Skip to main content
Top
Published in: Cancer Cell International 1/2019

Open Access 01-12-2019 | Breast Cancer | Review

Impaired redox regulation of estrogen metabolizing proteins is important determinant of human breast cancers

Authors: Smarajit Maiti, Aarifa Nazmeen

Published in: Cancer Cell International | Issue 1/2019

Login to get access

Abstract

Estrogen evidently involves critically in the pathogenesis of gynaecological-cancers. Reports reveal that interference in estrogen-signalling can influence cell-cycle associated regulatory-processes in female reproductive-organs. The major determinants that influence E2-signallings are estrogen-receptor (ER), estrogen-sulfotransferase (SULT1E1), sulfatase (STS), and a formylglycine-generating-enzyme (FGE) which regulates STS activity. The purpose of this mini review was to critically analyze the correlation between oxidative-threats and redox-regulation in the process of estrogen signalling. It is extensively investigated and reported that oxidative-stress is linked to cancer. But no definite mechanism has been explored till date. The adverse effects of oxidative-threat/free-radicals (like genotoxic-effects, gene-regulation, and mitochondrial impairment) have been linked to several diseases like diabetes/cardiovascular-syndrome/stroke and cancer. However, a significant correlation between oxidative-stress and gynaecological-cancers are repeatedly reported without pointing a definite mechanism. For the first time in our study we have investigated the relationship between oxidative stress and the regulation of estrogen via estrogen metabolizing proteins. Reports reveal that ER, SULT1E1, STS and FGE are target-molecules of oxidative-stress and may function differently in oxidizing and reducing environment. In addition, estrogen itself can induce oxidative-stress. This fact necessitates identifying the critical connecting events between oxidative-stress and regulation of estrogen-associated-molecules (ER, SULT1E1, STS, and FGE) that favors tumorigenesis/carcinogenesis. The current review focus is on unique redox-regulation of estrogen and its regulatory-molecules via oxidative-stress. This mechanistic-layout may identify new therapeutic-targets and open further scopes to treat gynecological-cancers more effectively.
Literature
1.
go back to reference Xu WH, Xiang YB, Ruan ZX, Zheng W, Cheng JR, Dai Q, Gao YT, Shu XO. Menstrual and reproductive factors and endometrial cancer risk: results from a population-based case–control study in urban Shanghai. Int J Cancer. 2004;108:613–9.CrossRefPubMed Xu WH, Xiang YB, Ruan ZX, Zheng W, Cheng JR, Dai Q, Gao YT, Shu XO. Menstrual and reproductive factors and endometrial cancer risk: results from a population-based case–control study in urban Shanghai. Int J Cancer. 2004;108:613–9.CrossRefPubMed
2.
go back to reference Zucchetto A, Serraino D, Polesel J, Negri E, De Paoli A, Dal Maso L, Montella M, La Vecchia C, Franceschi S, Talamini R. Hormone-related factors and gynecological conditions in relation to endometrial cancer risk. Eur J Cancer Prev. 2009;18:316–21.CrossRefPubMed Zucchetto A, Serraino D, Polesel J, Negri E, De Paoli A, Dal Maso L, Montella M, La Vecchia C, Franceschi S, Talamini R. Hormone-related factors and gynecological conditions in relation to endometrial cancer risk. Eur J Cancer Prev. 2009;18:316–21.CrossRefPubMed
3.
go back to reference Kaaks R, Berrino F, Key T, Rinaldi S, Dossus L, Biessy C, Secreto G, Amiano P, Bingham S, Boeing H, Bueno de Mesquita HB, Chang-Claude J, Clavel-Chapelon F, Fournier A, van Gils CH, Gonzalez CA, Gurrea AB, Critselis E, Khaw KT, Krogh V, Lahmann PH, Nagel G, Olsen A, Onland-Moret NC, Overvad K, Palli D, Panico S, Peeters P, Quiros JR, Roddam A, Thiebaut A, Tjonneland A, Chirlaque MD, Trichopoulou A, Trichopoulos D, Tumino R, Vineis P, Norat T, Ferrari P, Slimani N, Riboli E. Serum sex steroids in premenopausal women and breast cancer risk within the European Prospective Investigation into Cancer and Nutrition (EPIC). J Natl Cancer Inst. 2005;97:755–65.CrossRefPubMed Kaaks R, Berrino F, Key T, Rinaldi S, Dossus L, Biessy C, Secreto G, Amiano P, Bingham S, Boeing H, Bueno de Mesquita HB, Chang-Claude J, Clavel-Chapelon F, Fournier A, van Gils CH, Gonzalez CA, Gurrea AB, Critselis E, Khaw KT, Krogh V, Lahmann PH, Nagel G, Olsen A, Onland-Moret NC, Overvad K, Palli D, Panico S, Peeters P, Quiros JR, Roddam A, Thiebaut A, Tjonneland A, Chirlaque MD, Trichopoulou A, Trichopoulos D, Tumino R, Vineis P, Norat T, Ferrari P, Slimani N, Riboli E. Serum sex steroids in premenopausal women and breast cancer risk within the European Prospective Investigation into Cancer and Nutrition (EPIC). J Natl Cancer Inst. 2005;97:755–65.CrossRefPubMed
4.
go back to reference Castagnetta L, Granata OM, Cocciadiferro L, Saetta A, Polito L, Bronte G, Rizzo S, Campisi I, Agostara B, Carruba G. 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, Rizzo S, Campisi I, Agostara B, Carruba G. Sex steroids, carcinogenesis, and cancer progression. Ann N Y Acad Sci. 2004;1028:233–46.CrossRefPubMed
5.
go back to reference The National Toxicology Program (NTP). Federal Report on Carcinogens. 2002;177283–5. The National Toxicology Program (NTP). Federal Report on Carcinogens. 2002;177283–5.
6.
go back to reference Hofseth LJ, Raafat AM, Osuch JR, Pathak DR, Slomski CA, Haslam SZ. Hormone replacement therapy with estrogen or estrogen plus medroxyprogesterone acetate is associated with increased epithelial proliferation in the normal postmenopausal breast. J Clin Endocrinol Metab. 1999;84:4559–65.PubMed Hofseth LJ, Raafat AM, Osuch JR, Pathak DR, Slomski CA, Haslam SZ. Hormone replacement therapy with estrogen or estrogen plus medroxyprogesterone acetate is associated with increased epithelial proliferation in the normal postmenopausal breast. J Clin Endocrinol Metab. 1999;84:4559–65.PubMed
7.
go back to reference Bocchinfuso WP, Korach KS. Mammary gland development and tumorigenesis in estrogen receptor knockout mice. J Mammary Gland Biol Neoplasia. 1997;2:323–34.CrossRefPubMed Bocchinfuso WP, Korach KS. Mammary gland development and tumorigenesis in estrogen receptor knockout mice. J Mammary Gland Biol Neoplasia. 1997;2:323–34.CrossRefPubMed
8.
go back to reference Devanesan P, Santen RJ, Bocchinfuso WP, Korach KS, Rogan EG, Cavalieri E. Catechol estrogen metabolites and conjugates in mammary tumors and hyperplastic tissue from estrogen receptor-alpha knock-out (ERKO)/Wnt-1 mice: implications for initiation of mammary tumors. Carcinogenesis. 2001;22:1573–6.CrossRefPubMed Devanesan P, Santen RJ, Bocchinfuso WP, Korach KS, Rogan EG, Cavalieri E. Catechol estrogen metabolites and conjugates in mammary tumors and hyperplastic tissue from estrogen receptor-alpha knock-out (ERKO)/Wnt-1 mice: implications for initiation of mammary tumors. Carcinogenesis. 2001;22:1573–6.CrossRefPubMed
9.
go back to reference Chang XZ, Li DQ, Hou YF, Wu J, Lu JS, Di GH, Jin W, Ou ZL, Shen ZZ, Shao ZM. Identification of the functional role of peroxiredoxin 6 in the progression of breast cancer. Breast Cancer. 2007;9:R76.CrossRef Chang XZ, Li DQ, Hou YF, Wu J, Lu JS, Di GH, Jin W, Ou ZL, Shen ZZ, Shao ZM. Identification of the functional role of peroxiredoxin 6 in the progression of breast cancer. Breast Cancer. 2007;9:R76.CrossRef
10.
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
11.
go back to reference Takahashi K, Okada M, Ozaki T. Safety and efficacy of oestriol for symptoms of natural or surgically induced menopause. Hum Reprod. 2000;15:1028–36.CrossRefPubMed Takahashi K, Okada M, Ozaki T. Safety and efficacy of oestriol for symptoms of natural or surgically induced menopause. Hum Reprod. 2000;15:1028–36.CrossRefPubMed
12.
go back to reference Maggiolini M, Vivacqua A, Fasanella G, Recchia AG, Sisci D, Pezzi V. The G protein coupled receptor GPR30 mediates c-fos up-regulation by 17β-estradiol and phytoestrogens in breast cancer cells. J Biol Chem. 2004;279:27008–16.CrossRefPubMed Maggiolini M, Vivacqua A, Fasanella G, Recchia AG, Sisci D, Pezzi V. The G protein coupled receptor GPR30 mediates c-fos up-regulation by 17β-estradiol and phytoestrogens in breast cancer cells. J Biol Chem. 2004;279:27008–16.CrossRefPubMed
13.
go back to reference Nilsson S, Makela S, Treuter E, Tujague M, Thomsen J, Andersson G, Enmark E, Pettersson K, Warner M, Gustafsson JA. Mechanisms of estrogen action. Physiol Rev. 2001;81:1535–65.CrossRefPubMed Nilsson S, Makela S, Treuter E, Tujague M, Thomsen J, Andersson G, Enmark E, Pettersson K, Warner M, Gustafsson JA. Mechanisms of estrogen action. Physiol Rev. 2001;81:1535–65.CrossRefPubMed
14.
go back to reference Kazuhiro I, Kuniko Horie I, Satoshi I. Identification of estrogen-responsive genes based on the DNA binding properties of estrogen receptors using high-throughput sequencing technology. Acta Pharmacol Sin. 2015;36(1):24–31.CrossRef Kazuhiro I, Kuniko Horie I, Satoshi I. Identification of estrogen-responsive genes based on the DNA binding properties of estrogen receptors using high-throughput sequencing technology. Acta Pharmacol Sin. 2015;36(1):24–31.CrossRef
15.
go back to reference Lappano R, Rosano C, De Marco P, De Francesco EM, Pezzi V, Maggiolini M. Estriol acts as a GPR30 antagonist in estrogen receptor-negative breast cancer cells. Mol Cell Endocrinol. 2010;320:162–7010.CrossRefPubMed Lappano R, Rosano C, De Marco P, De Francesco EM, Pezzi V, Maggiolini M. Estriol acts as a GPR30 antagonist in estrogen receptor-negative breast cancer cells. Mol Cell Endocrinol. 2010;320:162–7010.CrossRefPubMed
16.
go back to reference El-Attar HA, Sheta MI. Hepatocyte growth factor profile with breast cancer. Indian J Pathol Microbiol. 2011;54:509–13.CrossRefPubMed El-Attar HA, Sheta MI. Hepatocyte growth factor profile with breast cancer. Indian J Pathol Microbiol. 2011;54:509–13.CrossRefPubMed
17.
go back to reference Girgert R, Emons G, Gründker C. Inactivation of GPR30 reduces growth of triple-negative breast cancer cells: possible application in targeted therapy. Breast Cancer Res Treat. 2012;134:199–205.CrossRefPubMedPubMedCentral Girgert R, Emons G, Gründker C. Inactivation of GPR30 reduces growth of triple-negative breast cancer cells: possible application in targeted therapy. Breast Cancer Res Treat. 2012;134:199–205.CrossRefPubMedPubMedCentral
18.
go back to reference Hao R, Bondesson M, Singh AV, Riu A, Mc Collum CW, Knudsen TB, Gorelick DA, Gustafsson JA. Identification of estrogen target genes during zebrafish embryonic development through transcriptomic analysis. PLoS ONE. 2013;8:11.CrossRef Hao R, Bondesson M, Singh AV, Riu A, Mc Collum CW, Knudsen TB, Gorelick DA, Gustafsson JA. Identification of estrogen target genes during zebrafish embryonic development through transcriptomic analysis. PLoS ONE. 2013;8:11.CrossRef
19.
go back to reference Yuriko K, Doi H, Ono Y, Urata Y, Goto S, Kitajima M, Miura K, Li TS, Masuzaki H. Estrogen deficiency heterogeneously affects tissue specific stem cells in mice. Sci Rep. 2015;5:12861.CrossRef Yuriko K, Doi H, Ono Y, Urata Y, Goto S, Kitajima M, Miura K, Li TS, Masuzaki H. Estrogen deficiency heterogeneously affects tissue specific stem cells in mice. Sci Rep. 2015;5:12861.CrossRef
20.
go back to reference Lamote I, Meyer E, Massart-Leen AM, Burvenich C. Sex steroids and growth factors in the regulation of mammary gland proliferation, differentiation, and involution. Steroids. 2004;69(3):145–59.CrossRefPubMed Lamote I, Meyer E, Massart-Leen AM, Burvenich C. Sex steroids and growth factors in the regulation of mammary gland proliferation, differentiation, and involution. Steroids. 2004;69(3):145–59.CrossRefPubMed
21.
go back to reference Serra R, Crowley MR. Mouse models of transforming growth factor beta impact in breast development and cancer. Endocr Relat Cancer. 2005;12:749–60.CrossRefPubMed Serra R, Crowley MR. Mouse models of transforming growth factor beta impact in breast development and cancer. Endocr Relat Cancer. 2005;12:749–60.CrossRefPubMed
22.
go back to reference Camacho PM. Evidence-based endocrinology, vol. 20. Philadelphia: Lippincott Williams & Wilkins; 2012. p. 98. Camacho PM. Evidence-based endocrinology, vol. 20. Philadelphia: Lippincott Williams & Wilkins; 2012. p. 98.
23.
go back to reference Kariagina A, Xie J, Leipprandt JR, Haslam SZ. Amphiregulin mediates estrogen, progesterone, and EGFR signaling in the normal rat mammary gland and in hormone-dependent rat mammary cancers. Horm Cancer. 2010;15:229–44.CrossRef Kariagina A, Xie J, Leipprandt JR, Haslam SZ. Amphiregulin mediates estrogen, progesterone, and EGFR signaling in the normal rat mammary gland and in hormone-dependent rat mammary cancers. Horm Cancer. 2010;15:229–44.CrossRef
24.
go back to reference Rawlings JS, Rosler KM, Harrison DA. The JAK/STAT signaling pathway. J Cell Sci. 2004;117:1281–3.CrossRefPubMed Rawlings JS, Rosler KM, Harrison DA. The JAK/STAT signaling pathway. J Cell Sci. 2004;117:1281–3.CrossRefPubMed
25.
go back to reference Jain A, Sharma G, Kushwah V, Garg NK, Kesharwani P, Ghoshal G, Singh B, Shivhare US, Jain S, Katare OP. Methotrexate and beta-carotene loaded-lipid polymer hybrid nanoparticles: a preclinical study for breast cancer. Nanomedicine. 2017;12(15):1851–72.CrossRefPubMed Jain A, Sharma G, Kushwah V, Garg NK, Kesharwani P, Ghoshal G, Singh B, Shivhare US, Jain S, Katare OP. Methotrexate and beta-carotene loaded-lipid polymer hybrid nanoparticles: a preclinical study for breast cancer. Nanomedicine. 2017;12(15):1851–72.CrossRefPubMed
26.
go back to reference Zhu B, Yu L, Yue Q. Co-delivery of vincristine and quercetin by nanocarriers for lymphoma combination chemotherapy. Biomed Pharmacother. 2017;91:287–94.CrossRefPubMed Zhu B, Yu L, Yue Q. Co-delivery of vincristine and quercetin by nanocarriers for lymphoma combination chemotherapy. Biomed Pharmacother. 2017;91:287–94.CrossRefPubMed
27.
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
28.
29.
go back to reference Chen X, Maiti S, Zhang J, Chen G. Nuclear receptor interactions in methotrexate induction of human dehydroepiandrosterone sulfotransferase (hSULT2A1). J Biochem Mol Toxicol. 2006;20(6):309–17.CrossRefPubMed Chen X, Maiti S, Zhang J, Chen G. Nuclear receptor interactions in methotrexate induction of human dehydroepiandrosterone sulfotransferase (hSULT2A1). J Biochem Mol Toxicol. 2006;20(6):309–17.CrossRefPubMed
30.
go back to reference Asi N, Mohammed K, Haydour Q, Gionfriddo MR, Vargas OL, Prokop LJ, Faubion SS, Murad MH. Progesterone vs. synthetic progestins and the risk of breast cancer: a systematic review and meta-analysis. Syst Rev. 2016;5(1):121.CrossRefPubMedPubMedCentral Asi N, Mohammed K, Haydour Q, Gionfriddo MR, Vargas OL, Prokop LJ, Faubion SS, Murad MH. Progesterone vs. synthetic progestins and the risk of breast cancer: a systematic review and meta-analysis. Syst Rev. 2016;5(1):121.CrossRefPubMedPubMedCentral
31.
go back to reference Salazar M, Lerma-Ortiz A, Hooks GM, Ashley AK, Ashley RL. Progestin-mediated activation of MAPK and AKT in nuclear progesterone receptor negative breast epithelial cells: the role of membrane progesterone receptors. Gene. 2016;91(1):6–13.CrossRef Salazar M, Lerma-Ortiz A, Hooks GM, Ashley AK, Ashley RL. Progestin-mediated activation of MAPK and AKT in nuclear progesterone receptor negative breast epithelial cells: the role of membrane progesterone receptors. Gene. 2016;91(1):6–13.CrossRef
32.
go back to reference Rojas PA, May M, Sequeira GR, Elia A, Alvarez M, Martínez P, Gonzalez P, Hewitt S, He X, Perou CM, Molinolo A, Gibbons L, Abba MC, Gass H, Lanari C. Progesterone receptor isoform ratio: a breast cancer prognostic and predictive factor for antiprogestin responsiveness. J Natl Cancer Inst. 2017;109(7):djw317.CrossRefPubMedCentral Rojas PA, May M, Sequeira GR, Elia A, Alvarez M, Martínez P, Gonzalez P, Hewitt S, He X, Perou CM, Molinolo A, Gibbons L, Abba MC, Gass H, Lanari C. Progesterone receptor isoform ratio: a breast cancer prognostic and predictive factor for antiprogestin responsiveness. J Natl Cancer Inst. 2017;109(7):djw317.CrossRefPubMedCentral
33.
go back to reference Knutson TP, Truong TH, Ma S, Brady NJ, Sullivan ME, Raj G, Schwertfeger KL, Lange CA. Posttranslationally modified progesterone receptors direct ligand-specific expression of breast cancer stem cell-associated gene programs. Hematol Oncol. 2017;10(1):89.CrossRef Knutson TP, Truong TH, Ma S, Brady NJ, Sullivan ME, Raj G, Schwertfeger KL, Lange CA. Posttranslationally modified progesterone receptors direct ligand-specific expression of breast cancer stem cell-associated gene programs. Hematol Oncol. 2017;10(1):89.CrossRef
34.
go back to reference Sato T, Tran TH, Peck AR, Girondo MA, Liu C, Goodman CR, Neilson LM, Freydin B, Chervoneva I, Hyslop T, Kovatich AJ. Prolactin suppresses a progestin-induced CK5-positive cell population in luminal breast cancer through inhibition of progestin-driven BCL6 expression. Oncogene. 2014;33(17):2215–24.CrossRefPubMed Sato T, Tran TH, Peck AR, Girondo MA, Liu C, Goodman CR, Neilson LM, Freydin B, Chervoneva I, Hyslop T, Kovatich AJ. Prolactin suppresses a progestin-induced CK5-positive cell population in luminal breast cancer through inhibition of progestin-driven BCL6 expression. Oncogene. 2014;33(17):2215–24.CrossRefPubMed
35.
go back to reference Fettig LM, McGinn O, Finlay-Schultz J, LaBarbera DV, Nordeen SK, Sartorius CA. Cross talk between progesterone receptors and retinoic acid receptors in regulation of cytokeratin 5-positive breast cancer cells. Oncogene. 2017;36:6074–84.CrossRefPubMedPubMedCentral Fettig LM, McGinn O, Finlay-Schultz J, LaBarbera DV, Nordeen SK, Sartorius CA. Cross talk between progesterone receptors and retinoic acid receptors in regulation of cytokeratin 5-positive breast cancer cells. Oncogene. 2017;36:6074–84.CrossRefPubMedPubMedCentral
36.
go back to reference Maiti S, Chen X, Chen G. All-trans retinoic acid induction of sulfotransferases. Basic Clin Pharmacol Toxicol. 2005;96(1):44–53.CrossRefPubMed Maiti S, Chen X, Chen G. All-trans retinoic acid induction of sulfotransferases. Basic Clin Pharmacol Toxicol. 2005;96(1):44–53.CrossRefPubMed
37.
go back to reference Cauley JA, Lucas FL, Kuller LH, Stone K, Browner W, Cummings SR. Elevated serum estradiol and testosterone concentrations are associated with a high risk for breast cancer. Study of Osteoporotic Fractures Research Group. Ann Intern Med. 1999;13:270–7.CrossRef Cauley JA, Lucas FL, Kuller LH, Stone K, Browner W, Cummings SR. Elevated serum estradiol and testosterone concentrations are associated with a high risk for breast cancer. Study of Osteoporotic Fractures Research Group. Ann Intern Med. 1999;13:270–7.CrossRef
38.
go back to reference Dimitrakakis C. Androgens and breast cancer in men and women. Endocrinol Metab Clin N Am. 2011;40:533–47.CrossRef Dimitrakakis C. Androgens and breast cancer in men and women. Endocrinol Metab Clin N Am. 2011;40:533–47.CrossRef
39.
go back to reference Glaser RL, Dimitrakakis C. Reduced breast cancer incidence in women treated with subcutaneous testosterone, or testosterone with anastrozole: a prospective, observational study. Maturitas. 2013;76:342–9.CrossRefPubMed Glaser RL, Dimitrakakis C. Reduced breast cancer incidence in women treated with subcutaneous testosterone, or testosterone with anastrozole: a prospective, observational study. Maturitas. 2013;76:342–9.CrossRefPubMed
40.
go back to reference Santen RJ, Brodie H, Simpson ER, Siiteri PK, Brodie A. History of aromatase: saga of an important biological mediator and therapeutic target. Endocr Rev. 2009;30:343–75.CrossRefPubMed Santen RJ, Brodie H, Simpson ER, Siiteri PK, Brodie A. History of aromatase: saga of an important biological mediator and therapeutic target. Endocr Rev. 2009;30:343–75.CrossRefPubMed
41.
go back to reference Sasano H, Suzuki T, Miki Y, Moriya T. Intracrinology of estrogens and androgens in breast carcinoma. J Steroid Biochem Mol Biol. 2008;108:181–5.CrossRefPubMed Sasano H, Suzuki T, Miki Y, Moriya T. Intracrinology of estrogens and androgens in breast carcinoma. J Steroid Biochem Mol Biol. 2008;108:181–5.CrossRefPubMed
42.
go back to reference Hickey TE, Robinson JLL, Carroll JS, Tilley WD. Minireview: the androgen receptor in breast tissues: growth inhibitor, tumor suppressor, oncogene. Mol Endocrinol. 2012;26:1252–67.CrossRefPubMedPubMedCentral Hickey TE, Robinson JLL, Carroll JS, Tilley WD. Minireview: the androgen receptor in breast tissues: growth inhibitor, tumor suppressor, oncogene. Mol Endocrinol. 2012;26:1252–67.CrossRefPubMedPubMedCentral
43.
go back to reference Suchar LA, Chang RL, Rosen RT, Lech J, Conney AH. High-performance liquid chromatography separation of hydroxylated estradiol metabolites: formation of estradiol metabolites by liver microsomes from male and female rats. J Pharmacol Exp Ther. 1995;272:197–206.PubMed Suchar LA, Chang RL, Rosen RT, Lech J, Conney AH. High-performance liquid chromatography separation of hydroxylated estradiol metabolites: formation of estradiol metabolites by liver microsomes from male and female rats. J Pharmacol Exp Ther. 1995;272:197–206.PubMed
44.
go back to reference Zhang F, Bolton JL. Synthesis of the equine estrogen metabolites 2-hydroxyequilin and 2-hydroxyequilenin. Chem Res Toxicol. 1999;12:200–3.CrossRefPubMed Zhang F, Bolton JL. Synthesis of the equine estrogen metabolites 2-hydroxyequilin and 2-hydroxyequilenin. Chem Res Toxicol. 1999;12:200–3.CrossRefPubMed
45.
go back to reference Esra B, Umit MS, Cansin S, Serpil E, Omer K. Oxidative stress and antioxidant defense. World Allergy Organ J. 2012;5(1):9–19.CrossRef Esra B, Umit MS, Cansin S, Serpil E, Omer K. Oxidative stress and antioxidant defense. World Allergy Organ J. 2012;5(1):9–19.CrossRef
46.
go back to reference Schutze N, Vollmer G, Knuppen R. Catecholestrogens are agonists of estrogen receptor dependent gene expression in MCF-7 cells. J Steroid Biochem Mol Biol. 1994;48:453–61.CrossRefPubMed Schutze N, Vollmer G, Knuppen R. Catecholestrogens are agonists of estrogen receptor dependent gene expression in MCF-7 cells. J Steroid Biochem Mol Biol. 1994;48:453–61.CrossRefPubMed
47.
go back to reference Zhu BT, Han GZ, Shim JY, Wen Y, Jiang XR. Quantitative structure–activity relationship of various endogenous estrogen metabolites for human estrogen receptor alpha and beta subtypes: insights into the structural determinants favoring a differential subtype binding. Endocrinology. 2006;147:4132–50.CrossRefPubMed Zhu BT, Han GZ, Shim JY, Wen Y, Jiang XR. Quantitative structure–activity relationship of various endogenous estrogen metabolites for human estrogen receptor alpha and beta subtypes: insights into the structural determinants favoring a differential subtype binding. Endocrinology. 2006;147:4132–50.CrossRefPubMed
48.
go back to reference Tsutsui T, Tamura Y, Hagiwara M, Miyachi T, Hikiba H, Kubo C, Barrett JC. Induction of mammalian cell transformation and genotoxicity by 2-methoxyestradiol, an endogenous metabolite of estrogen. Carcinogenesis. 2000;21:735–40.CrossRefPubMed Tsutsui T, Tamura Y, Hagiwara M, Miyachi T, Hikiba H, Kubo C, Barrett JC. Induction of mammalian cell transformation and genotoxicity by 2-methoxyestradiol, an endogenous metabolite of estrogen. Carcinogenesis. 2000;21:735–40.CrossRefPubMed
49.
go back to reference Sutherland TE, Schuliga M, Harris T, Eckhardt BL, Anderson RL, Quan L, Stewart A. G2-Methoxyestradiol is an estrogen receptor agonist that supports tumor growth in murine xenograft models of breast cancer. Clin Cancer Res. 2005;11:1722–32.CrossRefPubMed Sutherland TE, Schuliga M, Harris T, Eckhardt BL, Anderson RL, Quan L, Stewart A. G2-Methoxyestradiol is an estrogen receptor agonist that supports tumor growth in murine xenograft models of breast cancer. Clin Cancer Res. 2005;11:1722–32.CrossRefPubMed
50.
go back to reference Mobley JA, Brueggemeier RW. Estrogen receptor-mediated regulation of oxidative stress and DNA damage in breast cancer. Carcinogenesis. 2004;25:3–9.CrossRefPubMed Mobley JA, Brueggemeier RW. Estrogen receptor-mediated regulation of oxidative stress and DNA damage in breast cancer. Carcinogenesis. 2004;25:3–9.CrossRefPubMed
51.
go back to reference Geisler J, Sasano H, Chen S, Purohit A. Steroid sulfatase inhibitors: promising new tools for breast cancer therapy? J Steroid Biochem Mol Biol. 2011;125:39–45.CrossRefPubMed Geisler J, Sasano H, Chen S, Purohit A. Steroid sulfatase inhibitors: promising new tools for breast cancer therapy? J Steroid Biochem Mol Biol. 2011;125:39–45.CrossRefPubMed
52.
go back to reference Carlstrom K, Bergqvist A, Ljungberg O. Metabolism of estrone sulfate in endometriotic tissue and in uterine endometrium in proliferative and secretory cycle phase. Fertil Steril. 1988;49:229–33.CrossRefPubMed Carlstrom K, Bergqvist A, Ljungberg O. Metabolism of estrone sulfate in endometriotic tissue and in uterine endometrium in proliferative and secretory cycle phase. Fertil Steril. 1988;49:229–33.CrossRefPubMed
53.
go back to reference Santen RJ, Leszczynski D, Tilson-Mallet N, Feil PD, Wright C, Manni A, Santner SJ, Ann NY. Enzymatic control of estrogen production in human breast cancer: relative significance of aromatase versus sulfatase pathways. Acad Sci. 1986;464:126–37.CrossRef Santen RJ, Leszczynski D, Tilson-Mallet N, Feil PD, Wright C, Manni A, Santner SJ, Ann NY. Enzymatic control of estrogen production in human breast cancer: relative significance of aromatase versus sulfatase pathways. Acad Sci. 1986;464:126–37.CrossRef
55.
go back to reference Cavalieri E, Frenkel K, Liehr JG, Rogan E, Roy D. Chapter 4: estrogens as endogenous genotoxic agents—DNA adducts and mutations. J Natl Cancer Inst Monogr. 2000;27:75–94.CrossRef Cavalieri E, Frenkel K, Liehr JG, Rogan E, Roy D. Chapter 4: estrogens as endogenous genotoxic agents—DNA adducts and mutations. J Natl Cancer Inst Monogr. 2000;27:75–94.CrossRef
56.
go back to reference Bolton JL, Thatcher GRJ. Potential mechanisms of estrogen quinone carcinogenesis. Chem Res Toxicol. 2008;21:93–101.CrossRefPubMed Bolton JL, Thatcher GRJ. Potential mechanisms of estrogen quinone carcinogenesis. Chem Res Toxicol. 2008;21:93–101.CrossRefPubMed
57.
go back to reference Lavigne JA, Goodman JE, Fonong T, Odwin S, He P, Roberts DW, Yager JD. The effects of catechol-O-methyltransferase inhibition on estrogen metabolite and oxidative DNA damage levels in estradiol-treated MCF-7 cells. Cancer Res. 2001;61:7488–94.PubMed Lavigne JA, Goodman JE, Fonong T, Odwin S, He P, Roberts DW, Yager JD. The effects of catechol-O-methyltransferase inhibition on estrogen metabolite and oxidative DNA damage levels in estradiol-treated MCF-7 cells. Cancer Res. 2001;61:7488–94.PubMed
58.
go back to reference Banerjee S, Li SA, Li JJ. Induction of chromosome aberrations in Syrian hamster renal cortical cells by various estrogens. Mutat Res. 1994;311:191–7.CrossRefPubMed Banerjee S, Li SA, Li JJ. Induction of chromosome aberrations in Syrian hamster renal cortical cells by various estrogens. Mutat Res. 1994;311:191–7.CrossRefPubMed
59.
60.
go back to reference Sauer H, Wartenberg M, Hescheler J. Reactive oxygen species as intracellular messengers during cell growth and differentiation. Cell Physiol Biochem. 2001;11:173–86.CrossRefPubMed Sauer H, Wartenberg M, Hescheler J. Reactive oxygen species as intracellular messengers during cell growth and differentiation. Cell Physiol Biochem. 2001;11:173–86.CrossRefPubMed
61.
go back to reference Han X, Liehr JG. 8-Hydroxylation of guanine bases in kidney and liver DNA of hamsters treated with estradiol: role of free radicals in estrogen-induced carcinogenesis. Cancer Res. 1994;54:5515–7.PubMed Han X, Liehr JG. 8-Hydroxylation of guanine bases in kidney and liver DNA of hamsters treated with estradiol: role of free radicals in estrogen-induced carcinogenesis. Cancer Res. 1994;54:5515–7.PubMed
62.
go back to reference Mobley JA, Brueggemeier RW. Estrogen receptor-mediated regulation of oxidative stress and DNA damage in breast cancer. Carcinogenesis. 2004;25(1):3–9.CrossRefPubMed Mobley JA, Brueggemeier RW. Estrogen receptor-mediated regulation of oxidative stress and DNA damage in breast cancer. Carcinogenesis. 2004;25(1):3–9.CrossRefPubMed
63.
go back to reference Kalyanaraman B, Hintz P, Sealy RC. An electron spin resonance study of free radicals from catechol estrogens. In: Fed Proc, vol. 45, no. 10. 1986. p. 2477–84 (Review. PubMed PMID: 3017766). Kalyanaraman B, Hintz P, Sealy RC. An electron spin resonance study of free radicals from catechol estrogens. In: Fed Proc, vol. 45, no. 10. 1986. p. 2477–84 (Review. PubMed PMID: 3017766).
64.
go back to reference Ei-Bayoumy K. Evaluation of chemopreventive agents against breast cancer and proposed strategies for future clinical intervention trials. Carcinogenesis. 1994;15(11):2637–43.CrossRef Ei-Bayoumy K. Evaluation of chemopreventive agents against breast cancer and proposed strategies for future clinical intervention trials. Carcinogenesis. 1994;15(11):2637–43.CrossRef
65.
go back to reference Laura C, Bridgewater FC, Manning R, Steven RP. Base-specific arrest of in vitro DNA replication by carcinogenic chromium: relationship to DNA interstrand crosslinking. Carcinogenesis. 1994;15(11):2421–7.CrossRef Laura C, Bridgewater FC, Manning R, Steven RP. Base-specific arrest of in vitro DNA replication by carcinogenic chromium: relationship to DNA interstrand crosslinking. Carcinogenesis. 1994;15(11):2421–7.CrossRef
66.
go back to reference Surh YJ, Kundu JK, Na HK, Lee JS. Redox-sensitive transcription factors as prime targets for chemoprevention with anti-inflammatory and antioxidative phytochemicals. J Nutr. 2005;135:2993S–3001S.CrossRefPubMed Surh YJ, Kundu JK, Na HK, Lee JS. Redox-sensitive transcription factors as prime targets for chemoprevention with anti-inflammatory and antioxidative phytochemicals. J Nutr. 2005;135:2993S–3001S.CrossRefPubMed
67.
go back to reference Falany CN, Wheeler J, Oh TS, Falany JL. Steroid sulfation by expressed human cytosolic sulfotransferases. J Steroid Biochem Mol Biol. 1994;48:369–75.CrossRefPubMed Falany CN, Wheeler J, Oh TS, Falany JL. Steroid sulfation by expressed human cytosolic sulfotransferases. J Steroid Biochem Mol Biol. 1994;48:369–75.CrossRefPubMed
68.
go back to reference Maiti S, Chen G. Ethanol up-regulates phenol sulfotransferase (SULT1A1) and hydroxysteroid sulfotransferase (SULT2A1) in rat liver and intestine. Arch Physiol Biochem. 2015;121(2):68–74.CrossRefPubMed Maiti S, Chen G. Ethanol up-regulates phenol sulfotransferase (SULT1A1) and hydroxysteroid sulfotransferase (SULT2A1) in rat liver and intestine. Arch Physiol Biochem. 2015;121(2):68–74.CrossRefPubMed
69.
go back to reference Falany JL, Macrina N, Falany CN. Regulation of MCF-7 breast cancer cell growth by beta-estradiol sulfation. 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-estradiol sulfation. Breast Cancer Res Treat. 2002;74:167–76.CrossRefPubMed
70.
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
71.
go back to reference Anderson E, Howell A. Oestrogen sulfotransferases in malignant and normal human breast tissue. Endocr Relat Cancer. 1995;2:227–33.CrossRef Anderson E, Howell A. Oestrogen sulfotransferases in malignant and normal human breast tissue. Endocr Relat Cancer. 1995;2:227–33.CrossRef
72.
go back to reference Ji XW, Chen GP, Song Y, Hua M, Wang LJ, Li L, Yuan Y, Wang SY, Zhou TY, Lu W. Intratumoral estrogen sulfotransferase 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, Li L, Yuan Y, Wang SY, Zhou TY, Lu W. Intratumoral estrogen sulfotransferase induction contributes to the anti-breast cancer effects of the dithiocarbamate derivative TM208. Acta Pharmacol Sin. 2015;36:1246–55.CrossRefPubMedPubMedCentral
73.
go back to reference Maiti S, Zhang J, Chen G. Redox regulation of human estrogen sulfotransferase (hSULT1E1). Biochem Pharmacol. 2007;73:1474–81.CrossRefPubMed Maiti S, Zhang J, Chen G. Redox regulation of human estrogen sulfotransferase (hSULT1E1). Biochem Pharmacol. 2007;73:1474–81.CrossRefPubMed
74.
go back to reference Guo Y, Hu B, Huang H, Tsung A, Gaikwad NW, Xu M, Jiang M, Ren S, Fan J, Billiar TR, Huang M, Xie W. Estrogen sulfotransferase is an oxidative stress-responsive gene that gender-specifically affects liver ischemia/reperfusion injury. J Biol Chem. 2015;290(14):754–64. Guo Y, Hu B, Huang H, Tsung A, Gaikwad NW, Xu M, Jiang M, Ren S, Fan J, Billiar TR, Huang M, Xie W. Estrogen sulfotransferase is an oxidative stress-responsive gene that gender-specifically affects liver ischemia/reperfusion injury. J Biol Chem. 2015;290(14):754–64.
75.
go back to reference Yin Q, Fischer L, Noethling C, Schaefer WR. In vitro-assessment of putative antiprogestin activities of phytochemicals and synthetic UV absorbers in human endometrial Ishikawa cells. Gynecol Endocrinol. 2015;31:578–81.CrossRefPubMed Yin Q, Fischer L, Noethling C, Schaefer WR. In vitro-assessment of putative antiprogestin activities of phytochemicals and synthetic UV absorbers in human endometrial Ishikawa cells. Gynecol Endocrinol. 2015;31:578–81.CrossRefPubMed
76.
go back to reference Rogelsperger O, Wlcek K, Ekmekcioglu C, Humpeler S, Svoboda M, Königsberg R, Klimpfinger M, Jäger W, Thalhammer T. Melatonin receptors, melatonin metabolizing enzymes and cyclin D1 in human breast cancer. J Recept Signal Transduct Res. 2011;31:180–7.CrossRefPubMed Rogelsperger O, Wlcek K, Ekmekcioglu C, Humpeler S, Svoboda M, Königsberg R, Klimpfinger M, Jäger W, Thalhammer T. Melatonin receptors, melatonin metabolizing enzymes and cyclin D1 in human breast cancer. J Recept Signal Transduct Res. 2011;31:180–7.CrossRefPubMed
77.
go back to reference Sueyoshi T, Green WD, Vinal K, Woodrum TS, Moore R, Negishi M. Garlic extract diallyl sulfide (DAS) activates nuclear receptor CAR to induce the Sult1e1 gene in mouse liver. PLoS ONE. 2011;6:e21229.CrossRefPubMedPubMedCentral Sueyoshi T, Green WD, Vinal K, Woodrum TS, Moore R, Negishi M. Garlic extract diallyl sulfide (DAS) activates nuclear receptor CAR to induce the Sult1e1 gene in mouse liver. PLoS ONE. 2011;6:e21229.CrossRefPubMedPubMedCentral
78.
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;5:637–44.CrossRef Maiti S, Chen G. Tamoxifen induction of aryl sulfotransferase and hydroxysteroid sulfotransferase in male and female rat liver and intestine. Drug Metab Dispos. 2003;5:637–44.CrossRef
79.
go back to reference Atsriku C, Benz CC, Scott GK, Gibson BW, Baldwin MA. Quantification of cysteine oxidation in human estrogen receptor by mass spectrometry. Anal Chem. 2007;79:3083–90.CrossRefPubMedPubMedCentral Atsriku C, Benz CC, Scott GK, Gibson BW, Baldwin MA. Quantification of cysteine oxidation in human estrogen receptor by mass spectrometry. Anal Chem. 2007;79:3083–90.CrossRefPubMedPubMedCentral
80.
go back to reference Hayashi S, Hajiro-Nakanishi K, Makino Y, Eguchi H, Yodoi J, Tanaka H. Functional modulation of estrogen receptor by redox state with reference to thioredoxin as a mediator. Nucleic Acids Res. 1997;25:4035–40.CrossRefPubMedPubMedCentral Hayashi S, Hajiro-Nakanishi K, Makino Y, Eguchi H, Yodoi J, Tanaka H. Functional modulation of estrogen receptor by redox state with reference to thioredoxin as a mediator. Nucleic Acids Res. 1997;25:4035–40.CrossRefPubMedPubMedCentral
81.
go back to reference Stanway SJ, Delavault P, Purohit A. Steroid sulfatase: a new target for the endocrine therapy of breast cancer. Oncologist. 2007;12:370–4.CrossRefPubMed Stanway SJ, Delavault P, Purohit A. Steroid sulfatase: a new target for the endocrine therapy of breast cancer. Oncologist. 2007;12:370–4.CrossRefPubMed
82.
go back to reference Zaichuk D, Ivancic D, Scholtens D. Tissue-specific transcripts of human steroid sulfatase are under control of estrogen signaling pathways in breast carcinoma. J Steroid Biochem Mol Biol. 2007;105:76–84.CrossRefPubMed Zaichuk D, Ivancic D, Scholtens D. Tissue-specific transcripts of human steroid sulfatase are under control of estrogen signaling pathways in breast carcinoma. J Steroid Biochem Mol Biol. 2007;105:76–84.CrossRefPubMed
83.
go back to reference Secky L, Svoboda M, Klameth L, Bajna E, Hamilton G, Zeillinger R, Jager W, Thalhammer T. The sulfatase pathway for estrogen formation: targets for the treatment and diagnosis of hormone-associated tumors. J Drug Deliv. 2013;95:7605. Secky L, Svoboda M, Klameth L, Bajna E, Hamilton G, Zeillinger R, Jager W, Thalhammer T. The sulfatase pathway for estrogen formation: targets for the treatment and diagnosis of hormone-associated tumors. J Drug Deliv. 2013;95:7605.
84.
go back to reference Couse JF, Lindzey J, Grandien K, Gustafsson JA, Korach KS. Tissue distribution and quantitative analysis of estrogen receptor-alpha (ERα) and estrogen receptor-beta (ER-β) messenger ribonucleic acid in the wild-type and ERα-knockout mouse. Endocrinology. 1997;138:4613–21.CrossRefPubMed Couse JF, Lindzey J, Grandien K, Gustafsson JA, Korach KS. Tissue distribution and quantitative analysis of estrogen receptor-alpha (ERα) and estrogen receptor-beta (ER-β) messenger ribonucleic acid in the wild-type and ERα-knockout mouse. Endocrinology. 1997;138:4613–21.CrossRefPubMed
85.
go back to reference Dierks T, Lecca MR, Schlotterhose P, Schmidt B, von Figura K. Sequence determinants directing conversion of cysteine to formylglycine in eukaryotic sulfatases. EMBO J. 1999;8:2084–91.CrossRef Dierks T, Lecca MR, Schlotterhose P, Schmidt B, von Figura K. Sequence determinants directing conversion of cysteine to formylglycine in eukaryotic sulfatases. EMBO J. 1999;8:2084–91.CrossRef
86.
go back to reference Dierks T, Dickmanns A, Preusser-Kunze A, Schmidt B, Mariappan M, von Figura K, Ficner R. Rudolph MG Molecular basis for multiple sulfatase deficiency and mechanism for formylglycine generation of the human formylglycine-generating enzyme. Cell. 2005;121(4):541–52.CrossRefPubMed Dierks T, Dickmanns A, Preusser-Kunze A, Schmidt B, Mariappan M, von Figura K, Ficner R. Rudolph MG Molecular basis for multiple sulfatase deficiency and mechanism for formylglycine generation of the human formylglycine-generating enzyme. Cell. 2005;121(4):541–52.CrossRefPubMed
87.
go back to reference Dierks T, Lecca MR, Schmidt B, von Figura K. A novel protein modification generating an aldehyde group in sulfatases: its role in catalysis and disease. FEBS Lett. 1998;423:61–5.CrossRefPubMed Dierks T, Lecca MR, Schmidt B, von Figura K. A novel protein modification generating an aldehyde group in sulfatases: its role in catalysis and disease. FEBS Lett. 1998;423:61–5.CrossRefPubMed
88.
go back to reference Fey J, Balleininger M, Borissenko LV, Schmidt B, Von Figura K, Dierks T. Characterization of posttranslational formylglycine formation by luminal components of the endoplasmic reticulum. J Biol Chem. 2001;276:47021–8.CrossRefPubMed Fey J, Balleininger M, Borissenko LV, Schmidt B, Von Figura K, Dierks T. Characterization of posttranslational formylglycine formation by luminal components of the endoplasmic reticulum. J Biol Chem. 2001;276:47021–8.CrossRefPubMed
89.
go back to reference Preusser-Kunze A, Mariappan M, Schmidt B, Gande SL, Mutenda K, Wenzel D, von Figura K, Dierks T. Molecular characterization of the human Calpha-formylglycine-generating enzyme. J Biol Chem. 2005;280:14900–10.CrossRefPubMed Preusser-Kunze A, Mariappan M, Schmidt B, Gande SL, Mutenda K, Wenzel D, von Figura K, Dierks T. Molecular characterization of the human Calpha-formylglycine-generating enzyme. J Biol Chem. 2005;280:14900–10.CrossRefPubMed
90.
go back to reference Fraldi A, Biffi A, Lombardi A, Visigalli I, Pepe S, Settembre C, Nusco E, Auricchio A, Naldini L, Ballabio A, Cosma MP. SUMF1 enhances sulfatase activities in vivo in five sulfatase deficiencies. Biochem J. 2007;403:305–12.CrossRefPubMedPubMedCentral Fraldi A, Biffi A, Lombardi A, Visigalli I, Pepe S, Settembre C, Nusco E, Auricchio A, Naldini L, Ballabio A, Cosma MP. SUMF1 enhances sulfatase activities in vivo in five sulfatase deficiencies. Biochem J. 2007;403:305–12.CrossRefPubMedPubMedCentral
91.
go back to reference Dirk R, Andrea PK, Bernhard S, Kathrin G, Julia GW, Thomas D, von Kurt F, Markus GR. A general binding mechanism for all human sulfatases by the formylglycine-generating enzyme. Proc Natl Acad Sci USA. 2006;103:81–6.CrossRef Dirk R, Andrea PK, Bernhard S, Kathrin G, Julia GW, Thomas D, von Kurt F, Markus GR. A general binding mechanism for all human sulfatases by the formylglycine-generating enzyme. Proc Natl Acad Sci USA. 2006;103:81–6.CrossRef
92.
go back to reference Cecchi E, Lapi F, Vannacci A, Banchelli G, Mazzei T, Mugelli A. Increased levels of CA 125 and CA 19.9 serum tumour markers following cyclic combined hormone replacement therapy. J Clin Pharm Ther. 2009;34(1):129–32.CrossRefPubMed Cecchi E, Lapi F, Vannacci A, Banchelli G, Mazzei T, Mugelli A. Increased levels of CA 125 and CA 19.9 serum tumour markers following cyclic combined hormone replacement therapy. J Clin Pharm Ther. 2009;34(1):129–32.CrossRefPubMed
93.
go back to reference Nazmeen A, Maiti S, Mandal K, Roy SK, Ghosh TK, Sinha NK, Mandal K. Better predictive value of Cancer Antigen 125 (CA125) as biomarker in ovary and breast tumors and its correlation with the histopathological type/grade of the disease. Med Chem. 2017;13:796–804.CrossRefPubMed Nazmeen A, Maiti S, Mandal K, Roy SK, Ghosh TK, Sinha NK, Mandal K. Better predictive value of Cancer Antigen 125 (CA125) as biomarker in ovary and breast tumors and its correlation with the histopathological type/grade of the disease. Med Chem. 2017;13:796–804.CrossRefPubMed
94.
go back to reference Maiti S, Dutta SM, Baker SM, Zhang J, Narasaraju T, Liu L, Chen G. In vivo and in vitro oxidative regulation of rat aryl sulfotransferase IV (AST IV). J Biochem Mol Toxicol. 2005;19(2):109–18.CrossRefPubMed Maiti S, Dutta SM, Baker SM, Zhang J, Narasaraju T, Liu L, Chen G. In vivo and in vitro oxidative regulation of rat aryl sulfotransferase IV (AST IV). J Biochem Mol Toxicol. 2005;19(2):109–18.CrossRefPubMed
95.
go back to reference Maiti S, Grant S, Baker SM, Karanth S, Pope CN, Chen G. Stress regulation of sulfotransferases in male rat liver. Biochem Biophys Res Commun. 2004;323(1):235–41.CrossRefPubMed Maiti S, Grant S, Baker SM, Karanth S, Pope CN, Chen G. Stress regulation of sulfotransferases in male rat liver. Biochem Biophys Res Commun. 2004;323(1):235–41.CrossRefPubMed
96.
go back to reference Christopher CB, Christina Y. Ageing, oxidative stress and cancer: paradigms in parallax. Nat Rev Cancer. 2008;8(11):875–9.CrossRef Christopher CB, Christina Y. Ageing, oxidative stress and cancer: paradigms in parallax. Nat Rev Cancer. 2008;8(11):875–9.CrossRef
97.
go back to reference Clemons M, Goss P. Estrogen and the risk of breast cancer. Engl J Med. 2001;344(4):276–85.CrossRef Clemons M, Goss P. Estrogen and the risk of breast cancer. Engl J Med. 2001;344(4):276–85.CrossRef
Metadata
Title
Impaired redox regulation of estrogen metabolizing proteins is important determinant of human breast cancers
Authors
Smarajit Maiti
Aarifa Nazmeen
Publication date
01-12-2019
Publisher
BioMed Central
Published in
Cancer Cell International / Issue 1/2019
Electronic ISSN: 1475-2867
DOI
https://doi.org/10.1186/s12935-019-0826-x

Other articles of this Issue 1/2019

Cancer Cell International 1/2019 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