Skip to main content
Top
Published in: Breast Cancer Research and Treatment 1/2011

01-11-2011 | Review

Glucocorticoid receptor and breast cancer

Authors: Myriam Vilasco, Laudine Communal, Najat Mourra, Aurélie Courtin, Patricia Forgez, Anne Gompel

Published in: Breast Cancer Research and Treatment | Issue 1/2011

Login to get access

Abstract

Stress enhances glucocorticoid (GC) synthesis, which alters inflammation and immune responses, as well as cellular proliferation and apoptosis in a number of tissues. Increasingly, stress has been associated with cancer progression, and in particular in breast cancer. Consequently, an operational glucocorticoid receptor system in breast tissue influences breast cancer development. In this review, we summarize the data on the GC/GR system in normal and tumoral breast tissue. We also review the molecular mechanisms by which GCs control apoptosis and proliferation in breast cancer models and how GCs alter the chemotherapy of breast cancer treatment when used in combination. Finally, we discuss the participation of GR in breast tumorigenesis under hormone replacement therapy.
Literature
2.
go back to reference Kricker A et al (2009) Effects of life event stress and social support on the odds of a > or = 2 cm breast cancer. Cancer Causes Control 20(4):437–447PubMedCrossRef Kricker A et al (2009) Effects of life event stress and social support on the odds of a > or = 2 cm breast cancer. Cancer Causes Control 20(4):437–447PubMedCrossRef
3.
go back to reference Michael YL et al (2009) Influence of stressors on breast cancer incidence in the women’s health initiative. Health Psychol 28(2):137–146PubMedCrossRef Michael YL et al (2009) Influence of stressors on breast cancer incidence in the women’s health initiative. Health Psychol 28(2):137–146PubMedCrossRef
4.
go back to reference Hermes GL et al (2009) Social isolation dysregulates endocrine and behavioral stress while increasing malignant burden of spontaneous mammary tumors. Proc Natl Acad Sci USA 106(52):22393–22398PubMedCrossRef Hermes GL et al (2009) Social isolation dysregulates endocrine and behavioral stress while increasing malignant burden of spontaneous mammary tumors. Proc Natl Acad Sci USA 106(52):22393–22398PubMedCrossRef
5.
go back to reference Baschant U, Tuckermann J (2010) The role of the glucocorticoid receptor in inflammation and immunity. J Steroid Biochem Mol Biol 120(2–3):69–75PubMedCrossRef Baschant U, Tuckermann J (2010) The role of the glucocorticoid receptor in inflammation and immunity. J Steroid Biochem Mol Biol 120(2–3):69–75PubMedCrossRef
6.
go back to reference Wintermantel TM et al (2005) The epithelial glucocorticoid receptor is required for the normal timing of cell proliferation during mammary lobuloalveolar development but is dispensable for milk production. Mol Endocrinol 19(2):340–349PubMedCrossRef Wintermantel TM et al (2005) The epithelial glucocorticoid receptor is required for the normal timing of cell proliferation during mammary lobuloalveolar development but is dispensable for milk production. Mol Endocrinol 19(2):340–349PubMedCrossRef
7.
go back to reference Murtagh J et al (2004) Organization of mammary epithelial cells into 3D acinar structures requires glucocorticoid and JNK signaling. J Cell Biol 166(1):133–143PubMedCrossRef Murtagh J et al (2004) Organization of mammary epithelial cells into 3D acinar structures requires glucocorticoid and JNK signaling. J Cell Biol 166(1):133–143PubMedCrossRef
8.
go back to reference Courtin A et al. (2011) Glucocorticoid receptor activity discriminates between progesterone and medroxyprogesterone acetate effects in breast cells. Breast Cancer Res Treat. PMID:21336598 Courtin A et al. (2011) Glucocorticoid receptor activity discriminates between progesterone and medroxyprogesterone acetate effects in breast cells. Breast Cancer Res Treat. PMID:21336598
9.
10.
go back to reference Sorlie T et al (2001) Gene expression patterns of breast carcinomas distinguish tumor subclasses with clinical implications. Proc Natl Acad Sci USA 98(19):10869–10874PubMedCrossRef Sorlie T et al (2001) Gene expression patterns of breast carcinomas distinguish tumor subclasses with clinical implications. Proc Natl Acad Sci USA 98(19):10869–10874PubMedCrossRef
11.
go back to reference Richardson AL et al (2006) X chromosomal abnormalities in basal-like human breast cancer. Cancer Cell 9(2):121–132PubMedCrossRef Richardson AL et al (2006) X chromosomal abnormalities in basal-like human breast cancer. Cancer Cell 9(2):121–132PubMedCrossRef
12.
go back to reference Teulings FA, van Gilse HA (1977) Demonstration of glucocorticoid receptors in human mammary carcinomas. Horm Res 8(2):107–116PubMedCrossRef Teulings FA, van Gilse HA (1977) Demonstration of glucocorticoid receptors in human mammary carcinomas. Horm Res 8(2):107–116PubMedCrossRef
13.
go back to reference Allegra JC et al (1978) An association between steroid hormone receptors and response to cytotoxic chemotherapy in patients with metastatic breast cancer. Cancer Res 38(11 Pt 2):4299–4304PubMed Allegra JC et al (1978) An association between steroid hormone receptors and response to cytotoxic chemotherapy in patients with metastatic breast cancer. Cancer Res 38(11 Pt 2):4299–4304PubMed
14.
go back to reference Allegra JC et al (1979) Distribution, frequency, and quantitative analysis of estrogen, progesterone, androgen, and glucocorticoid receptors in human breast cancer. Cancer Res 39(5):1447–1454PubMed Allegra JC et al (1979) Distribution, frequency, and quantitative analysis of estrogen, progesterone, androgen, and glucocorticoid receptors in human breast cancer. Cancer Res 39(5):1447–1454PubMed
15.
go back to reference Moran TJ et al (2000) The glucocorticoid receptor mediates a survival signal in human mammary epithelial cells. Cancer Res 60(4):867–872PubMed Moran TJ et al (2000) The glucocorticoid receptor mediates a survival signal in human mammary epithelial cells. Cancer Res 60(4):867–872PubMed
16.
go back to reference Schorr K, Furth PA (2000) Induction of bcl-xL expression in mammary epithelial cells is glucocorticoid-dependent but not signal transducer and activator of transcription 5-dependent. Cancer Res 60(21):5950–5953PubMed Schorr K, Furth PA (2000) Induction of bcl-xL expression in mammary epithelial cells is glucocorticoid-dependent but not signal transducer and activator of transcription 5-dependent. Cancer Res 60(21):5950–5953PubMed
17.
go back to reference Mikosz CA et al (2001) Glucocorticoid receptor-mediated protection from apoptosis is associated with induction of the serine/threonine survival kinase gene, sgk-1. J Biol Chem 276(20):16649–16654PubMedCrossRef Mikosz CA et al (2001) Glucocorticoid receptor-mediated protection from apoptosis is associated with induction of the serine/threonine survival kinase gene, sgk-1. J Biol Chem 276(20):16649–16654PubMedCrossRef
18.
go back to reference Mattern J, Buchler MW, Herr I (2007) Cell cycle arrest by glucocorticoids may protect normal tissue and solid tumors from cancer therapy. Cancer Biol Ther 6(9):1345–1354PubMedCrossRef Mattern J, Buchler MW, Herr I (2007) Cell cycle arrest by glucocorticoids may protect normal tissue and solid tumors from cancer therapy. Cancer Biol Ther 6(9):1345–1354PubMedCrossRef
19.
go back to reference Lippman M, Bolan G, Huff K (1976) The effects of glucocorticoids and progesterone on hormone-responsive human breast cancer in long-term tissue culture. Cancer Res 36(12):4602–4609PubMed Lippman M, Bolan G, Huff K (1976) The effects of glucocorticoids and progesterone on hormone-responsive human breast cancer in long-term tissue culture. Cancer Res 36(12):4602–4609PubMed
20.
go back to reference Osborne CK et al (1979) Direct inhibition of growth and antagonism of insulin action by glucocorticoids in human breast cancer cells in culture. Cancer Res 39(7 Pt 1):2422–2428PubMed Osborne CK et al (1979) Direct inhibition of growth and antagonism of insulin action by glucocorticoids in human breast cancer cells in culture. Cancer Res 39(7 Pt 1):2422–2428PubMed
21.
go back to reference Huff KK et al (1988) Multihormonal regulation of insulin-like growth factor-I-related protein in MCF-7 human breast cancer cells. Mol Endocrinol 2(3):200–208PubMedCrossRef Huff KK et al (1988) Multihormonal regulation of insulin-like growth factor-I-related protein in MCF-7 human breast cancer cells. Mol Endocrinol 2(3):200–208PubMedCrossRef
22.
go back to reference Harris RA et al (1995) The induction of apoptosis in human mammary luminal epithelial cells by expression of activated c-neu and its abrogation by glucocorticoids. Br J Cancer 72(2):386–392PubMedCrossRef Harris RA et al (1995) The induction of apoptosis in human mammary luminal epithelial cells by expression of activated c-neu and its abrogation by glucocorticoids. Br J Cancer 72(2):386–392PubMedCrossRef
23.
go back to reference Wu W et al (2004) Microarray analysis reveals glucocorticoid-regulated survival genes that are associated with inhibition of apoptosis in breast epithelial cells. Cancer Res 64(5):1757–1764PubMedCrossRef Wu W et al (2004) Microarray analysis reveals glucocorticoid-regulated survival genes that are associated with inhibition of apoptosis in breast epithelial cells. Cancer Res 64(5):1757–1764PubMedCrossRef
24.
go back to reference Wu W et al (2005) Glucocorticoid receptor-induced MAPK phosphatase-1 (MPK-1) expression inhibits paclitaxel-associated MAPK activation and contributes to breast cancer cell survival. J Biol Chem 280(6):4117–4124PubMedCrossRef Wu W et al (2005) Glucocorticoid receptor-induced MAPK phosphatase-1 (MPK-1) expression inhibits paclitaxel-associated MAPK activation and contributes to breast cancer cell survival. J Biol Chem 280(6):4117–4124PubMedCrossRef
25.
go back to reference Pang D et al (2006) Dexamethasone decreases xenograft response to Paclitaxel through inhibition of tumor cell apoptosis. Cancer Biol Ther 5(8):933–940PubMedCrossRef Pang D et al (2006) Dexamethasone decreases xenograft response to Paclitaxel through inhibition of tumor cell apoptosis. Cancer Biol Ther 5(8):933–940PubMedCrossRef
26.
go back to reference Wang H et al (2004) Dexamethasone as a chemoprotectant in cancer chemotherapy: hematoprotective effects and altered pharmacokinetics and tissue distribution of carboplatin and gemcitabine. Cancer Chemother Pharmacol 53(6):459–467PubMedCrossRef Wang H et al (2004) Dexamethasone as a chemoprotectant in cancer chemotherapy: hematoprotective effects and altered pharmacokinetics and tissue distribution of carboplatin and gemcitabine. Cancer Chemother Pharmacol 53(6):459–467PubMedCrossRef
27.
go back to reference Wang H et al (2007) Dexamethasone as a chemosensitizer for breast cancer chemotherapy: potentiation of the antitumor activity of adriamycin, modulation of cytokine expression, and pharmacokinetics. Int J Oncol 30(4):947–953PubMed Wang H et al (2007) Dexamethasone as a chemosensitizer for breast cancer chemotherapy: potentiation of the antitumor activity of adriamycin, modulation of cytokine expression, and pharmacokinetics. Int J Oncol 30(4):947–953PubMed
28.
go back to reference Zhou J, Cidlowski JA (2005) The human glucocorticoid receptor: one gene, multiple proteins and diverse responses. Steroids 70(5–7):407–417PubMedCrossRef Zhou J, Cidlowski JA (2005) The human glucocorticoid receptor: one gene, multiple proteins and diverse responses. Steroids 70(5–7):407–417PubMedCrossRef
29.
go back to reference Miesfeld R et al (1984) Characterization of a steroid hormone receptor gene and mRNA in wild-type and mutant cells. Nature 312(5996):779–781PubMedCrossRef Miesfeld R et al (1984) Characterization of a steroid hormone receptor gene and mRNA in wild-type and mutant cells. Nature 312(5996):779–781PubMedCrossRef
30.
go back to reference Govindan MV et al (1985) Cloning of the human glucocorticoid receptor cDNA. Nucleic Acids Res 13(23):8293–8304PubMedCrossRef Govindan MV et al (1985) Cloning of the human glucocorticoid receptor cDNA. Nucleic Acids Res 13(23):8293–8304PubMedCrossRef
31.
go back to reference Galliher-Beckley AJ, Cidlowski JA (2009) Emerging roles of glucocorticoid receptor phosphorylation in modulating glucocorticoid hormone action in health and disease. IUBMB Life 61(10):979–986PubMedCrossRef Galliher-Beckley AJ, Cidlowski JA (2009) Emerging roles of glucocorticoid receptor phosphorylation in modulating glucocorticoid hormone action in health and disease. IUBMB Life 61(10):979–986PubMedCrossRef
32.
go back to reference Bodwell JE et al (1991) Identification of phosphorylated sites in the mouse glucocorticoid receptor. J Biol Chem 266(12):7549–7555PubMed Bodwell JE et al (1991) Identification of phosphorylated sites in the mouse glucocorticoid receptor. J Biol Chem 266(12):7549–7555PubMed
33.
go back to reference Krstic MD et al (1997) Mitogen-activated and cyclin-dependent protein kinases selectively and differentially modulate transcriptional enhancement by the glucocorticoid receptor. Mol Cell Biol 17(7):3947–3954PubMed Krstic MD et al (1997) Mitogen-activated and cyclin-dependent protein kinases selectively and differentially modulate transcriptional enhancement by the glucocorticoid receptor. Mol Cell Biol 17(7):3947–3954PubMed
34.
go back to reference Takabe S, Mochizuki K, Goda T (2008) De-phosphorylation of GR at Ser203 in nuclei associates with GR nuclear translocation and GLUT5 gene expression in Caco-2 cells. Arch Biochem Biophys 475(1):1–6PubMedCrossRef Takabe S, Mochizuki K, Goda T (2008) De-phosphorylation of GR at Ser203 in nuclei associates with GR nuclear translocation and GLUT5 gene expression in Caco-2 cells. Arch Biochem Biophys 475(1):1–6PubMedCrossRef
35.
go back to reference Miller AL et al (2005) p38 Mitogen-activated protein kinase (MAPK) is a key mediator in glucocorticoid-induced apoptosis of lymphoid cells: correlation between p38 MAPK activation and site-specific phosphorylation of the human glucocorticoid receptor at serine 211. Mol Endocrinol 19(6):1569–1583PubMedCrossRef Miller AL et al (2005) p38 Mitogen-activated protein kinase (MAPK) is a key mediator in glucocorticoid-induced apoptosis of lymphoid cells: correlation between p38 MAPK activation and site-specific phosphorylation of the human glucocorticoid receptor at serine 211. Mol Endocrinol 19(6):1569–1583PubMedCrossRef
36.
go back to reference Galliher-Beckley AJ et al (2008) Glycogen synthase kinase 3beta-mediated serine phosphorylation of the human glucocorticoid receptor redirects gene expression profiles. Mol Cell Biol 28(24):7309–7322PubMedCrossRef Galliher-Beckley AJ et al (2008) Glycogen synthase kinase 3beta-mediated serine phosphorylation of the human glucocorticoid receptor redirects gene expression profiles. Mol Cell Biol 28(24):7309–7322PubMedCrossRef
37.
go back to reference Itoh M et al (2002) Nuclear export of glucocorticoid receptor is enhanced by c-Jun N-terminal kinase-mediated phosphorylation. Mol Endocrinol 16(10):2382–2392PubMedCrossRef Itoh M et al (2002) Nuclear export of glucocorticoid receptor is enhanced by c-Jun N-terminal kinase-mediated phosphorylation. Mol Endocrinol 16(10):2382–2392PubMedCrossRef
38.
go back to reference Wang Z, Frederick J, Garabedian MJ (2002) Deciphering the phosphorylation “code” of the glucocorticoid receptor in vivo. J Biol Chem 277(29):26573–26580PubMedCrossRef Wang Z, Frederick J, Garabedian MJ (2002) Deciphering the phosphorylation “code” of the glucocorticoid receptor in vivo. J Biol Chem 277(29):26573–26580PubMedCrossRef
39.
go back to reference Chen W et al (2008) Glucocorticoid receptor phosphorylation differentially affects target gene expression. Mol Endocrinol 22(8):1754–1766PubMedCrossRef Chen W et al (2008) Glucocorticoid receptor phosphorylation differentially affects target gene expression. Mol Endocrinol 22(8):1754–1766PubMedCrossRef
40.
go back to reference Blind RD, Garabedian MJ (2008) Differential recruitment of glucocorticoid receptor phospho-isoforms to glucocorticoid-induced genes. J Steroid Biochem Mol Biol 109(1–2):150–157PubMedCrossRef Blind RD, Garabedian MJ (2008) Differential recruitment of glucocorticoid receptor phospho-isoforms to glucocorticoid-induced genes. J Steroid Biochem Mol Biol 109(1–2):150–157PubMedCrossRef
41.
go back to reference Wallace AD, Cidlowski JA (2001) Proteasome-mediated glucocorticoid receptor degradation restricts transcriptional signaling by glucocorticoids. J Biol Chem 276(46):42714–42721PubMedCrossRef Wallace AD, Cidlowski JA (2001) Proteasome-mediated glucocorticoid receptor degradation restricts transcriptional signaling by glucocorticoids. J Biol Chem 276(46):42714–42721PubMedCrossRef
42.
go back to reference Webster JC et al (1997) Mouse glucocorticoid receptor phosphorylation status influences multiple functions of the receptor protein. J Biol Chem 272(14):9287–9293PubMedCrossRef Webster JC et al (1997) Mouse glucocorticoid receptor phosphorylation status influences multiple functions of the receptor protein. J Biol Chem 272(14):9287–9293PubMedCrossRef
43.
go back to reference Lien HC et al (2006) Differential expression of glucocorticoid receptor in human breast tissues and related neoplasms. J Pathol 209(3):317–327PubMedCrossRef Lien HC et al (2006) Differential expression of glucocorticoid receptor in human breast tissues and related neoplasms. J Pathol 209(3):317–327PubMedCrossRef
44.
go back to reference Buxant F, Engohan-Aloghe C, Noel JC (2010) Estrogen receptor, progesterone receptor, and glucocorticoid receptor expression in normal breast tissue, breast in situ carcinoma, and invasive breast cancer. Appl Immunohistochem Mol Morphol 18(3):254–257PubMedCrossRef Buxant F, Engohan-Aloghe C, Noel JC (2010) Estrogen receptor, progesterone receptor, and glucocorticoid receptor expression in normal breast tissue, breast in situ carcinoma, and invasive breast cancer. Appl Immunohistochem Mol Morphol 18(3):254–257PubMedCrossRef
45.
go back to reference Horwitz KB, Costlow ME, McGuire WL (1975) MCF-7; a human breast cancer cell line with estrogen, androgen, progesterone, and glucocorticoid receptors. Steroids 26(6):785–795PubMedCrossRef Horwitz KB, Costlow ME, McGuire WL (1975) MCF-7; a human breast cancer cell line with estrogen, androgen, progesterone, and glucocorticoid receptors. Steroids 26(6):785–795PubMedCrossRef
46.
go back to reference Tronche F et al (1998) Genetic dissection of glucocorticoid receptor function in mice. Curr Opin Genet Dev 8(5):532–538PubMedCrossRef Tronche F et al (1998) Genetic dissection of glucocorticoid receptor function in mice. Curr Opin Genet Dev 8(5):532–538PubMedCrossRef
47.
go back to reference Reichardt HM et al (1998) DNA binding of the glucocorticoid receptor is not essential for survival. Cell 93(4):531–541PubMedCrossRef Reichardt HM et al (1998) DNA binding of the glucocorticoid receptor is not essential for survival. Cell 93(4):531–541PubMedCrossRef
48.
go back to reference Conde I et al (2008) Glucocorticoid receptor changes its cellular location with breast cancer development. Histol Histopathol 23(1):77–85PubMed Conde I et al (2008) Glucocorticoid receptor changes its cellular location with breast cancer development. Histol Histopathol 23(1):77–85PubMed
49.
go back to reference Belova L et al (2009) Glucocorticoid receptor expression in breast cancer associates with older patient age. Breast Cancer Res Treat 116(3):441–447PubMedCrossRef Belova L et al (2009) Glucocorticoid receptor expression in breast cancer associates with older patient age. Breast Cancer Res Treat 116(3):441–447PubMedCrossRef
50.
go back to reference Chen JQ, Brown TR, Yager JD (2008) Mechanisms of hormone carcinogenesis: evolution of views, role of mitochondria. Adv Exp Med Biol 630:1–18PubMedCrossRef Chen JQ, Brown TR, Yager JD (2008) Mechanisms of hormone carcinogenesis: evolution of views, role of mitochondria. Adv Exp Med Biol 630:1–18PubMedCrossRef
51.
go back to reference Lillberg K et al (2003) Stressful life events and risk of breast cancer in 10,808 women: a cohort study. Am J Epidemiol 157(5):415–423PubMedCrossRef Lillberg K et al (2003) Stressful life events and risk of breast cancer in 10,808 women: a cohort study. Am J Epidemiol 157(5):415–423PubMedCrossRef
52.
go back to reference Reiche EM, Nunes SO, Morimoto HK (2004) Stress, depression, the immune system, and cancer. Lancet Oncol 5(10):617–625PubMedCrossRef Reiche EM, Nunes SO, Morimoto HK (2004) Stress, depression, the immune system, and cancer. Lancet Oncol 5(10):617–625PubMedCrossRef
53.
54.
go back to reference Kruk J, Aboul-Enein HY (2004) Psychological stress and the risk of breast cancer: a case–control study. Cancer Detect Prev 28(6):399–408PubMedCrossRef Kruk J, Aboul-Enein HY (2004) Psychological stress and the risk of breast cancer: a case–control study. Cancer Detect Prev 28(6):399–408PubMedCrossRef
55.
go back to reference Morrison N, Eisman J (1993) Role of the negative glucocorticoid regulatory element in glucocorticoid repression of the human osteocalcin promoter. J Bone Miner Res 8(8):969–975PubMedCrossRef Morrison N, Eisman J (1993) Role of the negative glucocorticoid regulatory element in glucocorticoid repression of the human osteocalcin promoter. J Bone Miner Res 8(8):969–975PubMedCrossRef
56.
go back to reference De Bosscher K, Vanden Berghe W, Haegeman G (2003) The interplay between the glucocorticoid receptor and nuclear factor-kappaB or activator protein-1: molecular mechanisms for gene repression. Endocr Rev 24(4):488–522PubMedCrossRef De Bosscher K, Vanden Berghe W, Haegeman G (2003) The interplay between the glucocorticoid receptor and nuclear factor-kappaB or activator protein-1: molecular mechanisms for gene repression. Endocr Rev 24(4):488–522PubMedCrossRef
57.
go back to reference Messmer UK et al (2000) Suppression of apoptosis by glucocorticoids in glomerular endothelial cells: effects on proapoptotic pathways. Br J Pharmacol 129(8):1673–1683PubMedCrossRef Messmer UK et al (2000) Suppression of apoptosis by glucocorticoids in glomerular endothelial cells: effects on proapoptotic pathways. Br J Pharmacol 129(8):1673–1683PubMedCrossRef
58.
go back to reference Kerppola TK, Luk D, Curran T (1993) Fos is a preferential target of glucocorticoid receptor inhibition of AP-1 activity in vitro. Mol Cell Biol 13(6):3782–3791PubMed Kerppola TK, Luk D, Curran T (1993) Fos is a preferential target of glucocorticoid receptor inhibition of AP-1 activity in vitro. Mol Cell Biol 13(6):3782–3791PubMed
59.
go back to reference Scheinman RI et al (1995) Characterization of mechanisms involved in transrepression of NF-kappa B by activated glucocorticoid receptors. Mol Cell Biol 15(2):943–953PubMed Scheinman RI et al (1995) Characterization of mechanisms involved in transrepression of NF-kappa B by activated glucocorticoid receptors. Mol Cell Biol 15(2):943–953PubMed
60.
go back to reference Imai E et al (1993) Glucocorticoid receptor-cAMP response element-binding protein interaction and the response of the phosphoenolpyruvate carboxykinase gene to glucocorticoids. J Biol Chem 268(8):5353–5356PubMed Imai E et al (1993) Glucocorticoid receptor-cAMP response element-binding protein interaction and the response of the phosphoenolpyruvate carboxykinase gene to glucocorticoids. J Biol Chem 268(8):5353–5356PubMed
61.
62.
go back to reference Herr I et al (2007) Regulation of differential pro- and anti-apoptotic signaling by glucocorticoids. Apoptosis 12(2):271–291PubMedCrossRef Herr I et al (2007) Regulation of differential pro- and anti-apoptotic signaling by glucocorticoids. Apoptosis 12(2):271–291PubMedCrossRef
63.
go back to reference Feng Z et al (1995) Glucocorticoid and progesterone inhibit involution and programmed cell death in the mouse mammary gland. J Cell Biol 131(4):1095–1103PubMedCrossRef Feng Z et al (1995) Glucocorticoid and progesterone inhibit involution and programmed cell death in the mouse mammary gland. J Cell Biol 131(4):1095–1103PubMedCrossRef
64.
go back to reference Diamond MI et al (1990) Transcription factor interactions: selectors of positive or negative regulation from a single DNA element. Science 249(4974):1266–1272PubMedCrossRef Diamond MI et al (1990) Transcription factor interactions: selectors of positive or negative regulation from a single DNA element. Science 249(4974):1266–1272PubMedCrossRef
65.
go back to reference Shaulian E, Karin M (2002) AP-1 as a regulator of cell life and death. Nat Cell Biol 4(5):E131–E136PubMedCrossRef Shaulian E, Karin M (2002) AP-1 as a regulator of cell life and death. Nat Cell Biol 4(5):E131–E136PubMedCrossRef
66.
go back to reference Lu A et al (2002) Blockade of AP1 transactivation abrogates the abnormal expression of breast cancer-specific gene 1 in breast cancer cells. J Biol Chem 277(35):31364–31372PubMedCrossRef Lu A et al (2002) Blockade of AP1 transactivation abrogates the abnormal expression of breast cancer-specific gene 1 in breast cancer cells. J Biol Chem 277(35):31364–31372PubMedCrossRef
67.
go back to reference Bamberger AM et al (1999) Expression pattern of the AP-1 family in breast cancer: association of fosB expression with a well-differentiated, receptor-positive tumor phenotype. Int J Cancer 84(5):533–538PubMedCrossRef Bamberger AM et al (1999) Expression pattern of the AP-1 family in breast cancer: association of fosB expression with a well-differentiated, receptor-positive tumor phenotype. Int J Cancer 84(5):533–538PubMedCrossRef
68.
go back to reference Milde-Langosch K (2005) The Fos family of transcription factors and their role in tumourigenesis. Eur J Cancer 41(16):2449–2461PubMedCrossRef Milde-Langosch K (2005) The Fos family of transcription factors and their role in tumourigenesis. Eur J Cancer 41(16):2449–2461PubMedCrossRef
69.
go back to reference Ozaki T, Nakagawara A (2011) p53: the attractive tumor suppressor in the cancer research field. J Biomed Biotechnol 2011: 603925 Ozaki T, Nakagawara A (2011) p53: the attractive tumor suppressor in the cancer research field. J Biomed Biotechnol 2011: 603925
70.
go back to reference Sengupta S, Wasylyk B (2001) Ligand-dependent interaction of the glucocorticoid receptor with p53 enhances their degradation by Hdm2. Genes Dev 15(18):2367–2380PubMedCrossRef Sengupta S, Wasylyk B (2001) Ligand-dependent interaction of the glucocorticoid receptor with p53 enhances their degradation by Hdm2. Genes Dev 15(18):2367–2380PubMedCrossRef
71.
go back to reference Sengupta S et al (2000) Negative cross-talk between p53 and the glucocorticoid receptor and its role in neuroblastoma cells. EMBO J 19(22):6051–6064PubMedCrossRef Sengupta S et al (2000) Negative cross-talk between p53 and the glucocorticoid receptor and its role in neuroblastoma cells. EMBO J 19(22):6051–6064PubMedCrossRef
72.
go back to reference Moll UM, Riou G, Levine AJ (1992) Two distinct mechanisms alter p53 in breast cancer: mutation and nuclear exclusion. Proc Natl Acad Sci USA 89(15):7262–7266PubMedCrossRef Moll UM, Riou G, Levine AJ (1992) Two distinct mechanisms alter p53 in breast cancer: mutation and nuclear exclusion. Proc Natl Acad Sci USA 89(15):7262–7266PubMedCrossRef
73.
go back to reference Maiyar AC et al (1997) Repression of glucocorticoid receptor transactivation and DNA binding of a glucocorticoid response element within the serum/glucocorticoid-inducible protein kinase (sgk) gene promoter by the p53 tumor suppressor protein. Mol Endocrinol 11(3):312–329PubMedCrossRef Maiyar AC et al (1997) Repression of glucocorticoid receptor transactivation and DNA binding of a glucocorticoid response element within the serum/glucocorticoid-inducible protein kinase (sgk) gene promoter by the p53 tumor suppressor protein. Mol Endocrinol 11(3):312–329PubMedCrossRef
74.
go back to reference Maiyar AC et al (1996) p53 stimulates promoter activity of the sgk, serum/glucocorticoid-inducible serine/threonine protein kinase gene in rodent mammary epithelial cells. J Biol Chem 271(21):12414–12422PubMedCrossRef Maiyar AC et al (1996) p53 stimulates promoter activity of the sgk, serum/glucocorticoid-inducible serine/threonine protein kinase gene in rodent mammary epithelial cells. J Biol Chem 271(21):12414–12422PubMedCrossRef
75.
go back to reference Goya L et al (1993) Glucocorticoids induce a G1/G0 cell cycle arrest of Con8 rat mammary tumor cells that is synchronously reversed by steroid withdrawal or addition of transforming growth factor-alpha. Mol Endocrinol 7(9):1121–1132PubMedCrossRef Goya L et al (1993) Glucocorticoids induce a G1/G0 cell cycle arrest of Con8 rat mammary tumor cells that is synchronously reversed by steroid withdrawal or addition of transforming growth factor-alpha. Mol Endocrinol 7(9):1121–1132PubMedCrossRef
76.
go back to reference Urban G et al (2003) Identification of a functional link for the p53 tumor suppressor protein in dexamethasone-induced growth suppression. J Biol Chem 278(11):9747–9753PubMedCrossRef Urban G et al (2003) Identification of a functional link for the p53 tumor suppressor protein in dexamethasone-induced growth suppression. J Biol Chem 278(11):9747–9753PubMedCrossRef
77.
go back to reference Gutierrez MC et al (2005) Molecular changes in tamoxifen-resistant breast cancer: relationship between estrogen receptor, HER-2, and p38 mitogen-activated protein kinase. J Clin Oncol 23(11):2469–2476PubMedCrossRef Gutierrez MC et al (2005) Molecular changes in tamoxifen-resistant breast cancer: relationship between estrogen receptor, HER-2, and p38 mitogen-activated protein kinase. J Clin Oncol 23(11):2469–2476PubMedCrossRef
78.
go back to reference Whyte J et al (2009) Key signalling nodes in mammary gland development and cancer. Mitogen-activated protein kinase signalling in experimental models of breast cancer progression and in mammary gland development. Breast Cancer Res 11(5):209PubMedCrossRef Whyte J et al (2009) Key signalling nodes in mammary gland development and cancer. Mitogen-activated protein kinase signalling in experimental models of breast cancer progression and in mammary gland development. Breast Cancer Res 11(5):209PubMedCrossRef
79.
go back to reference Kee TH, Vit P, Melendez AJ (2005) Sphingosine kinase signalling in immune cells. Clin Exp Pharmacol Physiol 32(3):153–161PubMedCrossRef Kee TH, Vit P, Melendez AJ (2005) Sphingosine kinase signalling in immune cells. Clin Exp Pharmacol Physiol 32(3):153–161PubMedCrossRef
80.
go back to reference Ewing TM et al (1989) Regulation of epidermal growth factor receptor by progestins and glucocorticoids in human breast cancer cell lines. Int J Cancer 44(4):744–752PubMedCrossRef Ewing TM et al (1989) Regulation of epidermal growth factor receptor by progestins and glucocorticoids in human breast cancer cell lines. Int J Cancer 44(4):744–752PubMedCrossRef
81.
go back to reference Doll F, Pfeilschifter J, Huwiler A (2005) The epidermal growth factor stimulates sphingosine kinase-1 expression and activity in the human mammary carcinoma cell line MCF7. Biochim Biophys Acta 1738(1–3):72–81PubMed Doll F, Pfeilschifter J, Huwiler A (2005) The epidermal growth factor stimulates sphingosine kinase-1 expression and activity in the human mammary carcinoma cell line MCF7. Biochim Biophys Acta 1738(1–3):72–81PubMed
82.
go back to reference Grunberg SM et al (2009) Effectiveness of a single-day three-drug regimen of dexamethasone, palonosetron, and aprepitant for the prevention of acute and delayed nausea and vomiting caused by moderately emetogenic chemotherapy. Support Care Cancer 17(5):589–594PubMedCrossRef Grunberg SM et al (2009) Effectiveness of a single-day three-drug regimen of dexamethasone, palonosetron, and aprepitant for the prevention of acute and delayed nausea and vomiting caused by moderately emetogenic chemotherapy. Support Care Cancer 17(5):589–594PubMedCrossRef
83.
go back to reference Herrington JD, Jaskiewicz AD, Song J (2008) Randomized, placebo-controlled, pilot study evaluating aprepitant single dose plus palonosetron and dexamethasone for the prevention of acute and delayed chemotherapy-induced nausea and vomiting. Cancer 112(9):2080–2087PubMedCrossRef Herrington JD, Jaskiewicz AD, Song J (2008) Randomized, placebo-controlled, pilot study evaluating aprepitant single dose plus palonosetron and dexamethasone for the prevention of acute and delayed chemotherapy-induced nausea and vomiting. Cancer 112(9):2080–2087PubMedCrossRef
84.
go back to reference Lu YS et al (2005) Effects of glucocorticoids on the growth and chemosensitivity of carcinoma cells are heterogeneous and require high concentration of functional glucocorticoid receptors. World J Gastroenterol 11(40):6373–6380PubMed Lu YS et al (2005) Effects of glucocorticoids on the growth and chemosensitivity of carcinoma cells are heterogeneous and require high concentration of functional glucocorticoid receptors. World J Gastroenterol 11(40):6373–6380PubMed
85.
go back to reference Wang H et al (2004) Pretreatment with dexamethasone increases antitumor activity of carboplatin and gemcitabine in mice bearing human cancer xenografts: in vivo activity, pharmacokinetics, and clinical implications for cancer chemotherapy. Clin Cancer Res 10(5):1633–1644PubMedCrossRef Wang H et al (2004) Pretreatment with dexamethasone increases antitumor activity of carboplatin and gemcitabine in mice bearing human cancer xenografts: in vivo activity, pharmacokinetics, and clinical implications for cancer chemotherapy. Clin Cancer Res 10(5):1633–1644PubMedCrossRef
86.
go back to reference Balkwill F (2002) Tumor necrosis factor or tumor promoting factor? Cytokine Growth Factor Rev 13(2):135–141PubMedCrossRef Balkwill F (2002) Tumor necrosis factor or tumor promoting factor? Cytokine Growth Factor Rev 13(2):135–141PubMedCrossRef
87.
go back to reference Loda M et al (1996) Expression of mitogen-activated protein kinase phosphatase-1 in the early phases of human epithelial carcinogenesis. Am J Pathol 149(5):1553–1564PubMed Loda M et al (1996) Expression of mitogen-activated protein kinase phosphatase-1 in the early phases of human epithelial carcinogenesis. Am J Pathol 149(5):1553–1564PubMed
88.
go back to reference Magi-Galluzzi C et al (1997) Mitogen-activated protein kinase phosphatase 1 is overexpressed in prostate cancers and is inversely related to apoptosis. Lab Invest 76(1):37–51PubMed Magi-Galluzzi C et al (1997) Mitogen-activated protein kinase phosphatase 1 is overexpressed in prostate cancers and is inversely related to apoptosis. Lab Invest 76(1):37–51PubMed
89.
go back to reference Braunschweiger PG, Schiffer LM (1981) Antiproliferative effects of corticosteroids in C3H/HeJ mammary tumors and implications for sequential combination chemotherapy. Cancer Res 41(9 Pt 1):3324–3330PubMed Braunschweiger PG, Schiffer LM (1981) Antiproliferative effects of corticosteroids in C3H/HeJ mammary tumors and implications for sequential combination chemotherapy. Cancer Res 41(9 Pt 1):3324–3330PubMed
90.
go back to reference Stuhr LE et al (2006) High-dose, short-term, anti-inflammatory treatment with dexamethasone reduces growth and augments the effects of 5-fluorouracil on dimethyl-alpha-benzanthracene-induced mammary tumors in rats. Scand J Clin Lab Invest 66(6):477–486PubMedCrossRef Stuhr LE et al (2006) High-dose, short-term, anti-inflammatory treatment with dexamethasone reduces growth and augments the effects of 5-fluorouracil on dimethyl-alpha-benzanthracene-induced mammary tumors in rats. Scand J Clin Lab Invest 66(6):477–486PubMedCrossRef
91.
go back to reference Hall RE et al (1990) Steroid hormone receptor gene expression in human breast cancer cells: inverse relationship between oestrogen and glucocorticoid receptor messenger RNA levels. Int J Cancer 46(6):1081–1087PubMedCrossRef Hall RE et al (1990) Steroid hormone receptor gene expression in human breast cancer cells: inverse relationship between oestrogen and glucocorticoid receptor messenger RNA levels. Int J Cancer 46(6):1081–1087PubMedCrossRef
92.
go back to reference de Fazio A et al (1997) Antisense estrogen receptor RNA expression increases epidermal growth factor receptor gene expression in breast cancer cells. Cell Growth Differ 8(8):903–911 de Fazio A et al (1997) Antisense estrogen receptor RNA expression increases epidermal growth factor receptor gene expression in breast cancer cells. Cell Growth Differ 8(8):903–911
93.
go back to reference Krishnan AV, Swami S, Feldman D (2001) Estradiol inhibits glucocorticoid receptor expression and induces glucocorticoid resistance in MCF-7 human breast cancer cells. J Steroid Biochem Mol Biol 77(1):29–37PubMedCrossRef Krishnan AV, Swami S, Feldman D (2001) Estradiol inhibits glucocorticoid receptor expression and induces glucocorticoid resistance in MCF-7 human breast cancer cells. J Steroid Biochem Mol Biol 77(1):29–37PubMedCrossRef
94.
go back to reference Zhang Y et al (2009) Estrogen inhibits glucocorticoid action via protein phosphatase 5 (PP5)-mediated glucocorticoid receptor dephosphorylation. J Biol Chem 284(36):24542–24552PubMedCrossRef Zhang Y et al (2009) Estrogen inhibits glucocorticoid action via protein phosphatase 5 (PP5)-mediated glucocorticoid receptor dephosphorylation. J Biol Chem 284(36):24542–24552PubMedCrossRef
95.
go back to reference Gong H et al (2008) Glucocorticoids antagonize estrogens by glucocorticoid receptor-mediated activation of estrogen sulfotransferase. Cancer Res 68(18):7386–7393PubMedCrossRef Gong H et al (2008) Glucocorticoids antagonize estrogens by glucocorticoid receptor-mediated activation of estrogen sulfotransferase. Cancer Res 68(18):7386–7393PubMedCrossRef
96.
go back to reference Allegra JC et al (1979) Relationship between the progesterone, androgen, and glucocorticoid receptor and response rate to endocrine therapy in metastatic breast cancer. Cancer Res 39(6 Pt 1):1973–1979PubMed Allegra JC et al (1979) Relationship between the progesterone, androgen, and glucocorticoid receptor and response rate to endocrine therapy in metastatic breast cancer. Cancer Res 39(6 Pt 1):1973–1979PubMed
97.
go back to reference Wan Y et al (2001) Separable features of the ligand-binding domain determine the differential subcellular localization and ligand-binding specificity of glucocorticoid receptor and progesterone receptor. Mol Endocrinol 15(1):17–31PubMedCrossRef Wan Y et al (2001) Separable features of the ligand-binding domain determine the differential subcellular localization and ligand-binding specificity of glucocorticoid receptor and progesterone receptor. Mol Endocrinol 15(1):17–31PubMedCrossRef
98.
go back to reference Deroo BJ, Archer TK (2001) Glucocorticoid receptor-mediated chromatin remodeling in vivo. Oncogene 20(24):3039–3046PubMedCrossRef Deroo BJ, Archer TK (2001) Glucocorticoid receptor-mediated chromatin remodeling in vivo. Oncogene 20(24):3039–3046PubMedCrossRef
99.
go back to reference Li X et al (2003) Progesterone and glucocorticoid receptors recruit distinct coactivator complexes and promote distinct patterns of local chromatin modification. Mol Cell Biol 23(11):3763–3773PubMedCrossRef Li X et al (2003) Progesterone and glucocorticoid receptors recruit distinct coactivator complexes and promote distinct patterns of local chromatin modification. Mol Cell Biol 23(11):3763–3773PubMedCrossRef
100.
go back to reference van den Berg HW, Lynch M, Martin JH (1993) The relationship between affinity of progestins and antiprogestins for the progesterone receptor in breast cancer cells (ZR-PR-LT) and ability to down-regulate the receptor: evidence for heterospecific receptor modulation via the glucocorticoid receptor. Eur J Cancer 29A(12):1771–1775PubMed van den Berg HW, Lynch M, Martin JH (1993) The relationship between affinity of progestins and antiprogestins for the progesterone receptor in breast cancer cells (ZR-PR-LT) and ability to down-regulate the receptor: evidence for heterospecific receptor modulation via the glucocorticoid receptor. Eur J Cancer 29A(12):1771–1775PubMed
101.
go back to reference Wan Y, Nordeen SK (2003) Overlapping but distinct profiles of gene expression elicited by glucocorticoids and progestins. Recent Prog Horm Res 58:199–226PubMedCrossRef Wan Y, Nordeen SK (2003) Overlapping but distinct profiles of gene expression elicited by glucocorticoids and progestins. Recent Prog Horm Res 58:199–226PubMedCrossRef
102.
go back to reference Buser AC et al (2007) Progesterone receptor repression of prolactin/signal transducer and activator of transcription 5-mediated transcription of the beta-casein gene in mammary epithelial cells. Mol Endocrinol 21(1):106–125PubMedCrossRef Buser AC et al (2007) Progesterone receptor repression of prolactin/signal transducer and activator of transcription 5-mediated transcription of the beta-casein gene in mammary epithelial cells. Mol Endocrinol 21(1):106–125PubMedCrossRef
103.
go back to reference Quirk SJ et al (1985) Progestins specifically suppress alpha-lactalbumin synthesis and secretion. J Steroid Biochem 23(6A):901–905PubMedCrossRef Quirk SJ et al (1985) Progestins specifically suppress alpha-lactalbumin synthesis and secretion. J Steroid Biochem 23(6A):901–905PubMedCrossRef
104.
go back to reference Wiegratz I, Kuhl H (2004) Progestogen therapies: differences in clinical effects? Trends Endocrinol Metab 15(6):277–285PubMedCrossRef Wiegratz I, Kuhl H (2004) Progestogen therapies: differences in clinical effects? Trends Endocrinol Metab 15(6):277–285PubMedCrossRef
105.
go back to reference Poulin R et al (1991) Multiple actions of synthetic “progestins” on the growth of ZR-75–1 human breast cancer cells: an in vitro model for the simultaneous assay of androgen, progestin, estrogen, and glucocorticoid agonistic and antagonistic activities of steroids. Breast Cancer Res Treat 17(3):197–210PubMedCrossRef Poulin R et al (1991) Multiple actions of synthetic “progestins” on the growth of ZR-75–1 human breast cancer cells: an in vitro model for the simultaneous assay of androgen, progestin, estrogen, and glucocorticoid agonistic and antagonistic activities of steroids. Breast Cancer Res Treat 17(3):197–210PubMedCrossRef
106.
go back to reference Kontula K et al (1983) Binding of progestins to the glucocorticoid receptor. Correlation to their glucocorticoid-like effects on in vitro functions of human mononuclear leukocytes. Biochem Pharmacol 32(9):1511–1518PubMedCrossRef Kontula K et al (1983) Binding of progestins to the glucocorticoid receptor. Correlation to their glucocorticoid-like effects on in vitro functions of human mononuclear leukocytes. Biochem Pharmacol 32(9):1511–1518PubMedCrossRef
107.
go back to reference Parazzini F et al (1993) Treatment with tamoxifen and progestins for metastatic breast cancer in postmenopausal women: a quantitative review of published randomized clinical trials. Oncology 50(6):483–489PubMedCrossRef Parazzini F et al (1993) Treatment with tamoxifen and progestins for metastatic breast cancer in postmenopausal women: a quantitative review of published randomized clinical trials. Oncology 50(6):483–489PubMedCrossRef
108.
go back to reference Willemse PH et al (1990) Adrenal steroids as parameters of the bioavailability of MA and MPA. Eur J Cancer 26(3):359–362PubMedCrossRef Willemse PH et al (1990) Adrenal steroids as parameters of the bioavailability of MA and MPA. Eur J Cancer 26(3):359–362PubMedCrossRef
109.
110.
go back to reference Chlebowski RT et al (2010) Estrogen plus progestin and breast cancer incidence and mortality in postmenopausal women. JAMA 304(15):1684–1692PubMedCrossRef Chlebowski RT et al (2010) Estrogen plus progestin and breast cancer incidence and mortality in postmenopausal women. JAMA 304(15):1684–1692PubMedCrossRef
111.
go back to reference Hofseth LJ et al (1999) Hormone replacement therapy with estrogen or estrogen plus medroxyprogesterone acetate is associated with increased epithelial proliferation in the normal postmenopausal breast. J Clin Endocrinol Metab 84(12):4559–4565PubMedCrossRef Hofseth LJ et al (1999) Hormone replacement therapy with estrogen or estrogen plus medroxyprogesterone acetate is associated with increased epithelial proliferation in the normal postmenopausal breast. J Clin Endocrinol Metab 84(12):4559–4565PubMedCrossRef
112.
go back to reference Wood CE et al (2007) Effects of estradiol with micronized progesterone or medroxyprogesterone acetate on risk markers for breast cancer in postmenopausal monkeys. Breast Cancer Res Treat 101(2):125–134PubMedCrossRef Wood CE et al (2007) Effects of estradiol with micronized progesterone or medroxyprogesterone acetate on risk markers for breast cancer in postmenopausal monkeys. Breast Cancer Res Treat 101(2):125–134PubMedCrossRef
113.
go back to reference Murkes D et al (2011) Effects of percutaneous estradiol-oral progesterone versus oral conjugated equine estrogens-medroxyprogesterone acetate on breast cell proliferation and bcl-2 protein in healthy women. Fertil Steril 95(3):1188–1191PubMedCrossRef Murkes D et al (2011) Effects of percutaneous estradiol-oral progesterone versus oral conjugated equine estrogens-medroxyprogesterone acetate on breast cell proliferation and bcl-2 protein in healthy women. Fertil Steril 95(3):1188–1191PubMedCrossRef
114.
go back to reference Wood CE, Register TC, Cline JM (2009) Transcriptional profiles of progestogen effects in the postmenopausal breast. Breast Cancer Res Treat 114(2):233–242PubMedCrossRef Wood CE, Register TC, Cline JM (2009) Transcriptional profiles of progestogen effects in the postmenopausal breast. Breast Cancer Res Treat 114(2):233–242PubMedCrossRef
Metadata
Title
Glucocorticoid receptor and breast cancer
Authors
Myriam Vilasco
Laudine Communal
Najat Mourra
Aurélie Courtin
Patricia Forgez
Anne Gompel
Publication date
01-11-2011
Publisher
Springer US
Published in
Breast Cancer Research and Treatment / Issue 1/2011
Print ISSN: 0167-6806
Electronic ISSN: 1573-7217
DOI
https://doi.org/10.1007/s10549-011-1689-6

Other articles of this Issue 1/2011

Breast Cancer Research and Treatment 1/2011 Go to the issue
Webinar | 19-02-2024 | 17:30 (CET)

Keynote webinar | Spotlight on antibody–drug conjugates in cancer

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

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