S. Banerjee, K. L. Chambliss, C. Mineo, and P. W. Shaul, Recent insights into non-nuclear actions of estrogen receptor alpha, Steroids, vol.81, pp.64-73, 2014.

D. Gibson and P. Saunders, Estrogen dependent signaling in reproductive tissues-A role for estrogen receptors and estrogen related receptors, Mol Cell Endocrinol, vol.348, pp.361-372, 2012.

H. Qian, J. Xuan, Y. Liu, and G. Shi, Function of G-Protein-Coupled Estrogen Receptor-1. In Reproductive System Tumors, J Immunol Res, p.7128702, 2016.

X. T. Zhang and Z. Wang, Estrogen receptor-alpha variant, ER-alpha 36, is involved in tamoxifen resistance and estrogen hypersensitivity, Endocrinology, vol.154, pp.2013-1116, 1990.

X. T. Zhang, H. Deng, and Z. Y. Wang, Estrogen activation of the mitogen-activated protein kinase is mediated by ER-alpha36 in ER-positive breast cancer cells, J Steroid Biochem Mol Biol, vol.143, pp.434-443, 2014.

L. Yin, X. T. Zhang, X. W. Bian, Y. M. Guo, and Z. Y. Wang, Disruption of the ER-alpha36-EGFR/HER2 positive regulatory loops restores tamoxifen sensitivity in tamoxifen resistance breast cancer cells, PLoS ONE, vol.9, issue.9, p.107369, 2014.

A. Wallacides, A. Chesnel, H. Ajj, M. Chillet, S. Flament et al., Estrogens promote proliferation of the seminoma-like TCam-2 cell line through a GPER dependent ERalpha36 induction, Mol Cell Endocrinol, vol.350, pp.61-71, 2012.

H. Ajj, A. Chesnel, S. Pinel, F. Plenat, S. Flament et al., An alkylphenol mix promotes seminoma derived cell proliferation through an ERalpha36-mediated mechanism, PLoS ONE, vol.8, issue.4, 2013.
URL : https://hal.archives-ouvertes.fr/hal-00840880

, ER?36-dependent disruption of mammary epithelium, PLOS ONE, 2017.

A. M. Soto and C. Sonnenschein, Environmental causes of cancer: endocrine disruptors as carcinogens, Nat Rev Endocrinol, vol.6, issue.7, pp.363-70, 2010.

D. Trichopoulos, Is breast cancer initiated in utero?, Epidemiology, vol.1, issue.2, pp.95-101, 1990.

T. J. Murray, M. V. Maini, A. A. Ucci, C. Sonnenschein, and A. M. Soto, Induction of mammary gland ductal hyperplasias and carcinoma in situ following fetal BPA exposure, Reprod Toxicol, vol.23, issue.3, pp.383-90, 2007.

O. Yang, H. L. Kim, J. I. Weon, and Y. R. Seo, Endocrine-disrupting chemicals: review of toxicological mechanisms using molecular pathway analysis, J Cancer Prev, vol.20, issue.1, pp.12-24, 2015.

P. Homas and J. Dong, Binding and activation of the seven-transmembrane estrogen receptor GPR30 by environmental estrogens: a potential novel mechanism of endocrine disruption, J Steroid Biochem Mol Biol, vol.102, issue.1-5, pp.175-184, 2006.

. Who/unep/ilo, Integrated Risk Assessment: Nonylphenol Case Study, Geneva: International Programme on Chemical Safety, 2004.

R. E. Chapin, J. Delaney, Y. Wang, L. Lanning, B. Davis et al., he efects of 4-nonylphenol in rats: a multigeneration reproduction study, Toxicol Sci, vol.52, issue.1, pp.80-91, 1999.

R. W. Tyl, C. B. Myers, M. C. Marr, N. P. Castillo, J. C. Seely et al., hreegeneration evaluation of dietary para-nonylphenol in CD-(Sprague-Dawley) rats, Toxicol Sci, vol.92, issue.1, pp.295-310, 2006.

R. Acevedo, P. G. Parnell, H. Villanueva, L. M. Chapman, T. Gimenez et al., he contribution of hepatic steroid metabolism to serum estradiol and estriol concentrations in nonylphenol-treated MMTVneu mice and its potential efects on breast cancer incidence and latency, J Appl Toxicol, vol.25, issue.5, pp.339-53, 2005.

H. J. Moon, S. Y. Han, J. H. Shin, I. H. Kang, T. S. Kim et al., Gestational exposure to nonylphenol causes precocious mammary gland development in female rat ofspring, J Reprod Dev, vol.53, issue.2, pp.333-377, 2007.

A. M. Soto, H. Justicia, J. W. Wray, and C. Sonnenschein, p-Nonyl-phenol: an estrogenic xenobiotic released from "modiied" polystyrene, Environ Health Perspect, vol.92, pp.167-73, 1991.

T. Raecker, B. Boehme, R. M. Guenther, and K. , Endocrine disrupting nonyland octylphenol in infant food in Germany: considerable daily intake of nonylphenol for babies, Chemosphere, vol.82, issue.11, pp.1533-1573, 2011.

H. Ajj, A. Chesnel, S. Pinel, F. Plenat, S. Flament et al., An alkylphenol mix promotes seminoma derived cell proliferation through an ERalpha36-mediated mechanism, PLoS One, vol.8, issue.4, p.61758, 2013.
URL : https://hal.archives-ouvertes.fr/hal-00840880

M. Manikkam, C. Guerrero-bosagna, R. Tracey, M. M. Haque, and M. K. Skinner, Transgenerational actions of environmental compounds on reproductive disease and identiication of epigenetic biomarkers of ancestral exposures, PLoS One, vol.7, issue.2, p.31901, 2012.

Z. Wang, X. Zhang, P. Shen, B. W. Loggie, Y. Chang et al., Identiication, cloning, and expression of human ER?36, a novel variant of human ER?66, Biochem Biophys Res Commun, vol.336, issue.4, pp.1023-1030, 2005.

C. Hiebaut, C. Chamard-jovenin, A. Chesnel, C. Morel, E. H. Djermoune et al., Mammary epithelial cell phenotype disruption in vitro and in vivo through ERalpha36 overexpression, PLoS One, vol.12, issue.3, p.173931, 2017.

V. Gayrard, M. Z. Lacroix, S. H. Collet, C. Viguié, A. Bousquet-melou et al., High bioavailability of bisphenol A from sublingual exposure, Environ Health Perspect, vol.121, pp.951-957, 2013.

J. Odum, I. T. Pyrah, A. R. Soames, J. R. Foster, V. Miller et al., Efects of p-nonylphenol (NP) and diethylstilboestrol (DES) on the Alderley Park (Alpk) rat: comparison of mammary gland and uterus sensitivity following oral gavage or implanted mini-pumps, J Appl Toxicol, vol.19, issue.5, pp.367-78, 1999.

J. Odum, I. T. Pyrah, J. R. Foster, V. Miller, J. P. Joiner et al., Comparative activities of p-nonylphenol and diethylstilbestrol in noble rat mammary gland and uterotrophic assays, Regul Toxicol Pharmacol, vol.29, issue.2, pp.184-95, 1999.

, Frontiers in Endocrinology | www.frontiersin.org October, vol.8, p.272, 2017.

L. N. Vandenberg, M. V. Maini, P. R. Wadia, C. Sonnenschein, B. S. Rubin et al., Exposure to environmentally relevant doses of the xenoestrogen bisphenol-A alters development of the fetal mouse mammary gland, Endocrinology, vol.148, issue.1, pp.116-143, 2007.

J. B. Tylcz, K. El-alaoui-lasmaili, E. H. Djermoune, N. Homas, B. Faivre et al., Data-driven modeling and characterization of anti-angiogenic molecule efects on tumoral vascular density, Biomed Signal Process Control, vol.20, pp.52-60, 2015.

A. F. Frangi, W. J. Niessen, K. L. Vincken, and M. A. Viergever, Multiscale vessel enhancement iltering, Proceedings from MICCAI'98, vol.1496, pp.130-137, 1998.

P. Howroyd, R. Hoyle-hacker, O. Lyght, D. Williams, and E. Kleymenova, Morphology of the fetal rat testis preserved in diferent ixatives, Toxicol Pathol, vol.33, issue.2, pp.300-304, 2005.

H. Mi, X. Huang, A. Muruganujan, H. Tang, C. Mills et al., PANTHER version 11: expanded annotation data from Gene Ontology and Reactome pathways, and data analysis tool enhancements, Nucleic Acids Res, pp.331-339, 2016.

V. K. Mootha, C. M. Lindgren, K. F. Eriksson, A. Subramanian, S. Sihag et al., PGC-1alpha-responsive genes involved in oxidative phosphorylation are coordinately downregulated in human diabetes, Nat Genet, vol.34, issue.3, pp.267-73, 2003.

A. Subramanian, P. Tamayo, V. K. Mootha, S. Mukherjee, B. L. Ebert et al., Gene set enrichment analysis: a knowledge-based approach for interpreting genome-wide expression proiles, Proc Natl Acad Sci U S A, vol.102, issue.43, pp.15545-50, 2005.

H. D. Soule, T. M. Maloney, S. R. Wolman, W. D. Peterson, R. Brenz et al., Isolation and characterization of a spontaneously immortalized human breast epithelial cell line, MCF-10, Cancer Res, vol.50, issue.18, pp.6075-86, 1990.

D. G. Altman and J. M. Bland, Standard deviations and standard errors, BMJ, vol.331, p.903, 2005.

T. M. Ong, B. Song, H. W. Qian, Z. L. Wu, and W. Z. Whong, Detection of genomic instability in lung cancer tissues by random ampliied polymorphic DNA analysis, Carcinogenesis, vol.19, issue.1, pp.233-238, 1998.

S. Papadopoulos, T. Benter, G. Anastassiou, M. Pape, G. S. Bornfeld et al., Assessment of genomic instability in breast cancer and uveal melanoma by random ampliied polymorphic DNA analysis, Int J Cancer, vol.99, issue.2, pp.193-200, 2002.

J. B. Colerangle and D. Roy, Exposure of environmental estrogenic compound nonlyphenol to noble rats alters cell-cycle kinetics in the mammary gland, Endocrine, vol.4, issue.2, pp.115-137, 1996.

S. Jenkins, J. Wang, I. Eltoum, R. Desmond, and C. A. Lamartiniere, Chronic oral exposure to bisphenol A results in a nonmonotonic dose response in mammary carcinogenesis and metastasis in MMTV-erbB2 mice, Environ Health Perspect, vol.119, pp.1604-1613, 2011.

A. M. Betancourt, J. Wang, S. Jenkins, J. Mobley, J. Russo et al., Altered carcinogenesis and proteome in mammary glands of rats ater prepubertal exposures to the hormonally active chemicals bisphenol A and genistein, J Nutr, vol.142, issue.7, pp.1382-1390, 2012.

J. H. Peng, F. Zhang, H. X. Zhang, and H. Y. Fan, Prepubertal octylphenol exposure up-regulate BRCA1 expression, down-regulate ER alpha expression and reduce rat mammary tumorigenesis, Cancer Epidemiol, vol.33, issue.1, pp.51-56, 2009.

B. L. Sprague, A. Trentham-dietz, C. J. Hedman, J. Wang, J. D. Hemming et al., Circulating serum xenoestrogens and mammographic breast density, Breast Cancer Res, vol.15, issue.3, p.45, 2013.

T. Paulose, L. Speroni, C. Sonnenschein, and A. M. Soto, Estrogens in the wrong place at the wrong time: fetal BPA exposure and mammary cancer, Reprod Toxicol, vol.54, pp.58-65, 2015.

P. Duan, C. Hu, H. J. Butler, C. Quan, W. Chen et al., 4-Nonylphenol induces disruption of spermatogenesis associated with oxidative stress-related apoptosis by targeting p53-Bcl-2/Bax-Fas/FasL signaling, Environ Toxicol, vol.32, issue.3, pp.739-53, 2016.

L. N. Vandenberg, M. V. Maini, C. M. Schaeberle, A. A. Ucci, C. Sonnenschein et al., Perinatal exposure to the xenoestrogen bisphenol-A induces mammary intraductal hyperplasias in adult CD-1 mice, Reprod Toxicol, vol.26, issue.3-4, pp.210-219, 2008.

M. M. Dobrzy?ska, Male-mediated F1 efects in mice exposed to nonylphenol or to a combination of X-rays and nonylphenol, Drug Chem Toxicol, vol.35, issue.1, pp.36-42, 2012.

S. Frassinetti, C. Barberio, L. Caltavuturo, F. Fava, D. Gioia et al., Genotoxicity of 4-nonylphenol and nonylphenol ethoxylate mixtures by the use of Saccharomyces cerevisiae D7 mutation assay and use of this text to evaluate the eiciency of biodegradation treatments, Ecotoxicol Environ Saf, vol.74, issue.3, pp.253-261, 2011.

M. Forte, D. Lorenzo, M. Carrizzo, A. Valiante, S. Vecchione et al., Nonylphenol efects on human prostate non tumorigenic cells, Toxicology, pp.357-358, 2016.

T. Negishi, Y. Ishii, S. Kyuwa, Y. Kuroda, and Y. Yoshikawa, Inhibition of staurosporine-induced neuronal cell death by BPA and nonylphenol in primary cultured rat hippocampal and cortical neurons, Neurosci Lett, vol.353, issue.2, pp.99-102, 2003.

C. Liu, Y. Sun, Y. Song, T. Saito, and M. Kurasaki, Nonylphenol diethoxylate inhibits apoptosis induced in PC12 cells, Environ Toxicol, issue.11, pp.1389-98, 2016.

P. R. Wadia, N. J. Cabaton, M. D. Borrero, B. S. Rubin, C. Sonnenschein et al., Low-dose BPA exposure alters the mesenchymal and epithelial transcriptomes of the mouse fetal mammary gland, PLoS One, vol.8, issue.5, p.6390202, 2013.

Z. Li, H. Zhang, M. Gibson, and J. Li, An evaluation on combination efects of phenolic endocrine disruptors by ER binding assay, Toxicol In Vitro, vol.26, issue.6, pp.769-74, 2012.

Z. Y. Wang and L. Yin, ERalpha36: a new player in human breast cancer, Mol Cell Endocrinol, vol.418, issue.3, pp.193-206, 2015.

S. Omarjee, J. Jacquemetton, C. Poulard, R. N. Dejaegere, A. Chebaro et al., he molecular mechanisms underlying the ER?-36-mediated signaling in breast cancer, Oncogene, vol.36, issue.18, pp.2503-2517, 2017.

C. Chamard-jovenin, A. C. Jung, A. Chesnel, J. Abecassis, S. Flament et al., From ER?66 to ER?36: a generic method for validating a prognosis marker of breast tumor progression, BMC Syst Biol, vol.9, p.28, 2015.

M. Szostakowska, A. Tr?bi?ska-stryjewska, E. A. Grzybowska, and A. Fabisiewicz, Resistance to endocrine therapy in breast cancer: Molecular mechanisms and future goals, Breast Cancer Res. Treat, vol.173, pp.489-497, 2019.

, Int. J. Mol. Sci, vol.20, p.2637, 2019.

Q. Wang, J. Jiang, G. Ying, X. Xie, X. Zhang et al., Tamoxifen enhances stemness and promotes metastasis of ER?36+ breast cancer by upregulating ALDH1A1 in cancer cells, Cell Res, vol.28, pp.336-358, 2018.

Z. Wang, X. Zhang, P. Shen, B. W. Loggie, Y. Chang et al., A variant of estrogen receptor-?, hER-?36: Transduction of estrogen-and antiestrogen-dependent membrane-initiated mitogenic signaling, Proc. Natl. Acad. Sci, vol.103, pp.9063-9068, 2006.

X. Zhang and Z. Wang, Estrogen receptor-? variant, ER-?36, is involved in tamoxifen resistance and estrogen hypersensitivity, Endocrinology, vol.154, 1990.

G. Li, J. Zhang, K. Jin, K. He, Y. Zheng et al., Estrogen receptor-?36 is involved in development of acquired tamoxifen resistance via regulating the growth status switch in breast cancer cells, Mol. Oncol, vol.7, pp.611-624, 2013.

Z. Wang, X. Zhang, P. Shen, B. W. Loggie, Y. Chang et al., Identification, cloning, and expression of human estrogen receptor-alpha36, a novel variant of human estrogen receptor-alpha66, Biochem. Biophys. Res. Commun, vol.336, pp.1023-1027, 2005.

R. A. Chaudhri, A. Hadadi, K. S. Lobachev, Z. Schwartz, and B. D. Boyan, Estrogen receptor-alpha 36 mediates the anti-apoptotic effect of estradiol in triple negative breast cancer cells via a membrane-associated mechanism, Biochim. Biophys. Acta, vol.1843, pp.2796-2806, 2014.

S. Omarjee, J. Jacquemetton, C. Poulard, N. Rochel, A. Dejaegere et al., The molecular mechanisms underlying the ER?-36-mediated signaling in breast cancer, vol.36, pp.2503-2514, 2017.

R. A. Chaudhri, N. Schwartz, K. Elbaradie, Z. Schwartz, and B. D. Boyan, Role of ER?36 in membrane-associated signaling by estrogen, Steroids, vol.81, pp.74-80, 2014.

C. Thiebaut, C. Chamard-jovenin, A. Chesnel, C. Morel, E. Djermoune et al., Mammary epithelial cell phenotype disruption in vitro and in vivo through ERalpha36 overexpression, PLoS ONE, vol.12, 2017.
URL : https://hal.archives-ouvertes.fr/hal-01492524

Z. Wang and L. Yin, Estrogen receptor alpha-36 (ER-?36): A new player in human breast cancer, Mol. Cell. Endocrinol, vol.418, pp.193-206, 2015.

K. So?tysik and P. Czekaj, ER?36-Another piece of the estrogen puzzle, Eur. J. Cell Biol, vol.94, pp.611-625, 2015.

C. Chamard-jovenin, A. C. Jung, A. Chesnel, J. Abecassis, S. Flament et al., From ER?66 to ER?36: A generic method for validating a prognosis marker of breast tumor progression, BMC Syst. Biol, vol.9, p.28, 2015.

Y. Zou, L. Ding, M. Coleman, and Z. Wang, Estrogen receptor-alpha (ER-?) suppresses expression of its variant ER-?36, FEBS Lett, vol.583, pp.1368-1374, 2009.

L. Yin and Z. Wang, Roles of the ER-?36-EGFR/HER2 positive regulatory loops in tamoxifen resistance, Steroids, vol.111, pp.95-99, 2016.

C. Chamard-jovenin, C. Thiebaut, A. Chesnel, E. Bresso, C. Morel et al., Low-Dose Alkylphenol Exposure Promotes Mammary Epithelium Alterations and Transgenerational Developmental Defects, But Does Not Enhance Tumorigenic Behavior of Breast Cancer Cells

K. Pileti? and T. Kunej, MicroRNA epigenetic signatures in human disease, Arch. Toxicol, vol.90, pp.2405-2419, 2016.

M. Yan, X. Li, D. Tong, C. Han, R. Zhao et al., Jin, X. miR-136 suppresses tumor invasion and metastasis by targeting RASAL2 in triple-negative breast cancer, Oncol. Rep, vol.36, pp.65-71, 2016.

J. Huan, L. Xing, Q. Lin, H. Xui, and X. Qin, Long noncoding RNA CRNDE activates Wnt/?-catenin signaling pathway through acting as a molecular sponge of microRNA-136 in human breast cancer, Am. J. Transl. Res, vol.9, pp.1977-1989, 2017.

T. Guo and G. Pan, MicroRNA-136 functions as a tumor suppressor in osteosarcoma via regulating metadherin, Cancer Biomark. Sect. Dis. Markers, vol.22, pp.79-87, 2018.

G. Deng, Y. Gao, Z. Cen, J. He, B. Cao et al., miR-136-5p Regulates the Inflammatory Response by Targeting the IKK?/NF-?B/A20 Pathway After Spinal Cord Injury, Cell. Physiol. Biochem. Int. J. Exp. Cell. Physiol. Biochem. Pharmacol, vol.50, pp.512-524, 2018.

, Int. J. Mol. Sci, vol.20, p.2637, 2019.

Z. Xie, T. Li, B. Gan, X. Gao, L. Gao et al., Investigation of miR-136-5p key target genes and pathways in lung squamous cell cancer based on TCGA database and bioinformatics analysis, Pathol. Res. Pract, vol.214, pp.644-654, 2018.

P. Chen, L. Zhao, X. Pan, L. Jin, C. Lin et al., Tumor suppressor microRNA-136-5p regulates the cellular function of renal cell carcinoma, Oncol. Lett, vol.15, pp.5995-6002, 2018.

T. Li, X. Gao, L. Gao, B. Gan, Z. Xie et al., Role of upregulated miR-136-5p in lung adenocarcinoma: A study of 1242 samples utilizing bioinformatics analysis, Pathol. Res. Pract, vol.214, pp.750-766, 2018.

A. Kozomara, S. Griffiths-jones, and . Mirbase, Annotating high confidence microRNAs using deep sequencing data, Nucleic Acids Res, vol.42, pp.68-73, 2014.

A. Kozomara, S. Griffiths-jones, and . Mirbase, Integrating microRNA annotation and deep-sequencing data, Nucleic Acids Res, vol.39, pp.152-157, 2011.

S. Griffiths-jones, R. J. Grocock, S. Van-dongen, A. Bateman, and A. J. Enright, miRBase: microRNA sequences, targets and gene nomenclature, Nucleic Acids Res, vol.34, pp.140-144, 2006.

S. Griffiths-jones, H. K. Saini, S. Van-dongen, A. J. Enright, and . Mirbase, Tools for microRNA genomics, Nucleic Acids Res, vol.36, pp.154-158, 2008.

B. Panwar, G. S. Omenn, and Y. Guan, miRmine: A database of human miRNA expression profiles, Bioinform. Oxf. Engl, vol.33, pp.1554-1560, 2017.

L. F. Gulyaeva and N. E. Kushlinskiy, Regulatory mechanisms of microRNA expression, J. Transl. Med, vol.14, p.143, 2016.

T. Eggermann, G. Perez-de-nanclares, E. R. Maher, I. K. Temple, Z. Tümer et al., Imprinting disorders: A group of congenital disorders with overlapping patterns of molecular changes affecting imprinted loci, Clin. Epigenetics, vol.7, issue.123, 2015.
URL : https://hal.archives-ouvertes.fr/hal-01231437

A. Uppal, S. C. Wightman, S. Mallon, G. Oshima, S. P. Pitroda et al., 14q32-encoded microRNAs mediate an oligometastatic phenotype, Oncotarget, vol.6, pp.3540-3552, 2015.

Z. Xu, B. Huang, J. Liu, X. Wu, N. Luo et al., Combinatorial anti-proliferative effects of tamoxifen and naringenin: The role of four estrogen receptor subtypes, Toxicology, vol.410, pp.231-246, 2018.

H. Ajj, A. Chesnel, S. Pinel, F. Plénat, S. Flament et al., An alkylphenol mix promotes seminoma derived cell proliferation through an ERalpha36-mediated mechanism, PLoS ONE, vol.8, 2013.
URL : https://hal.archives-ouvertes.fr/hal-00840880

E. A. Ariazi, J. C. Taylor, M. A. Black, E. Nicolas, M. J. Slifker et al., A New Role for ER?: Silencing via DNA Methylation of Basal, Stem Cell, and EMT Genes, Mol. Cancer Res, vol.15, pp.152-164, 2017.

R. A. Chaudhri, R. Olivares-navarrete, N. Cuenca, A. Hadadi, B. D. Boyan et al., Membrane estrogen signaling enhances tumorigenesis and metastatic potential of breast cancer cells via estrogen receptor-?36 (ER?36), J. Biol. Chem, vol.287, pp.7169-7181, 2012.

L. Shi, B. Dong, Z. Li, Y. Lu, T. Ouyang et al., Expression of ER-{alpha}36, a novel variant of estrogen receptor {alpha}, and resistance to tamoxifen treatment in breast cancer, J. Clin. Oncol, vol.27, pp.3423-3429, 2009.

T. Fleischer, X. Tekpli, A. Mathelier, S. Wang, D. Nebdal et al., DNA methylation at enhancers identifies distinct breast cancer lineages, Nat. Commun, vol.8, p.1379, 2017.

X. Xue, Y. A. Yang, A. Zhang, K. Fong, J. Kim et al., LncRNA HOTAIR enhances ER signaling and confers tamoxifen resistance in breast cancer, Oncogene, vol.35, pp.2746-2755, 2016.

S. Borges, H. Döppler, E. A. Perez, C. A. Andorfer, Z. Sun et al., Pharmacologic reversion of epigenetic silencing of the PRKD1 promoter blocks breast tumor cell invasion and metastasis, Breast Cancer Res, vol.15, 2013.

J. Yu, B. Qin, A. M. Moyer, S. Nowsheen, T. Liu et al., DNA methyltransferase expression in triple-negative breast cancer predicts sensitivity to decitabine, J. Clin. Invest, vol.128, pp.2376-2388, 2018.

, Int. J. Mol. Sci, vol.20, p.2637, 2019.

C. A. Pitta, P. Papageorgis, C. Charalambous, and A. I. Constantinou, Reversal of ER-? silencing by chromatin modifying agents overrides acquired tamoxifen resistance, Cancer Lett, vol.337, pp.167-176, 2013.

Y. Zhu, S. Shao, H. Pan, Z. Cheng, and X. Rui, MicroRNA-136 inhibits prostate cancer cell proliferation and invasion by directly targeting mitogen-activated protein kinase kinase 4, Mol. Med. Rep, vol.17, pp.4803-4810, 2018.

D. Li, X. Fei, Y. Dong, C. Cheng, Y. Yang et al., The long non-coding RNA CRNDE acts as a ceRNA and promotes glioma malignancy by preventing miR-136-5p-mediated downregulation of Bcl-2 and Wnt2, Oncotarget, vol.8, pp.88163-88178, 2017.

A. Singh, E. Willems, A. Singh, I. M. Ong, and A. K. Verma, Ultraviolet radiation-induced differential microRNA expression in the skin of hairless SKH1 mice, a widely used mouse model for dermatology research, Oncotarget, vol.7, pp.84924-84937, 2016.

A. Wallacides, A. Chesnel, H. Ajj, M. Chillet, S. Flament et al., Estrogens promote proliferation of the seminoma-like TCam-2 cell line through a GPER-dependent ER?36 induction, Mol. Cell. Endocrinol, vol.350, pp.61-71, 2012.

M. V. Iorio, M. Ferracin, C. Liu, A. Veronese, R. Spizzo et al., MicroRNA gene expression deregulation in human breast cancer, Cancer Res, vol.65, pp.7065-7070, 2005.

S. Paszek, N. Gab?o, E. Barna?, M. Szybka, J. Morawiec et al., Zawlik, I. Dysregulation of microRNAs in triple-negative breast cancer, Ginekol. Pol, vol.88, pp.530-536, 2017.

C. Braicu, L. Raduly, G. Morar-bolba, R. Cojocneanu, A. Jurj et al., Aberrant miRNAs expressed in HER-2 negative breast cancers patient, J. Exp. Clin. Cancer Res, vol.37, 2018.

Y. Sathipati, S. Ho, and S. , Identifying a miRNA signature for predicting the stage of breast cancer, Sci. Rep, vol.8, 2018.

D. Cheng, H. He, and B. Liang, A three-microRNA signature predicts clinical outcome in breast cancer patients, Eur. Rev. Med. Pharmacol. Sci, vol.22, pp.6386-6395, 2018.

M. Kagami, M. J. O'sullivan, A. J. Green, Y. Watabe, O. Arisaka et al., The IG-DMR and the MEG3-DMR at human chromosome 14q32.2: Hierarchical interaction and distinct functional properties as imprinting control centers

L. Zhu, Y. Liu, Q. Chen, G. Yu, J. Chen et al., Long-Noncoding RNA Colorectal Neoplasia Differentially Expressed Gene as a Potential Target to Upregulate the Expression of IRX5 by miR-136-5P to Promote Oncogenic Properties in Hepatocellular Carcinoma, Cell. Physiol. Biochem, vol.50, pp.2229-2248, 2018.

B. Li, F. Wang, X. Li, S. Sun, Y. Shen et al., Hsa_circ_0008309 May Be a Potential Biomarker for Oral Squamous Cell Carcinoma, Dis. Markers, vol.7496890, 2018.

C. Duan, Y. Liu, Y. Li, H. Chen, X. Liu et al., Sulfasalazine alters microglia phenotype by competing endogenous RNA effect of miR-136-5p and long non-coding RNA HOTAIR in cuprizone-induced demyelination, Biochem. Pharmacol, vol.155, pp.110-123, 2018.

M. Dai, S. Li, and X. Qin, Colorectal neoplasia differentially expressed: A long noncoding RNA with an imperative role in cancer, OncoTargets Ther, vol.11, pp.3755-3763, 2018.

W. Wang, F. Yuan, and J. Xu, The prognostic role of long noncoding RNA CRNDE in cancer patients: A systematic review and meta-analysis, Neoplasma, vol.66, pp.73-82, 2019.

C. Lin, L. Liu, J. B. Addison, W. F. Wonderlin, A. V. Ivanov et al., A KLF4-miRNA-206 autoregulatory feedback loop can promote or inhibit protein translation depending upon cell context, Mol. Cell. Biol, vol.31, pp.2513-2527, 2011.

L. H. Sobin and C. H. Wittekind, TNM classification of malignant tumors

D. G. Altman and J. M. Bland, CrossRef] REFERENCES and performed the functional analysis that follows the covariates selection with armada. Stefano Cairo and Olivier Déas, XenTech, Genopole -Campus, vol.331, p.903, 2005.

I. Aurélien-de-reynies, J. Bieche, and . Métral-from-the-carte, Identité des Tumeurs program provided the access to transcriptomic raw data and biological samples (ERa66+, PR+, PR-datasets). The statistical methodology has been developed by Anne Gégout-Petit and Aurélie Muller-Gueudin, and thanks to Yaojie Shi and Hafid Chakir during their Master internships. References Antanavi?i?t?, Ieva, Mikalayeva, Valeryia, Ceslevi?ien?, Ieva, Mila?i?t?, Gintar?, Skeberdis, Vytenis Arvydas and Bordel, Scientific reports, vol.7, issue.1, p.7820, 2017.

J. Aubert, . Bar-hen, . Avner, J. Daudin, R. et al., , 2004.

, Determination of the differentially expressed genes in microarray experiments using local fdr, BMC bioinformatics, vol.5, issue.1, p.1

. Bar-hen, . Avner, J. Daudin, R. , and S. , Comparaisons multiples pour les microarrays, Journal de la Société Française de Statistique, vol.146, issue.1-2, pp.45-62, 2005.
URL : https://hal.archives-ouvertes.fr/hal-01219538

Y. Benjamini and Y. Hochberg, Controlling the false discovery rate: a practical and powerful approach to multiple testing, Journal of the Royal Statistical Society, 1995.

, Series B (Methodological, pp.289-300

Y. Benjamini and D. Yekutieli, The control of the false discovery rate in multiple testing under dependency. The Annals of Statistics, pp.1165-1188, 2001.

C. E. Bonferroni, Teoria statistica delle classi e calcolo delle probabilita, 1936.

D. Causeur, . Friguet, . Chloé, M. Houee-bigot, and M. Kloareg, , 2011.

, Factor analysis for multiple testing (famt): an R package for large-scale significance testing under dependence, Journal of Statistical Software, vol.40, issue.14, p.19

. Chamard-jovenin, J. Clémence, A. C. Chesnel, . Amand, . Abecassis et al., From ER?66 to ER?36: a generic method for validating a prognosis marker of breast tumor progression, BMC systems biology, vol.9, issue.1, p.28, 2015.

M. Chavent, . Kuentz, . Vanessa, . Liquet, . Benoit et al., Clustofvar: an R package for the clustering of variables, Journal of Statistical Software, vol.50, pp.91-116, 2012.
URL : https://hal.archives-ouvertes.fr/hal-00646805

S. Dudoit, M. J. Van-der-laan, and K. S. Pollard, Multiple testing. part I. single-step procedures for control of general type I error rates, Statistical Applications in Genetics and Molecular Biology, vol.3, issue.1, pp.1-69, 2004.

. Dudoit, Y. Sandrine, Y. Hwa, M. J. Callow, and T. P. Speed, Statistical methods for identifying differentially expressed genes in replicated cDNA microarray experiments, Statistica sinica, pp.111-139, 2002.

B. Efron, Local false discovery rates. Division of Biostatistics, 2005.

C. Friguet, Impact of dependence in large-scale multiple testing, 2012.
URL : https://hal.archives-ouvertes.fr/tel-00539741

C. Friguet and D. Causeur, Estimation of the proportion of true null hy-REFERENCES potheses in high-dimensional data under dependence, Computational Statistics & Data Analysis, vol.55, issue.9, pp.2665-2676, 2011.

C. Friguet, . Kloareg, and D. Causeur, A factor model approach to multiple testing under dependence, Journal of the American Statistical Association, vol.104, issue.488, pp.1406-1415, 2009.
URL : https://hal.archives-ouvertes.fr/hal-00458049

R. Genuer, J. Poggi, and C. Tuleau-malot, Variable selection using random forests, Pattern Recognition Letters, vol.31, issue.14, pp.2225-2236, 2010.
URL : https://hal.archives-ouvertes.fr/hal-00755489

O. P. Günther, . Shin, . Heesun, R. T. Ng, . Mcmaster et al., Novel multivariate methods for integration of genomics and proteomics data: applications in a kidney transplant rejection study, Omics: a journal of integrative biology, vol.18, issue.11, pp.682-695, 2014.

L. Cao, . Kim-anh, S. Boitard, and P. Besse, Sparse PLS discriminant analysis: biologically relevant feature selection and graphical displays for multiclass problems, BMC bioinformatics, vol.12, issue.1, p.1, 2011.
URL : https://hal.archives-ouvertes.fr/hal-00959981

L. Quesne, J. Caldas, and C. , Micro-rnas and breast cancer, Molecular oncology, vol.4, issue.3, pp.230-241, 2010.

L. Meier, S. Van-de-geer, and P. Bühlmann, The group lasso for logistic regression, Journal of the Royal Statistical Society: Series B (Statistical Methodology), vol.70, issue.1, pp.53-71, 2008.

N. Meinshausen and P. Bühlmann, High-dimensional graphs and variable selection with the lasso, The Annals of Statistics, pp.1436-1462, 2006.

A. Muller-gueudin and A. Gégout-petit, , 2019.

Y. Shi, Microarray data analysis : feature selection, clustering and prediction, pp.1-40, 2016.

J. D. Storey, A direct approach to false discovery rates, Journal of the Royal Statistical Society: Series B (Statistical Methodology), vol.64, issue.3, pp.479-498, 2002.

J. D. Storey, The positive false discovery rate: a bayesian interpretation and the q-value, The Annals of Statistics, pp.2013-2035, 2003.

J. D. Storey and R. Tibshirani, Statistical significance for genomewide studies, Proceedings of the National Academy of Sciences, vol.100, issue.16, pp.9440-9445, 2003.

Y. Su, . Murali, . Tm, . Pavlovic, . Vladimir et al., Rankgene: identification of diagnostic genes based on expression data, Bioinformatics, vol.19, issue.12, pp.1578-1579, 2003.

M. Tenenhaus, V. Vinzi, . Esposito, Y. Chatelin, and C. Lauro, PLS path modeling, Computational Statistics & Data Analysis, vol.48, issue.1, pp.159-205, 2005.
URL : https://hal.archives-ouvertes.fr/hal-00869374

R. Tibshirani, Regression shrinkage and selection via the lasso, Journal of the Royal Statistical Society. Series B (Methodological, pp.267-288, 1996.

Z. Wang, . Zhang, . Xintian, . Shen, . Peng et al., Identification, cloning, and expression of human estrogen receptor-?36, a novel variant of human estrogen receptor-?66, the Oncogenetics laboratory, INSERM U735, Institut Curie-Hôpital-Centre René Huguenin, vol.336, pp.1023-1027, 2005.

, The measurement of ER?36 expression in each tumor (Step 1: clinical data completion) has been done as described in Thiebaut and others, 2017.

, Assays were performed at least in triplicate, and the mean values were used to calculate expression levels

M. Guedj, . Marisa, D. Laëtitia, . Reynies, . Aurélien et al., A refined molecular taxonomy of breast cancer, Oncogene, vol.31, issue.9, p.1196, 2012.
URL : https://hal.archives-ouvertes.fr/inserm-00611752

F. Reyal, C. Guyader, . Decraene, . Charles, C. Lucchesi et al., Molecular profiling of patient-derived breast cancer xenografts, R11. traductionnelles des histones, comme les HDACs ou les HMTs, vol.14, 2012.
URL : https://hal.archives-ouvertes.fr/inserm-00675301

. Muñoz-de-toro, Soto ont mis en évidence que l'exposition foetale ou néonatale au BPA perturbe la signalisation oestrogénique et conduit à l'apparition d'altérations structurales de la glande mammaire, En parallèle, étant donné que différentes études menées par l'équipe d'Ana M, 2005.

U. Toutefois and . Étude-de-huan, Au total, nous avons mis en évidence 12 micro-ARNs susceptibles d'interagir avec la région 3'UTR de l'ARNm de ce variant et nous avons validé l'implication d'hsa-miR-136-5p dans la régulation négative de l'expression d'ER?36. A l'heure actuelle, les études disponibles dans la littérature indiquent que le niveau d'expression de hsa-miR-136-5p ne constitue pas un marqueur pronostic suffisant pour prédire le devenir des patientes, Pour finir, nous avons exploré au cours de ces travaux les mécanismes de régulation posttranscriptionnelle de l'expression d, 2017.

B. Chen, Ainsi, l'analyse de ces ARNs qui se comportent comme, 2018.

B. Chen, Par exemple, le répertoire en micro-ARNs dans des testicules de rats adultes est fortement perturbé après une exposition chronique (du jour de la conception à l'âge adulte) à de faibles doses d'un mélange d'alkylphénols et de phtalates, 2016.

T. Dans-le-même-ordre-d'idée, ont montré que le BPA module l'expression de plusieurs micro-ARNs, et notamment du micro-ARN oncogénique hsa-miR-21, 2012.

F. Acconcia, P. Ascenzi, G. Fabozzi, P. Visca, and M. Marino, S-palmitoylation modulates human estrogen receptor-alpha functions, Biochem. Biophys. Res. Commun, vol.316, pp.878-883, 2004.

B. D. Adams, H. Furneaux, and B. A. White, The micro-ribonucleic acid (miRNA) miR-206 targets the human estrogen receptor-alpha (ERalpha) and represses ERalpha messenger RNA and protein expression in breast cancer cell lines, Mol. Endocrinol. Baltim. Md, vol.21, pp.1132-1147, 2007.

A. B. Adomas, S. A. Grimm, C. Malone, M. Takaku, J. K. Sims et al., Breast tumor specific mutation in GATA3 affects physiological mechanisms regulating transcription factor turnover, BMC Cancer, vol.14, p.278, 2014.

H. Ajj, A. Chesnel, S. Pinel, F. Plénat, S. Flament et al., An alkylphenol mix promotes seminoma derived cell proliferation through an ERalpha36-mediated mechanism, PLoS ONE, vol.8, 2013.
URL : https://hal.archives-ouvertes.fr/hal-00840880

A. Alavian-ghavanini and J. Rüegg, Understanding Epigenetic Effects of Endocrine Disrupting Chemicals: From Mechanisms to Novel Test Methods, Basic Clin. Pharmacol. Toxicol, vol.122, pp.38-45, 2018.

M. Al-hajj, M. S. Wicha, A. Benito-hernandez, S. J. Morrison, C. et al., Prospective identification of tumorigenic breast cancer cells, Proc. Natl. Acad. Sci. U. S. A, vol.100, pp.3983-3988, 2003.

H. Al-nakhle, L. Smith, S. Bell, P. Burns, M. Cummings et al., Regulation of estrogen receptor ?1 expression in breast cancer by epigenetic modification of the 5? regulatory region, Int. J. Oncol, vol.43, 2013.

F. Aloraifi, T. Mcdevitt, R. Martiniano, J. Mcgreevy, R. Mclaughlin et al., Detection of novel germline mutations for breast cancer in non-BRCA1/2 families, FEBS J, vol.282, pp.3424-3437, 2015.

S. Amaral, W. Schroth, S. Kugler, P. Fritz, W. Simon et al., The promoter C specific ERalpha isoform is associated with tamoxifen outcome in breast cancer, Breast Cancer Res. Treat, vol.118, pp.323-331, 2009.

I. Antanavi?i?t?, V. Mikalayeva, I. Ceslevi?ien?, G. Mila?i?t?, V. A. Skeberdis et al., Transcriptional hallmarks of cancer cell lines reveal an emerging role of branched chain amino acid catabolism, 2017.

A. Aranda and A. Pascual, Nuclear hormone receptors and gene expression, Physiol. Rev, vol.81, pp.1269-1304, 2001.

E. A. Ariazi, J. C. Taylor, M. A. Black, E. Nicolas, M. J. Slifker et al., A New Role for ER?: Silencing via DNA Methylation of Basal, Stem Cell, and EMT Genes, Mol. Cancer Res, vol.15, pp.152-164, 2017.

H. Armanious, P. Gelebart, J. Mackey, Y. Ma, L. et al., STAT3 upregulates the protein expression and transcriptional activity of ?-catenin in breast cancer, Int. J. Clin. Exp. Pathol, vol.3, pp.654-664, 2010.

L. S. Aroeira, A. Aguilera, J. A. Sánchez-tomero, M. A. Bajo, G. Peso et al., Epithelial to Mesenchymal Transition and Peritoneal Membrane Failure in Peritoneal Dialysis Patients: Pathologic Significance and Potential Therapeutic Interventions, J. Am. Soc. Nephrol, vol.18, pp.2004-2013, 2007.

M. K. Asiedu, J. N. Ingle, M. D. Behrens, D. C. Radisky, and K. L. Knutson, TGF?/TNF?-Mediated Epithelial-Mesenchymal Transition Generates Breast Cancer Stem Cells with a Claudin-Low Phenotype, Cancer Res, vol.71, pp.4707-4719, 2011.

M. Asselin-labat, K. D. Sutherland, H. Barker, R. Thomas, M. Shackleton et al., Gata-3 is an essential regulator of mammarygland morphogenesis and luminal-cell differentiation, Nat. Cell Biol, vol.9, pp.201-209, 2007.

S. De-assis, A. Warri, M. I. Cruz, O. Laja, Y. Tian et al., High-fat or ethinyl-oestradiol intake during pregnancy increases mammary cancer risk in several generations of offspring, Nat. Commun, vol.3, p.1053, 2012.

P. Augereau, F. Miralles, V. Cavaillès, C. Gaudelet, M. Parker et al., Characterization of the proximal estrogen-responsive element of human cathepsin D gene, Mol. Endocrinol. Baltim. Md, vol.8, pp.693-703, 1994.

A. Ayyanan, O. Laribi, S. Schuepbach-mallepell, C. Schrick, M. Gutierrez et al., Perinatal Exposure to Bisphenol A Increases Adult Mammary Gland Progesterone Response and Cell Number, Mol. Endocrinol, vol.25, pp.1915-1923, 2011.

S. Balasenthil and R. K. Vadlamudi, Functional Interactions between the Estrogen Receptor Coactivator PELP1/MNAR and Retinoblastoma Protein, J. Biol. Chem, vol.278, pp.22119-22127, 2003.

M. P. Ball, J. B. Li, Y. Gao, J. Lee, E. Leproust et al., Targeted and genome-scale methylomics reveals gene body signatures in human cell lines, Nat. Biotechnol, vol.27, pp.361-368, 2009.

K. Banerjee and H. Resat, Constitutive activation of STAT3 in breast cancer cells: A review, Int. J. Cancer, vol.138, pp.2570-2578, 2016.

S. Baulande, A. Criqui, and M. Duthieuw, Les microARN circulants, une nouvelle classe de biomarqueurs pour la médecine. médecine/sciences, vol.30, pp.289-296, 2014.

L. Benetatos, E. Hatzimichael, E. Londin, G. Vartholomatos, P. Loher et al., The microRNAs within the DLK1-DIO3 genomic region: involvement in disease pathogenesis, Cell. Mol. Life Sci. CMLS, vol.70, pp.795-814, 2013.

M. A. Bennesch and D. Picard, Minireview: Tipping the Balance: Ligand-Independent Activation of Steroid Receptors, Mol. Endocrinol, vol.29, pp.349-363, 2015.

D. K. Biswas, A. P. Cruz, E. Gansberger, and A. B. Pardee, Epidermal growth factor-induced nuclear factor kappa B activation: A major pathway of cell-cycle progression in estrogen-receptor negative breast cancer cells, Proc. Natl. Acad. Sci. U. S. A, vol.97, pp.8542-8547, 2000.

W. P. Bocchinfuso, J. K. Lindzey, S. C. Hewitt, J. A. Clark, P. H. Myers et al., Induction of mammary gland development in estrogen receptor-alpha knockout mice, Endocrinology, vol.141, pp.2982-2994, 2000.

W. Bourguet, P. Germain, and H. Gronemeyer, Nuclear receptor ligand-binding domains: three-dimensional structures, molecular interactions and pharmacological implications, Trends Pharmacol. Sci, vol.21, pp.381-388, 2000.

D. W. Brann, Q. Zhang, R. Wang, V. B. Mahesh, and R. K. Vadlamudi, PELP1 -A Novel Estrogen Receptor-Interacting Protein, Mol. Cell. Endocrinol, vol.290, pp.2-7, 2008.

F. Bray, J. Ferlay, I. Soerjomataram, R. L. Siegel, L. A. Torre et al., Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries, CA. Cancer J. Clin, vol.68, pp.394-424, 2018.

C. Brisken and B. Malley, Hormone action in the mammary gland, Cold Spring Harb. Perspect. Biol, vol.2, p.3178, 2010.

W. H. Brooks and Y. Renaudineau, Epigenetics and autoimmune diseases: the X chromosomenucleolus nexus, Front. Genet, vol.6, 2015.
URL : https://hal.archives-ouvertes.fr/hal-01132409

S. E. Bulun, D. Chen, I. Moy, D. C. Brooks, and H. Zhao, Aromatase, breast cancer and obesity: a complex interaction, Trends Endocrinol. Metab. TEM, vol.23, pp.83-89, 2012.

J. Buñay, E. Larriba, R. D. Moreno, and J. Mazo, , 2017.

M. L. Burness and M. S. Wicha, Tamoxifen and ER?36: Fertilizing the seeds of breast cancer metastasis, Cell Res, vol.28, pp.391-392, 2018.

L. Caizzi, G. Ferrero, S. Cutrupi, F. Cordero, C. Ballaré et al., Genome-wide activity of unliganded estrogen receptor-? in breast cancer cells, Proc. Natl. Acad. Sci. U. S. A, vol.111, pp.4892-4897, 2014.

R. A. Campbell, P. Bhat-nakshatri, N. M. Patel, D. Constantinidou, S. Ali et al., , 2001.

, Phosphatidylinositol 3-Kinase/AKT-mediated Activation of Estrogen Receptor ?: A NEW MODEL FOR ANTI-ESTROGEN RESISTANCE, J. Biol. Chem, vol.276, pp.9817-9824

C. Carmeci, D. A. Thompson, H. Z. Ring, U. Francke, and R. J. Weigel, Identification of a gene (GPR30) with homology to the G-protein-coupled receptor superfamily associated with estrogen receptor expression in breast cancer, Genomics, vol.45, pp.607-617, 1997.

S. Carreau, C. Bois, L. Zanatta, F. R. Silva, H. Bouraima-lelong et al., , 2011.

, Estrogen signaling in testicular cells, Life Sci, vol.89, pp.584-587

J. S. Carroll, Mechanisms of oestrogen receptor (ER) gene regulation in breast cancer, Eur. J. Endocrinol, vol.175, pp.41-49, 2016.

J. S. Carroll, X. S. Liu, A. S. Brodsky, W. Li, C. A. Meyer et al., Chromosome-wide mapping of estrogen receptor binding reveals long-range regulation requiring the forkhead protein FoxA1, Cell, vol.122, pp.33-43, 2005.

J. S. Carroll, T. E. Hickey, G. A. Tarulli, M. Williams, and W. D. Tilley, Deciphering the divergent roles of progestogens in breast cancer, Nat. Rev. Cancer, vol.17, pp.54-64, 2017.

L. Castellano, G. Giamas, J. Jacob, R. C. Coombes, W. Lucchesi et al., The estrogen receptor-?-induced microRNA signature regulates itself and its transcriptional response, Proc. Natl. Acad. Sci. U. S. A, vol.106, pp.15732-15737, 2009.

A. Castet, A. Boulahtouf, G. Versini, S. Bonnet, P. Augereau et al., Multiple domains of the Receptor-Interacting Protein 140 contribute to transcription inhibition, Nucleic Acids Res, vol.32, pp.1957-1966, 2004.

A. Castet, S. Carascossa, V. Duong, P. Augereau, S. Jalaguier et al., Rôle de RIP140 dans la signalisation hormonale. médecine/sciences 21, pp.273-278, 2005.

S. Catalano, I. Barone, C. Giordano, P. Rizza, H. Qi et al., Rapid Estradiol/ER? Signaling Enhances Aromatase Enzymatic Activity in Breast Cancer Cells, Mol. Endocrinol, vol.23, pp.1634-1645, 2009.

S. Chakraborty, N. Jaiswal, and A. Nag, Biology of FOXM1 and its emerging role in cancer therapy, J. Proteins Proteomics, vol.5, 2014.

C. Chamard-jovenin, A. C. Jung, A. Chesnel, J. Abecassis, S. Flament et al., From ER?66 to ER?36: a generic method for validating a prognosis marker of breast tumor progression, BMC Syst. Biol, vol.9, p.28, 2015.

K. L. Chambliss, I. S. Yuhanna, R. G. Anderson, M. E. Mendelsohn, and P. W. Shaul, ERbeta has nongenomic action in caveolae, Mol. Endocrinol. Baltim. Md, vol.16, pp.938-946, 2002.

S. Chandarlapaty, D. Chen, W. He, P. Sung, A. Samoila et al., Prevalence of ESR1 Mutations in Cell-Free DNA and Outcomes in Metastatic Breast Cancer: A Secondary Analysis of the BOLERO-2 Clinical Trial, JAMA Oncol, vol.2, pp.1310-1315, 2016.

D. H. Chang, G. Cattoretti, and K. L. Calame, The dynamic expression pattern of B lymphocyte induced maturation protein-1 (Blimp-1) during mouse embryonic development, Mech. Dev, vol.117, pp.305-309, 2002.

A. K. Charles and P. D. Darbre, Combinations of parabens at concentrations measured in human breast tissue can increase proliferation of MCF-7 human breast cancer cells, J. Appl. Toxicol, vol.33, pp.390-398, 2013.

A. Chatterjee, E. J. Rodger, and M. R. Eccles, Epigenetic drivers of tumourigenesis and cancer metastasis, Semin. Cancer Biol, vol.51, pp.149-159, 2018.

R. A. Chaudhri, R. Olivares-navarrete, N. Cuenca, A. Hadadi, B. D. Boyan et al., , 2012.

, Membrane estrogen signaling enhances tumorigenesis and metastatic potential of breast cancer cells via estrogen receptor-?36 (ER?36), J. Biol. Chem, vol.287, pp.7169-7181

R. A. Chaudhri, A. Hadadi, K. S. Lobachev, Z. Schwartz, and B. D. Boyan, Estrogen receptoralpha 36 mediates the anti-apoptotic effect of estradiol in triple negative breast cancer cells via a membrane-associated mechanism, Biochim. Biophys. Acta, vol.1843, pp.2796-2806, 2014.

R. A. Chaudhri, N. Schwartz, K. Elbaradie, Z. Schwartz, and B. D. Boyan, Role of ER?36 in membrane-associated signaling by estrogen, Steroids, vol.81, pp.74-80, 2014.

B. Chen, H. Tang, X. Liu, P. Liu, L. Yang et al., miR-22 as a prognostic factor targets glucose transporter protein type 1 in breast cancer, Cancer Lett, vol.356, pp.410-417, 2015.

B. Chen, W. Wei, X. Huang, X. Xie, Y. Kong et al., circEPSTI1 as a Prognostic Marker and Mediator of Triple-Negative Breast Cancer Progression, Theranostics, vol.8, pp.4003-4015, 2018.

P. Chen, P. Chen, L. Zhao, L. Zhao, X. Pan et al., Tumor suppressor microRNA 136 5p regulates the cellular function of renal cell carcinoma, Oncol. Lett, vol.15, pp.5995-6002, 2018.

G. S. Chetrite, J. Cortes-prieto, J. C. Philippe, F. Wright, and J. R. Pasqualini, Comparison of estrogen concentrations, estrone sulfatase and aromatase activities in normal, and in cancerous, human breast tissues, J. Steroid Biochem. Mol. Biol, vol.72, pp.23-27, 2000.

A. Chiche, Study of mammary stem and progenitor cells and of their contribution in tumorigenesis : role of transcriptional factors Myc and p53, 2012.
URL : https://hal.archives-ouvertes.fr/tel-00924981

I. Chu, K. Blackwell, S. Chen, and J. Slingerland, The dual ErbB1/ErbB2 inhibitor, lapatinib (GW572016), cooperates with tamoxifen to inhibit both cell proliferation-and estrogen-dependent gene expression in antiestrogen-resistant breast cancer, Cancer Res, vol.65, pp.18-25, 2005.

S. S. Chung, C. Aroh, and J. V. Vadgama, Constitutive Activation of STAT3 Signaling Regulates hTERT and Promotes Stem Cell-Like Traits in Human Breast Cancer Cells, PLOS ONE, vol.8, p.83971, 2013.

C. H. Clarke, A. M. Norfleet, M. S. Clarke, C. S. Watson, K. A. Cunningham et al.,

, Perimembrane localization of the estrogen receptor alpha protein in neuronal processes of cultured hippocampal neurons, Neuroendocrinology, vol.71, pp.34-42

M. Clarke, R. Collins, S. Darby, C. Davies, P. Elphinstone et al., Effects of radiotherapy and of differences in the extent of surgery for early breast cancer on local recurrence and 15-year survival: an overview of the randomised trials, Lancet Lond. Engl, vol.366, pp.2087-2106, 2005.

C. Bennink, H. J. Holinka, C. F. Diczfalusy, and E. , Estetrol review: profile and potential clinical applications, Climacteric J. Int. Menopause Soc, vol.11, issue.1, pp.47-58, 2008.

F. Coin and J. Egly, Formation du complexe d'initiation de la transcription : des facteurs généraux aux complexes qui déstabilisent la chromatine. médecine/sciences 16, p.593, 2000.

L. Connelly, W. Barham, R. Pigg, L. Saint-jean, T. Sherrill et al., Activation of nuclear factor-kappa B in mammary epithelium promotes milk loss during mammary development and infection, J. Cell. Physiol, vol.222, pp.73-81, 2010.

A. Cvoro, C. Tzagarakis-foster, D. Tatomer, S. Paruthiyil, M. S. Fox et al., Distinct roles of unliganded and liganded estrogen receptors in transcriptional repression, Mol. Cell, vol.21, pp.555-564, 2006.

N. C. D'amato, M. A. Gordon, B. Babbs, N. S. Spoelstra, C. Butterfield et al., Cooperative Dynamics of AR and ER Activity in Breast Cancer, Mol. Cancer Res. MCR, vol.14, pp.1054-1067, 2016.

C. Davies, H. Pan, J. Godwin, R. Gray, R. Arriagada et al., Long-term effects of continuing adjuvant tamoxifen to 10 years versus stopping at 5 years after diagnosis of oestrogen receptor-positive breast cancer: ATLAS, a randomised trial, Lancet Lond. Engl, vol.381, pp.805-816, 2013.

E. C. Deconinck, L. A. Mcpherson, and R. J. Weigel, Transcriptional regulation of estrogen receptor in breast carcinomas, Mol. Cell. Biol, vol.15, pp.2191-2196, 1995.

D. G. Denardo, H. Kim, S. Hilsenbeck, V. Cuba, A. Tsimelzon et al., Global Gene Expression Analysis of Estrogen Receptor Transcription Factor Cross Talk in Breast Cancer: Identification of Estrogen-Induced/Activator Protein-1-Dependent Genes, Mol. Endocrinol, vol.19, pp.362-378, 2005.

D. G. Denardo, V. L. Cuba, H. Kim, K. Wu, A. V. Lee et al., Estrogen receptor DNA binding is not required for estrogen-induced breast cell growth, Mol. Cell. Endocrinol, vol.277, pp.13-25, 2007.

H. Deng, X. Huang, J. Fan, L. Wang, Q. Xia et al., A variant of estrogen receptor-?, ER-?36 is expressed in human gastric cancer and is highly correlated with lymph node metastasis, Oncol. Rep, vol.24, pp.171-176, 2010.

H. Denis, . Ndlovu, N. Matladi, and F. Fuks, Regulation of mammalian DNA methyltransferases: a route to new mechanisms, EMBO Rep, vol.12, pp.647-656, 2011.

A. Derghal, M. Djelloul, J. Trouslard, and L. Mounien, An Emerging Role of micro-RNA in the Effect of the Endocrine Disruptors, Front. Neurosci, vol.10, 2016.
URL : https://hal.archives-ouvertes.fr/hal-01471933

A. E. Dhamad, Z. Zhou, J. Zhou, and Y. Du, Systematic Proteomic Identification of the Heat Shock Proteins (Hsp) that Interact with Estrogen Receptor Alpha (ER?) and Biochemical Characterization of the ER?-Hsp70 Interaction, PloS One, vol.11, p.160312, 2016.

E. Dhimolea, P. R. Wadia, T. J. Murray, M. L. Settles, J. D. Treitman et al., Prenatal Exposure to BPA Alters the Epigenome of the Rat Mammary Gland and Increases the Propensity to Neoplastic Development, PLoS ONE, vol.9, 2014.

W. Ding, J. Ren, H. Ren, W. , and D. , Long Noncoding RNA HOTAIR Modulates MiR-206-mediated Bcl-w Signaling to Facilitate Cell Proliferation in, Breast Cancer. Sci. Rep, vol.7, p.17261, 2017.

J. Dittmer, Breast cancer stem cells: Features, key drivers and treatment options, Semin. Cancer Biol, vol.53, pp.59-74, 2018.

M. Dolled-filhart, R. L. Camp, D. P. Kowalski, B. L. Smith, and D. L. Rimm, Tissue microarray analysis of signal transducers and activators of transcription 3 (Stat3) and phospho-Stat3 (Tyr705) in node-negative breast cancer shows nuclear localization is associated with a better prognosis, Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res, vol.9, pp.594-600, 2003.

M. D. Driscoll, G. Sathya, M. Muyan, C. M. Klinge, R. Hilf et al., Sequence requirements for estrogen receptor binding to estrogen response elements, J. Biol. Chem, vol.273, pp.29321-29330, 1998.

M. J. Duffy, N. Harbeck, M. Nap, R. Molina, A. Nicolini et al., Clinical use of biomarkers in breast cancer: Updated guidelines from the European Group on Tumor Markers (EGTM), Eur. J. Cancer Oxf. Engl, vol.75, pp.284-298, 1990.

D. Eckert, S. Buhl, S. Weber, R. Jäger, and H. Schorle, The AP-2 family of transcription factors, Genome Biol, vol.6, p.246, 2005.

J. Eeckhoute, E. K. Keeton, M. Lupien, S. A. Krum, J. S. Carroll et al., Positive cross-regulatory loop ties GATA-3 to estrogen receptor alpha expression in breast cancer, Cancer Res, vol.67, pp.6477-6483, 2007.

M. Ehrlich, L. , and M. , DNA methylation and differentiation: silencing, upregulation and modulation of gene expression, Epigenomics, vol.5, pp.553-568, 2013.

P. Eroles, A. Bosch, A. Pérez-fidalgo, J. Lluch, and A. , Molecular biology in breast cancer: Intrinsic subtypes and signaling pathways, Cancer Treat. Rev, vol.38, pp.698-707, 2012.

S. Fallahpour, T. Navaneelan, P. De, and A. Borgo, Breast cancer survival by molecular subtype: a population-based analysis of cancer registry data, CMAJ Open, vol.5, pp.734-739, 2017.

A. Farooq, Structural and Functional Diversity of Estrogen Receptor Ligands, Curr. Top. Med. Chem, vol.15, pp.1372-1384, 2015.

Y. Feng, M. Spezia, S. Huang, C. Yuan, Z. Zeng et al., Breast cancer development and progression: Risk factors, cancer stem cells, signaling pathways, genomics, and molecular pathogenesis, Genes Dis, vol.5, pp.77-106, 2018.

E. J. Filardo, T. , and P. , Minireview: G protein-coupled estrogen receptor-1, GPER-1: its mechanism of action and role in female reproductive cancer, renal and vascular physiology, Endocrinology, vol.153, pp.2953-2962, 2012.

G. Flouriot, H. Brand, S. Denger, R. Metivier, M. Kos et al., Identification of a new isoform of the human estrogen receptor-alpha (hER-?) that is encoded by distinct transcripts and that is able to repress hER-? activation function 1, EMBO J, vol.19, pp.4688-4700, 2000.

E. M. Fox, R. J. Davis, and M. A. Shupnik, ER? in Breast Cancer -Onlooker, Passive Player, or Active Protector?, Steroids, vol.73, pp.1039-1051, 2008.

R. C. Friedman, K. K. Farh, .. Burge, C. B. Bartel, and D. P. , Most mammalian mRNAs are conserved targets of microRNAs, Genome Res, vol.19, pp.92-105, 2009.

N. Friedrichs, R. Jäger, E. Paggen, C. Rudlowski, S. Merkelbach-bruse et al., Distinct spatial expression patterns of AP-2alpha and AP-2gamma in non-neoplastic human breast and breast cancer, Mod. Pathol. Off. J. U. S. Can. Acad. Pathol. Inc, vol.18, pp.431-438, 2005.

Y. G. Gangloff, C. Romier, S. Thuault, S. Werten, D. et al., The histone fold is a key structural motif of transcription factor TFIID, Trends Biochem. Sci, vol.26, pp.250-257, 2001.
URL : https://hal.archives-ouvertes.fr/hal-02126931

H. Gao, X. Song, T. Kang, B. Yan, L. Feng et al., Long noncoding RNA CRNDE functions as a competing endogenous RNA to promote metastasis and oxaliplatin resistance by sponging miR-136 in colorectal cancer, OncoTargets Ther, vol.10, pp.205-216, 2017.

D. A. Gibson and P. T. Saunders, Estrogen dependent signaling in reproductive tissues -a role for estrogen receptors and estrogen related receptors, Mol. Cell. Endocrinol, vol.348, pp.361-372, 2012.

V. Giguère, To ERR in the estrogen pathway, Trends Endocrinol. Metab. TEM, vol.13, pp.220-225, 2002.

V. Giguère, Transcriptional control of energy homeostasis by the estrogen-related receptors, Endocr. Rev, vol.29, pp.677-696, 2008.

V. Giguère, N. Yang, P. Segui, and R. M. Evans, Identification of a new class of steroid hormone receptors, Nature, vol.331, pp.91-94, 1988.

C. Ginestier, M. H. Hur, E. Charafe-jauffret, F. Monville, J. Dutcher et al., ALDH1 is a marker of normal and malignant human mammary stem cells and a predictor of poor clinical outcome, Cell Stem Cell, vol.1, pp.555-567, 2007.
URL : https://hal.archives-ouvertes.fr/hal-01431968

C. Ginestier, H. Korkaya, G. Dontu, D. Birnbaum, M. Wicha et al., The cancer stem cell: The breast cancer driver. médecine/sciences 23, pp.1133-1142, 2008.
URL : https://hal.archives-ouvertes.fr/hal-01431965

C. Giordano, Y. Cui, I. Barone, S. Ando, M. A. Mancini et al., , 2010.

, Growth factor-induced resistance to tamoxifen is associated with a mutation of estrogen receptor alpha and its phosphorylation at serine 305, Breast Cancer Res. Treat, vol.119, pp.71-85

W. Gong, J. Yu, Q. Wang, S. Li, J. Song et al., Estrogen-related receptor (ERR) protects against puromycin aminonucleoside-induced podocyte apoptosis by targeting PI3K/Akt signaling, Int. J. Biochem. Cell Biol, vol.78, pp.75-86, 2016.

E. Gonzalez-de-valdivia, S. Broselid, R. Kahn, B. Olde, and L. M. Leeb-lundberg, G protein-coupled estrogen receptor 1 (GPER1)/GPR30 increases ERK1/2 activity through PDZ motifdependent and -independent mechanisms, J. Biol. Chem, vol.292, pp.9932-9943, 2017.

G. Goode, B. R. Ballard, H. C. Manning, M. L. Freeman, Y. Kang et al., , 2013.

, Knockdown of aberrantly upregulated aryl hydrocarbon receptor reduces tumor growth and metastasis of MDA-MB-231 human breast cancer cell line, Int. J. Cancer J. Int. Cancer, vol.133, pp.2769-2780

P. E. Goss, J. N. Ingle, K. I. Pritchard, N. J. Robert, H. Muss et al., Extending Aromatase-Inhibitor Adjuvant Therapy to 10 Years, N. Engl. J. Med, vol.375, pp.209-219, 2016.

K. F. Grandien, A. Berkenstam, S. Nilsson, and J. A. Gustafsson, Localization of DNase I hypersensitive sites in the human oestrogen receptor gene correlates with the transcriptional activity of two differentially used promoters, J. Mol. Endocrinol, vol.10, pp.269-277, 1993.

G. L. Greene, P. Gilna, M. Waterfield, A. Baker, Y. Hort et al., Sequence and expression of human estrogen receptor complementary DNA, Science, vol.231, pp.1150-1154, 1986.

C. J. Gruber, W. Tschugguel, C. Schneeberger, and J. C. Huber, Production and Actions of Estrogens, N. Engl. J. Med, vol.346, pp.340-352, 2002.

T. Gudjonsson, M. C. Adriance, M. D. Sternlicht, O. W. Petersen, and M. J. Bissell, , 2005.

, Myoepithelial Cells: Their Origin and Function in Breast Morphogenesis and Neoplasia, J. Mammary Gland Biol. Neoplasia, vol.10, pp.261-272

M. Guedj, L. Marisa, A. De-reynies, B. Orsetti, R. Schiappa et al., A refined molecular taxonomy of breast cancer, Oncogene, vol.31, pp.1196-1206, 2012.
URL : https://hal.archives-ouvertes.fr/inserm-00611752

S. Guo and G. E. Sonenshein, Forkhead box transcription factor FOXO3a regulates estrogen receptor alpha expression and is repressed by the Her-2/neu/phosphatidylinositol 3-kinase/Akt signaling pathway, Mol. Cell. Biol, vol.24, pp.8681-8690, 2004.

T. Guo and G. Pan, MicroRNA-136 functions as a tumor suppressor in osteosarcoma via regulating metadherin, Cancer Biomark. Sect. Dis. Markers, vol.22, pp.79-87, 2018.

M. Guo, M. Wang, H. Deng, X. Zhang, and Z. Wang, A novel anticancer agent Broussoflavonol B downregulates estrogen receptor (ER)-?36 expression and inhibits growth of ERnegative breast cancer MDA-MB-231 cells, Eur. J. Pharmacol, vol.714, pp.56-64, 2013.

Y. Guo, P. Yu, Z. Liu, Y. Maimaiti, C. Chen et al.,

, Prognostic and clinicopathological value of GATA binding protein 3 in breast cancer: A systematic review and meta-analysis, PLOS ONE, vol.12, 174843.

B. A. Gusterson and T. Stein, Human breast development, Semin. Cell Dev. Biol, vol.23, pp.567-573, 2012.

M. Häggström and D. Richfield, Diagram of the pathways of human steroidogenesis, WikiJournal Med, vol.1, p.5, 2014.

L. Haldosén, C. Zhao, and K. Dahlman-wright, Estrogen receptor beta in breast cancer, Mol. Cell. Endocrinol, vol.382, pp.665-672, 2014.

D. Hanahan and R. A. Weinberg, Hallmarks of Cancer: The Next Generation, Cell, vol.144, pp.646-674, 2011.

H. Hao, M. Salazar, K. M. Kelley, A. Shatnawi, S. Mukherjee et al., Estrogen-induced and TAFII30-mediated gene repression by direct recruitment of the estrogen receptor and co-repressors to the core promoter and its reversal by tamoxifen, Oncogene, vol.26, pp.7872-7884, 2007.

S. M. Hawkins and M. M. Matzuk, Menstrual Cycle: Basic Biology, Ann. N. Y. Acad. Sci, vol.1135, pp.10-18, 2008.

T. He, A. Zuo, J. Wang, and P. Zhao, Organochlorine pesticides accumulation and breast cancer: A hospital-based case-control study, Tumour Biol. J. Int. Soc. Oncodevelopmental Biol, 2017.

M. M. Heckler, H. Thakor, C. C. Schafer, and R. B. Riggins, ERK/MAPK regulates ERR expression, transcriptional activity and receptor-mediated tamoxifen resistance in ER+ breast cancer, FEBS J, vol.281, pp.2431-2442, 2014.

D. M. Heery, E. Kalkhoven, S. Hoare, and M. G. Parker, A signature motif in transcriptional co-activators mediates binding to nuclear receptors, Nature, vol.387, pp.733-736, 1997.

N. Heldring, T. Pawson, D. Mcdonnell, E. Treuter, J. Gustafsson et al., , 2007.

, Structural Insights into Corepressor Recognition by Antagonist-bound Estrogen Receptors, J. Biol. Chem, vol.282, pp.10449-10455

C. Hennequin and A. Fourquet, Controverse sur l'irradiation de la chaîne mammaire interne dans le cancer du sein, Cancer/radiothérapie, vol.18, pp.351-355, 2014.

J. R. Hens and J. J. Wysolmerski, Key stages of mammary gland development: Molecular mechanisms involved in the formation of the embryonic mammary gland, Breast Cancer Res, vol.7, pp.220-224, 2005.

L. Hilakivi-clarke, Maternal exposure to diethylstilbestrol during pregnancy and increased breast cancer risk in daughters, Breast Cancer Res. BCR, vol.16, p.208, 2014.

C. F. Holinka, E. Diczfalusy, C. Bennink, and H. J. , Estetrol: a unique steroid in human pregnancy, J. Steroid Biochem. Mol. Biol, vol.110, pp.138-143, 2008.

F. Holst, Estrogen receptor alpha gene amplification in breast cancer: 25 years of debate, World J. Clin. Oncol, vol.7, pp.160-173, 2016.

H. Hong, L. Yang, and M. R. Stallcup, Hormone-independent transcriptional activation and coactivator binding by novel orphan nuclear receptor ERR3, J. Biol. Chem, vol.274, pp.22618-22626, 1999.

N. Honma, R. Horii, T. Iwase, S. Saji, M. Younes et al., Clinical importance of estrogen receptor-beta evaluation in breast cancer patients treated with adjuvant tamoxifen therapy, J. Clin. Oncol. Off. J. Am. Soc. Clin. Oncol, vol.26, pp.3727-3734, 2008.

T. A. Hopp, H. L. Weiss, I. S. Parra, Y. Cui, C. K. Osborne et al., Low levels of estrogen receptor beta protein predict resistance to tamoxifen therapy in breast cancer, Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res, vol.10, pp.7490-7499, 2004.

B. Horard and J. Vanacker, Estrogen receptor-related receptors: orphan receptors desperately seeking a ligand, J. Mol. Endocrinol, vol.31, pp.349-357, 2003.

L. Hsu, N. Chu, Y. Lin, and S. Kao, G-Protein Coupled Estrogen Receptor in Breast Cancer, Int. J. Mol. Sci, vol.20, p.306, 2019.

H. Hua, H. Zhang, Q. Kong, and Y. Jiang, Mechanisms for estrogen receptor expression in human cancer, Exp. Hematol. Oncol, vol.7, 2018.

J. Huan, L. Xing, Q. Lin, H. Xui, and X. Qin, Long noncoding RNA CRNDE activates Wnt/?catenin signaling pathway through acting as a molecular sponge of microRNA-136 in human breast cancer, Am. J. Transl. Res, vol.9, pp.1977-1989, 2017.

K. Hughes and C. J. Watson, The Multifaceted Role of STAT3 in Mammary Gland Involution and Breast Cancer, Int. J. Mol. Sci, vol.19, 2018.

A. Hurtado, K. A. Holmes, T. R. Geistlinger, I. R. Hutcheson, R. I. Nicholson et al., Regulation of ERBB2 by oestrogen receptor-PAX2 determines response to tamoxifen, Nature, vol.456, pp.663-666, 2008.

N. H. Ing, D. A. Massuto, and L. A. Jaeger, Estradiol up-regulates AUF1p45 binding to stabilizing regions within the 3'-untranslated region of estrogen receptor alpha mRNA, J. Biol. Chem, vol.283, pp.1764-1772, 2008.

W. V. Ingman and S. A. Robertson, Mammary gland development in transforming growth factor beta1 null mutant mice: systemic and epithelial effects, Biol. Reprod, vol.79, pp.711-717, 2008.

J. L. Inman, C. Robertson, J. D. Mott, and M. J. Bissell, Mammary gland development: cell fate specification, stem cells and the microenvironment, Dev. Camb. Engl, vol.142, pp.1028-1042, 2015.

X. Jacq, C. Brou, Y. Lutz, I. Davidson, P. Chambon et al., Human TAFII30 is present in a distinct TFIID complex and is required for transcriptional activation by the estrogen receptor, Cell, vol.79, pp.107-117, 1994.

A. Javed and A. Lteif, Development of the Human Breast, Semin. Plast. Surg, vol.27, pp.5-12, 2013.

K. Jéhannin-ligier, Projection de l'incidence et de la mortalité par cancer en France métropolitaine en 2017, 2017.

E. V. Jensen, On the mechanism of estrogen action, Perspect. Biol. Med, vol.6, pp.47-59, 1962.

R. Jeselsohn, G. Buchwalter, C. De-angelis, M. Brown, and R. Schiff, ESR1 mutations as a mechanism for acquired endocrine resistance in breast cancer, Nat. Rev. Clin. Oncol, vol.12, pp.573-583, 2015.

R. Jeselsohn, J. S. Bergholz, M. Pun, M. Cornwell, W. Liu et al., Allele-Specific Chromatin Recruitment and Therapeutic Vulnerabilities of ESR1 Activating Mutations, Cancer Cell, vol.33, pp.173-186, 2018.

M. Jia, K. Dahlman-wright, and J. Gustafsson, Estrogen receptor alpha and beta in health and disease, Best Pract. Res. Clin. Endocrinol. Metab, vol.29, pp.557-568, 2015.

H. Jiang, R. Teng, Q. Wang, X. Zhang, H. Wang et al., Transcriptional analysis of estrogen receptor alpha variant mRNAs in colorectal cancers and their matched normal colorectal tissues, J. Steroid Biochem. Mol. Biol, vol.112, pp.20-24, 2008.

Y. Jiang, Y. E. Liu, A. Lu, A. Gupta, I. D. Goldberg et al., Stimulation of estrogen receptor signaling by gamma synuclein, Cancer Res, vol.63, pp.3899-3903, 2003.

N. Josso, R. , and R. , La cellule de Sertoli, une cellule endocrine. médecine/sciences 11, 1995.

M. Kagami, M. J. O'sullivan, A. J. Green, Y. Watabe, O. Arisaka et al., The IG-DMR and the MEG3-DMR at Human Chromosome 14q32.2: Hierarchical Interaction and Distinct Functional Properties as Imprinting Control Centers, PLoS Genet, vol.6, 2010.

D. Kalaitzidis and T. D. Gilmore, Transcription factor cross-talk: the estrogen receptor and NF-?B, Trends Endocrinol. Metab, vol.16, pp.46-52, 2005.

B. T. Kalet, S. R. Anglin, A. Handschy, L. E. O'donoghue, C. Halsey et al., Transcription factor Ets1 cooperates with estrogen receptor ? to stimulate estradioldependent growth in breast cancer cells and tumors, PloS One, vol.8, p.68815, 2013.

L. Kang, X. Zhang, Y. Xie, Y. Tu, D. Wang et al., Involvement of Estrogen Receptor Variant ER-?36, Not GPR30, Nongenomic Estrogen Signaling. Mol. Endocrinol, vol.24, pp.709-721, 2010.

L. Kang, Y. Guo, X. Zhang, J. Meng, and Z. Wang, A positive cross-regulation of HER2 and ER-?36 controls ALDH1 positive breast cancer cells, J. Steroid Biochem. Mol. Biol, vol.127, pp.262-268, 2011.

L. Kang, L. Wang, and Z. Wang, Opposite regulation of estrogen receptor-? and its variant ER-?36 by the Wilms' tumor suppressor WT1, Oncol. Lett, vol.2, pp.337-341, 2011.

I. Kastrati, S. Semina, B. Gordon, and E. Smart, Insights into how phosphorylation of estrogen receptor at serine 305 modulates tamoxifen activity in breast cancer, Mol. Cell. Endocrinol, vol.483, pp.97-101, 2019.

M. Keaveney, J. Klug, M. T. Dawson, P. V. Nestor, J. G. Neilan et al., , 1991.

D. Khan, A. Ahmed, and S. , The Immune System Is a Natural Target for Estrogen Action: Opposing Effects of Estrogen in Two Prototypical Autoimmune Diseases, Front. Immunol, vol.6, 2016.

S. Khosla, M. J. Oursler, and D. G. Monroe, Estrogen and the skeleton, Trends Endocrinol. Metab. TEM, vol.23, pp.576-581, 2012.

J. Kim, L. N. Petz, Y. S. Ziegler, J. R. Wood, S. J. Potthoff et al., Regulation of the estrogen-responsive pS2 gene in MCF-7 human breast cancer cells, J. Steroid Biochem. Mol. Biol, vol.74, pp.157-168, 2000.

V. Kirn, L. Strake, F. Thangarajah, L. Richters, H. Eischeid et al., ESR1-promoter-methylation status in primary breast cancer and its corresponding metastases, Clin. Exp. Metastasis, vol.35, pp.707-712, 2018.

L. Klein-hitpass, M. Kaling, and G. U. Ryffel, Synergism of closely adjacent estrogenresponsive elements increases their regulatory potential, J. Mol. Biol, vol.201, pp.537-544, 1988.

C. M. Klinge, Estrogen receptor interaction with estrogen response elements, Nucleic Acids Res, vol.29, pp.2905-2919, 2001.

M. Kok, C. Holm-wigerup, M. Hauptmann, R. Michalides, O. Stål et al., Estrogen receptor-alpha phosphorylation at serine-118 and tamoxifen response in breast cancer, J. Natl. Cancer Inst, vol.101, pp.1725-1729, 2009.

S. L. Kong, G. Li, S. L. Loh, W. Sung, and E. T. Liu, Cellular reprogramming by the conjoint action of ER?, FOXA1, and GATA3 to a ligand-inducible growth state, Mol. Syst. Biol, vol.7, p.526, 2011.

M. Kos, G. Reid, S. Denger, and F. Gannon, Minireview: genomic organization of the human ERalpha gene promoter region, Mol. Endocrinol. Baltim. Md, vol.15, pp.2057-2063, 2001.

H. Kouros-mehr, E. M. Slorach, M. D. Sternlicht, and Z. Werb, GATA-3 maintains the differentiation of the luminal cell fate in the mammary gland, Cell, vol.127, pp.1041-1055, 2006.

S. Kovats, Estrogen receptors regulate innate immune cells and signaling pathways, Cell. Immunol, vol.294, pp.63-69, 2015.

V. Krishnan, W. Porter, M. Santostefano, X. Wang, and S. Safe, Molecular mechanism of inhibition of estrogen-induced cathepsin D gene expression by 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) in MCF-7 cells, Mol. Cell. Biol, vol.15, pp.6710-6719, 1995.

G. G. Kuiper, E. Enmark, M. Pelto-huikko, S. Nilsson, and J. A. Gustafsson, Cloning of a novel receptor expressed in rat prostate and ovary, Proc. Natl. Acad. Sci. U. S. A, vol.93, pp.5925-5930, 1996.

R. Kumar and E. B. Thompson, The structure of the nuclear hormone receptors, Steroids, vol.64, pp.310-319, 1999.

R. Kumar, M. N. Zakharov, S. H. Khan, R. Miki, H. Jang et al., The Dynamic Structure of the Estrogen Receptor, J. Amino Acids, 2011.

N. J. Lakhani, M. A. Sarkar, J. Venitz, and W. D. Figg, 2-Methoxyestradiol, a promising anticancer agent, Pharmacotherapy, vol.23, pp.165-172, 2003.

V. Laudet, Evolution of the nuclear receptor superfamily: early diversification from an ancestral orphan receptor, J. Mol. Endocrinol, vol.19, pp.207-226, 1997.

J. Le-quesne and C. Caldas, Micro-RNAs and breast cancer, Mol. Oncol, vol.4, pp.230-241, 2010.

L. Romancer, M. Poulard, C. Cohen, P. Sentis, S. Renoir et al., Cracking the estrogen receptor's posttranslational code in breast tumors, Endocr. Rev, vol.32, pp.597-622, 2011.

E. Y. Lee and W. J. Muller, Oncogenes and Tumor Suppressor Genes, Cold Spring Harb. Perspect. Biol, vol.2, 2010.

T. I. Lee, Y. , and R. A. , Transcription of eukaryotic protein-coding genes, Annu. Rev. Genet, vol.34, pp.77-137, 2000.

D. Li, X. Fei, Y. Dong, C. Cheng, Y. Yang et al., The long non-coding RNA CRNDE acts as a ceRNA and promotes glioma malignancy by preventing miR-136-5p-mediated downregulation of Bcl-2 and Wnt2, Oncotarget, 2017.

L. Li, M. P. Haynes, and J. R. Bender, Plasma membrane localization and function of the estrogen receptor alpha variant (ER46) in human endothelial cells, Proc. Natl. Acad. Sci. U. S. A, vol.100, pp.4807-4812, 2003.

T. Li, X. Gao, L. Gao, B. Gan, Z. Xie et al., Role of upregulated miR-136-5p in lung adenocarcinoma: A study of 1242 samples utilizing bioinformatics analysis, Pathol. Res. Pract, vol.214, pp.750-766, 2018.

M. A. Lillo-osuna, J. Garcia-lopez, I. El-ayachi, I. Fatima, A. B. Khalid et al., Activation of Estrogen Receptor Alpha by Decitabine Inhibits Osteosarcoma Growth and Metastasis, Cancer Res, vol.79, pp.1054-1068, 2019.

A. H. Lin, R. W. Li, E. Y. Ho, G. P. Leung, S. W. Leung et al.,

, Differential Ligand Binding Affinities of Human Estrogen Receptor-? Isoforms, PLoS ONE, vol.8

H. Lin, T. Dai, H. Xiong, X. Zhao, X. Chen et al., , 2010.

, Unregulated miR-96 Induces Cell Proliferation in Human Breast Cancer by Downregulating Transcriptional Factor FOXO3a, PLoS ONE, vol.5

S. Lin, L. Yan, X. Zhang, J. Yuan, M. Li et al., , 2010.

, ER-?36, a Variant of ER-?, Promotes Tamoxifen Agonist Action in Endometrial Cancer Cells via the MAPK/ERK and PI3K/Akt Pathways, PLoS ONE, vol.5

Z. Lin, P. Yin, S. Reierstad, M. O'halloran, V. J. Coon et al., Adenosine A1 receptor, a target and regulator of estrogen receptoralpha action, mediates the proliferative effects of estradiol in breast cancer, Oncogene, vol.29, pp.1114-1122, 2010.

Y. Lipovka and J. P. Konhilas, The complex nature of oestrogen signalling in breast cancer: enemy or ally?, Biosci. Rep, vol.36, 2016.

Y. E. Liu, W. Pu, Y. Jiang, D. Shi, R. Dackour et al., Chaperoning of estrogen receptor and induction of mammary gland proliferation by neuronal protein synuclein gamma, Oncogene, vol.26, pp.2115-2125, 2007.

W. Lou, J. Liu, B. Ding, L. Xu, F. et al., Identification of chemoresistance-associated miRNAs in breast cancer, Cancer Manag. Res, vol.10, pp.4747-4757, 2018.

S. Low, H. Zembutsu, and Y. Nakamura, Breast cancer: The translation of big genomic data to cancer precision medicine, Cancer Sci, vol.109, pp.497-506, 2018.

Q. Lu, D. C. Pallas, H. K. Surks, W. E. Baur, M. E. Mendelsohn et al., Striatin assembles a membrane signaling complex necessary for rapid, nongenomic activation of endothelial NO synthase by estrogen receptor alpha, Proc. Natl. Acad. Sci. U. S. A, vol.101, pp.17126-17131, 2004.

B. Ma and M. O. Hottiger, Crosstalk between Wnt/?-Catenin and NF-?B Signaling Pathway during, Inflammation. Front. Immunol, vol.7, 2016.

H. Macias and L. Hinck, Mammary Gland Development, Wiley Interdiscip. Rev. Dev. Biol, vol.1, pp.533-557, 2012.

P. A. Madureira, R. Varshochi, D. Constantinidou, R. E. Francis, R. C. Coombes et al., The Forkhead box M1 protein regulates the transcription of the estrogen receptor alpha in breast cancer cells, J. Biol. Chem, vol.281, pp.25167-25176, 2006.

R. Maekawa, S. Sato, M. Okada, L. Lee, I. Tamura et al., Tissue-Specific Expression of Estrogen Receptor 1 Is Regulated by DNA Methylation in a T-DMR, Mol. Endocrinol. Baltim. Md, vol.30, pp.335-347, 2016.

L. Magnani, E. B. Ballantyne, X. Zhang, and M. Lupien, PBX1 Genomic Pioneer Function Drives ER? Signaling Underlying Progression in Breast Cancer, PLOS Genet, vol.7, p.1002368, 2011.

G. K. Malhotra, X. Zhao, H. Band, and V. Band, Histological, molecular and functional subtypes of breast cancers, Cancer Biol. Ther, vol.10, pp.955-960, 2010.

O. Maller, H. Martinson, and P. Schedin, Extracellular matrix composition reveals complex and dynamic stromal-epithelial interactions in the mammary gland, J. Mammary Gland Biol. Neoplasia, vol.15, pp.301-318, 2010.

B. Manavathi, O. Dey, V. N. Gajulapalli, R. S. Bhatia, S. Bugide et al., Derailed Estrogen Signaling and Breast Cancer: An Authentic Couple, Endocr. Rev, vol.34, pp.1-32, 2013.

M. Manikkam, C. Guerrero-bosagna, R. Tracey, M. M. Haque, and M. K. Skinner, , 2012.

A. Markiewicz, M. We?nicka-ja?kiewicz, J. Skokowski, J. Ja?kiewicz, J. Szade et al., Prognostic significance of ESR1 amplification and ESR1 PvuII, CYP2C19*2, UGT2B15*2 polymorphisms in breast cancer patients, PloS One, vol.8, 2013.

L. L. Marotta, V. Almendro, A. Marusyk, M. Shipitsin, J. Schemme et al., The JAK2/STAT3 signaling pathway is required for growth of CD44+CD24-stem cell-like breast cancer cells in human tumors, J. Clin. Invest, vol.121, pp.2723-2735, 2011.

W. A. Marshall and J. M. Tanner, Variations in pattern of pubertal changes in girls, Arch. Dis. Child, vol.44, pp.291-303, 1969.

J. Matthews and J. Gustafsson, Estrogen receptor and aryl hydrocarbon receptor signaling pathways, Nucl. Recept, 2006.

J. R. Matthews, C. , and A. R. , , 2005.

J. Mcbryan, J. Howlin, S. Napoletano, M. , and F. , Amphiregulin: role in mammary gland development and breast cancer, J. Mammary Gland Biol. Neoplasia, vol.13, pp.159-169, 2008.

I. J. Mcewan, The Nuclear Receptor Superfamily at Thirty, Methods Mol. Biol. Clifton NJ, vol.1443, pp.3-9, 2016.

L. A. Mcpherson and R. J. Weigel, AP2alpha and AP2gamma: a comparison of binding site specificity and trans-activation of the estrogen receptor promoter and single site promoter constructs, Nucleic Acids Res, vol.27, pp.4040-4049, 1999.

L. A. Mcpherson, V. R. Baichwal, and R. J. Weigel, Identification of ERF-1 as a member of the AP2 transcription factor family, Proc. Natl. Acad. Sci. U. S. A, vol.94, pp.4342-4347, 1997.

R. Mehra, S. Varambally, L. Ding, R. Shen, M. S. Sabel et al., Identification of GATA3 as a breast cancer prognostic marker by global gene expression meta-analysis, Cancer Res, vol.65, pp.11259-11264, 2005.

L. P. Menasce, G. R. White, C. J. Harrison, and J. M. Boyle, Localization of the estrogen receptor locus (ESR) to chromosome 6q25.1 by FISH and a simple post-FISH banding technique, Genomics, vol.17, pp.263-265, 1993.

M. E. Menezes, S. K. Das, L. Emdad, J. J. Windle, X. Y. Wang et al., Genetically engineered mice as experimental tools to dissect the critical events in breast cancer, Adv. Cancer Res, vol.121, pp.331-382, 2014.

K. M. Menon and A. Goldstrohm, Post-transcriptional Mechanisms in Endocrine Regulation, 2015.

J. L. Merlin, Les inhibiteurs de tyrosine kinase en oncologie -Tyrosine kinase inhibitors in oncology, vol.22, pp.51-62, 2008.

L. Meunier, B. Siddeek, A. Vega, N. Lakhdari, L. Inoubli et al., Perinatal programming of adult rat germ cell death after exposure to xenoestrogens: role of microRNA miR-29 family in the down-regulation of DNA methyltransferases and Mcl-1, Endocrinology, vol.153, pp.1936-1947, 2012.

K. C. Miranda, T. Huynh, Y. Tay, Y. Ang, W. Tam et al., A pattern-based method for the identification of MicroRNA binding sites and their corresponding heteroduplexes, Cell, vol.126, pp.1203-1217, 2006.

A. Misawa and S. Inoue, Estrogen-Related Receptors in Breast Cancer and Prostate Cancer, Front. Endocrinol, vol.6, p.83, 2015.

H. Mohammed, I. A. Russell, R. Stark, O. M. Rueda, T. E. Hickey et al., Progesterone receptor modulates ER? action in breast cancer, Nature, vol.523, pp.313-317, 2015.

J. Mohandass, S. Ravichandran, K. Srilakshmi, C. P. Rajadurai, S. Sanmugasamy et al.,

, BCDB A database for breast cancer research and information. Bioinformation, vol.5, pp.1-3

S. Morales, M. Monzo, and A. Navarro, Epigenetic regulation mechanisms of microRNA expression, Biomol. Concepts, vol.8, 2017.

K. Moriarty, K. H. Kim, and J. R. Bender, Estrogen Receptor-Mediated Rapid Signaling, Endocrinology, vol.147, pp.5557-5563, 2006.

S. Mosselman, J. Polman, and R. Dijkema, ER beta: identification and characterization of a novel human estrogen receptor, FEBS Lett, vol.392, pp.49-53, 1996.

M. Moumen, A. Chiche, S. Cagnet, V. Petit, K. Raymond et al., The mammary myoepithelial cell, Int. J. Dev. Biol, vol.55, pp.763-771, 2011.

S. O. Mueller, J. A. Clark, P. H. Myers, and K. S. Korach, Mammary gland development in adult mice requires epithelial and stromal estrogen receptor alpha, Endocrinology, vol.143, pp.2357-2365, 2002.

M. Muñoz-de-toro, C. M. Markey, P. R. Wadia, E. H. Luque, B. S. Rubin et al., Perinatal Exposure to Bisphenol-A Alters Peripubertal Mammary Gland Development in Mice, Endocrinology, vol.146, pp.4138-4147, 2005.

E. A. Musgrove and R. L. Sutherland, Biological determinants of endocrine resistance in breast cancer, Nat. Rev. Cancer, vol.9, pp.631-643, 2009.

A. Nagel, J. Szade, M. Iliszko, J. Elzanowska, M. Welnicka-jaskiewicz et al., Clinical and Biological Significance of ESR1 Gene Alteration and Estrogen Receptors Isoforms Expression in Breast Cancer Patients, Int. J. Mol. Sci, vol.20, 2019.

A. Naimi, P. Rajabi, H. Ghasemibasir, A. Eftekhari, and F. Mohammadizadeh, Expression of basal and luminal cytokeratins in breast cancer and their correlation with clinicopathological prognostic variables, Indian J. Med. Sci, vol.63, p.152, 2009.

O. Najim, M. Huizing, K. Papadimitriou, X. B. Trinh, P. Pauwels et al., The prevalence of estrogen receptor-1 mutation in advanced breast cancer: The estrogen receptor one study (EROS1), Cancer Treat. Res. Commun, vol.19, p.100123, 2019.

A. Nebbioso, F. P. Tambaro, C. Dell'aversana, A. , and L. , Cancer epigenetics: Moving forward, PLoS Genet, vol.14, p.1007362, 2018.

E. F. Need, V. Atashgaran, W. V. Ingman, and P. Dasari, Hormonal regulation of the immune microenvironment in the mammary gland, J. Mammary Gland Biol. Neoplasia, vol.19, pp.229-239, 2014.

K. W. Nettles, G. Gil, J. Nowak, R. Métivier, V. B. Sharma et al., CBP Is a dosage-dependent regulator of nuclear factor-kappaB suppression by the estrogen receptor, Mol. Endocrinol. Baltim. Md, vol.22, pp.263-272, 2008.

M. C. Neville, T. B. Mcfadden, and I. Forsyth, Hormonal Regulation of Mammary Differentiation and Milk Secretion, J. Mammary Gland Biol. Neoplasia, 2002.

E. E. Nilsson, M. D. Anway, J. Stanfield, and M. K. Skinner, Transgenerational epigenetic effects of the endocrine disruptor vinclozolin on pregnancies and female adult onset disease, Reprod. Camb. Engl, vol.135, pp.713-721, 2008.

A. M. Norfleet, M. L. Thomas, B. Gametchu, and C. S. Watson, , 1999.

T. Obsil and V. Obsilova, Structure/function relationships underlying regulation of FOXO transcription factors, Oncogene, vol.27, pp.2263-2275, 2008.

S. Ogawa, S. Inoue, T. Watanabe, A. Orimo, T. Hosoi et al., , 1998.

, Molecular cloning and characterization of human estrogen receptor betacx: a potential inhibitor ofestrogen action in human, Nucleic Acids Res, vol.26, pp.3505-3512

R. O'lone, M. C. Frith, E. K. Karlsson, and U. Hansen, Genomic Targets of Nuclear Estrogen Receptors, Mol. Endocrinol, vol.18, pp.1859-1875, 2004.

S. Omarjee, J. Jacquemetton, C. Poulard, N. Rochel, A. Dejaegere et al., The molecular mechanisms underlying the ER?-36-mediated signaling in breast cancer, Oncogene, vol.36, pp.2503-2514, 2017.

Y. Omoto and H. Iwase, , 2015.

A. C. Panda, Circular RNAs Act as miRNA Sponges, Adv. Exp. Med. Biol, vol.1087, pp.67-79, 2018.

D. P. Pandey and D. Picard, miR-22 inhibits estrogen signaling by directly targeting the estrogen receptor alpha mRNA, Mol. Cell. Biol, vol.29, pp.3783-3790, 2009.

M. Panneerselvam, K. Muthu, and K. Ramadas, , 2015.

, Mol. Biosyst, vol.11, pp.2998-3010

B. Panwar, G. S. Omenn, and Y. Guan, miRmine: a database of human miRNA expression profiles, Bioinforma. Oxf. Engl, vol.33, pp.1554-1560, 2017.

A. Pavlopoulou, D. A. Spandidos, and I. Michalopoulos, Human cancer databases (Review), 2015.

, Oncol. Rep, vol.33, pp.3-18

J. Pellikainen, A. Naukkarinen, K. Ropponen, J. Rummukainen, V. Kataja et al., Expression of HER2 and its association with AP-2 in breast cancer, Eur. J. Cancer, vol.40, pp.1485-1495, 2004.

L. Penolazzi, E. Lambertini, S. Giordano, V. Sollazzo, G. Traina et al., , 2004.

, Methylation analysis of the promoter F of estrogen receptor alpha gene: effects on the level of transcription on human osteoblastic cells, J. Steroid Biochem. Mol. Biol, vol.91, pp.1-9

C. M. Perou, T. Sørlie, M. B. Eisen, M. Van-de-rijn, S. S. Jeffrey et al., Molecular portraits of human breast tumours, Nature, vol.406, pp.747-752, 2000.

A. A. Peters, G. Buchanan, C. Ricciardelli, T. Bianco-miotto, M. M. Centenera et al., Androgen receptor inhibits estrogen receptor-alpha activity and is prognostic in breast cancer, Cancer Res, vol.69, pp.6131-6140, 2009.

R. J. Pietras and C. M. Szego, Endometrial cell calcium and oestrogen action, Nature, vol.253, p.357, 1975.

L. Piggott, A. Silva, T. Robinson, A. Santiago-gómez, B. M. Simões et al., Acquired Resistance of ER-Positive Breast Cancer to Endocrine Treatment Confers an Adaptive Sensitivity to TRAIL through Posttranslational Downregulation of c-FLIP, Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res, vol.24, pp.2452-2463, 2018.

M. Ponglikitmongkol, S. Green, C. , and P. , Genomic organization of the human oestrogen receptor gene, EMBO J, vol.7, pp.3385-3388, 1988.

C. Poulard, J. Jacquemetton, O. Trédan, P. A. Cohen, J. Vendrell et al., Oestrogen Non-Genomic Signalling is Activated in Tamoxifen-Resistant Breast Cancer, Int. J. Mol. Sci, vol.20, 2019.

J. B. Powell, G. D. Goode, and S. E. Eltom, The Aryl Hydrocarbon Receptor: A Target for Breast Cancer Therapy, J. Cancer Ther, vol.4, pp.1177-1186, 2013.

W. B. Pratt and D. O. Toft, Regulation of signaling protein function and trafficking by the hsp90/hsp70-based chaperone machinery, Exp. Biol. Med. Maywood NJ, vol.228, pp.111-133, 2003.

P. Pryzbylkowski, O. Obajimi, and J. C. Keen, Trichostatin A and 5 Aza-2' deoxycytidine decrease estrogen receptor mRNA stability in ER positive MCF7 cells through modulation of HuR, Breast Cancer Res. Treat, vol.111, pp.15-25, 2008.

M. Pupo, A. Vivacqua, I. Perrotta, A. Pisano, S. Aquila et al., The nuclear localization signal is required for nuclear GPER translocation and function in breast Cancer-Associated Fibroblasts (CAFs), Mol. Cell. Endocrinol, vol.376, pp.23-32, 2013.

M. Pupo, M. Maggiolini, and A. M. Musti, GPER Mediates Non-Genomic Effects of Estrogen, Methods Mol. Biol. Clifton NJ, vol.1366, pp.471-488, 2016.

M. Pupo, A. Bodmer, M. Berto, M. Maggiolini, P. Dietrich et al., A genetic polymorphism repurposes the G-protein coupled and membrane-associated estrogen receptor GPER to a transcription factor-like molecule promoting paracrine signaling between stroma and breast carcinoma cells, Oncotarget, vol.8, pp.46728-46744, 2017.

M. Puzianowska-kuznicka, E. Pawlik-pachucka, M. Owczarz, M. Budzi?ska, and J. Polosak, , 2013.

, Small-Molecule Hormones: Molecular Mechanisms of Action, Int. J. Endocrinol, 2013.

H. Qian, J. Xuan, Y. Liu, and G. Shi, Function of G-Protein-Coupled Estrogen Receptor-1 in Reproductive System Tumors, J. Immunol. Res, 2016.

J. Rao, X. Jiang, Y. Wang, C. , and B. , Advances in the understanding of the structure and function of ER-?36,a novel variant of human estrogen receptor-alpha, J. Steroid Biochem. Mol. Biol, vol.127, pp.231-237, 2011.

M. Razandi, A. Pedram, E. M. Rosen, and E. R. Levin, BRCA1 Inhibits Membrane Estrogen and Growth Factor Receptor Signaling to Cell Proliferation in Breast Cancer, Mol. Cell. Biol, vol.24, pp.5900-5913, 2004.

T. R. Rebbeck, N. D. Kauff, and S. M. Domchek, Meta-analysis of risk reduction estimates associated with risk-reducing salpingo-oophorectomy in BRCA1 or BRCA2 mutation carriers, J. Natl. Cancer Inst, vol.101, pp.80-87, 2009.

G. Reid, M. R. Hübner, R. Métivier, H. Brand, S. Denger et al., Cyclic, proteasome-mediated turnover of unliganded and liganded ERalpha on responsive promoters is an integral feature of estrogen signaling, Mol. Cell, vol.11, pp.695-707, 2003.

C. M. Revankar, D. F. Cimino, L. A. Sklar, J. B. Arterburn, and E. R. Prossnitz, A Transmembrane Intracellular Estrogen Receptor Mediates Rapid Cell Signaling, vol.307, p.6, 2005.

F. Reyal, C. Guyader, C. Decraene, C. Lucchesi, N. Auger et al., Molecular profiling of patient-derived breast cancer xenografts, Breast Cancer Res, vol.14, p.11, 2012.
URL : https://hal.archives-ouvertes.fr/inserm-00675301

R. Rezaei, Z. Wu, Y. Hou, F. W. Bazer, and G. Wu, Amino acids and mammary gland development: nutritional implications for milk production and neonatal growth, J. Anim. Sci. Biotechnol, vol.7, 2016.

A. Reznikov, Hormonal impact on tumor growth and progression, Exp. Oncol, vol.37, pp.162-172, 2015.

G. W. Robinson, Cooperation of signalling pathways in embryonic mammary gland development, Nat. Rev. Genet, vol.8, pp.963-972, 2007.

D. R. Robinson, Y. Wu, P. Vats, F. Su, R. J. Lonigro et al., Activating ESR1 mutations in hormone-resistant metastatic breast cancer, Nat. Genet, vol.45, pp.1446-1451, 2013.

V. Rochira and C. Carani, Aromatase deficiency in men: a clinical perspective, Nat. Rev. Endocrinol, vol.5, pp.559-568, 2009.

K. M. Rodgers, J. O. Udesky, R. A. Rudel, and J. G. Brody, Environmental chemicals and breast cancer: An updated review of epidemiological literature informed by biological mechanisms, Environ. Res, vol.160, pp.152-182, 2018.

M. Sabbah, K. I. Kang, L. Tora, and G. Redeuilh, Oestrogen receptor facilitates the formation of preinitiation complex assembly: involvement of the general transcription factor TFIIB, Biochem. J, vol.336, pp.639-646, 1998.

S. Safe, K. , and K. , Nonclassical genomic ER/Sp and ER/AP-1 signaling pathways, J. Mol. Endocrinol, vol.41, pp.263-275, 2008.

T. Sakakura, Y. Suzuki, and R. Shiurba, Mammary stroma in development and carcinogenesis, J. Mammary Gland Biol. Neoplasia, vol.18, pp.189-197, 2013.

H. Samavat and M. S. Kurzer, Estrogen metabolism and breast cancer, Cancer Lett, vol.356, pp.231-243, 2015.

H. Sasano, H. Nagura, N. Harada, Y. Goukon, and M. Kimura, Immunolocalization of aromatase and other steroidogenic enzymes in human breast disorders, Hum. Pathol, vol.25, pp.530-535, 1994.

T. Sato, L. M. Neilson, A. R. Peck, C. Liu, T. H. Tran et al., Signal transducer and activator of transcription-3 and breast cancer prognosis, Am. J. Cancer Res, vol.1, pp.347-355, 2011.

R. S. Savkur and T. P. Burris, The coactivator LXXLL nuclear receptor recognition motif, J. Pept. Res, vol.63, pp.207-212, 2004.

V. Scarabin-carré, M. Canonico, S. Brailly-tabard, S. Trabado, P. Ducimetière et al., High Level of Plasma Estradiol as a New Predictor of Ischemic Arterial Disease in Older Postmenopausal Women: The Three City Cohort Study, 2012.

, J. Am. Heart Assoc, vol.1

T. T. Schug, A. Janesick, B. Blumberg, and J. J. Heindel, Endocrine Disrupting Chemicals and Disease Susceptibility, J. Steroid Biochem. Mol. Biol, vol.127, pp.204-215, 2011.

M. Schulster, A. M. Bernie, and R. Ramasamy, The role of estradiol in male reproductive function, Asian J. Androl, vol.18, pp.435-440, 2016.

E. R. Schuur, L. A. Mcpherson, G. P. Yang, and R. J. Weigel, Genomic structure of the promoters of the human estrogen receptor-alpha gene demonstrate changes in chromatin structure induced by AP2gamma, J. Biol. Chem, vol.276, pp.15519-15526, 2001.

D. D. Seachrist, K. W. Bonk, S. Ho, G. S. Prins, A. M. Soto et al., A Review of the Carcinogenic Potential of Bisphenol A, Reprod. Toxicol. Elmsford N, vol.59, pp.167-182, 2016.

H. Selye, Correlations between the chemical structure and the pharmacological actions of the steroids, Endocrinology, vol.30, pp.437-453, 1942.

L. F. Sempere, Celebrating 25 Years of MicroRNA Research: From Discovery to Clinical Application, Int. J. Mol. Sci, vol.20, 1987.

G. N. Sharma, R. Dave, J. Sanadya, P. Sharma, and K. K. Sharma, Various types and management of breast cancer: an overview, J. Adv. Pharm. Technol. Res, vol.1, pp.109-126, 2010.

A. Shehu, C. Albarracin, Y. S. Devi, K. Luther, J. Halperin et al., The stimulation of HSD17B7 expression by estradiol provides a powerful feedforward mechanism for estradiol biosynthesis in breast cancer cells, Mol. Endocrinol. Baltim. Md, vol.25, pp.754-766, 2011.

L. Shi, B. Dong, Z. Li, Y. Lu, T. Ouyang et al., Expression of ER-?36, a Novel Variant of Estrogen Receptor ?, and Resistance to Tamoxifen Treatment in Breast Cancer, J. Clin. Oncol, vol.27, pp.3423-3429, 2009.

Y. E. Shi, Y. Chen, R. Dackour, L. Potters, S. Wang et al., , 2010.

, Synuclein gamma stimulates membrane-initiated estrogen signaling by chaperoning estrogen receptor (ER)-alpha36, a variant of ER-alpha, Am. J. Pathol, vol.177, pp.964-973

R. Siersbaek, S. Kumar, and J. S. Carroll, Signaling pathways and steroid receptors modulating estrogen receptor ? function in breast cancer, Genes Dev, vol.32, pp.1141-1154, 2018.

K. So?tysik and P. Czekaj, ER?36--Another piece of the estrogen puzzle, Eur. J. Cell Biol, vol.94, pp.611-625, 2015.

C. Stresemann and F. Lyko, Modes of action of the DNA methyltransferase inhibitors azacytidine and decitabine, Int. J. Cancer, vol.123, pp.8-13, 2008.

M. Suetsugi, L. Su, K. Karlsberg, Y. Yuan, C. et al., Flavone and isoflavone phytoestrogens are agonists of estrogen-related receptors, Mol. Cancer Res. MCR, vol.1, pp.981-991, 2003.

L. Sun, J. Wang, L. Zhang, X. Li, and D. Shen, Expression of ER-?36, a novel variant of estrogen receptor in endometrial carcinoma and its clinical significance, Gynecol. Obstet. Invest, vol.75, pp.68-72, 2013.

C. M. Szego, J. S. Davis, W. Tan, B. Liu, S. Qu et al., Adenosine 3',5'-monophosphate in rat uterus: acute elevation by estrogen, Proc. Natl. Acad. Sci. U. S. A, vol.58, pp.2735-2742, 1967.

K. Tanimoto, H. Eguchi, T. Yoshida, K. Hajiro-nakanishi, and S. Hayashi, Regulation of estrogen receptor alpha gene mediated by promoter B responsible for its enhanced expressionin human breast cancer, Nucleic Acids Res, vol.27, pp.903-909, 1999.

T. Tanos and C. Brisken, What Signals Operate in the Mammary Niche?, Breast Dis, vol.29, pp.69-82, 2008.

J. R. Tata, One hundred years of hormones, EMBO Rep, vol.6, pp.490-496, 2005.

Y. Tay, J. Zhang, A. M. Thomson, B. Lim, R. et al., MicroRNAs to Nanog, Oct4 and Sox2 coding regions modulate embryonic stem cell differentiation, Nature, vol.455, pp.1124-1128, 2008.

A. Teymourzadeh, S. Mansouri, L. Farahmand, A. Hosseinzade, M. et al., ER-?36 Interactions With Cytosolic Molecular Network in Acquired Tamoxifen Resistance, Clin. Breast Cancer, vol.17, pp.403-407, 2017.

P. Thomas, D. , and J. , Binding and activation of the seven-transmembrane estrogen receptor GPR30 by environmental estrogens: a potential novel mechanism of endocrine disruption, J. Steroid Biochem. Mol. Biol, vol.102, pp.175-179, 2006.

P. Thomas, Y. Pang, E. J. Filardo, D. , and J. , Identity of an estrogen membrane receptor coupled to a G protein in human breast cancer cells, Endocrinology, vol.146, pp.624-632, 2005.

J. W. Thornton, Evolution of vertebrate steroid receptors from an ancestral estrogen receptor by ligand exploitation and serial genome expansions, Proc. Natl. Acad. Sci. U. S. A, vol.98, pp.5671-5676, 2001.

S. L. Tilghman, M. R. Bratton, H. C. Segar, E. C. Martin, L. V. Rhodes et al., Endocrine disruptor regulation of microRNA expression in breast carcinoma cells, PloS One, vol.7, 2012.

S. Q. To, V. Cheung, K. A. Lazarus, K. C. Knower, and C. D. Clyne, Estradiol regulates Tumor Necrosis Factor-? expression and secretion in Estrogen Receptor positive breast cancer cells, Mol. Cell. Endocrinol, vol.394, pp.21-28, 2014.

G. B. Tremblay, D. Bergeron, and V. Giguere, 4-Hydroxytamoxifen is an isoform-specific inhibitor of orphan estrogen-receptor-related (ERR) nuclear receptors beta and gamma, Endocrinology, vol.142, pp.4572-4575, 2001.

K. Tsuboi, T. Nagatomo, T. Gohno, T. Higuchi, S. Sasaki et al., Single CpG site methylation controls estrogen receptor gene transcription and correlates with hormone therapy resistance, J. Steroid Biochem. Mol. Biol, vol.171, pp.209-217, 2017.

G. Turashvili and E. Brogi, Tumor Heterogeneity in Breast Cancer, 2017.

B. C. Turner, J. Zhang, A. A. Gumbs, M. G. Maher, L. Kaplan et al., Expression of AP-2 transcription factors in human breast cancer correlates with the regulation of multiple growth factor signalling pathways, Cancer Res, vol.58, pp.5466-5472, 1998.

S. Vacher, P. Castagnet, W. Chemlali, F. Lallemand, D. Meseure et al., High AHR expression in breast tumors correlates with expression of genes from several signaling pathways namely inflammation and endogenous tryptophan metabolism, PLoS ONE, vol.13, 2018.

F. Velloso, A. Bianco, J. O. Farias, N. E. Torres, P. Y. Ferruzo et al., The crossroads of breast cancer progression: insights into the modulation of major signaling pathways, Onco Targets Ther, vol.10, pp.5491-5524, 2017.

J. M. Veltmaat, A. A. Mailleux, J. P. Thiery, and S. Bellusci, Mouse embryonic mammogenesis as a model for the molecular regulation of pattern formation, Differ. Res. Biol. Divers, vol.71, pp.1-17, 2003.

J. E. Visvader, Keeping abreast of the mammary epithelial hierarchy and breast tumorigenesis, 2009.

, Genes Dev, vol.23, pp.2563-2577

J. E. Visvader and J. Stingl, Mammary stem cells and the differentiation hierarchy: current status and perspectives, Genes Dev, vol.28, pp.1143-1158, 2014.

A. Vivacqua, E. Romeo, P. De-marco, E. M. De-francesco, S. Abonante et al., , 2012.

, GPER mediates the Egr-1 expression induced by 17?-estradiol and 4-hydroxitamoxifen in breast and endometrial cancer cells, Breast Cancer Res. Treat, vol.133, pp.1025-1035

V. Roemeling, C. A. Mathias, L. Dawson, . Harrison, L. Marlow et al., Estrogen receptor mRNA directed therapy for triple negative breast cancer, 2014.

P. Vrta?nik, B. Ostanek, S. Mencej-bedra?, M. , and J. , The many faces of estrogen signaling, Biochem. Medica, vol.24, pp.329-342, 2014.

K. Wagner, A. Krempler, A. A. Triplett, Y. Qi, N. M. George et al., Impaired alveologenesis and maintenance of secretory mammary epithelial cells in Jak2 conditional knockout mice, Mol. Cell. Biol, vol.24, pp.5510-5520, 2004.

N. Wajapeyee and K. Somasundaram, Cell cycle arrest and apoptosis induction by activator protein 2alpha (AP-2alpha) and the role of p53 and p21WAF1/CIP1 in AP-2alpha-mediated growth inhibition, J. Biol. Chem, vol.278, pp.52093-52101, 2003.

H. Wang, H. Yeh, W. Huang, C. Lin, W. Su et al., Activation of the signal transducer and activator of transcription 3 pathway up-regulates estrogen receptor-beta expression in lung adenocarcinoma cells, Mol. Endocrinol. Baltim. Md, vol.25, pp.1145-1158, 2011.

J. Wang, J. Li, R. Fang, S. Xie, L. Wang et al., Expression of ER?36 in gastric cancer samples and their matched normal tissues, Oncol. Lett, vol.3, pp.172-175, 2012.

L. Wang, H. Li, S. Yang, W. Ma, M. Liu et al., , 2016.

, Cyanidin-3-o-glucoside directly binds to ER?36 and inhibits EGFR-positive triple-negative breast cancer, Oncotarget, vol.7, pp.68864-68882

Q. Wang, W. Zhang, J. Yang, Y. Liu, Z. Yan et al., , 2015.

, High ER?36 Expression Level and Membrane Location Predict Poor Prognosis in Renal Cell Carcinoma: Medicine (Baltimore) 94, e1048

Q. Wang, J. Jiang, G. Ying, X. Xie, X. Zhang et al.,

S. Wang, Y. Yao, M. Yao, P. Fu, W. et al., Interleukin-22 promotes triple negative breast cancer cells migration and paclitaxel resistance through JAK-STAT3/MAPKs/AKT signaling pathways, Biochem. Biophys. Res. Commun, vol.503, pp.1605-1609, 2018.

X. Wang, K. Belguise, C. F. O'neill, N. Sánchez-morgan, M. Romagnoli et al., RelB NF-?B Represses Estrogen Receptor ? Expression via Induction of the Zinc Finger Protein Blimp1, Mol. Cell. Biol, vol.29, pp.3832-3844, 2009.

X. Wang, N. Zheng, J. Dong, X. Wang, L. Liu et al., Estrogen receptor-?36 is involved in icaritin induced growth inhibition of triple-negative breast cancer cells, J. Steroid Biochem, 2017.

, Mol. Biol, vol.171, pp.318-327

Z. Wang, X. Zhang, P. Shen, B. W. Loggie, Y. Chang et al., Identification, cloning, and expression of human estrogen receptor-alpha36, a novel variant of human estrogen receptor-alpha66, Biochem. Biophys. Res. Commun, vol.336, pp.1023-1027, 2005.

Z. Wang, X. Zhang, P. Shen, B. W. Loggie, Y. Chang et al., A variant of estrogen receptor-?, hER-?36: Transduction of estrogen-and antiestrogen-dependent membrane-initiated mitogenic signaling, Proc. Natl. Acad. Sci. U. S. A, vol.103, pp.9063-9068, 2006.

C. J. Watson, Involution: apoptosis and tissue remodelling that convert the mammary gland from milk factory to a quiescent organ, Breast Cancer Res. BCR, vol.8, p.203, 2006.

C. J. Watson and K. Neoh, The Stat family of transcription factors have diverse roles in mammary gland development, Semin. Cell Dev. Biol, vol.19, pp.401-406, 2008.

P. Webb, P. Nguyen, C. Valentine, G. N. Lopez, G. R. Kwok et al., The Estrogen Receptor Enhances AP-1 Activity by Two Distinct Mechanisms with Different Requirements for Receptor Transactivation Functions, Mol. Endocrinol, vol.13, pp.1672-1685, 1999.

D. R. Welch, Tumor heterogeneity 2016, Cancer Res, vol.76, pp.4-6, 2016.

R. E. White, Estrogen and vascular function, Vascul. Pharmacol, vol.38, pp.73-80, 2002.

B. S. Wiseman, M. D. Sternlicht, L. R. Lund, C. M. Alexander, J. Mott et al., Site-specific inductive and inhibitory activities of MMP-2 and MMP-3 orchestrate mammary gland branching morphogenesis, J. Cell Biol, vol.162, pp.1123-1133, 2003.

H. Xie, M. Sun, X. Liao, L. Yuan, Z. Sheng et al., Estrogen receptor ?36 mediates a bone-sparing effect of 17?-estrodiol in postmenopausal women, J. Bone Miner. Res, vol.26, pp.156-168, 2011.

Z. Xie, T. Li, B. Gan, X. Gao, L. Gao et al., Investigation of miR-136-5p key target genes and pathways in lung squamous cell cancer based on TCGA database and bioinformatics analysis, Pathol. Res. Pract, vol.214, pp.644-654, 2018.

H. Yamashita, M. Nishio, S. Kobayashi, Y. Ando, H. Sugiura et al., Phosphorylation of estrogen receptor alpha serine 167 is predictive of response to endocrine therapy and increases postrelapse survival in metastatic breast cancer, Breast Cancer Res. BCR, vol.7, pp.753-764, 2005.

M. Yan, X. Li, D. Tong, C. Han, R. Zhao et al., miR-136 suppresses tumor invasion and metastasis by targeting RASAL2 in triple-negative breast cancer, Oncol. Rep, vol.36, pp.65-71, 2016.

Y. Yan, L. Yu, L. Castro, and D. Dixon, ER?36, a variant of estrogen receptor ?, is predominantly localized in mitochondria of human uterine smooth muscle and leiomyoma cells, PLoS ONE, vol.12, 2017.

S. Yang, M. Hou, F. Chen, F. Ou-yang, Y. Wu et al., , 2016.

X. Yang, H. Wang, and B. Jiao, Mammary gland stem cells and their application in breast cancer, Oncotarget, vol.8, pp.10675-10691, 2016.

P. Ya?ar, G. Ayaz, S. D. User, G. Güpür, and M. Muyan, Molecular mechanism of estrogenestrogen receptor signaling, Reprod. Med. Biol, vol.16, pp.4-20, 2017.

Y. I. Yeom, G. Fuhrmann, C. E. Ovitt, A. Brehm, K. Ohbo et al., Germline regulatory element of Oct-4 specific for the totipotent cycle of embryonal cells, vol.14, 1996.

P. Yi, M. D. Driscoll, J. Huang, S. Bhagat, R. Hilf et al., The effects of estrogen-responsive element-and ligand-induced structural changes on the recruitment of cofactors and transcriptional responses by ER alpha and ER beta, Mol. Endocrinol. Baltim. Md, vol.16, pp.674-693, 2002.

L. Yin, X. Zhang, X. Bian, Y. Guo, and Z. Wang, Disruption of the ER-?36-EGFR/HER2 positive regulatory loops restores tamoxifen sensitivity in tamoxifen resistance breast cancer cells, PloS One, vol.9, 2014.

B. York and B. W. Malley, Steroid Receptor Coactivator (SRC) Family: Masters of Systems Biology, J. Biol. Chem, vol.285, pp.38743-38750, 2010.

T. Yoshida, H. Eguchi, K. Nakachi, K. Tanimoto, Y. Higashi et al., Distinct mechanisms of loss of estrogen receptor alpha gene expression in human breast cancer: methylation of the gene and alteration of trans-acting factors, Carcinogenesis, vol.21, pp.2193-2201, 2000.

T. Yoshimizu, N. Sugiyama, M. D. Felice, Y. I. Yeom, K. Ohbo et al., Germline-specific expression of the Oct-4/green fluorescent protein (GFP) transgene in mice, Dev. Growth Differ, vol.41, pp.675-684, 1999.

J. Yu, C. Angelin-duclos, J. Greenwood, J. Liao, C. et al., Transcriptional Repression by Blimp-1 (PRDI-BF1) Involves Recruitment of Histone Deacetylase, Mol. Cell. Biol, vol.20, pp.2592-2603, 2000.

Q. C. Yu, E. M. Verheyen, and Y. A. Zeng, Mammary Development and Breast Cancer: A Wnt Perspective, Cancers, vol.8, 2016.

F. Yuan, W. , and W. , MicroRNA-802 suppresses breast cancer proliferation through downregulation of FoxM1, Mol. Med. Rep, vol.12, pp.4647-4651, 2015.

X. Zeng, L. Liu, L. Lü, and Q. Zou, Prediction of potential disease-associated microRNAs using structural perturbation method, Bioinforma. Oxf. Engl, vol.34, pp.2425-2432, 2018.

H. Zhang, L. , and R. , STAT3 in Cancer-Friend or Foe? Cancers, vol.6, pp.1408-1440, 2014.

C. Zhang, X. Luo, L. Liu, S. Guo, W. Zhao et al.,

, Myocardin-related transcription factor A is up-regulated by 17?-estradiol and promotes migration of MCF-7 breast cancer cells via transactivation of MYL9 and CYR61, Acta Biochim. Biophys. Sin, vol.45, pp.921-927

J. Zhang, S. Brewer, J. Huang, W. , and T. , Overexpression of transcription factor AP-2alpha suppresses mammary gland growth and morphogenesis, Dev. Biol, vol.256, pp.127-145, 2003.

J. Zhang, G. Li, Z. Li, X. Yu, Y. Zheng et al., , 2012.

, Estrogen-independent effects of ER-?36 in ER-negative breast cancer, Steroids, vol.77, pp.666-673

J. Zhang, C. Zhou, H. Jiang, L. Liang, W. Shi et al., ZEB1 induces ER-? promoter hypermethylation and confers antiestrogen resistance in breast cancer, Cell Death Dis, vol.8, p.2732, 2017.

S. Zhang, C. Qin, and S. H. Safe, Flavonoids as aryl hydrocarbon receptor agonists/antagonists: effects of structure and cell context, Environ. Health Perspect, vol.111, pp.1877-1882, 2003.

S. Zhang, C. Qiu, L. Wang, Q. Liu, and J. Du, The elevated level of ER?36 is correlated with nodal metastasis and poor prognosis in lung adenocarcinoma, Steroids, vol.87, pp.39-45, 2014.

X. Zhang, N. Tang, T. J. Hadden, and A. K. Rishi, Akt, FoxO and regulation of apoptosis, Biochim. Biophys. Acta, vol.1813, pp.1978-1986, 2011.

X. Zhang, L. Kang, L. Ding, S. Vranic, Z. Gatalica et al., A Positive Feedback Loop of ER-?36/EGFR Promotes Malignant Growth of ER-negative Breast Cancer Cells, Oncogene, vol.30, pp.770-780, 2011.

X. Zhang, Y. Li, D. Wang, W. , and X. , miR-22 suppresses tumorigenesis and improves radiosensitivity of breast cancer cells by targeting Sirt1, Biol. Res, vol.50, p.27, 2017.

X. Zhang, L. Ding, L. Kang, and Z. Wang, Involvement of ER-?36, Src, EGFR and STAT5 in the biphasic estrogen signaling of ER-negative breast cancer cells, Oncol. Rep, vol.27, pp.2057-2065, 2012.

C. Zhao, K. Yasui, C. J. Lee, H. Kurioka, Y. Hosokawa et al., Elevated expression levels of NCOA3, TOP1, and TFAP2C in breast tumors as predictors of poor prognosis, Cancer, vol.98, pp.18-23, 2003.

C. Zhao, J. Matthews, M. Tujague, J. Wan, A. Ström et al., Estrogen receptor beta2 negatively regulates the transactivation of estrogen receptor alpha in human breast cancer cells, Cancer Res, vol.67, pp.3955-3962, 2007.

C. Zhao, K. Dahlman-wright, and J. Gustafsson, Estrogen receptor beta: an overview and update, Nucl. Recept. Signal, vol.6, p.3, 2008.

Y. Zheng, J. Zhang, Z. Xu, J. Sheng, X. Zhang et al., Quantitative profiles of the mRNAs of ER-? and its novel variant ER-?36 in breast cancers and matched normal tissues*, J. Zhejiang Univ. Sci. B, vol.11, pp.144-150, 2010.

H. Zhu, Y. Huang, H. Su, Y. Ma, Y. Tao et al., Identification of a novel human estrogen receptor-? splice variant able to enhance malignant biological behaviors of breast cancer cells, Oncol. Lett, vol.15, pp.5339-5344, 2018.

L. Zhu, J. Zou, Y. Zhao, X. Jiang, Y. Wang et al., ER-?36 mediates cisplatin resistance in breast cancer cells through EGFR/HER-2/ERK signaling pathway, J. Exp. Clin. Cancer Res. CR, vol.37, p.123, 2018.

Y. Zhu, S. Shao, H. Pan, Z. Cheng, and X. Rui, MicroRNA 136 inhibits prostate cancer cell proliferation and invasion by directly targeting mitogen activated protein kinase kinase 4, Mol. Med. Rep, vol.17, pp.4803-4810, 2018.

S. Zimmerman, Get to Know Your Sex Hormones, 2019.

Y. Zou, L. Ding, M. Coleman, W. , and Z. , Estrogen receptor-alpha (ER-?) suppresses expression of its variant ER-?36, FEBS Lett, vol.583, pp.1368-1374, 2009.

W. Zwart, A. Griekspoor, V. Berno, K. Lakeman, K. Jalink et al., PKA-induced resistance to tamoxifen is associated with an altered orientation of ERalpha towards co-activator SRC-1, EMBO J, vol.26, pp.3534-3544, 2007.