Azizi Samir A, Carrier A, Isnardon D, Cecchinelli B, et al. TP53INP1s and homeodomain-interacting protein kinase-2 (HIPK2) are partners in regulating p53 activity. J Biol Chem. 2003;278:37722–9.CrossRefPubMed
53.
go back to reference Karin M, Yamamoto Y, Wang QM. The IKK NF-kappa B system: a treasure trove for drug development. Nat Rev Drug Discov. 2004;3:17–26.CrossRefPubMed Karin M, Yamamoto Y, Wang QM. The IKK NF-kappa B system: a treasure trove for drug development. Nat Rev Drug Discov. 2004;3:17–26.CrossRefPubMed
54.
go back to reference Nakanishi C, Toi M. Nuclear factor-κB inhibitors as sensitizers to anticancer drugs. Nat Rev Cancer. 2005;5:297–309.CrossRefPubMed Nakanishi C, Toi M. Nuclear factor-κB inhibitors as sensitizers to anticancer drugs. Nat Rev Cancer. 2005;5:297–309.CrossRefPubMed
55.
go back to reference Tong Z, Wu X, Kehrer JP. Increased expression of the lipocalin 24p3 as an apoptotic mechanism for MK886. Biochem J. 2003;372:203–10.CrossRefPubMed Tong Z, Wu X, Kehrer JP. Increased expression of the lipocalin 24p3 as an apoptotic mechanism for MK886. Biochem J. 2003;372:203–10.CrossRefPubMed
56.
go back to reference Sykes SM, Mellert HS, Holbert MA, Li K, Marmorstein R, et al. Acetylation of the p53 DNA-binding domain regulates apoptosis induction. Mol Cell. 2006;24:841–51.CrossRefPubMed Sykes SM, Mellert HS, Holbert MA, Li K, Marmorstein R, et al. Acetylation of the p53 DNA-binding domain regulates apoptosis induction. Mol Cell. 2006;24:841–51.CrossRefPubMed
57.
go back to reference Tang Y, Luo J, Zhang W, Gu W. Tip60-dependent acetylation of p53 modulates the decision between cell-cycle arrest and apoptosis. Mol Cell. 2006;24:827–39.CrossRefPubMed Tang Y, Luo J, Zhang W, Gu W. Tip60-dependent acetylation of p53 modulates the decision between cell-cycle arrest and apoptosis. Mol Cell. 2006;24:827–39.CrossRefPubMed
58.
go back to reference Huang TT, Wuerzberger-Davis SM, Wu ZH, Miyamoto S. Sequential modification of NEMO/IKK gamma by SUMO-1 and ubiquitin mediates NF-kappa B activation by genotoxic stress. Cell. 2003;115:565–76.CrossRefPubMed Huang TT, Wuerzberger-Davis SM, Wu ZH, Miyamoto S. Sequential modification of NEMO/IKK gamma by SUMO-1 and ubiquitin mediates NF-kappa B activation by genotoxic stress. Cell. 2003;115:565–76.CrossRefPubMed
Metadata
Title
Arsenic trioxide induces apoptosis in NB-4, an acute promyelocytic leukemia cell line, through up-regulation of p73 via suppression of nuclear factor kappa B-mediated inhibition of p73 transcription and prevention of NF-κB-mediated induction of XIAP, cIAP2, BCL-XL and survivin
Authors
Majid Momeny
Majid Zakidizaji
Reza Ghasemi
Ahmad R. Dehpour
Maryam Rahimi_Balaei
Yassan Abdolazimi
Ardeshir Ghavamzadeh
Kamran Alimoghaddam
Seyed H. Ghaffari
Publication date
01-09-2010
Publisher
Springer US
Published in
Medical Oncology / Issue 3/2010
Print ISSN: 1357-0560
Electronic ISSN: 1559-131X
DOI
https://doi.org/10.1007/s12032-009-9294-9

Other articles of this Issue 3/2010

Medical Oncology 3/2010 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.