Skip to main content
Top
Published in: Cancer Cell International 1/2020

Open Access 01-12-2020 | Breast Cancer | Primary research

Breast cancer pathogenesis is linked to the intra-tumoral estrogen sulfotransferase (hSULT1E1) expressions regulated by cellular redox dependent Nrf-2/NFκβ interplay

Authors: Aarifa Nazmeen, Guangping Chen, Tamal Kanti Ghosh, Smarajit Maiti

Published in: Cancer Cell International | Issue 1/2020

Login to get access

Abstract

Background

Estrogen sulfotransferase catalyzes conjugation of sulfuryl-group to estradiol/estrone and regulates E2 availability/activity via estrogen-receptor or non-receptor mediated pathways. Sulfoconjugated estrogen fails to bind estrogen-receptor (ER). High estrogen is a known carcinogen in postmenopausal women. Reports reveal a potential redox-regulation of hSULT1E1/E2-signalling. Further, oxidatively-regulated nuclear-receptor-factor 2 (Nrf2) and NFκβ in relation to hSULT1E1/E2 could be therapeutic-target via cellular redox-modification.

Methods

Here, oxidative stress-regulated SULT1E1-expression was analyzed in human breast carcinoma-tissues and in rat xenografted with human breast-tumor. Tumor and its surrounding tissues were obtained from the district-hospital. Intracellular redox-environment of tumors was screened with some in vitro studies. RT-PCR and western blotting was done for SULT1E1 expression. Immunohistochemistry was performed to analyze SULT1E1/Nrf2/NFκβ localization. Tissue-histoarchitecture/DNA-stability (comet assay) studies were done.

Results

Oxidative-stress induces SULT1E1 via Nrf2/NFκβ cooperatively in tumor-pathogenesis to maintain the required proliferative-state under enriched E2-environment. Higher malondialdehyde/non-protein-soluble-thiol with increased superoxide-dismutase/glutathione-peroxidase/catalase activities was noticed. SULT1E1 expression and E2-level were increased in tumor-tissue compared to their corresponding surrounding-tissues.

Conclusions

It may be concluded that tumors maintain a sustainable oxidative-stress through impaired antioxidants as compared to the surrounding. Liver-tissues from xenografted rat manifested similar E2/antioxidant dysregulations favoring pre-tumorogenic environment.
Literature
1.
go back to reference Ferlay J, et. al. Cancer incidence and mortality worldwide: IARC cancer base no. 11. Lyon, France: International Agency for Research on Cancer; 2014. http://globocan.iarc.fr. Accessed 16 Jan 2015. GLOBOCAN 2012 v1.1. Ferlay J, et. al. Cancer incidence and mortality worldwide: IARC cancer base no. 11. Lyon, France: International Agency for Research on Cancer; 2014. http://​globocan.​iarc.​fr. Accessed 16 Jan 2015. GLOBOCAN 2012 v1.1.
2.
go back to reference Ghoncheh M, Pournamdar Z, Salehiniya H. Incidence and mortality and epidemiology of breast cancer in the world. Asian Pac J Cancer Prev. 2016;17(S3):43–6.CrossRefPubMed Ghoncheh M, Pournamdar Z, Salehiniya H. Incidence and mortality and epidemiology of breast cancer in the world. Asian Pac J Cancer Prev. 2016;17(S3):43–6.CrossRefPubMed
3.
go back to reference Ferley J, Soerjomataram II, Ervik M, Dikshit R, Eser S. Cancer incidence and mortality worldwide: IARC cancer base no. 11. Lyon, France: International Agency for Research on Cancer; 2013. GLOBOCAN 2012 v1. 0. Ferley J, Soerjomataram II, Ervik M, Dikshit R, Eser S. Cancer incidence and mortality worldwide: IARC cancer base no. 11. Lyon, France: International Agency for Research on Cancer; 2013. GLOBOCAN 2012 v1. 0.
5.
go back to reference Hortobagyi GN, de la Garza Salazar J, Pritchard K, et al. The global breast cancer burden: variations in epidemiology and survival. Clin Breast Cancer. 2005;6(5):391–401.CrossRefPubMed Hortobagyi GN, de la Garza Salazar J, Pritchard K, et al. The global breast cancer burden: variations in epidemiology and survival. Clin Breast Cancer. 2005;6(5):391–401.CrossRefPubMed
6.
go back to reference Winters S, Martin C, Murphy D, Shokar NK. Breast cancer epidemiology, prevention, and screening. Prog Mol Biol Transl Sci. 2017;151:1–32.CrossRefPubMed Winters S, Martin C, Murphy D, Shokar NK. Breast cancer epidemiology, prevention, and screening. Prog Mol Biol Transl Sci. 2017;151:1–32.CrossRefPubMed
7.
go back to reference Ito H, Matsuo K. Molecular epidemiology and possible real-world applications in breast cancer. Breast Cancer. 2016;23(1):33–8.CrossRefPubMed Ito H, Matsuo K. Molecular epidemiology and possible real-world applications in breast cancer. Breast Cancer. 2016;23(1):33–8.CrossRefPubMed
8.
go back to reference Olopade OI, Weber BI. Toward molecular characterization of individuals at increased risk for breast cancer: part I. PPO updates. Princ Pract Oncol. 1998;12:1–12. Olopade OI, Weber BI. Toward molecular characterization of individuals at increased risk for breast cancer: part I. PPO updates. Princ Pract Oncol. 1998;12:1–12.
9.
go back to reference Revillion F, Bonneterre J, Peyrat JP. ERBB2 oncogene in human breast cancer and its clinical significance. Eur J Cancer. 1998;34:791–808.CrossRefPubMed Revillion F, Bonneterre J, Peyrat JP. ERBB2 oncogene in human breast cancer and its clinical significance. Eur J Cancer. 1998;34:791–808.CrossRefPubMed
10.
go back to reference Hormonal contraception and post-menopausal hormonal therapy. In: IARC monographs on the evaluation of carcinogenic risks to humans, vol 72. Lyon: WHO IARC Press; 1999. p. 399–530. Hormonal contraception and post-menopausal hormonal therapy. In: IARC monographs on the evaluation of carcinogenic risks to humans, vol 72. Lyon: WHO IARC Press; 1999. p. 399–530.
11.
go back to reference The National Toxicology Program (NTP). Federal Report on Carcinogens. 2002;177283–177285. The National Toxicology Program (NTP). Federal Report on Carcinogens. 2002;177283–177285.
12.
go back to reference Moore SC, Matthews CE, Ou Shu X, et al. Endogenous estrogens, estrogen metabolites, and breast cancer risk in postmenopausal Chinese women. J Natl Cancer Inst. 2016;108(10):djw103.CrossRefPubMedCentral Moore SC, Matthews CE, Ou Shu X, et al. Endogenous estrogens, estrogen metabolites, and breast cancer risk in postmenopausal Chinese women. J Natl Cancer Inst. 2016;108(10):djw103.CrossRefPubMedCentral
13.
go back to reference Castagnetta L, Granata OM, Cocciadiferro L, Saetta A, Polito L, Bronte G, et al. Sex steroids, carcinogenesis, and cancer progression. Ann N Y Acad Sci. 2004;1028:233–46.CrossRefPubMed Castagnetta L, Granata OM, Cocciadiferro L, Saetta A, Polito L, Bronte G, et al. Sex steroids, carcinogenesis, and cancer progression. Ann N Y Acad Sci. 2004;1028:233–46.CrossRefPubMed
14.
go back to reference Susan S, Julie T. Effect of estrogens on skin aging and the potential role of SERMs. Clin Interv Aging. 2007;2:283–97.CrossRef Susan S, Julie T. Effect of estrogens on skin aging and the potential role of SERMs. Clin Interv Aging. 2007;2:283–97.CrossRef
15.
go back to reference Kanda N, Watanabe S. 17-Beta-estradiol enhances the production of nerve growth factor in THP-1-derived macrophages or peripheral blood monocyte-derived macrophages. J Invest Dermatol. 2003;121:771–80.CrossRefPubMed Kanda N, Watanabe S. 17-Beta-estradiol enhances the production of nerve growth factor in THP-1-derived macrophages or peripheral blood monocyte-derived macrophages. J Invest Dermatol. 2003;121:771–80.CrossRefPubMed
16.
go back to reference O’Lone R, Frith MC, Karlsson EK, Hansen U. Genomic targets of nuclear estrogen receptors. Mol Endocrinol. 2004;18:1859–75.CrossRefPubMed O’Lone R, Frith MC, Karlsson EK, Hansen U. Genomic targets of nuclear estrogen receptors. Mol Endocrinol. 2004;18:1859–75.CrossRefPubMed
17.
go back to reference Carlstrom K, Bergqvist A, Ljungberg O. Metabolism of estronesulfate in endometriotic tissue and in uterine endometrium in proliferative and secretory cycle phase. Feril Steril. 1988;49:229–33.CrossRef Carlstrom K, Bergqvist A, Ljungberg O. Metabolism of estronesulfate in endometriotic tissue and in uterine endometrium in proliferative and secretory cycle phase. Feril Steril. 1988;49:229–33.CrossRef
18.
go back to reference Hobkirk R. Steroid sulfotransferases and steroid sulfatases: characteristics and biological roles. Can J Biochem Cell Biol. 1985;63:1127–44.CrossRefPubMed Hobkirk R. Steroid sulfotransferases and steroid sulfatases: characteristics and biological roles. Can J Biochem Cell Biol. 1985;63:1127–44.CrossRefPubMed
19.
go back to reference Aksoy IA, Wood TC, Weinshilboum R. Human liver estrogensulfotransferase: identification by cDNA cloning and expression. Biochem Biophys Res Commun. 1994;200:1621–9.CrossRefPubMed Aksoy IA, Wood TC, Weinshilboum R. Human liver estrogensulfotransferase: identification by cDNA cloning and expression. Biochem Biophys Res Commun. 1994;200:1621–9.CrossRefPubMed
20.
go back to reference Falany JL, Macrina N, Falany CN. Regulation of MCF-7 breast cancer cell growth by beta-estradiolsulfation. Breast Cancer Res Treat. 2002;74:167–76.CrossRefPubMed Falany JL, Macrina N, Falany CN. Regulation of MCF-7 breast cancer cell growth by beta-estradiolsulfation. Breast Cancer Res Treat. 2002;74:167–76.CrossRefPubMed
21.
go back to reference Falany JL, Falany CN. Regulation of estrogen activity by sulfation in MCF-7 human breast cancer cells. Oncol Res. 1997;9:589–96.PubMed Falany JL, Falany CN. Regulation of estrogen activity by sulfation in MCF-7 human breast cancer cells. Oncol Res. 1997;9:589–96.PubMed
22.
go back to reference Anderson E, Howell A. Oestrogen sulfotransferases in malignant and normal human breast tissue. Endocr Related Cancer. 1995;2:227–33.CrossRef Anderson E, Howell A. Oestrogen sulfotransferases in malignant and normal human breast tissue. Endocr Related Cancer. 1995;2:227–33.CrossRef
23.
go back to reference Ji XW, Chen GP, Song Y, Hua M, Wang LJ, et al. Intratumoralestrogensulfotransferase induction contributes to the anti-breast cancer effects of the dithiocarbamate derivative TM208. Acta Pharmacol Sin. 2015;36:1246–55.CrossRefPubMedPubMedCentral Ji XW, Chen GP, Song Y, Hua M, Wang LJ, et al. Intratumoralestrogensulfotransferase induction contributes to the anti-breast cancer effects of the dithiocarbamate derivative TM208. Acta Pharmacol Sin. 2015;36:1246–55.CrossRefPubMedPubMedCentral
24.
go back to reference Wu YJ, Muldoon LL, Dickey DT, Lewin SJ, Varallyay CG, et al. Cyclophosphamide enhances human tumor growth in nude rat xenograftedtumor Models. Neoplasia. 2009;11:187–95.CrossRefPubMedPubMedCentral Wu YJ, Muldoon LL, Dickey DT, Lewin SJ, Varallyay CG, et al. Cyclophosphamide enhances human tumor growth in nude rat xenograftedtumor Models. Neoplasia. 2009;11:187–95.CrossRefPubMedPubMedCentral
25.
go back to reference Ligresti A, Moriello AS, Starowicz K, et al. Antitumor activity of plant Cannabinoids with emphasis on the effect of cannabidiol on human breast carcinoma. J Pharmacol Exp Ther. 2006;318:1375–87.CrossRefPubMed Ligresti A, Moriello AS, Starowicz K, et al. Antitumor activity of plant Cannabinoids with emphasis on the effect of cannabidiol on human breast carcinoma. J Pharmacol Exp Ther. 2006;318:1375–87.CrossRefPubMed
27.
28.
go back to reference Christine JW, Joseph JC. Measurement of superoxide dismutase, catalase, and glutathione peroxidase in cultured cells and tissue. Nat Protoc. 2010;5:51–66.CrossRef Christine JW, Joseph JC. Measurement of superoxide dismutase, catalase, and glutathione peroxidase in cultured cells and tissue. Nat Protoc. 2010;5:51–66.CrossRef
29.
go back to reference Maiti S, Zhang J, Chen G. Redox regulation of human estrogensulfotransferase (hSULT1E1). Biochem Pharmacol. 2007;73:1474–81.CrossRefPubMed Maiti S, Zhang J, Chen G. Redox regulation of human estrogensulfotransferase (hSULT1E1). Biochem Pharmacol. 2007;73:1474–81.CrossRefPubMed
30.
go back to reference Acharyya N, Chattopadhyay S, Maiti S. Chemoprevention against arsenic-induced mutagenic DNA breakage and apoptotic liver damage in rat via antioxidant and SOD1 upregulation by green tea (Camellia sinensis) which recovers broken DNA resulted from arsenic-H2O2 related in vitro oxidant stress. J Environ Sci Health C Environ Carcinog Ecotoxicol. 2014;32:338–61.CrossRef Acharyya N, Chattopadhyay S, Maiti S. Chemoprevention against arsenic-induced mutagenic DNA breakage and apoptotic liver damage in rat via antioxidant and SOD1 upregulation by green tea (Camellia sinensis) which recovers broken DNA resulted from arsenic-H2O2 related in vitro oxidant stress. J Environ Sci Health C Environ Carcinog Ecotoxicol. 2014;32:338–61.CrossRef
31.
go back to reference Maiti S, Nazmeen A. Impaired redox regulation of estrogen metabolizing proteins is important determinant of human breast cancers. Cancer Cell Int. 2019;15(19):111.CrossRef Maiti S, Nazmeen A. Impaired redox regulation of estrogen metabolizing proteins is important determinant of human breast cancers. Cancer Cell Int. 2019;15(19):111.CrossRef
32.
go back to reference Yue W, Wang JP, Li Y, et al. Effects of estrogen on breast cancer development: role of estrogen receptor independent mechanisms. Int J Cancer. 2010;127:1748–57.CrossRefPubMedPubMedCentral Yue W, Wang JP, Li Y, et al. Effects of estrogen on breast cancer development: role of estrogen receptor independent mechanisms. Int J Cancer. 2010;127:1748–57.CrossRefPubMedPubMedCentral
33.
go back to reference Manning HC, Buck JR, Cook RS. Mouse models of breast cancer: platforms for discovering precision imaging diagnostics and future cancer medicine. J Nucl Med. 2016;1:60S–8S.CrossRef Manning HC, Buck JR, Cook RS. Mouse models of breast cancer: platforms for discovering precision imaging diagnostics and future cancer medicine. J Nucl Med. 2016;1:60S–8S.CrossRef
34.
go back to reference Kaaks R, Rinaldi S, Key TJ, et al. Postmenopausal serum androgens, oestrogens and breast cancer risk: the European prospective investigation into cancer and nutrition. Endocr Relat Cancer. 2005;12:1071–82.CrossRefPubMed Kaaks R, Rinaldi S, Key TJ, et al. Postmenopausal serum androgens, oestrogens and breast cancer risk: the European prospective investigation into cancer and nutrition. Endocr Relat Cancer. 2005;12:1071–82.CrossRefPubMed
35.
go back to reference Nazmeen A, Maiti S. Oxidant stress induction and signaling in xenografted (human breast cancer-tissues) plus estradiol treated or N-ethyl-N-nitrosourea treated female rats via altered estrogen sulfotransferase (rSULT1E1) expressions and SOD1/catalase regulations. Mol Biol Rep. 2018;45(6):2571–84.CrossRefPubMed Nazmeen A, Maiti S. Oxidant stress induction and signaling in xenografted (human breast cancer-tissues) plus estradiol treated or N-ethyl-N-nitrosourea treated female rats via altered estrogen sulfotransferase (rSULT1E1) expressions and SOD1/catalase regulations. Mol Biol Rep. 2018;45(6):2571–84.CrossRefPubMed
36.
go back to reference Maiti S, Chen G. Methotrexate is a novel inducer of rat liver and intestinal sulfotransferases. Arch Biochem Biophys. 2003;418(2):161–8.CrossRefPubMed Maiti S, Chen G. Methotrexate is a novel inducer of rat liver and intestinal sulfotransferases. Arch Biochem Biophys. 2003;418(2):161–8.CrossRefPubMed
37.
go back to reference Maiti S, Chen G. Tamoxifen induction of aryl sulfotransferase and hydroxysteroid sulfotransferase in male and female rat liver and intestine. Drug Metab Dispos. 2003;31(5):637–44.CrossRefPubMed Maiti S, Chen G. Tamoxifen induction of aryl sulfotransferase and hydroxysteroid sulfotransferase in male and female rat liver and intestine. Drug Metab Dispos. 2003;31(5):637–44.CrossRefPubMed
38.
go back to reference zu Schwabedissen HEM, Tirona RG, Yip CS, Ho RH, Kim RB, et al. Interplay between the nuclear receptor PXR and the uptake transporter OATP1A2 selectively enhances estrogen effects in breast cancer. Cancer Res. 2008;68:9338–47.CrossRef zu Schwabedissen HEM, Tirona RG, Yip CS, Ho RH, Kim RB, et al. Interplay between the nuclear receptor PXR and the uptake transporter OATP1A2 selectively enhances estrogen effects in breast cancer. Cancer Res. 2008;68:9338–47.CrossRef
39.
go back to reference Park SK, Yim DS, Yoon KS, et al. Combined effect of GSTM1, GSTT1, and COMT genotypes in individual breast cancer risk. Breast Cancer Res Treat. 2004;88:55–62.CrossRefPubMed Park SK, Yim DS, Yoon KS, et al. Combined effect of GSTM1, GSTT1, and COMT genotypes in individual breast cancer risk. Breast Cancer Res Treat. 2004;88:55–62.CrossRefPubMed
40.
go back to reference Jerome F, Strauss R, Barbieri L. The synthesis and metabolism of steroid hormones. Yen Jaffe’s Reprod Endocrinol. 2014;7:66–92. Jerome F, Strauss R, Barbieri L. The synthesis and metabolism of steroid hormones. Yen Jaffe’s Reprod Endocrinol. 2014;7:66–92.
41.
go back to reference Gilligan LC, Gondal A, Tang V, et al. Estrone sulfate transport and steroid sulfatase activity in colorectal cancer: implications for hormone replacement therapy. Front Pharmacol. 2017;8:103.CrossRefPubMedPubMedCentral Gilligan LC, Gondal A, Tang V, et al. Estrone sulfate transport and steroid sulfatase activity in colorectal cancer: implications for hormone replacement therapy. Front Pharmacol. 2017;8:103.CrossRefPubMedPubMedCentral
42.
go back to reference Bulun SE, Cheng YH, Pavone ME, et al. Beta-hydroxysteroid dehydrogenase-2 deficiency and progesterone resistance in endometriosis. Semin Reprod Med. 2010;28:44–50.CrossRefPubMedPubMedCentral Bulun SE, Cheng YH, Pavone ME, et al. Beta-hydroxysteroid dehydrogenase-2 deficiency and progesterone resistance in endometriosis. Semin Reprod Med. 2010;28:44–50.CrossRefPubMedPubMedCentral
43.
go back to reference Pasqualini JR. Estrogensulfotransferases in breast and endometrial cancers. Ann N Y Acad Sci. 2009;1155:88–98.CrossRefPubMed Pasqualini JR. Estrogensulfotransferases in breast and endometrial cancers. Ann N Y Acad Sci. 2009;1155:88–98.CrossRefPubMed
45.
go back to reference Utsumi T, Yoshimura N, Takeuchi S, et al. Steroid sulfatase expression is an independent predictor of recurrence in human breast cancer. Cancer Res. 1999;59:377–81.PubMed Utsumi T, Yoshimura N, Takeuchi S, et al. Steroid sulfatase expression is an independent predictor of recurrence in human breast cancer. Cancer Res. 1999;59:377–81.PubMed
46.
go back to reference Poisson DP, Song D, Luu-The V, et al. Pelletier expression of estrogen sulfotransferase 1E1 and steroid sulfatase in breast cancer: a immunohistochemical study. Breast Cancer. 2009;3:9–21. Poisson DP, Song D, Luu-The V, et al. Pelletier expression of estrogen sulfotransferase 1E1 and steroid sulfatase in breast cancer: a immunohistochemical study. Breast Cancer. 2009;3:9–21.
47.
go back to reference Xu Y, Lin X, Xu J, Jing H, Qin Y, Li Y. SULT1E1 inhibits cell proliferation and invasion by activating PPARγ in breast cancer. J Cancer. 2018;9:1078–87.CrossRefPubMedPubMedCentral Xu Y, Lin X, Xu J, Jing H, Qin Y, Li Y. SULT1E1 inhibits cell proliferation and invasion by activating PPARγ in breast cancer. J Cancer. 2018;9:1078–87.CrossRefPubMedPubMedCentral
48.
go back to reference Mobley JA, BhatA S, Brueggemeier RW. Measurement of oxidative DNA damage by catechol estrogens and analogues in vitro. Chem Res Toxicol. 1999;12:270–7.CrossRefPubMed Mobley JA, BhatA S, Brueggemeier RW. Measurement of oxidative DNA damage by catechol estrogens and analogues in vitro. Chem Res Toxicol. 1999;12:270–7.CrossRefPubMed
49.
go back to reference Chang M, Zhang F, Shen L, et al. Inhibition of glutathione S-transferase activity by the quinoidmetabo-lites of equine estrogens. Chem Res Toxicol. 1998;11:758–65.CrossRefPubMed Chang M, Zhang F, Shen L, et al. Inhibition of glutathione S-transferase activity by the quinoidmetabo-lites of equine estrogens. Chem Res Toxicol. 1998;11:758–65.CrossRefPubMed
50.
go back to reference Sarabia SF, Zhu BT, Kurosawa T, Tohma M, Liehr JG. Mechanism of cytochrome P450-catalyzed aromatic hydroxylation of estrogens. Chem Res Toxicol. 1997;10:767.CrossRefPubMed Sarabia SF, Zhu BT, Kurosawa T, Tohma M, Liehr JG. Mechanism of cytochrome P450-catalyzed aromatic hydroxylation of estrogens. Chem Res Toxicol. 1997;10:767.CrossRefPubMed
51.
go back to reference Rygiel TP. The Rac activator Tiam1 prevents keratinocyte apoptosis by controlling ROS mediated ERK phosphorylation. J Cell Sci. 2008;121:1183–92.CrossRefPubMed Rygiel TP. The Rac activator Tiam1 prevents keratinocyte apoptosis by controlling ROS mediated ERK phosphorylation. J Cell Sci. 2008;121:1183–92.CrossRefPubMed
52.
go back to reference Zhou J, Chen Y, Lang JY, Lu JJ, Ding J. Salvicine inactivates beta 1 integrin and inhibits adhesion of MDA-MB-435 cells to fibronectin via reactive oxygen species signalling. Mol Cancer Res. 2008;6:194–204.CrossRefPubMed Zhou J, Chen Y, Lang JY, Lu JJ, Ding J. Salvicine inactivates beta 1 integrin and inhibits adhesion of MDA-MB-435 cells to fibronectin via reactive oxygen species signalling. Mol Cancer Res. 2008;6:194–204.CrossRefPubMed
53.
go back to reference Mazzio EA, Soliman KF. Glioma cell antioxidant capacity relative to reactive oxygen species produced by dopamine. J Appl Toxicol. 2004;24:99–106.CrossRefPubMed Mazzio EA, Soliman KF. Glioma cell antioxidant capacity relative to reactive oxygen species produced by dopamine. J Appl Toxicol. 2004;24:99–106.CrossRefPubMed
54.
go back to reference Zhang Y, Zhao W, Zhang HJ, Domann FE, Oberley LW. Overexpression of copper zinc superoxide dismutase suppresses human glioma cell growth. Cancer Res. 2002;62:1205–12.PubMed Zhang Y, Zhao W, Zhang HJ, Domann FE, Oberley LW. Overexpression of copper zinc superoxide dismutase suppresses human glioma cell growth. Cancer Res. 2002;62:1205–12.PubMed
55.
go back to reference Glorieux C, Calderon PB. Catalase down-regulation in cancer cells exposed to arsenic trioxide is involved in their increased sensitivity to a pro-oxidant treatment. Cancer Cell Int. 2018;18:24.CrossRefPubMedPubMedCentral Glorieux C, Calderon PB. Catalase down-regulation in cancer cells exposed to arsenic trioxide is involved in their increased sensitivity to a pro-oxidant treatment. Cancer Cell Int. 2018;18:24.CrossRefPubMedPubMedCentral
56.
go back to reference Maynard S, Schurman SH, Harboe C, de Souza-Pinto NC, Bohr VA. Base excision repair of oxidative DNA damage and association with cancer and aging. Carcinogenesis. 2009;30:2–10.CrossRefPubMed Maynard S, Schurman SH, Harboe C, de Souza-Pinto NC, Bohr VA. Base excision repair of oxidative DNA damage and association with cancer and aging. Carcinogenesis. 2009;30:2–10.CrossRefPubMed
57.
go back to reference Burdon RH, Gill V, Rice-Evans C. Oxidative stress and tumor cell proliferation. Free Radic Res Commun. 1990;11:65–76.CrossRefPubMed Burdon RH, Gill V, Rice-Evans C. Oxidative stress and tumor cell proliferation. Free Radic Res Commun. 1990;11:65–76.CrossRefPubMed
58.
go back to reference Wang M, Kirk JS, Venkataraman S, et al. Manganese superoxide dismutase suppresses hypoxic induction of hypoxia inducible factor-1alpha and vascular endothelial growth factor. Oncogene. 2005;24:8154–66.CrossRefPubMed Wang M, Kirk JS, Venkataraman S, et al. Manganese superoxide dismutase suppresses hypoxic induction of hypoxia inducible factor-1alpha and vascular endothelial growth factor. Oncogene. 2005;24:8154–66.CrossRefPubMed
59.
60.
go back to reference Meng TC, Fukada T, Tonks NK. Reversible oxidation and inactivation of protein tyrosine phosphatases in vivo. Mol Cell. 2002;9:387–99.CrossRefPubMed Meng TC, Fukada T, Tonks NK. Reversible oxidation and inactivation of protein tyrosine phosphatases in vivo. Mol Cell. 2002;9:387–99.CrossRefPubMed
61.
go back to reference Brunet A, Bonni A, Zigmond MJ, et al. Akt promotes cell survival by phosphorylating and inhibiting a Forkhead transcription factor. Cell. 1999;96:857–68.CrossRefPubMed Brunet A, Bonni A, Zigmond MJ, et al. Akt promotes cell survival by phosphorylating and inhibiting a Forkhead transcription factor. Cell. 1999;96:857–68.CrossRefPubMed
62.
go back to reference Limaye V, Li X, Hahn C, et al. Sphingosine kinase-1 enhances endothelial cell survival through PECAM-1dependent activation of PI-3K/Akt and regulation of Bcl-2 family members. Blood. 2005;105:3169–77.CrossRefPubMed Limaye V, Li X, Hahn C, et al. Sphingosine kinase-1 enhances endothelial cell survival through PECAM-1dependent activation of PI-3K/Akt and regulation of Bcl-2 family members. Blood. 2005;105:3169–77.CrossRefPubMed
63.
go back to reference Parkash J, Felty Q, Roy D. Estrogen exerts a spatial and temporal influence on reactive oxygen species generation that precedes calcium uptake in high-capacity mitochondria: implications for rapid nongenomicsignaling of cell growth. Biochemistry. 2006;45:2872–81.CrossRefPubMed Parkash J, Felty Q, Roy D. Estrogen exerts a spatial and temporal influence on reactive oxygen species generation that precedes calcium uptake in high-capacity mitochondria: implications for rapid nongenomicsignaling of cell growth. Biochemistry. 2006;45:2872–81.CrossRefPubMed
64.
go back to reference Storz P. Mitochondrial ROS–radical detoxification, mediated by protein kinase D. Trends Cell Biol. 2007;17:13–8.CrossRefPubMed Storz P. Mitochondrial ROS–radical detoxification, mediated by protein kinase D. Trends Cell Biol. 2007;17:13–8.CrossRefPubMed
65.
go back to reference Guo Y, Hu B, Huang H, et al. Estrogen sulfotransferase is an oxidative stress-responsive gene that gender-specifically affects liver ischemia/reperfusion injury. J Biol Chem. 2015;290:14754–64.CrossRefPubMedPubMedCentral Guo Y, Hu B, Huang H, et al. Estrogen sulfotransferase is an oxidative stress-responsive gene that gender-specifically affects liver ischemia/reperfusion injury. J Biol Chem. 2015;290:14754–64.CrossRefPubMedPubMedCentral
66.
go back to reference Malhotra D, Portales-Casamar E, Singh A, et al. Global mapping of binding sites for Nrf2 identifies novel targets in cell survival response through ChIP-Seq profiling and network analysis. Nucleic Acids Res. 2010;38:5718–34.CrossRefPubMedPubMedCentral Malhotra D, Portales-Casamar E, Singh A, et al. Global mapping of binding sites for Nrf2 identifies novel targets in cell survival response through ChIP-Seq profiling and network analysis. Nucleic Acids Res. 2010;38:5718–34.CrossRefPubMedPubMedCentral
67.
go back to reference Agyeman AS, Chaerkady R, Shaw PG, et al. Transcriptomic and proteomic profiling of KEAP1 disrupted and sulforaphane-treated human breast epithelial cells reveals common expression profiles. Breast Cancer Res Treat. 2012;132:175–87.CrossRefPubMed Agyeman AS, Chaerkady R, Shaw PG, et al. Transcriptomic and proteomic profiling of KEAP1 disrupted and sulforaphane-treated human breast epithelial cells reveals common expression profiles. Breast Cancer Res Treat. 2012;132:175–87.CrossRefPubMed
68.
go back to reference Lu K, Alcivar AL, Ma J, et al. NRF2 induction supporting breast cancer cell survival is enabled by oxidative stress-induced DPP3-KEAP1 interaction. Cancer Res. 2017;77:2881–92.CrossRefPubMedPubMedCentral Lu K, Alcivar AL, Ma J, et al. NRF2 induction supporting breast cancer cell survival is enabled by oxidative stress-induced DPP3-KEAP1 interaction. Cancer Res. 2017;77:2881–92.CrossRefPubMedPubMedCentral
70.
go back to reference Sporn MB, Liby KT. NRF2 and cancer: the good, the bad and the importance of context. Nat Rev Cancer. 2012;12:564–71.CrossRefPubMed Sporn MB, Liby KT. NRF2 and cancer: the good, the bad and the importance of context. Nat Rev Cancer. 2012;12:564–71.CrossRefPubMed
71.
go back to reference Gong H, Jarzynka MJ, Cole TJ, et al. Glucocorticoids antagonize estrogens by glucocorticoid receptor-mediated activation of estrogen sulfotransferase. Cancer Res. 2008;68:7386–93.CrossRefPubMedPubMedCentral Gong H, Jarzynka MJ, Cole TJ, et al. Glucocorticoids antagonize estrogens by glucocorticoid receptor-mediated activation of estrogen sulfotransferase. Cancer Res. 2008;68:7386–93.CrossRefPubMedPubMedCentral
72.
go back to reference Okamoto K, Tanaka H, Ogawa H, et al. Redox-dependent regulation of nuclear import of the glucocorticoid receptor. J Biol Chem. 1999;274:10363–71.CrossRefPubMed Okamoto K, Tanaka H, Ogawa H, et al. Redox-dependent regulation of nuclear import of the glucocorticoid receptor. J Biol Chem. 1999;274:10363–71.CrossRefPubMed
73.
go back to reference Yamashita Y, Ueyama T, Nishi T, et al. Nrf2-inducing anti-oxidation stress response in the rat liver—new beneficial effect of lansoprazole. PLoS ONE. 2014;9:e97419.CrossRefPubMedPubMedCentral Yamashita Y, Ueyama T, Nishi T, et al. Nrf2-inducing anti-oxidation stress response in the rat liver—new beneficial effect of lansoprazole. PLoS ONE. 2014;9:e97419.CrossRefPubMedPubMedCentral
74.
go back to reference Fu J, Fang H, Paulsen M, Ljungman M, Kocarek TA, Runge-Morris M. Regulation of estrogen sulfotransferase expression by confluence of MCF-10A breast epithelial cells: role of the aryl hydrocarbon receptor. J Pharmacol Exp Ther. 2011;339:597–606.CrossRefPubMedPubMedCentral Fu J, Fang H, Paulsen M, Ljungman M, Kocarek TA, Runge-Morris M. Regulation of estrogen sulfotransferase expression by confluence of MCF-10A breast epithelial cells: role of the aryl hydrocarbon receptor. J Pharmacol Exp Ther. 2011;339:597–606.CrossRefPubMedPubMedCentral
75.
go back to reference Wu JP, Chang LW, Yao HT, et al. Involvement of oxidative stress and activation of aryl hydrocarbon receptor in elevation of CYP1A1 expression and activity in lung cells and tissues by arsenic: an in vitro and in vivo study. Toxicol Sci. 2009;107:385–93.CrossRefPubMed Wu JP, Chang LW, Yao HT, et al. Involvement of oxidative stress and activation of aryl hydrocarbon receptor in elevation of CYP1A1 expression and activity in lung cells and tissues by arsenic: an in vitro and in vivo study. Toxicol Sci. 2009;107:385–93.CrossRefPubMed
77.
go back to reference Morgan MJ, Liu ZG. Crosstalk of reactive oxygen species and NF-κB signaling. Cell Res. 2011;21:103–15.CrossRefPubMed Morgan MJ, Liu ZG. Crosstalk of reactive oxygen species and NF-κB signaling. Cell Res. 2011;21:103–15.CrossRefPubMed
78.
go back to reference Srivastava S, Matsuda M, Hou Z, et al. Receptor activator of NF-kappaB ligand induction via Jak2 and Stat5a in mammary epithelial cells. J Biol Chem. 2003;278:46171–8.CrossRefPubMed Srivastava S, Matsuda M, Hou Z, et al. Receptor activator of NF-kappaB ligand induction via Jak2 and Stat5a in mammary epithelial cells. J Biol Chem. 2003;278:46171–8.CrossRefPubMed
79.
go back to reference Keyomarsi K, Tucker SL, Buchholz TA, et al. Cyclin E and survival in patients with breast cancer. N Engl J Med. 2002;347:1566–75.CrossRefPubMed Keyomarsi K, Tucker SL, Buchholz TA, et al. Cyclin E and survival in patients with breast cancer. N Engl J Med. 2002;347:1566–75.CrossRefPubMed
80.
go back to reference Vakkala M, Kahlos K, Lakari E, Pääkkö P, Kinnula V, Soini Y. Inducible nitric oxide synthase expression, apoptosis, and angiogenesis in in situ and invasive breast carcinomas. Clin Cancer Res. 2000;6:2408–16.PubMed Vakkala M, Kahlos K, Lakari E, Pääkkö P, Kinnula V, Soini Y. Inducible nitric oxide synthase expression, apoptosis, and angiogenesis in in situ and invasive breast carcinomas. Clin Cancer Res. 2000;6:2408–16.PubMed
81.
go back to reference Half E, Tang XM, Gwyn K, Sahin A, Wathen K, Sinicrope FA. Cyclooxygenase-2 expression in human breast cancers and adjacent ductal carcinoma in situ. Cancer Res. 2002;62:1676–81.PubMed Half E, Tang XM, Gwyn K, Sahin A, Wathen K, Sinicrope FA. Cyclooxygenase-2 expression in human breast cancers and adjacent ductal carcinoma in situ. Cancer Res. 2002;62:1676–81.PubMed
82.
go back to reference Skobe M, Hawighorst T, Jackson DG, et al. Induction of tumor lymphangiogenesis by VEGF-C promotes breast cancer metastasis. Nat Med. 2001;7:192–8.CrossRefPubMed Skobe M, Hawighorst T, Jackson DG, et al. Induction of tumor lymphangiogenesis by VEGF-C promotes breast cancer metastasis. Nat Med. 2001;7:192–8.CrossRefPubMed
83.
go back to reference Liao BC, Hsieh CW, Liu YC, Tzeng TT, Sun YW, Wung BS. Cinnamaldehyde inhibits the tumor necrosis factor-alpha-induced expression of cell adhesion molecules in endothelial cells by suppressing NF-kappaB activation: effects upon IkappaB and Nrf2. Toxicol Appl Pharmacol. 2008;229:161–71.CrossRefPubMed Liao BC, Hsieh CW, Liu YC, Tzeng TT, Sun YW, Wung BS. Cinnamaldehyde inhibits the tumor necrosis factor-alpha-induced expression of cell adhesion molecules in endothelial cells by suppressing NF-kappaB activation: effects upon IkappaB and Nrf2. Toxicol Appl Pharmacol. 2008;229:161–71.CrossRefPubMed
84.
go back to reference Banning A, Brigelius-Flohé R. NF-kappaB, Nrf2, and HO-1 interplay in redox-regulated VCAM-1 expression. Antioxid Redox Signal. 2005;7:889–99.CrossRefPubMed Banning A, Brigelius-Flohé R. NF-kappaB, Nrf2, and HO-1 interplay in redox-regulated VCAM-1 expression. Antioxid Redox Signal. 2005;7:889–99.CrossRefPubMed
85.
go back to reference Rushworth SA, MacEwan DJ. HO-1 underlies resistance of AML cells to TNF-induced apoptosis. Blood. 2008;111:3793–801.CrossRefPubMed Rushworth SA, MacEwan DJ. HO-1 underlies resistance of AML cells to TNF-induced apoptosis. Blood. 2008;111:3793–801.CrossRefPubMed
86.
go back to reference Petrache I, Otterbein LE, Alam J, Wiegand GW, Choi AM. Heme oxygenase-1 inhibits TNF-alpha-induced apoptosis in cultured fibroblasts. Am J Physiol Lung Cell Mol Physiol. 2000;278:L312–9.CrossRefPubMed Petrache I, Otterbein LE, Alam J, Wiegand GW, Choi AM. Heme oxygenase-1 inhibits TNF-alpha-induced apoptosis in cultured fibroblasts. Am J Physiol Lung Cell Mol Physiol. 2000;278:L312–9.CrossRefPubMed
87.
go back to reference Liu GH, Qu J, Shen X. NF-kappaB/p65 antagonizes Nrf2-ARE pathway by depriving CBP from Nrf2 and facilitating recruitment of HDAC3 to MafK. Biochim Biophys Acta. 2008;1783:713–27.CrossRefPubMed Liu GH, Qu J, Shen X. NF-kappaB/p65 antagonizes Nrf2-ARE pathway by depriving CBP from Nrf2 and facilitating recruitment of HDAC3 to MafK. Biochim Biophys Acta. 2008;1783:713–27.CrossRefPubMed
89.
go back to reference Yoshida GJ. Therapeutic strategies of drug repositioning targeting autophagy to induce cancer cell death: from pathophysiology to treatment. J Hematol Oncol. 2017;10(1):67.CrossRefPubMedPubMedCentral Yoshida GJ. Therapeutic strategies of drug repositioning targeting autophagy to induce cancer cell death: from pathophysiology to treatment. J Hematol Oncol. 2017;10(1):67.CrossRefPubMedPubMedCentral
90.
go back to reference Kenney PA, Wszolek MF, Rieger-Christ KM, et al. Novel ZEB1 expression in bladder tumorigenesis. BJU Int. 2011;107(4):656–63.CrossRefPubMed Kenney PA, Wszolek MF, Rieger-Christ KM, et al. Novel ZEB1 expression in bladder tumorigenesis. BJU Int. 2011;107(4):656–63.CrossRefPubMed
91.
go back to reference Wu Z, Zhang L, Xu S, Lin Y, Yin W, Lu J, Sha R, Sheng X, Zhou L, Lu J. Predictive and prognostic value of ZEB1 protein expression in breast cancer patients with neoadjuvant chemotherapy. Cancer Cell Int. 2019;19:78.CrossRefPubMedPubMedCentral Wu Z, Zhang L, Xu S, Lin Y, Yin W, Lu J, Sha R, Sheng X, Zhou L, Lu J. Predictive and prognostic value of ZEB1 protein expression in breast cancer patients with neoadjuvant chemotherapy. Cancer Cell Int. 2019;19:78.CrossRefPubMedPubMedCentral
92.
go back to reference Mohammad N, Singh SV, Malvi P, et al. Strategy to enhance efficacy of doxorubicin in solid tumor cells by methyl-β-cyclodextrin: involvement of p53 and Fas receptor ligand complex. Sci Rep. 2015;5:11853.CrossRefPubMedPubMedCentral Mohammad N, Singh SV, Malvi P, et al. Strategy to enhance efficacy of doxorubicin in solid tumor cells by methyl-β-cyclodextrin: involvement of p53 and Fas receptor ligand complex. Sci Rep. 2015;5:11853.CrossRefPubMedPubMedCentral
93.
go back to reference Haque MM, Desai KV. Pathways to endocrine therapy resistance in breast cancer. Front Endocrinol. 2019;10:573.CrossRef Haque MM, Desai KV. Pathways to endocrine therapy resistance in breast cancer. Front Endocrinol. 2019;10:573.CrossRef
94.
go back to reference Kisková T, Mungenast F, Suváková M, Jäger W, Thalhammer T. Future aspects for cannabinoids in breast cancer therapy. Int J Mol Sci. 2019;20(7):1673.CrossRefPubMedCentral Kisková T, Mungenast F, Suváková M, Jäger W, Thalhammer T. Future aspects for cannabinoids in breast cancer therapy. Int J Mol Sci. 2019;20(7):1673.CrossRefPubMedCentral
95.
go back to reference Selli C, Sims AH. Neoadjuvant therapy for breast cancer as a model for translational research. Breast Cancer. 2019;13:1178223419829072.PubMedPubMedCentral Selli C, Sims AH. Neoadjuvant therapy for breast cancer as a model for translational research. Breast Cancer. 2019;13:1178223419829072.PubMedPubMedCentral
Metadata
Title
Breast cancer pathogenesis is linked to the intra-tumoral estrogen sulfotransferase (hSULT1E1) expressions regulated by cellular redox dependent Nrf-2/NFκβ interplay
Authors
Aarifa Nazmeen
Guangping Chen
Tamal Kanti Ghosh
Smarajit Maiti
Publication date
01-12-2020
Publisher
BioMed Central
Published in
Cancer Cell International / Issue 1/2020
Electronic ISSN: 1475-2867
DOI
https://doi.org/10.1186/s12935-020-1153-y

Other articles of this Issue 1/2020

Cancer Cell International 1/2020 Go to the issue
Webinar | 19-02-2024 | 17:30 (CET)

Keynote webinar | Spotlight on antibody–drug conjugates in cancer

Antibody–drug conjugates (ADCs) are novel agents that have shown promise across multiple tumor types. Explore the current landscape of ADCs in breast and lung cancer with our experts, and gain insights into the mechanism of action, key clinical trials data, existing challenges, and future directions.

Dr. Véronique Diéras
Prof. Fabrice Barlesi
Developed by: Springer Medicine