Skip to main content
Erschienen in: Discover Oncology 4/2017

02.06.2017 | Original Paper

Estrogen Receptor-β Modulation of the ERα-p53 Loop Regulating Gene Expression, Proliferation, and Apoptosis in Breast Cancer

verfasst von: Wenwen Lu, Benita S. Katzenellenbogen

Erschienen in: Discover Oncology | Ausgabe 4/2017

Einloggen, um Zugang zu erhalten

Abstract

Estrogen receptor α (ERα) is a crucial transcriptional regulator in breast cancer, but estrogens mediate their effects through two estrogen receptors, ERα and ERβ, subtypes that have contrasting regulatory actions on gene expression and the survival and growth of breast cancer cells. Here, we examine the impact of ERβ on the ERα-p53 loop in breast cancer. We found that ERβ attenuates ERα-induced cell proliferation, increases apoptosis, and reverses transcriptional activation and repression by ERα. Further, ERβ physically interacts with p53, reduces ERα-p53 binding, and antagonizes ERα-p53-mediated transcriptional regulation. ERα directs SUV39H1/H2 and histone H3 lys9 trimethylation (H3K9me3) heterochromatin assembly at estrogen-repressed genes to silence p53-activated transcription. The copresence of ERβ in ERα-positive cells abrogates the H3K9me3 repressive heterochromatin conformation by downregulating SUV39H1 and SUV39H2, thereby releasing the ERα-induced transcriptional block. Furthermore, the presence of ERβ stimulates accumulation of histone H3 lys4 trimethylation (H3K4me3) and RNA polymerase II (RNA Pol II) on ERα-repressed genes, inducing H3K4me3-associated epigenetic activation of the transcription of these repressed genes that can promote p53-based tumor suppression. ERβ also reduced corepressor N-CoR and SMRT recruitment by ERα that could attenuate the crosstalk between ERα and p53. Overall, our data reveal a novel mechanism for ERβ’s anti-proliferative and pro-apoptotic effects in breast cancer cells involving p53 and epigenetic changes in histone methylation that underlie gene regulation of these cellular activities.
Anhänge
Nur mit Berechtigung zugänglich
Literatur
1.
Zurück zum Zitat Pettersson K, Gustafsson JA (2001) Role of estrogen receptor beta in estrogen action. Annu Rev Physiol 63:165–192CrossRefPubMed Pettersson K, Gustafsson JA (2001) Role of estrogen receptor beta in estrogen action. Annu Rev Physiol 63:165–192CrossRefPubMed
2.
Zurück zum Zitat Stanford JL, Szklo M, Brinton LA (1986) Estrogen receptors and breast cancer. Epidemiol Rev 8:42–59CrossRefPubMed Stanford JL, Szklo M, Brinton LA (1986) Estrogen receptors and breast cancer. Epidemiol Rev 8:42–59CrossRefPubMed
3.
Zurück zum Zitat Katzenellenbogen BS, Frasor J (2004) Therapeutic targeting in the estrogen receptor hormonal pathway. Semin Oncol 31:28–38CrossRefPubMed Katzenellenbogen BS, Frasor J (2004) Therapeutic targeting in the estrogen receptor hormonal pathway. Semin Oncol 31:28–38CrossRefPubMed
4.
Zurück zum Zitat Kurebayashi J, Otsuki T, Kunisue H, Tanaka K, Yamamoto S, Sonoo H (2000) Expression levels of estrogen receptor-alpha, estrogen receptor-beta, coactivators, and corepressors in breast cancer. Clin Cancer Res 6:512–518PubMed Kurebayashi J, Otsuki T, Kunisue H, Tanaka K, Yamamoto S, Sonoo H (2000) Expression levels of estrogen receptor-alpha, estrogen receptor-beta, coactivators, and corepressors in breast cancer. Clin Cancer Res 6:512–518PubMed
5.
Zurück zum Zitat Saji S, Hirose M, Toi M (2005) Clinical significance of estrogen receptor beta in breast cancer. Cancer Chemother Pharmacol 56(Suppl 1):21–26CrossRefPubMed Saji S, Hirose M, Toi M (2005) Clinical significance of estrogen receptor beta in breast cancer. Cancer Chemother Pharmacol 56(Suppl 1):21–26CrossRefPubMed
6.
Zurück zum Zitat el-Deiry WS, Tokino T, Velculescu VE, Levy DB, Parsons R, Trent JM et al (1993) WAF1, a potential mediator of p53 tumor suppression. Cell 75:817–825CrossRefPubMed el-Deiry WS, Tokino T, Velculescu VE, Levy DB, Parsons R, Trent JM et al (1993) WAF1, a potential mediator of p53 tumor suppression. Cell 75:817–825CrossRefPubMed
7.
Zurück zum Zitat Chang EC, Frasor J, Komm B, Katzenellenbogen BS (2006) Impact of estrogen receptor beta on gene networks regulated by estrogen receptor alpha in breast cancer cells. Endocrinology 147:4831–4842CrossRefPubMed Chang EC, Frasor J, Komm B, Katzenellenbogen BS (2006) Impact of estrogen receptor beta on gene networks regulated by estrogen receptor alpha in breast cancer cells. Endocrinology 147:4831–4842CrossRefPubMed
8.
Zurück zum Zitat Gong P, Madak-Erdogan Z, Li J, Cheng J, Greenlief CM, Helferich W et al (2014) Transcriptomic analysis identifies gene networks regulated by estrogen receptor alpha (ERalpha) and ERbeta that control distinct effects of different botanical estrogens. Nucl Recept Signal 12:e001PubMedPubMedCentral Gong P, Madak-Erdogan Z, Li J, Cheng J, Greenlief CM, Helferich W et al (2014) Transcriptomic analysis identifies gene networks regulated by estrogen receptor alpha (ERalpha) and ERbeta that control distinct effects of different botanical estrogens. Nucl Recept Signal 12:e001PubMedPubMedCentral
9.
Zurück zum Zitat Lindberg MK, Moverare S, Skrtic S, Gao H, Dahlman-Wright K, Gustafsson JA et al (2003) Estrogen receptor (ER)-beta reduces ERalpha-regulated gene transcription, supporting a “ying yang” relationship between ERalpha and ERbeta in mice. Mol Endocrinol 17:203–208CrossRefPubMed Lindberg MK, Moverare S, Skrtic S, Gao H, Dahlman-Wright K, Gustafsson JA et al (2003) Estrogen receptor (ER)-beta reduces ERalpha-regulated gene transcription, supporting a “ying yang” relationship between ERalpha and ERbeta in mice. Mol Endocrinol 17:203–208CrossRefPubMed
10.
Zurück zum Zitat Madak-Erdogan Z, Charn TH, Jiang Y, Liu ET, Katzenellenbogen JA, Katzenellenbogen BS (2013) Integrative genomics of gene and metabolic regulation by estrogen receptors alpha and beta, and their coregulators. Mol Syst Biol 9:676CrossRefPubMedPubMedCentral Madak-Erdogan Z, Charn TH, Jiang Y, Liu ET, Katzenellenbogen JA, Katzenellenbogen BS (2013) Integrative genomics of gene and metabolic regulation by estrogen receptors alpha and beta, and their coregulators. Mol Syst Biol 9:676CrossRefPubMedPubMedCentral
11.
Zurück zum Zitat Paruthiyil S, Parmar H, Kerekatte V, Cunha GR, Firestone GL, Leitman DC (2004) Estrogen receptor beta inhibits human breast cancer cell proliferation and tumor formation by causing a G2 cell cycle arrest. Cancer Res 64:423–428CrossRefPubMed Paruthiyil S, Parmar H, Kerekatte V, Cunha GR, Firestone GL, Leitman DC (2004) Estrogen receptor beta inhibits human breast cancer cell proliferation and tumor formation by causing a G2 cell cycle arrest. Cancer Res 64:423–428CrossRefPubMed
12.
Zurück zum Zitat Strom A, Hartman J, Foster JS, Kietz S, Wimalasena J, Gustafsson JA (2004) Estrogen receptor beta inhibits 17beta-estradiol-stimulated proliferation of the breast cancer cell line T47D. Proc Natl Acad Sci U S A 101:1566–1571CrossRefPubMedPubMedCentral Strom A, Hartman J, Foster JS, Kietz S, Wimalasena J, Gustafsson JA (2004) Estrogen receptor beta inhibits 17beta-estradiol-stimulated proliferation of the breast cancer cell line T47D. Proc Natl Acad Sci U S A 101:1566–1571CrossRefPubMedPubMedCentral
13.
Zurück zum Zitat Tovar C, Rosinski J, Filipovic Z, Higgins B, Kolinsky K, Hilton H et al (2006) Small-molecule MDM2 antagonists reveal aberrant p53 signaling in cancer: implications for therapy. Proc Natl Acad Sci U S A 103:1888–1893CrossRefPubMedPubMedCentral Tovar C, Rosinski J, Filipovic Z, Higgins B, Kolinsky K, Hilton H et al (2006) Small-molecule MDM2 antagonists reveal aberrant p53 signaling in cancer: implications for therapy. Proc Natl Acad Sci U S A 103:1888–1893CrossRefPubMedPubMedCentral
14.
Zurück zum Zitat Shangary S, Wang S (2009) Small-molecule inhibitors of the MDM2-p53 protein-protein interaction to reactivate p53 function: a novel approach for cancer therapy. Annu Rev Pharmacol Toxicol 49:223–241CrossRefPubMedPubMedCentral Shangary S, Wang S (2009) Small-molecule inhibitors of the MDM2-p53 protein-protein interaction to reactivate p53 function: a novel approach for cancer therapy. Annu Rev Pharmacol Toxicol 49:223–241CrossRefPubMedPubMedCentral
15.
Zurück zum Zitat Vassilev LT, Vu BT, Graves B, Carvajal D, Podlaski F, Filipovic Z et al (2004) In vivo activation of the p53 pathway by small-molecule antagonists of MDM2. Science 303:844–848CrossRefPubMed Vassilev LT, Vu BT, Graves B, Carvajal D, Podlaski F, Filipovic Z et al (2004) In vivo activation of the p53 pathway by small-molecule antagonists of MDM2. Science 303:844–848CrossRefPubMed
16.
Zurück zum Zitat Selivanova G (2010) Therapeutic targeting of p53 by small molecules. Semin Cancer Biol 20:46–56CrossRefPubMed Selivanova G (2010) Therapeutic targeting of p53 by small molecules. Semin Cancer Biol 20:46–56CrossRefPubMed
17.
Zurück zum Zitat Bailey ST, Shin H, Westerling T, Liu XS, Brown M (2012) Estrogen receptor prevents p53-dependent apoptosis in breast cancer. Proc Natl Acad Sci U S A 109:18060–18065CrossRefPubMedPubMedCentral Bailey ST, Shin H, Westerling T, Liu XS, Brown M (2012) Estrogen receptor prevents p53-dependent apoptosis in breast cancer. Proc Natl Acad Sci U S A 109:18060–18065CrossRefPubMedPubMedCentral
18.
Zurück zum Zitat Konduri SD, Medisetty R, Liu W, Kaipparettu BA, Srivastava P, Brauch H et al (2010) Mechanisms of estrogen receptor antagonism toward p53 and its implications in breast cancer therapeutic response and stem cell regulation. Proc Natl Acad Sci U S A 107:15081–15086CrossRefPubMedPubMedCentral Konduri SD, Medisetty R, Liu W, Kaipparettu BA, Srivastava P, Brauch H et al (2010) Mechanisms of estrogen receptor antagonism toward p53 and its implications in breast cancer therapeutic response and stem cell regulation. Proc Natl Acad Sci U S A 107:15081–15086CrossRefPubMedPubMedCentral
19.
Zurück zum Zitat Liu W, Konduri SD, Bansal S, Nayak BK, Rajasekaran SA, Karuppayil SM et al (2006) Estrogen receptor-alpha binds p53 tumor suppressor protein directly and represses its function. J Biol Chem 281:9837–9840CrossRefPubMed Liu W, Konduri SD, Bansal S, Nayak BK, Rajasekaran SA, Karuppayil SM et al (2006) Estrogen receptor-alpha binds p53 tumor suppressor protein directly and represses its function. J Biol Chem 281:9837–9840CrossRefPubMed
21.
Zurück zum Zitat Yamamoto M, Hosoda M, Nakano K, Jia S, Hatanaka KC, Takakuwa E et al (2014) p53 accumulation is a strong predictor of recurrence in estrogen receptor-positive breast cancer patients treated with aromatase inhibitors. Cancer Sci 105:81–88CrossRefPubMed Yamamoto M, Hosoda M, Nakano K, Jia S, Hatanaka KC, Takakuwa E et al (2014) p53 accumulation is a strong predictor of recurrence in estrogen receptor-positive breast cancer patients treated with aromatase inhibitors. Cancer Sci 105:81–88CrossRefPubMed
22.
Zurück zum Zitat Li B, Carey M, Workman JL (2007) The role of chromatin during transcription. Cell 128:707–719CrossRefPubMed Li B, Carey M, Workman JL (2007) The role of chromatin during transcription. Cell 128:707–719CrossRefPubMed
23.
Zurück zum Zitat Chang EC, Charn TH, Park SH, Helferich WG, Komm B, Katzenellenbogen JA et al (2008) Estrogen receptors alpha and beta as determinants of gene expression: influence of ligand, dose, and chromatin binding. Mol Endocrinol 22:1032–1043CrossRefPubMedPubMedCentral Chang EC, Charn TH, Park SH, Helferich WG, Komm B, Katzenellenbogen JA et al (2008) Estrogen receptors alpha and beta as determinants of gene expression: influence of ligand, dose, and chromatin binding. Mol Endocrinol 22:1032–1043CrossRefPubMedPubMedCentral
24.
Zurück zum Zitat Bergamaschi A, Christensen BL, Katzenellenbogen BS (2011) Reversal of endocrine resistance in breast cancer: interrelationships among 14-3-3zeta, FOXM1, and a gene signature associated with mitosis. Breast Cancer Res 13:R70CrossRefPubMedPubMedCentral Bergamaschi A, Christensen BL, Katzenellenbogen BS (2011) Reversal of endocrine resistance in breast cancer: interrelationships among 14-3-3zeta, FOXM1, and a gene signature associated with mitosis. Breast Cancer Res 13:R70CrossRefPubMedPubMedCentral
25.
Zurück zum Zitat Choi I, Ko C, Park-Sarge OK, Nie R, Hess RA, Graves C et al (2001) Human estrogen receptor beta-specific monoclonal antibodies: characterization and use in studies of estrogen receptor beta protein expression in reproductive tissues. Mol Cell Endocrinol 181:139–150CrossRefPubMed Choi I, Ko C, Park-Sarge OK, Nie R, Hess RA, Graves C et al (2001) Human estrogen receptor beta-specific monoclonal antibodies: characterization and use in studies of estrogen receptor beta protein expression in reproductive tissues. Mol Cell Endocrinol 181:139–150CrossRefPubMed
26.
Zurück zum Zitat Dennis G Jr, Sherman BT, Hosack DA, Yang J, Gao W, Lane HC et al (2003) DAVID: database for annotation, visualization, and integrated discovery. Genome Biol 4:P3CrossRefPubMed Dennis G Jr, Sherman BT, Hosack DA, Yang J, Gao W, Lane HC et al (2003) DAVID: database for annotation, visualization, and integrated discovery. Genome Biol 4:P3CrossRefPubMed
27.
Zurück zum Zitat Huang da W, Sherman BT, Lempicki RA (2009) Systematic and integrative analysis of large gene lists using DAVID bioinformatics resources. Nat Protoc 4:44–57CrossRefPubMed Huang da W, Sherman BT, Lempicki RA (2009) Systematic and integrative analysis of large gene lists using DAVID bioinformatics resources. Nat Protoc 4:44–57CrossRefPubMed
28.
Zurück zum Zitat Lv WW, Wei HM, Wang DL, Ni JQ, Sun FL (2012) Depletion of histone deacetylase 3 antagonizes PI3K-mediated overgrowth of drosophila organs through the acetylation of histone H4 at lysine 16. J Cell Sci 125:5369–5378CrossRefPubMed Lv WW, Wei HM, Wang DL, Ni JQ, Sun FL (2012) Depletion of histone deacetylase 3 antagonizes PI3K-mediated overgrowth of drosophila organs through the acetylation of histone H4 at lysine 16. J Cell Sci 125:5369–5378CrossRefPubMed
29.
Zurück zum Zitat Nelson AW, Groen AJ, Miller JL, Warren AY, Holmes KA, Tarulli GA et al (2017) Comprehensive assessment of estrogen receptor beta antibodies in cancer cell line models and tissue reveals critical limitations in reagent specificity. Mol Cell Endocrinol 440:138–150CrossRefPubMedPubMedCentral Nelson AW, Groen AJ, Miller JL, Warren AY, Holmes KA, Tarulli GA et al (2017) Comprehensive assessment of estrogen receptor beta antibodies in cancer cell line models and tissue reveals critical limitations in reagent specificity. Mol Cell Endocrinol 440:138–150CrossRefPubMedPubMedCentral
30.
Zurück zum Zitat Rhodes DR, Yu J, Shanker K, Deshpande N, Varambally R, Ghosh D et al (2004) ONCOMINE: a cancer microarray database and integrated data-mining platform. Neoplasia 6:1–6CrossRefPubMedPubMedCentral Rhodes DR, Yu J, Shanker K, Deshpande N, Varambally R, Ghosh D et al (2004) ONCOMINE: a cancer microarray database and integrated data-mining platform. Neoplasia 6:1–6CrossRefPubMedPubMedCentral
31.
Zurück zum Zitat Katzenellenbogen BS, Kendra KL, Norman MJ, Berthois Y (1987) Proliferation, hormonal responsiveness, and estrogen receptor content of MCF-7 human breast cancer cells grown in the short-term and long-term absence of estrogens. Cancer Res 47:4355–4360PubMed Katzenellenbogen BS, Kendra KL, Norman MJ, Berthois Y (1987) Proliferation, hormonal responsiveness, and estrogen receptor content of MCF-7 human breast cancer cells grown in the short-term and long-term absence of estrogens. Cancer Res 47:4355–4360PubMed
32.
Zurück zum Zitat Calza S, Hall P, Auer G, Bjohle J, Klaar S, Kronenwett U et al (2006) Intrinsic molecular signature of breast cancer in a population-based cohort of 412 patients. Breast Cancer Res 8:R34CrossRefPubMedPubMedCentral Calza S, Hall P, Auer G, Bjohle J, Klaar S, Kronenwett U et al (2006) Intrinsic molecular signature of breast cancer in a population-based cohort of 412 patients. Breast Cancer Res 8:R34CrossRefPubMedPubMedCentral
33.
Zurück zum Zitat Kao KJ, Chang KM, Hsu HC (2011) Huang AT correlation of microarray-based breast cancer molecular subtypes and clinical outcomes: implications for treatment optimization. BMC Cancer 11:143CrossRefPubMedPubMedCentral Kao KJ, Chang KM, Hsu HC (2011) Huang AT correlation of microarray-based breast cancer molecular subtypes and clinical outcomes: implications for treatment optimization. BMC Cancer 11:143CrossRefPubMedPubMedCentral
34.
Zurück zum Zitat van de Vijver MJ, He YD, van’t Veer LJ, Dai H, Hart AA, Voskuil DW et al (2002) A gene-expression signature as a predictor of survival in breast cancer. N Engl J Med 347:1999–2009CrossRefPubMed van de Vijver MJ, He YD, van’t Veer LJ, Dai H, Hart AA, Voskuil DW et al (2002) A gene-expression signature as a predictor of survival in breast cancer. N Engl J Med 347:1999–2009CrossRefPubMed
35.
Zurück zum Zitat Nikulenkov F, Spinnler C, Li H, Tonelli C, Shi Y, Turunen M et al (2012) Insights into p53 transcriptional function via genome-wide chromatin occupancy and gene expression analysis. Cell Death Differ 19:1992–2002CrossRefPubMedPubMedCentral Nikulenkov F, Spinnler C, Li H, Tonelli C, Shi Y, Turunen M et al (2012) Insights into p53 transcriptional function via genome-wide chromatin occupancy and gene expression analysis. Cell Death Differ 19:1992–2002CrossRefPubMedPubMedCentral
36.
Zurück zum Zitat Janky R, Verfaillie A, Imrichova H, Van de Sande B, Standaert L, Christiaens V et al (2014) iRegulon: from a gene list to a gene regulatory network using large motif and track collections. PLoS Comput Biol 10:e1003731CrossRefPubMedPubMedCentral Janky R, Verfaillie A, Imrichova H, Van de Sande B, Standaert L, Christiaens V et al (2014) iRegulon: from a gene list to a gene regulatory network using large motif and track collections. PLoS Comput Biol 10:e1003731CrossRefPubMedPubMedCentral
37.
Zurück zum Zitat Mungamuri SK, Benson EK, Wang S, Gu W, Lee SW, Aaronson SA (2012) p53-mediated heterochromatin reorganization regulates its cell fate decisions. Nat Struct Mol Biol 19:478–484 S471CrossRefPubMedPubMedCentral Mungamuri SK, Benson EK, Wang S, Gu W, Lee SW, Aaronson SA (2012) p53-mediated heterochromatin reorganization regulates its cell fate decisions. Nat Struct Mol Biol 19:478–484 S471CrossRefPubMedPubMedCentral
38.
Zurück zum Zitat Lehnertz B, Ueda Y, Derijck AA, Braunschweig U, Perez-Burgos L, Kubicek S et al (2003) Suv39h-mediated histone H3 lysine 9 methylation directs DNA methylation to major satellite repeats at pericentric heterochromatin. Curr Biol 13:1192–1200CrossRefPubMed Lehnertz B, Ueda Y, Derijck AA, Braunschweig U, Perez-Burgos L, Kubicek S et al (2003) Suv39h-mediated histone H3 lysine 9 methylation directs DNA methylation to major satellite repeats at pericentric heterochromatin. Curr Biol 13:1192–1200CrossRefPubMed
39.
Zurück zum Zitat Perissi V, Jepsen K, Glass CK, Rosenfeld MG (2010) Deconstructing repression: evolving models of co-repressor action. Nat Rev Genet 11:109–123CrossRefPubMed Perissi V, Jepsen K, Glass CK, Rosenfeld MG (2010) Deconstructing repression: evolving models of co-repressor action. Nat Rev Genet 11:109–123CrossRefPubMed
40.
Zurück zum Zitat Stossi F, Likhite VS, Katzenellenbogen JA, Katzenellenbogen BS (2006) Estrogen-occupied estrogen receptor represses cyclin G2 gene expression and recruits a repressor complex at the cyclin G2 promoter. J Biol Chem 281:16272–16278CrossRefPubMed Stossi F, Likhite VS, Katzenellenbogen JA, Katzenellenbogen BS (2006) Estrogen-occupied estrogen receptor represses cyclin G2 gene expression and recruits a repressor complex at the cyclin G2 promoter. J Biol Chem 281:16272–16278CrossRefPubMed
41.
Zurück zum Zitat Lauberth SM, Nakayama T, Wu X, Ferris AL, Tang Z, Hughes SH et al (2013) H3K4me3 interactions with TAF3 regulate preinitiation complex assembly and selective gene activation. Cell 152:1021–1036CrossRefPubMedPubMedCentral Lauberth SM, Nakayama T, Wu X, Ferris AL, Tang Z, Hughes SH et al (2013) H3K4me3 interactions with TAF3 regulate preinitiation complex assembly and selective gene activation. Cell 152:1021–1036CrossRefPubMedPubMedCentral
42.
Zurück zum Zitat Frasor J, Danes JM, Komm B, Chang KC, Lyttle CR, Katzenellenbogen BS (2003) Profiling of estrogen up- and down-regulated gene expression in human breast cancer cells: insights into gene networks and pathways underlying estrogenic control of proliferation and cell phenotype. Endocrinology 144:4562–4574CrossRefPubMed Frasor J, Danes JM, Komm B, Chang KC, Lyttle CR, Katzenellenbogen BS (2003) Profiling of estrogen up- and down-regulated gene expression in human breast cancer cells: insights into gene networks and pathways underlying estrogenic control of proliferation and cell phenotype. Endocrinology 144:4562–4574CrossRefPubMed
43.
Zurück zum Zitat Green KA, Carroll JS (2007) Oestrogen-receptor-mediated transcription and the influence of co-factors and chromatin state. Nat Rev Cancer 7:713–722CrossRefPubMed Green KA, Carroll JS (2007) Oestrogen-receptor-mediated transcription and the influence of co-factors and chromatin state. Nat Rev Cancer 7:713–722CrossRefPubMed
44.
Zurück zum Zitat Metivier R, Penot G, Hubner MR, Reid G, Brand H, Kos M et al (2003) Estrogen receptor-alpha directs ordered, cyclical, and combinatorial recruitment of cofactors on a natural target promoter. Cell 115:751–763CrossRefPubMed Metivier R, Penot G, Hubner MR, Reid G, Brand H, Kos M et al (2003) Estrogen receptor-alpha directs ordered, cyclical, and combinatorial recruitment of cofactors on a natural target promoter. Cell 115:751–763CrossRefPubMed
45.
Zurück zum Zitat Shang Y, Hu X, DiRenzo J, Lazar MA, Brown M (2000) Cofactor dynamics and sufficiency in estrogen receptor-regulated transcription. Cell 103:843–852CrossRefPubMed Shang Y, Hu X, DiRenzo J, Lazar MA, Brown M (2000) Cofactor dynamics and sufficiency in estrogen receptor-regulated transcription. Cell 103:843–852CrossRefPubMed
46.
Zurück zum Zitat Stossi F, Madak-Erdogan Z, Katzenellenbogen BS (2009) Estrogen receptor alpha represses transcription of early target genes via p300 and CtBP1. Mol Cell Biol 29:1749–1759CrossRefPubMedPubMedCentral Stossi F, Madak-Erdogan Z, Katzenellenbogen BS (2009) Estrogen receptor alpha represses transcription of early target genes via p300 and CtBP1. Mol Cell Biol 29:1749–1759CrossRefPubMedPubMedCentral
47.
Zurück zum Zitat Paruthiyil S, Cvoro A, Tagliaferri M, Cohen I, Shtivelman E, Leitman DC (2011) Estrogen receptor beta causes a G2 cell cycle arrest by inhibiting CDK1 activity through the regulation of cyclin B1, GADD45A, and BTG2. Breast Cancer Res Treat 129:777–784CrossRefPubMed Paruthiyil S, Cvoro A, Tagliaferri M, Cohen I, Shtivelman E, Leitman DC (2011) Estrogen receptor beta causes a G2 cell cycle arrest by inhibiting CDK1 activity through the regulation of cyclin B1, GADD45A, and BTG2. Breast Cancer Res Treat 129:777–784CrossRefPubMed
48.
Zurück zum Zitat Dey P, Strom A, Gustafsson JA (2014) Estrogen receptor beta upregulates FOXO3a and causes induction of apoptosis through PUMA in prostate cancer. Oncogene 33:4213–4225CrossRefPubMed Dey P, Strom A, Gustafsson JA (2014) Estrogen receptor beta upregulates FOXO3a and causes induction of apoptosis through PUMA in prostate cancer. Oncogene 33:4213–4225CrossRefPubMed
49.
50.
51.
Zurück zum Zitat Taverna SD, Li H, Ruthenburg AJ, Allis CD, Patel DJ (2007) How chromatin-binding modules interpret histone modifications: lessons from professional pocket pickers. Nat Struct Mol Biol 14:1025–1040CrossRefPubMedPubMedCentral Taverna SD, Li H, Ruthenburg AJ, Allis CD, Patel DJ (2007) How chromatin-binding modules interpret histone modifications: lessons from professional pocket pickers. Nat Struct Mol Biol 14:1025–1040CrossRefPubMedPubMedCentral
52.
Zurück zum Zitat Peters AH, Mermoud JE, O’Carroll D, Pagani M, Schweizer D, Brockdorff N et al (2002) Histone H3 lysine 9 methylation is an epigenetic imprint of facultative heterochromatin. Nat Genet 30:77–80CrossRefPubMed Peters AH, Mermoud JE, O’Carroll D, Pagani M, Schweizer D, Brockdorff N et al (2002) Histone H3 lysine 9 methylation is an epigenetic imprint of facultative heterochromatin. Nat Genet 30:77–80CrossRefPubMed
53.
Zurück zum Zitat Barski A, Cuddapah S, Cui K, Roh TY, Schones DE, Wang Z et al (2007) High-resolution profiling of histone methylations in the human genome. Cell 129:823–837CrossRefPubMed Barski A, Cuddapah S, Cui K, Roh TY, Schones DE, Wang Z et al (2007) High-resolution profiling of histone methylations in the human genome. Cell 129:823–837CrossRefPubMed
54.
Zurück zum Zitat Guenther MG, Levine SS, Boyer LA, Jaenisch R, Young RA (2007) A chromatin landmark and transcription initiation at most promoters in human cells. Cell 130:77–88CrossRefPubMedPubMedCentral Guenther MG, Levine SS, Boyer LA, Jaenisch R, Young RA (2007) A chromatin landmark and transcription initiation at most promoters in human cells. Cell 130:77–88CrossRefPubMedPubMedCentral
55.
Zurück zum Zitat Czvitkovich S, Sauer S, Peters AH, Deiner E, Wolf A, Laible G et al (2001) Over-expression of the SUV39H1 histone methyltransferase induces altered proliferation and differentiation in transgenic mice. Mech Dev 107:141–153CrossRefPubMed Czvitkovich S, Sauer S, Peters AH, Deiner E, Wolf A, Laible G et al (2001) Over-expression of the SUV39H1 histone methyltransferase induces altered proliferation and differentiation in transgenic mice. Mech Dev 107:141–153CrossRefPubMed
56.
Zurück zum Zitat Vousden KH, Lu X (2002) Live or let die: the cell’s response to p53. Nat Rev Cancer 2:594–604CrossRefPubMed Vousden KH, Lu X (2002) Live or let die: the cell’s response to p53. Nat Rev Cancer 2:594–604CrossRefPubMed
57.
Zurück zum Zitat Olivier M, Langerod A, Carrieri P, Bergh J, Klaar S, Eyfjord J et al (2006) The clinical value of somatic TP53 gene mutations in 1,794 patients with breast cancer. Clin Cancer Res 12:1157–1167CrossRefPubMed Olivier M, Langerod A, Carrieri P, Bergh J, Klaar S, Eyfjord J et al (2006) The clinical value of somatic TP53 gene mutations in 1,794 patients with breast cancer. Clin Cancer Res 12:1157–1167CrossRefPubMed
58.
Zurück zum Zitat Vinh-Hung V, Verschraegen C (2004) Breast-conserving surgery with or without radiotherapy: pooled-analysis for risks of ipsilateral breast tumor recurrence and mortality. J Natl Cancer Inst 96:115–121CrossRefPubMed Vinh-Hung V, Verschraegen C (2004) Breast-conserving surgery with or without radiotherapy: pooled-analysis for risks of ipsilateral breast tumor recurrence and mortality. J Natl Cancer Inst 96:115–121CrossRefPubMed
59.
Zurück zum Zitat Whelan TJ, Julian J, Wright J, Jadad AR, Levine ML (2000) Does locoregional radiation therapy improve survival in breast cancer? A meta-analysis. J Clin Oncol 18:1220–1229CrossRefPubMed Whelan TJ, Julian J, Wright J, Jadad AR, Levine ML (2000) Does locoregional radiation therapy improve survival in breast cancer? A meta-analysis. J Clin Oncol 18:1220–1229CrossRefPubMed
60.
Zurück zum Zitat Ma R, Karthik GM, Lovrot J, Haglund F, Rosin G, Katchy A et al (2017) Estrogen receptor beta as a therapeutic target in breast cancer stem cells. J Natl Cancer Inst 109:1–14CrossRefPubMed Ma R, Karthik GM, Lovrot J, Haglund F, Rosin G, Katchy A et al (2017) Estrogen receptor beta as a therapeutic target in breast cancer stem cells. J Natl Cancer Inst 109:1–14CrossRefPubMed
Metadaten
Titel
Estrogen Receptor-β Modulation of the ERα-p53 Loop Regulating Gene Expression, Proliferation, and Apoptosis in Breast Cancer
verfasst von
Wenwen Lu
Benita S. Katzenellenbogen
Publikationsdatum
02.06.2017
Verlag
Springer US
Erschienen in
Discover Oncology / Ausgabe 4/2017
Print ISSN: 1868-8497
Elektronische ISSN: 2730-6011
DOI
https://doi.org/10.1007/s12672-017-0298-1

Weitere Artikel der Ausgabe 4/2017

Discover Oncology 4/2017 Zur Ausgabe

Update Onkologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.