Skip to main content
Top
Published in: Breast Cancer Research and Treatment 2/2010

01-11-2010 | Brief Report

Inhibition of estrogen signaling activates the NRF2 pathway in breast cancer

Authors: Yuan Yao, Angela M. H. Brodie, Nancy E. Davidson, Thomas W. Kensler, Qun Zhou

Published in: Breast Cancer Research and Treatment | Issue 2/2010

Login to get access

Abstract

Exposure to higher levels of estrogen produces genotoxic metabolites that can stimulate mammary tumorigenesis. Induction of NF-E2-related factor 2 (NRF2)-dependent detoxifying enzymes (e.g., NAD(P)H-quinone oxidoreductase 1 (NQO1)) is considered an important mechanism of protection against estrogen-associated carcinogenesis because they would facilitate removal of toxic estrogens. Here, we studied the impact of estrogen-receptor (ER) signaling on NRF2-dependent gene transcription. In luciferase assay experiments using the 5-flanking region of the human NQO1 gene promoter, we observe that ERα ligand-binding domain (LBD) is required for estrogen inhibition of NQO1 promoter activity in estrogen-dependent breast cancer cells. Chromatin immunoprecipitation (ChIP) assay shows that estrogen recruits ERα and a class III histone deacetylase SIRT1 at the NQO1 promoter, leading to inhibition of NQO1 transcription. Inhibition of ERα expression by the antiestrogen shikonin reverses the inhibitory effect of estrogen on NQO1 expression. As a consequence, a chemoprevention study was undertaken to monitor the impact of shikonin on DNA lesions and tumor growth. Treatment of MCF-7 breast cancer cells with shikonin inhibits estrogen-induced 8-hydroxy-2-deoxyguanosine (8-OHdG), a marker of DNA damage. NQO1 deficiency promotes estrogen-dependent tumor formation, and shikonin inhibits estrogen-dependent tumor growth in an NQO1-dependent manner in MCF-7 xenografts. These results suggest that estrogen-receptor signaling pathway has an inhibitory effect on NRF2-dependent enzymes. Moreover, shikonin reverses the inhibitory effects of estrogen on this pathway and may contribute to breast cancer prevention.
Literature
1.
2.
go back to reference Cavalieri EL, Stack DE, Devanesan PD, Todorovic R, Dwivedy I, Higginbotham S, Johansson SL, Patil KD, Gross ML, Gooden JK, Ramanathan R, Cerny RL, Rogan EG (1997) Molecular origin of cancer: catechol estrogen-3,4-quinones as endogenous tumor initiators. Proc Natl Acad Sci USA 94:10937–10942CrossRefPubMed Cavalieri EL, Stack DE, Devanesan PD, Todorovic R, Dwivedy I, Higginbotham S, Johansson SL, Patil KD, Gross ML, Gooden JK, Ramanathan R, Cerny RL, Rogan EG (1997) Molecular origin of cancer: catechol estrogen-3,4-quinones as endogenous tumor initiators. Proc Natl Acad Sci USA 94:10937–10942CrossRefPubMed
3.
go back to reference Yue W, Santen RJ, Wang JP, Li Y, Verderame MF, Bocchinfuso WP, Korach KS, Devanesan P, Todorovic R, Rogan EG, Cavalieri EL (2003) Genotoxic metabolites of estradiol in breast: potential mechanism of estradiol induced carcinogenesis. J Steroid Biochem Mol Biol 86:477–486CrossRefPubMed Yue W, Santen RJ, Wang JP, Li Y, Verderame MF, Bocchinfuso WP, Korach KS, Devanesan P, Todorovic R, Rogan EG, Cavalieri EL (2003) Genotoxic metabolites of estradiol in breast: potential mechanism of estradiol induced carcinogenesis. J Steroid Biochem Mol Biol 86:477–486CrossRefPubMed
4.
go back to reference Wang Z, Wijewickrama GT, Peng KW, Dietz BM, Yuan L, van Breemen RB, Bolton JL, Thatcher GR (2009) Estrogen receptor alpha enhances the rate of oxidative DNA damage by targeting an equine estrogen catechol metabolite to the nucleus. J Biol Chem 284:8633–8642CrossRefPubMed Wang Z, Wijewickrama GT, Peng KW, Dietz BM, Yuan L, van Breemen RB, Bolton JL, Thatcher GR (2009) Estrogen receptor alpha enhances the rate of oxidative DNA damage by targeting an equine estrogen catechol metabolite to the nucleus. J Biol Chem 284:8633–8642CrossRefPubMed
5.
go back to reference Kensler TW, Wakabayashi N, Biswal S (2007) Cell survival responses to environmental stresses via the Keap1-Nrf2-ARE pathway. Annu Rev Pharmacol Toxicol 47:89–116CrossRefPubMed Kensler TW, Wakabayashi N, Biswal S (2007) Cell survival responses to environmental stresses via the Keap1-Nrf2-ARE pathway. Annu Rev Pharmacol Toxicol 47:89–116CrossRefPubMed
6.
go back to reference Kensler TW, Wakabayashi N (2010) Nrf2: friend or foe for chemoprevention? Carcinogenesis 31:90–99CrossRefPubMed Kensler TW, Wakabayashi N (2010) Nrf2: friend or foe for chemoprevention? Carcinogenesis 31:90–99CrossRefPubMed
7.
go back to reference Kwak MK, Kensler TW (2010) Targeting NRF2 signaling for cancer chemoprevention. Toxicol Appl Pharmacol 244:66–76CrossRefPubMed Kwak MK, Kensler TW (2010) Targeting NRF2 signaling for cancer chemoprevention. Toxicol Appl Pharmacol 244:66–76CrossRefPubMed
8.
go back to reference Montano MM, Chaplin LJ, Deng H, Mesia-Vela S, Gaikwad N, Zahid M, Rogan E (2007) Protective roles of quinone reductase and tamoxifen against estrogen-induced mammary. Oncogene 26:3587–3590CrossRefPubMed Montano MM, Chaplin LJ, Deng H, Mesia-Vela S, Gaikwad N, Zahid M, Rogan E (2007) Protective roles of quinone reductase and tamoxifen against estrogen-induced mammary. Oncogene 26:3587–3590CrossRefPubMed
9.
go back to reference Sripathy SP, Chaplin LJ, Gaikwad NW, Rogan EG, Montano MM (2008) hPMC2 is required for recruiting an ERbeta coactivator complex to mediate transcriptional upregulation of NQO1 and protection against oxidative DNA damage by tamoxifen. Oncogene 27:6376–6384CrossRefPubMed Sripathy SP, Chaplin LJ, Gaikwad NW, Rogan EG, Montano MM (2008) hPMC2 is required for recruiting an ERbeta coactivator complex to mediate transcriptional upregulation of NQO1 and protection against oxidative DNA damage by tamoxifen. Oncogene 27:6376–6384CrossRefPubMed
10.
go back to reference Ansell PJ, Espinosa-Nicholas C, Curran EM, Judy BM, Philips BJ, Hannink M, Lubahn DB (2004) In vitro and in vivo regulation of antioxidant response element-dependent gene expression by estrogens. Endocrinology 145:311–317CrossRefPubMed Ansell PJ, Espinosa-Nicholas C, Curran EM, Judy BM, Philips BJ, Hannink M, Lubahn DB (2004) In vitro and in vivo regulation of antioxidant response element-dependent gene expression by estrogens. Endocrinology 145:311–317CrossRefPubMed
11.
go back to reference Ansell PJ, Lo SC, Newton LG, Espinosa-Nicholas C, Zhang DD, Liu JH, Hannink M, Lubahn DB (2005) Repression of cancer protective genes by 17beta-estradiol: ligand-dependent interaction between human Nrf2 and estrogen receptor alpha. Mol Cell Endocrinol 243:27–34CrossRefPubMed Ansell PJ, Lo SC, Newton LG, Espinosa-Nicholas C, Zhang DD, Liu JH, Hannink M, Lubahn DB (2005) Repression of cancer protective genes by 17beta-estradiol: ligand-dependent interaction between human Nrf2 and estrogen receptor alpha. Mol Cell Endocrinol 243:27–34CrossRefPubMed
12.
go back to reference Montano MM, Jaiswal AK, Katzenellenbogen BS (1998) Transcriptional regulation of the human quinone reductase gene by antiestrogen-liganded estrogen receptor-alpha and estrogen receptor-beta. J Biol Chem 273:25443–25449CrossRefPubMed Montano MM, Jaiswal AK, Katzenellenbogen BS (1998) Transcriptional regulation of the human quinone reductase gene by antiestrogen-liganded estrogen receptor-alpha and estrogen receptor-beta. J Biol Chem 273:25443–25449CrossRefPubMed
13.
go back to reference Yao Y, Zhou Q (2010) A novel antiestrogen agent Shikonin inhibits estrogen-dependent gene transcription in human breast cancer cells. Breast Cancer Res Treat 121:233–240CrossRefPubMed Yao Y, Zhou Q (2010) A novel antiestrogen agent Shikonin inhibits estrogen-dependent gene transcription in human breast cancer cells. Breast Cancer Res Treat 121:233–240CrossRefPubMed
14.
go back to reference Singh A, Misra V, Thimmulappa RK, Lee H, Ames S, Hoque MO, Herman JG, Baylin SB, Sidransky D, Gabrielson E, Brock MV, Biswal S (2006) Dysfunctional KEAP1-NRF2 interaction in non-small-cell lung cancer. PLoS Med 3:e420CrossRefPubMed Singh A, Misra V, Thimmulappa RK, Lee H, Ames S, Hoque MO, Herman JG, Baylin SB, Sidransky D, Gabrielson E, Brock MV, Biswal S (2006) Dysfunctional KEAP1-NRF2 interaction in non-small-cell lung cancer. PLoS Med 3:e420CrossRefPubMed
15.
go back to reference Keen JC, Zhou Q, Park BH, Pettit C, Mack KM, Blair B, Brenner K, Davidson NE (2005) Protein phosphatase 2A (PP2A) regulates estrogen receptor alpha (ER) expression through modulation of ER mRNA stability. J Biol Chem 280:29519–29524CrossRefPubMed Keen JC, Zhou Q, Park BH, Pettit C, Mack KM, Blair B, Brenner K, Davidson NE (2005) Protein phosphatase 2A (PP2A) regulates estrogen receptor alpha (ER) expression through modulation of ER mRNA stability. J Biol Chem 280:29519–29524CrossRefPubMed
16.
go back to reference Kim MY, Woo EM, Chong YT, Homenko DR, Kraus WL (2006) Acetylation of estrogen receptor alpha by p300 at lysines 266 and 268 enhances the deoxyribonucleic acid binding and transactivation activities of the receptor. Mol Endocrinol 20:1479–1493CrossRefPubMed Kim MY, Woo EM, Chong YT, Homenko DR, Kraus WL (2006) Acetylation of estrogen receptor alpha by p300 at lysines 266 and 268 enhances the deoxyribonucleic acid binding and transactivation activities of the receptor. Mol Endocrinol 20:1479–1493CrossRefPubMed
17.
go back to reference Kressler D, Hock MB, Kralli A (2007) Coactivators PGC-1beta and SRC-1 interact functionally to promote the agonist activity of the selective estrogen receptor modulator tamoxifen. J Biol Chem 282:26897–26907CrossRefPubMed Kressler D, Hock MB, Kralli A (2007) Coactivators PGC-1beta and SRC-1 interact functionally to promote the agonist activity of the selective estrogen receptor modulator tamoxifen. J Biol Chem 282:26897–26907CrossRefPubMed
18.
go back to reference Yao Y, Li H, Gu Y, Davidson NE, Zhou Q (2010) Inhibition of SIRT1 deacetylase suppresses estrogen receptor signaling. Carcinogenesis 31:382–387CrossRefPubMed Yao Y, Li H, Gu Y, Davidson NE, Zhou Q (2010) Inhibition of SIRT1 deacetylase suppresses estrogen receptor signaling. Carcinogenesis 31:382–387CrossRefPubMed
19.
go back to reference De Iuliis GN, Thomson LK, Mitchell LA, Finnie JM, Koppers AJ, Hedges A, Nixon B, Aitken RJ (2009) DNA damage in human spermatozoa is highly correlated with the efficiency of chromatin remodeling and the formation of 8-hydroxy-2′-deoxyguanosine, a marker of oxidative stress. Biol Reprod 81:517–524CrossRefPubMed De Iuliis GN, Thomson LK, Mitchell LA, Finnie JM, Koppers AJ, Hedges A, Nixon B, Aitken RJ (2009) DNA damage in human spermatozoa is highly correlated with the efficiency of chromatin remodeling and the formation of 8-hydroxy-2′-deoxyguanosine, a marker of oxidative stress. Biol Reprod 81:517–524CrossRefPubMed
20.
go back to reference Long BJ, Jelovac D, Handratta V, Thiantanawat A, MacPherson N, Ragaz J, Goloubeva OG, Brodie AM (2004) Therapeutic strategies using the aromatase inhibitor letrozole and tamoxifen in a breast cancer model. J Natl Cancer Inst 96:456–465CrossRefPubMed Long BJ, Jelovac D, Handratta V, Thiantanawat A, MacPherson N, Ragaz J, Goloubeva OG, Brodie AM (2004) Therapeutic strategies using the aromatase inhibitor letrozole and tamoxifen in a breast cancer model. J Natl Cancer Inst 96:456–465CrossRefPubMed
21.
go back to reference Gottardis MM, Robinson SP, Jordan VC (1998) Estradiol-stimulated growth of MCF-7 tumors implanted in athymic mice: a model to study the tumoristatic action of tamoxifen. J Steroid Biochem 30:311–314CrossRef Gottardis MM, Robinson SP, Jordan VC (1998) Estradiol-stimulated growth of MCF-7 tumors implanted in athymic mice: a model to study the tumoristatic action of tamoxifen. J Steroid Biochem 30:311–314CrossRef
22.
go back to reference Brodie A, Jelovac D, Macedo L, Sabnis G, Tilghman S, Goloubeva O (2005) Therapeutic observations in MCF-7 aromatase xenografts. Clin Cancer Res 11:884s–888sPubMed Brodie A, Jelovac D, Macedo L, Sabnis G, Tilghman S, Goloubeva O (2005) Therapeutic observations in MCF-7 aromatase xenografts. Clin Cancer Res 11:884s–888sPubMed
23.
go back to reference Asher G, Lotem J, Sachs L, Kahana C, Shaul Y (2002) Mdm-2 and ubiquitin-independent p53 proteasomal degradation regulated by NQO1. Proc Natl Acad Sci USA 99:13125–13130CrossRefPubMed Asher G, Lotem J, Sachs L, Kahana C, Shaul Y (2002) Mdm-2 and ubiquitin-independent p53 proteasomal degradation regulated by NQO1. Proc Natl Acad Sci USA 99:13125–13130CrossRefPubMed
24.
go back to reference Tsvetkov P, Asher G, Reiss V, Shaul Y, Sachs L, Lotem J (2005) Inhibition of NAD(P)H:quinone oxidoreductase 1 activity and induction of p53 degradation by the natural phenolic compound curcumin. Proc Natl Acad Sci USA 102:5535–5540CrossRefPubMed Tsvetkov P, Asher G, Reiss V, Shaul Y, Sachs L, Lotem J (2005) Inhibition of NAD(P)H:quinone oxidoreductase 1 activity and induction of p53 degradation by the natural phenolic compound curcumin. Proc Natl Acad Sci USA 102:5535–5540CrossRefPubMed
25.
go back to reference Bianco NR, Perry G, Smith MA, Templeton DJ, Montano MM (2003) Functional implications of antiestrogen induction of quinone reductase: inhibition of estrogen-induced deoxyribonucleic acid damage. Mol Endocrinol 17:1344–1355CrossRefPubMed Bianco NR, Perry G, Smith MA, Templeton DJ, Montano MM (2003) Functional implications of antiestrogen induction of quinone reductase: inhibition of estrogen-induced deoxyribonucleic acid damage. Mol Endocrinol 17:1344–1355CrossRefPubMed
26.
go back to reference Yang H, Zhou P, Huang H, Chen D, Ma N, Cui QC, Shen S, Dong W, Zhang X, Lian W, Wang X, Dou QP, Liu J (2009) Shikonin exerts antitumor activity via proteasome inhibition and cell death induction in vitro and in vivo. Int J Cancer 124:2450–2459CrossRefPubMed Yang H, Zhou P, Huang H, Chen D, Ma N, Cui QC, Shen S, Dong W, Zhang X, Lian W, Wang X, Dou QP, Liu J (2009) Shikonin exerts antitumor activity via proteasome inhibition and cell death induction in vitro and in vivo. Int J Cancer 124:2450–2459CrossRefPubMed
27.
go back to reference Fagerholm R, Hofstetter B, Tommiska J, Aaltonen K, Vrtel R, Syrjäkoski K, Kallioniemi A, Kilpivaara O, Mannermaa A, Kosma VM, Uusitupa M, Eskelinen M, Kataja V, Aittomäki K, von Smitten K, Heikkilä P, Lukas J, Holli K, Bartkova J, Blomqvist C, Bartek J, Nevanlinna H (2008) NAD(P)H:quinone oxidoreductase 1 NQO1*2 genotype (P187S) is a strong prognostic and predictive factor in breast cancer. Nat Genet 40:844–853CrossRefPubMed Fagerholm R, Hofstetter B, Tommiska J, Aaltonen K, Vrtel R, Syrjäkoski K, Kallioniemi A, Kilpivaara O, Mannermaa A, Kosma VM, Uusitupa M, Eskelinen M, Kataja V, Aittomäki K, von Smitten K, Heikkilä P, Lukas J, Holli K, Bartkova J, Blomqvist C, Bartek J, Nevanlinna H (2008) NAD(P)H:quinone oxidoreductase 1 NQO1*2 genotype (P187S) is a strong prognostic and predictive factor in breast cancer. Nat Genet 40:844–853CrossRefPubMed
29.
go back to reference Fisher B, Costantino JP, Wickerham DL, Redmond CK, Kavanah M, Cronin WM, Vogel V, Robidoux A, Dimitrov N, Atkins J, Daly M, Wieand S, Tan-Chiu E, Ford L, Wolmark N (1998) Tamoxifen for prevention of breast cancer: report of the National Surgical Adjuvant Breast and Bowel Project P-1 Study. J Natl Cancer Inst 90:1371–1388CrossRefPubMed Fisher B, Costantino JP, Wickerham DL, Redmond CK, Kavanah M, Cronin WM, Vogel V, Robidoux A, Dimitrov N, Atkins J, Daly M, Wieand S, Tan-Chiu E, Ford L, Wolmark N (1998) Tamoxifen for prevention of breast cancer: report of the National Surgical Adjuvant Breast and Bowel Project P-1 Study. J Natl Cancer Inst 90:1371–1388CrossRefPubMed
30.
go back to reference Takimoto GS, Graham JD, Jackson TA, Tung L, Powell RL, Horwitz LD, Horwitz KB (1999) Tamoxifen resistant breast cancer: coregulators determine the direction of transcription by antagonist-occupied steroid receptors. J Steroid Biochem Mol Biol 69:45–50CrossRefPubMed Takimoto GS, Graham JD, Jackson TA, Tung L, Powell RL, Horwitz LD, Horwitz KB (1999) Tamoxifen resistant breast cancer: coregulators determine the direction of transcription by antagonist-occupied steroid receptors. J Steroid Biochem Mol Biol 69:45–50CrossRefPubMed
31.
go back to reference Bown D (1995) Encyclopaedia of herbs and their uses. Dorling Kindersley, London Bown D (1995) Encyclopaedia of herbs and their uses. Dorling Kindersley, London
32.
go back to reference Chen X, Yang L, Oppenheim JJ, Howard MZ (2002) Cellular pharmacology studies of shikonin derivatives. Phytother Res 16:199–209CrossRefPubMed Chen X, Yang L, Oppenheim JJ, Howard MZ (2002) Cellular pharmacology studies of shikonin derivatives. Phytother Res 16:199–209CrossRefPubMed
Metadata
Title
Inhibition of estrogen signaling activates the NRF2 pathway in breast cancer
Authors
Yuan Yao
Angela M. H. Brodie
Nancy E. Davidson
Thomas W. Kensler
Qun Zhou
Publication date
01-11-2010
Publisher
Springer US
Published in
Breast Cancer Research and Treatment / Issue 2/2010
Print ISSN: 0167-6806
Electronic ISSN: 1573-7217
DOI
https://doi.org/10.1007/s10549-010-1023-8

Other articles of this Issue 2/2010

Breast Cancer Research and Treatment 2/2010 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