Browse > Article
http://dx.doi.org/10.1007/s43188-022-00150-4

Induction of synergistic apoptosis by tetramethoxystilbene and nutlin-3a in human cervical cancer cells  

An, Hong-Gyu (College of Pharmacy, Chung-Ang University)
Shin, Sangyun (College of Pharmacy, Chung-Ang University)
Lee, Boyoung (College of Pharmacy, Chung-Ang University)
Kwon, Yeonju (College of Pharmacy, Chung-Ang University)
Kwon, Tae-Uk (College of Pharmacy, Chung-Ang University)
Kwon, Yeo-Jung (College of Pharmacy, Chung-Ang University)
Chun, Young-Jin (College of Pharmacy, Chung-Ang University)
Publication Information
Toxicological Research / v.38, no.4, 2022 , pp. 591-600 More about this Journal
Abstract
2,4,3',5'-Tetramethoxystilbene (TMS) is a selective inhibitor of cytochrome P450 1B1 to block the conversion from estradiol to 4-OH-estradiol. Several studies suggested that TMS may act as a potent anti-cancer agent for hormone-related cancer including cervical cancer. Nutlin-3a is a cis-imidazoline analog that interferes with the interaction between mouse double minute 2 homolog (MDM2) and the tumor suppressor p53. The purpose of the study was to compare the cytotoxic effect of TMS and nutlin-3a treatment individually and in combination in HeLa cells. To assess the potential synergistic effects between TMS and nutlin-3a, low concentrations of TMS and nutlin-3a were simultaneously treated in HeLa cells. Based on cell viability, apoptosis assays, and the increase in cleaved caspase-3 and poly (ADP-ribose) polymerase cleavage, it was demonstrated that the combination with TMS and nutlin-3a exerts a synergistic effect on cancer cell death. Isobologram analysis of HeLa cells noted synergism between TMS and nutlin-3a. The combined treatment increased the expression of mitochondrial pro-apoptotic factors such as Bax and Bak, and decreased the expression of the XIAP. In addition, combination treatment significantly enhanced the translocation of AIF to the nucleus in HeLa cells. In conclusion, the results demonstrate that the combination of TMS and nutlin-3a induces synergistic apoptosis in HeLa cells, suggesting the possibility that this combination can be applied as a novel therapeutic strategy for cervical cancer.
Keywords
Tetramethoxystilbene; CYP1B1; nutlin-3a; p53; Combination treatment;
Citations & Related Records
Times Cited By KSCI : 2  (Citation Analysis)
연도 인용수 순위
1 Wang X, Simpson ER, Brown KA (2015) p53: protection against tumor growth beyond effects on cell cycle and apoptosis. Cancer Res 75:5001-5007. https://doi.org/10.1158/0008-5472.CAN-15-0563   DOI
2 Joerger AC, Fersht AR (2016) The p53 pathway: origins, inactivation in cancer, and emerging therapeutic approaches. Annu Rev Biochem 85:375-404. https://doi.org/10.1146/annurev-biochem-060815-014710   DOI
3 Barbarotto E, Corallini F, Rimondi E et al (2008) Differential effects of chemotherapeutic drugs versus the MDM-2 antagonist nutlin-3 on cell cycle progression and induction of apoptosis in SKW6.4 lymphoblastoid B-cells. J Cell Biochem 104:595-605. https://doi.org/10.1002/jcb.21649   DOI
4 Guengerich FP (2020) A history of the roles of cytochrome P450 enzymes in the toxicity of drugs. Toxicol Res 37:1-23. https://doi.org/10.1007/s43188-020-00056-z   DOI
5 Stanley A, Ashrafi GH, Seddon AM, Modjtahedi H (2017) Synergistic effects of various Her inhibitors in combination with IGF-1R, C-MET and Src targeting agents in breast cancer cell lines. Sci Rep 7:3964. https://doi.org/10.1038/s41598-017-04301-8   DOI
6 Hong B, van den Heuvel AP, Prabhu VV, Zhang S, El-Deiry WS (2014) Targeting tumor suppressor p53 for cancer therapy: strategies, challenges and opportunities. Curr Drug Targets 15:80-89. https://doi.org/10.2174/1389450114666140106101412   DOI
7 Huang B, Deo D, Xia M, Vassilev LT (2009) Pharmacologic p53 activation blocks cell cycle progression but fails to induce senescence in epithelial cancer cells. Mol Cancer Res 7:1497-1509. https://doi.org/10.1158/1541-7786.MCR-09-0144   DOI
8 Susin SA, Lorenzo HK, Zamzami N et al (1999) Molecular characterization of mitochondrial apoptosis-inducing factor. Nature 397:441-446. https://doi.org/10.1038/17135   DOI
9 Lipton SA, Bossy-Wetzel E (2002) Dueling activities of AIF in cell death versus survival: DNA binding and redox activity. Cell 111:147-150. https://doi.org/10.1016/s0092-8674(02)01046-2   DOI
10 Daugas E, Susin SA, Zamzami N et al (2000) Mitochondrionuclear translocation of AIF in apoptosis and necrosis. FASEB J 14:729-739. https://doi.org/10.1096/fasebj.14.5.729   DOI
11 Loeffler M, Daugas E, Susin SA et al (2001) Dominant cell death induction by extramitochondrially targeted apoptosisinducing factor. FASEB J 15:758-767. https://doi.org/10.1096/fj.00-0388com   DOI
12 Mondesire WH, Jian W, Zhang H et al (2004) Targeting mammalian target of rapamycin synergistically enhances chemotherapy-induced cytotoxicity in breast cancer cells. Clin Cancer Res 10:7031-7042. https://doi.org/10.1158/1078-0432.CCR-04-0361   DOI
13 DeVita VT Jr, Young RC, Canellos GP (1975) Combination versus single agent chemotherapy: a review of the basis for selection of drug treatment of cancer. Cancer 35:98-110. https://doi.org/10.1002/1097-0142(197501)35:198::aidcncr28203501153.0.co;2-b   DOI
14 Fan C, Zheng W, Fu X, Li X, Wong YS, Chen T (2014) Enhancement of auranofin-induced lung cancer cell apoptosis by selenocystine, a natural inhibitor of TrxR1 in vitro and in vivo. Cell Death Dis 5:e1191. https://doi.org/10.1038/cddis.2014.132   DOI
15 Kojima K, Konopleva M, Samudio IJ et al (2005) MDM2 antagonists induce p53-dependent apoptosis in AML: implications for leukemia therapy. Blood 106:3150-3159. https://doi.org/10.1182/blood-2005-02-0553   DOI
16 Deben C, Wouters A, Op de Beeck K et al (2015) The MDM2-inhibitor nutlin-3 synergizes with cisplatin to induce p53 dependent tumor cell apoptosis in non-small cell lung cancer. Oncotarget 6:22666-22679. https://doi.org/10.1158/1541-7786.MCR-09-0144   DOI
17 Carter BZ, Mak DH, Schober WD et al (2010) Simultaneous activation of p53 and inhibition of XIAP enhance the activation of apoptosis signaling pathways in AML. Blood 115:306-314. https://doi.org/10.1182/blood-2009-03-212563   DOI
18 Kwon YJ, Baek HS, Ye DJ, Shin S, Kim D, Chun YJ (2016) CYP1B1 enhances cell proliferation and metastasis through induction of EMT and activation of Wnt/β-catenin signaling via sp1 upregulation. PLoS One 11:e0151598. https://doi.org/10.1371/journal.pone.0151598   DOI
19 Baek HS, Kwon YJ, Ye DJ, Cho E, Kwon TU, Chun YJ (2019) CYP1B1 prevents proteasome-mediated XIAP degradation by inducing PKCε activation and phosphorylation of XIAP. Biochim Biophys Acta Mol Cell Res 1866:118553. https://doi.org/10.1016/j.bbamcr.2019.118553   DOI
20 Park HS, Aiyar SE, Fan P et al (2007) Effects of tetramethoxystilbene on hormone-resistant breast cancer cells: biological and biochemical mechanisms of action. Cancer Res 67:5717-5726. https://doi.org/10.1158/0008-5472.CAN-07-0056   DOI
21 Yan C, Huang D, Zhang Y (2011) The involvement of ROS overproduction and mitochondrial dysfunction in PBDE-47-induced apoptosis on Jurkat cells. Exp Toxicol Pathol 63:413-417. https://doi.org/10.1016/j.etp.2010.02.018   DOI
22 Yu SW, Andrabi SA, Wang H et al (2006) Apoptosis-inducing factor mediates poly(ADP-ribose) (PAR) polymer-induced cell death. Proc Natl Acad Sci USA 103:18314-18319. https://doi.org/10.1073/pnas.0606528103   DOI
23 Kuganesan N, Dlamini S, Tillekeratne LMV, Taylor WR (2021) Tumor suppressor p53 promotes ferroptosis in oxidative stress conditions independent of modulation of ferroptosis by p21, CDKs, RB, and E2F. J Biol Chem 297:101365. https://doi.org/10.1016/j.jbc.2021.101365   DOI
24 Delavallee L, Mathiah N, Cabon L et al (2020) Mitochondrial AIF loss causes metabolic reprogramming, caspase-independent cell death blockade, embryonic lethality, and perinatal hydrocephalus. Mol Metab 40:101027. https://doi.org/10.1016/j.molmet.2020.101027   DOI
25 Kwon YJ, Shin S, Chun YJ (2021) Biological roles of cytochrome P450 1A1, 1A2, and 1B1 enzymes. Arch Pharm Res 44:63-83. https://doi.org/10.1007/s12272-021-01306-w   DOI
26 Tallarida RJ (2001) Drug synergism: its detection and applications. J Pharmacol Exp Ther 298:865-872. https://jpet.aspetjournals.org/content/298/3/865.long
27 Schimmer AD, Dalili S, Batey RA, Riedl SJ (2006) Targeting XIAP for the treatment of malignancy. Cell Death Differ 13:179-188. https://doi.org/10.1038/sj.cdd.4401826   DOI
28 Murray GI (2000) The role of cytochrome P450 in tumour development and progression and its potential in therapy. J Pathol 192:419-426. https://doi.org/10.1002/1096-9896(2000)9999:9999::AID-PATH7503.0.CO;2-0   DOI
29 Chun YJ, Kim S, Kim D, Lee SK, Guengerich FP (2001) A new selective and potent inhibitor of human cytochrome P450 1B1 and its application to antimutagenesis. Cancer Res 61:8164-8170. https://aacrjournals.org/cancerres/article/61/22/8164/508230/ANew-Selective-and-Potent-Inhibitor-of-Human
30 Kim SW, Jung HK, Kim MY (2008) Induction of p27kip1 by 2,4,3',5'-tetramethoxystilbene is regulated by protein phosphatase 2A-dependent Akt dephosphorylation in PC-3 prostate cancer cells. Arch Pharm Res 31:1187-1194. https://doi.org/10.1007/s12272-001-1287-1   DOI
31 Cosentino K, Garcia-Saez AJ (2017) Bax and bak pores: are we closing the circle. Trends Cell Biol 27:266-275. https://doi.org/10.1016/j.tcb.2016.11.004   DOI
32 Kuwana T, Mackey MR, Perkins G et al (2002) Bid, Bax, and lipids cooperate to form supramolecular openings in the outer mitochondrial membrane. Cell 111:331-342. https://doi.org/10.1016/s0092-8674(02)01036-x   DOI
33 Schimmer AD (2004) Inhibitor of apoptosis proteins: translating basic knowledge into clinical practice. Cancer Res 64:7183-7190. https://doi.org/10.1158/0008-5472.CAN-04-1918   DOI
34 Kocik J, Machula M, Wisniewska A, Surmiak E, Holak TA, Skalniak L (2019) Helping the released guardian: drug combinations for supporting the anticancer activity of HDM2 (MDM2) antagonists. Cancers (Basel) 11:1014. https://doi.org/10.3390/cancers11071014   DOI
35 Davis JR, Mossalam M, Lim CS (2013) Controlled access of p53 to the nucleus regulates its proteasomal degradation by MDM2. Mol Pharm 10:1340-1349. https://doi.org/10.1021/mp300543t   DOI
36 Khoo KH, Verma CS, Lane DP (2014) Drugging the p53 pathway: understanding the route to clinical efficacy. Nat Rev Drug Discov 13:217-236. https://doi.org/10.1038/nrd4236   DOI
37 Nakamura M, Obata T, Daikoku T, Fujiwara H (2019) The association and significance of p53 in gynecologic cancers: the potential of targeted therapy. Int J Mol Sci 20:5482. https://doi.org/10.3390/ijms20215482   DOI
38 Tsao CC, Corn PG (2010) MDM-2 antagonists induce p53-dependent cell cycle arrest but not cell death in renal cancer cell lines. Cancer Biol Ther 10:1315-1325. https://doi.org/10.4161/cbt.10.12.13612   DOI
39 Tonsing-Carter E, Bailey BJ, Saadatzadeh MR et al (2015) Potentiation of carboplatin-mediated DNA damage by the mdm2 modulator nutlin-3a in a humanized orthotopic breast-to-lung metastatic model. Mol Cancer Ther 14:2850-2863. https://doi.org/10.1158/1535-7163.MCT-15-0237   DOI
40 Turner N, Moretti E, Siclari O et al (2013) Targeting triple negative breast cancer: is p53 the answer? Cancer Treat Rev 39:541-550. https://doi.org/10.1016/j.ctrv.2012.12.001   DOI
41 Chun YJ, Lee SK, Kim MY (2005) Modulation of human cytochrome P450 1B1 expression by 2,4,3',5'-tetramethoxystilbene. Drug Metab Dispos 33:1771-1776. https://doi.org/10.1124/dmd.105.006502   DOI
42 Yu SW, Wang H, Poitras MF et al (2002) Mediation of poly (ADP-ribose) polymerase-1-dependent cell death by apoptosisinducing factor. Science 297:259-263. https://doi.org/10.1126/science.1072221   DOI
43 Shi Y (2004) Caspase activation, inhibition, and reactivation: a mechanistic view. Protein Sci 13:1979-1987. https://doi.org/10.1110/ps.04789804   DOI
44 Sung H, Ferlay J, Siegel RL et al (2021) Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin 71:209-249. https://doi.org/10.3322/caac.21660   DOI
45 Marth C, Landoni F, Mahner S, McCormack M, Gonzalez-Martin A, Colombo N (2017) Cervical cancer: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up. Ann Oncol 28:72-83. https://doi.org/10.1093/annonc/mdx220   DOI
46 Wrzesien-Kus A, Smolewski P, Sobczak-Pluta A, Wierzbowska A, Robak T (2004) The inhibitor of apoptosis protein family and its antagonists in acute leukemias. Apoptosis 9:705-715. https://doi.org/10.1023/B:APPT.0000045788.61012.b2   DOI
47 Tovar C, Rosinski J, Filipovic Z et al (2006) Small-molecule MDM2 antagonists reveal aberrant p53 signaling in cancer: implications for therapy. Proc Natl Acad Sci USA 103:1888-1893. https://doi.org/10.1073/pnas.0507493103   DOI
48 Ohnstad HO, Paulsen EB, Noordhuis P et al (2011) MDM2 antagonist nutlin-3a potentiates antitumour activity of cytotoxic drugs in sarcoma cell lines. BMC Cancer 11:211:1-11. https://doi.org/10.1186/1471-2407-11-211   DOI
49 Kwon YJ, Cho NH, Ye DJ, Baek HS, Ryu YS, Chun YJ (2018) Cytochrome P450 1B1 promotes cancer cell survival via specificity protein 1 (Sp1)-mediated suppression of death receptor 4. J Toxicol Environ Health A 81:278-287. https://doi.org/10.1080/15287394.2018.1440186   DOI
50 Hong M, Park N, Chun YJ (2014) Role of annexin a5 on mitochondria-dependent apoptosis induced by tetramethoxystilbene in human breast cancer cells. Biomol Ther (Seoul) 22:519-524. https://doi.org/10.4062/biomolther.2014.112   DOI
51 Urbano A, Lakshmanan U, Choo PH et al (2005) AIF suppresses chemical stress-induced apoptosis and maintains the transformed state of tumor cells. EMBO J 24:2815-2826. https://doi.org/10.1038/sj.emboj.7600746   DOI
52 Palenski TL, Sorenson CM, Jefcoate CR, Sheibani N (2013) Lack of Cyp1b1 promotes the proliferative and migratory phenotype of perivascular supporting cells. Lab Invest 93:646-662. https://doi.org/10.1038/labinvest.2013.55   DOI