Skip to main content
Top
Published in: Breast Cancer Research and Treatment 1/2008

01-01-2008 | Preclinical Study

Farnesol, a mevalonate pathway intermediate, stimulates MCF-7 breast cancer cell growth through farnesoid-X-receptor-mediated estrogen receptor activation

Authors: Fabrice Journe, Guy Laurent, Carole Chaboteaux, Denis Nonclercq, Virginie Durbecq, Denis Larsimont, Jean-Jacques Body

Published in: Breast Cancer Research and Treatment | Issue 1/2008

Login to get access

Abstract

Farnesoid X receptor (FXR) is a metabolic nuclear receptor expressed in the liver and traditionally considered as a bile acid sensor. Yet, FXR has been recently demonstrated in other tissues and cells, such as the kidneys, the adrenals, and arterial smooth muscle cells. Immunohistochemical data reported in this study point to the expression of FXR in human breast cancer. In addition, FXR expression was also found by Western blotting and immunofluorescence microscopy in breast-cancer-derived cell lines MCF-7 (estrogen receptor [ER]-positive) and MDA-MB-231 (ER-negative). The FXR activator farnesol, a mevalonate pathway intermediate, exerts a mitogenic effect on MCF-7 cells. The growth stimulation is completely suppressed by antiestrogens. In contrast, MDA-MB-231 cells appear farnesol-insensitive, suggesting an involvement of ER in farnesol mitogenicity. In accordance with this interpretation, farnesol induces in MCF-7 cells a decrease of ER level, consistent with a phenomenon of receptor downregulation. Farnesol also increases progesterone receptor (PgR) expression in MCF-7 cells and stimulates ER-mediated gene transactivation in MVLN cells (MCF-7 cells stably transfected with an ER reporter gene). Of note, both effects of farnesol on ER expression and activity are completely suppressed by antiestrogens. In addition, farnesol-induced PgR is markedly reduced by FXR gene silencing (siRNA), demonstrating the involvement of FXR in the estrogenic effects of farnesol. Finally, coimmunoprecipitation experiments (FXR immunoprecipitation followed by Western blot analysis of ER in the immunoprecipitate) produced definite evidence that FXR interacts with ER. Altogether, these observations reveal the hitherto unreported presence of FXR in breast cancer and show that the latter receptor functionally interacts with ER. The occurrence of such a crosstalk calls for some caution regarding the pharmacological use of FXR agonists.
Literature
1.
go back to reference Dumitrescu RG, Cotarla I (2005) Understanding breast cancer risk—where do we stand in 2005? J Cell Mol Med 9(1):208–221PubMedCrossRef Dumitrescu RG, Cotarla I (2005) Understanding breast cancer risk—where do we stand in 2005? J Cell Mol Med 9(1):208–221PubMedCrossRef
2.
go back to reference Jensen EV, Jordan VC (2003) The estrogen receptor: a model for molecular medicine. Clin Cancer Res 9(6):1980–1989PubMed Jensen EV, Jordan VC (2003) The estrogen receptor: a model for molecular medicine. Clin Cancer Res 9(6):1980–1989PubMed
3.
go back to reference Singh RR, Kumar R (2005) Steroid hormone receptor signaling in tumorigenesis. J Cell Biochem 96(3):490–505PubMedCrossRef Singh RR, Kumar R (2005) Steroid hormone receptor signaling in tumorigenesis. J Cell Biochem 96(3):490–505PubMedCrossRef
4.
go back to reference Althuis MD, Fergenbaum JH, Garcia-Closas M, Brinton LA, Madigan MP, Sherman ME (2004) Etiology of hormone receptor-defined breast cancer: a systematic review of the literature. Cancer Epidemiol Biomarkers Prev 13(10):1558–1568PubMed Althuis MD, Fergenbaum JH, Garcia-Closas M, Brinton LA, Madigan MP, Sherman ME (2004) Etiology of hormone receptor-defined breast cancer: a systematic review of the literature. Cancer Epidemiol Biomarkers Prev 13(10):1558–1568PubMed
5.
go back to reference Jordan VC (2004) Selective estrogen receptor modulation: concept and consequences in cancer. Cancer Cell 5(3):207–213PubMedCrossRef Jordan VC (2004) Selective estrogen receptor modulation: concept and consequences in cancer. Cancer Cell 5(3):207–213PubMedCrossRef
6.
go back to reference Geisler J, Lonning PE (2005) Aromatase inhibition: translation into a successful therapeutic approach. Clin Cancer Res 11(8):2809–2821PubMedCrossRef Geisler J, Lonning PE (2005) Aromatase inhibition: translation into a successful therapeutic approach. Clin Cancer Res 11(8):2809–2821PubMedCrossRef
7.
go back to reference Gronemeyer H, Gustafsson JA, Laudet V (2004) Principles for modulation of the nuclear receptor superfamily. Nat Rev Drug Discov 3(11):950–964PubMedCrossRef Gronemeyer H, Gustafsson JA, Laudet V (2004) Principles for modulation of the nuclear receptor superfamily. Nat Rev Drug Discov 3(11):950–964PubMedCrossRef
8.
go back to reference Driggers PH, Segars JH (2002) Estrogen action and cytoplasmic signaling pathways. Part II. The role of growth factors and phosphorylation in estrogen signaling. Trends Endocrinol Metab 13(10):422–427PubMedCrossRef Driggers PH, Segars JH (2002) Estrogen action and cytoplasmic signaling pathways. Part II. The role of growth factors and phosphorylation in estrogen signaling. Trends Endocrinol Metab 13(10):422–427PubMedCrossRef
9.
go back to reference Ohtake F, Takeyama K, Matsumoto T, Kitagawa H, Yamamoto Y, Nohara K, Tohyama C, Krust A, Mimura J, Chambon P et al (2003) Modulation of oestrogen receptor signalling by association with the activated dioxin receptor. Nature 423(6939):545–550PubMedCrossRef Ohtake F, Takeyama K, Matsumoto T, Kitagawa H, Yamamoto Y, Nohara K, Tohyama C, Krust A, Mimura J, Chambon P et al (2003) Modulation of oestrogen receptor signalling by association with the activated dioxin receptor. Nature 423(6939):545–550PubMedCrossRef
10.
go back to reference Clarke RB, Anderson E, Howell A (2004) Steroid receptors in human breast cancer. Trends Endocrinol Metab 15(7):316–323PubMedCrossRef Clarke RB, Anderson E, Howell A (2004) Steroid receptors in human breast cancer. Trends Endocrinol Metab 15(7):316–323PubMedCrossRef
11.
go back to reference Francis GA, Fayard E, Picard F, Auwerx J (2003) Nuclear receptors and the control of metabolism. Annu Rev Physiol 65:261–311PubMedCrossRef Francis GA, Fayard E, Picard F, Auwerx J (2003) Nuclear receptors and the control of metabolism. Annu Rev Physiol 65:261–311PubMedCrossRef
12.
go back to reference Forman BM, Goode E, Chen J, Oro AE, Bradley DJ, Perlmann T, Noonan DJ, Burka LT, McMorris T, Lamph WW et al (1995) Identification of a nuclear receptor that is activated by farnesol metabolites. Cell 81(5):687–693PubMedCrossRef Forman BM, Goode E, Chen J, Oro AE, Bradley DJ, Perlmann T, Noonan DJ, Burka LT, McMorris T, Lamph WW et al (1995) Identification of a nuclear receptor that is activated by farnesol metabolites. Cell 81(5):687–693PubMedCrossRef
13.
go back to reference Nishimaki-Mogami T, Une M, Fujino T, Sato Y, Tamehiro N, Kawahara Y, Shudo K, Inoue K (2004) Identification of intermediates in the bile acid synthetic pathway as ligands for the farnesoid X receptor. J Lipid Res 45(8):1538–1545PubMedCrossRef Nishimaki-Mogami T, Une M, Fujino T, Sato Y, Tamehiro N, Kawahara Y, Shudo K, Inoue K (2004) Identification of intermediates in the bile acid synthetic pathway as ligands for the farnesoid X receptor. J Lipid Res 45(8):1538–1545PubMedCrossRef
14.
go back to reference Makishima M (2005) Nuclear receptors as targets for drug development: regulation of cholesterol and bile acid metabolism by nuclear receptors. J Pharmacol Sci 97(2):177–183PubMedCrossRef Makishima M (2005) Nuclear receptors as targets for drug development: regulation of cholesterol and bile acid metabolism by nuclear receptors. J Pharmacol Sci 97(2):177–183PubMedCrossRef
15.
go back to reference Rogers MJ (2003) New insights into the molecular mechanisms of action of bisphosphonates. Curr Pharm Des 9(32):2643–2658PubMedCrossRef Rogers MJ (2003) New insights into the molecular mechanisms of action of bisphosphonates. Curr Pharm Des 9(32):2643–2658PubMedCrossRef
16.
go back to reference Body JJ (2006) Bisphosphonates for malignancy-related bone disease: current status, future developments. Support Care Cancer 14(5):408–418PubMedCrossRef Body JJ (2006) Bisphosphonates for malignancy-related bone disease: current status, future developments. Support Care Cancer 14(5):408–418PubMedCrossRef
17.
go back to reference Allred DC, Harvey JM, Berardo M, Clark GM (1998) Prognostic and predictive factors in breast cancer by immunohistochemical analysis. Mod Pathol 11(2):155–168PubMed Allred DC, Harvey JM, Berardo M, Clark GM (1998) Prognostic and predictive factors in breast cancer by immunohistochemical analysis. Mod Pathol 11(2):155–168PubMed
18.
go back to reference Demirpence E, Duchesne MJ, Badia E, Gagne D, Pons M (1993) MVLN cells: a bioluminescent MCE-7-derived cell line to study the modulation of estrogenic activity. J Steroid Biochem Mol Biol 46(3):355–364PubMedCrossRef Demirpence E, Duchesne MJ, Badia E, Gagne D, Pons M (1993) MVLN cells: a bioluminescent MCE-7-derived cell line to study the modulation of estrogenic activity. J Steroid Biochem Mol Biol 46(3):355–364PubMedCrossRef
19.
go back to reference Devleeschouwer N, Legros N, Olea-Serrano N, Paridaens R, Leclercq G (1987) Estrogen conjugates and serum factors mediating the estrogenic trophic effect on MCF-7 cell growth. Cancer Res 47(22):5883–5887PubMed Devleeschouwer N, Legros N, Olea-Serrano N, Paridaens R, Leclercq G (1987) Estrogen conjugates and serum factors mediating the estrogenic trophic effect on MCF-7 cell growth. Cancer Res 47(22):5883–5887PubMed
20.
go back to reference Journe F, Body JJ, Leclercq G, Nonclercq D, Laurent G (2004) Estrogen responsiveness of IBEP-2, a new human cell line derived from breast carcinoma. Breast Cancer Res Treat 86(1):39–53PubMedCrossRef Journe F, Body JJ, Leclercq G, Nonclercq D, Laurent G (2004) Estrogen responsiveness of IBEP-2, a new human cell line derived from breast carcinoma. Breast Cancer Res Treat 86(1):39–53PubMedCrossRef
21.
go back to reference Brohee R, Nonclercq D, Journe DN, Toubeau G, Falmagne P, Leclercq G, Heuson-Stiennon JA, Laurent G (2000) Demonstration of estrogen receptors and of estrogen responsiveness in the HKT-1097 cell line derived from diethylstilbestrol-induced kidney tumors. In Vitro Cell Dev Biol Anim 36(10):640–649PubMedCrossRef Brohee R, Nonclercq D, Journe DN, Toubeau G, Falmagne P, Leclercq G, Heuson-Stiennon JA, Laurent G (2000) Demonstration of estrogen receptors and of estrogen responsiveness in the HKT-1097 cell line derived from diethylstilbestrol-induced kidney tumors. In Vitro Cell Dev Biol Anim 36(10):640–649PubMedCrossRef
22.
go back to reference Lee MV, Fong EM, Singer FR, Guenette RS (2001) Bisphosphonate treatment inhibits the growth of prostate cancer cells. Cancer Res 61(6):2602–2608PubMed Lee MV, Fong EM, Singer FR, Guenette RS (2001) Bisphosphonate treatment inhibits the growth of prostate cancer cells. Cancer Res 61(6):2602–2608PubMed
23.
go back to reference Bauer DC (2003) HMG CoA reductase inhibitors and the skeleton: a comprehensive review. Osteoporos Int 14(4):273–282PubMedCrossRef Bauer DC (2003) HMG CoA reductase inhibitors and the skeleton: a comprehensive review. Osteoporos Int 14(4):273–282PubMedCrossRef
24.
go back to reference Swales KE, Korbonits M, Carpenter R, Walsh DT, Warner TD, Bishop-Bailey D (2006) The farnesoid X receptor is expressed in breast cancer and regulates apoptosis and aromatase expression. Cancer Res 66(20):10120–10126PubMedCrossRef Swales KE, Korbonits M, Carpenter R, Walsh DT, Warner TD, Bishop-Bailey D (2006) The farnesoid X receptor is expressed in breast cancer and regulates apoptosis and aromatase expression. Cancer Res 66(20):10120–10126PubMedCrossRef
25.
go back to reference Silva J, Dasgupta S, Wang G, Krishnamurthy K, Ritter E, Bieberich E (2006) Lipids isolated from bone induce the migration of human breast cancer cells. J Lipid Res 47(4):724–733PubMedCrossRef Silva J, Dasgupta S, Wang G, Krishnamurthy K, Ritter E, Bieberich E (2006) Lipids isolated from bone induce the migration of human breast cancer cells. J Lipid Res 47(4):724–733PubMedCrossRef
26.
go back to reference Lange CA, Shen T, Horwitz KB (2000) Phosphorylation of human progesterone receptors at serine-294 by mitogen-activated protein kinase signals their degradation by the 26S proteasome. Proc Natl Acad Sci USA 97(3):1032–1037PubMedCrossRef Lange CA, Shen T, Horwitz KB (2000) Phosphorylation of human progesterone receptors at serine-294 by mitogen-activated protein kinase signals their degradation by the 26S proteasome. Proc Natl Acad Sci USA 97(3):1032–1037PubMedCrossRef
27.
go back to reference Lew JL, Zhao A, Yu J, Huang L, De Pedro N, Pelaez F, Wright SD, Cui J (2004) The farnesoid X receptor controls gene expression in a ligand- and promoter-selective fashion. J Biol Chem 279(10):8856–8861PubMedCrossRef Lew JL, Zhao A, Yu J, Huang L, De Pedro N, Pelaez F, Wright SD, Cui J (2004) The farnesoid X receptor controls gene expression in a ligand- and promoter-selective fashion. J Biol Chem 279(10):8856–8861PubMedCrossRef
28.
go back to reference Davies GF, McFie PJ, Khandelwal RL, Roesler WJ (2002) Unique ability of troglitazone to up-regulate peroxisome proliferator-activated receptor-gamma expression in hepatocytes. J Pharmacol Exp Ther 300(1):72–77PubMedCrossRef Davies GF, McFie PJ, Khandelwal RL, Roesler WJ (2002) Unique ability of troglitazone to up-regulate peroxisome proliferator-activated receptor-gamma expression in hepatocytes. J Pharmacol Exp Ther 300(1):72–77PubMedCrossRef
29.
go back to reference Habeos I, Ziros PG, Psyrogiannis A, Vagenakis AG, Papavassiliou AG (2005) Statins and transcriptional regulation: the FXR connection. Biochem Biophys Res Commun 334(2):601–605PubMedCrossRef Habeos I, Ziros PG, Psyrogiannis A, Vagenakis AG, Papavassiliou AG (2005) Statins and transcriptional regulation: the FXR connection. Biochem Biophys Res Commun 334(2):601–605PubMedCrossRef
30.
go back to reference Leclercq G, Lacroix M, Laios I, Laurent G (2006) Estrogen receptor alpha: impact of ligands on intracellular shuttling and turnover rate in breast cancer cells. Curr Cancer Drug Targets 6(1):39–64PubMedCrossRef Leclercq G, Lacroix M, Laios I, Laurent G (2006) Estrogen receptor alpha: impact of ligands on intracellular shuttling and turnover rate in breast cancer cells. Curr Cancer Drug Targets 6(1):39–64PubMedCrossRef
31.
go back to reference Baker PR, Wilton JC, Jones CE, Stenzel DJ, Watson N, Smith GJ (1992) Bile acids influence the growth, oestrogen receptor and oestrogen-regulated proteins of MCF-7 human breast cancer cells. Br J Cancer 65(4):566–572PubMed Baker PR, Wilton JC, Jones CE, Stenzel DJ, Watson N, Smith GJ (1992) Bile acids influence the growth, oestrogen receptor and oestrogen-regulated proteins of MCF-7 human breast cancer cells. Br J Cancer 65(4):566–572PubMed
32.
go back to reference Safe S, Wormke M, Samudio I (2000) Mechanisms of inhibitory aryl hydrocarbon receptor-estrogen receptor crosstalk in human breast cancer cells. J Mammary Gland Biol Neoplasia 5(3):295–306PubMedCrossRef Safe S, Wormke M, Samudio I (2000) Mechanisms of inhibitory aryl hydrocarbon receptor-estrogen receptor crosstalk in human breast cancer cells. J Mammary Gland Biol Neoplasia 5(3):295–306PubMedCrossRef
33.
go back to reference Wormke M, Stoner M, Saville B, Safe S (2000) Crosstalk between estrogen receptor alpha and the aryl hydrocarbon receptor in breast cancer cells involves unidirectional activation of proteasomes. FEBS Lett 478(1–2):109–112PubMedCrossRef Wormke M, Stoner M, Saville B, Safe S (2000) Crosstalk between estrogen receptor alpha and the aryl hydrocarbon receptor in breast cancer cells involves unidirectional activation of proteasomes. FEBS Lett 478(1–2):109–112PubMedCrossRef
34.
go back to reference Wormke M, Stoner M, Saville B, Walker K, Abdelrahim M, Burghardt R, Safe S (2003) The aryl hydrocarbon receptor mediates degradation of estrogen receptor alpha through activation of proteasomes. Mol Cell Biol 23(6):1843–1855PubMedCrossRef Wormke M, Stoner M, Saville B, Walker K, Abdelrahim M, Burghardt R, Safe S (2003) The aryl hydrocarbon receptor mediates degradation of estrogen receptor alpha through activation of proteasomes. Mol Cell Biol 23(6):1843–1855PubMedCrossRef
35.
go back to reference Pollenz RS (2002) The mechanism of AH receptor protein down-regulation (degradation) and its impact on AH receptor-mediated gene regulation. Chem Biol Interact 141(1–2):41–61PubMedCrossRef Pollenz RS (2002) The mechanism of AH receptor protein down-regulation (degradation) and its impact on AH receptor-mediated gene regulation. Chem Biol Interact 141(1–2):41–61PubMedCrossRef
36.
go back to reference Costarelli V, Sanders TA (2002) Plasma deoxycholic acid concentration is elevated in postmenopausal women with newly diagnosed breast cancer. Eur J Clin Nutr 56(9):925–927PubMedCrossRef Costarelli V, Sanders TA (2002) Plasma deoxycholic acid concentration is elevated in postmenopausal women with newly diagnosed breast cancer. Eur J Clin Nutr 56(9):925–927PubMedCrossRef
37.
go back to reference Raju U, Levitz M, Javitt NB (1990) Bile acids in human breast cyst fluid: the identification of lithocholic acid. J Clin Endocrinol Metab 70(4):1030–1034PubMedCrossRef Raju U, Levitz M, Javitt NB (1990) Bile acids in human breast cyst fluid: the identification of lithocholic acid. J Clin Endocrinol Metab 70(4):1030–1034PubMedCrossRef
38.
go back to reference Javitt NB, Budai K, Miller DG, Cahan AC, Raju U, Levitz M (1994) Breast-gut connection: origin of chenodeoxycholic acid in breast cyst fluid. Lancet 343(8898):633–635PubMedCrossRef Javitt NB, Budai K, Miller DG, Cahan AC, Raju U, Levitz M (1994) Breast-gut connection: origin of chenodeoxycholic acid in breast cyst fluid. Lancet 343(8898):633–635PubMedCrossRef
39.
go back to reference Costarelli V, Sanders TA (2002) Plasma bile acids and risk of breast cancer. IARC Sci Publ 156:305–306PubMed Costarelli V, Sanders TA (2002) Plasma bile acids and risk of breast cancer. IARC Sci Publ 156:305–306PubMed
40.
go back to reference Zhang Y, Lee FY, Barrera G, Lee H, Vales C, Gonzalez FJ, Willson TM, Edwards PA (2006) Activation of the nuclear receptor FXR improves hyperglycemia and hyperlipidemia in diabetic mice. Proc Natl Acad Sci USA 103(4):1006–1011PubMedCrossRef Zhang Y, Lee FY, Barrera G, Lee H, Vales C, Gonzalez FJ, Willson TM, Edwards PA (2006) Activation of the nuclear receptor FXR improves hyperglycemia and hyperlipidemia in diabetic mice. Proc Natl Acad Sci USA 103(4):1006–1011PubMedCrossRef
41.
go back to reference Claudel T, Sturm E, Kuipers F, Staels B (2004) The farnesoid X receptor: a novel drug target? Expert Opin Investig Drugs 13(9):1135–1148PubMedCrossRef Claudel T, Sturm E, Kuipers F, Staels B (2004) The farnesoid X receptor: a novel drug target? Expert Opin Investig Drugs 13(9):1135–1148PubMedCrossRef
42.
go back to reference Pellicciari R, Costantino G, Fiorucci S (2005) Farnesoid X receptor: from structure to potential clinical applications. J Med Chem 48(17):5383–5403PubMedCrossRef Pellicciari R, Costantino G, Fiorucci S (2005) Farnesoid X receptor: from structure to potential clinical applications. J Med Chem 48(17):5383–5403PubMedCrossRef
43.
go back to reference Bishop-Bailey D (2004) FXR as a novel therapeutic target for vascular disease. Drug News Perspect 17(8):499–504PubMedCrossRef Bishop-Bailey D (2004) FXR as a novel therapeutic target for vascular disease. Drug News Perspect 17(8):499–504PubMedCrossRef
Metadata
Title
Farnesol, a mevalonate pathway intermediate, stimulates MCF-7 breast cancer cell growth through farnesoid-X-receptor-mediated estrogen receptor activation
Authors
Fabrice Journe
Guy Laurent
Carole Chaboteaux
Denis Nonclercq
Virginie Durbecq
Denis Larsimont
Jean-Jacques Body
Publication date
01-01-2008
Publisher
Springer US
Published in
Breast Cancer Research and Treatment / Issue 1/2008
Print ISSN: 0167-6806
Electronic ISSN: 1573-7217
DOI
https://doi.org/10.1007/s10549-007-9535-6

Other articles of this Issue 1/2008

Breast Cancer Research and Treatment 1/2008 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