Skip to main content Accessibility help
×
Hostname: page-component-76fb5796d-45l2p Total loading time: 0 Render date: 2024-04-25T14:41:00.725Z Has data issue: false hasContentIssue false

32 - Androgens and the androgen receptor (AR)

from Part 2.3 - Molecular pathways underlying carcinogenesis: nuclear receptors

Published online by Cambridge University Press:  05 February 2015

Nicole L. Moore
Affiliation:
Dame Roma Mitchell Cancer Research Laboratories, Adelaide University/Hanson Institute, Adelaide, South Australia
Margaret M. Centenera
Affiliation:
Dame Roma Mitchell Cancer Research Laboratories, Adelaide University/Hanson Institute, Adelaide, South Australia
Lisa M. Butler
Affiliation:
Dame Roma Mitchell Cancer Research Laboratories, Adelaide University/Hanson Institute, Adelaide, South Australia
Theresa E. Hickey
Affiliation:
Dame Roma Mitchell Cancer Research Laboratories, Adelaide University/Hanson Institute, Adelaide, South Australia
Wayne D. Tilley
Affiliation:
Dame Roma Mitchell Cancer Research Laboratories, Adelaide University/Hanson Institute, Adelaide, South Australia
Edward P. Gelmann
Affiliation:
Columbia University, New York
Charles L. Sawyers
Affiliation:
Memorial Sloan-Kettering Cancer Center, New York
Frank J. Rauscher, III
Affiliation:
The Wistar Institute Cancer Centre, Philadelphia
Get access

Summary

Introduction

Androgens are sex steroid hormones that are produced in both males and females, albeit in different amounts. Androgens are essential for development and maintenance of the male phenotype (1), and are also important in females, where they can either act directly via the androgen receptor (AR) or indirectly as metabolic precursors for estrogen biosynthesis (2). The majority of circulating androgens in males and females are produced by the gonads, under hypothalamic–pituitary regulation, and adrenal glands. The major circulating androgens are dehydroepiandrosterone, DHEA sulfate, androstenedione, and testosterone, which can all be metabolized to other androgens or to estrogens. Testosterone and its intracrine metabolite, 5α-dihydrotestosterone (DHT), are the most potent androgenic hormones, as they bind with high affinity to the AR. Many organs in both males and females are sensitive to androgen action and, depending on the tissue-specific context, androgens regulate multiple cellular processes, including proliferation, differentiation, apoptosis, metabolism, secretory responses, and the synthesis of lipids and fatty acids (3). Accordingly, deregulation of AR expression and/or function is associated with a diverse range of clinical conditions, including androgen insensitivity syndrome (AIS), a form of motor neuron disease known as Kennedy's disease, male and female pattern baldness, hirsutism, acne, male infertility, polycystic ovary syndrome, benign prostatic hyperplasia, and breast and prostate cancer. In particular, the AR is considered an oncogene in the prostate as it plays a critical role at all stages of prostate carcinogenesis, and consequently, inhibition or abrogation of androgen signaling is the goal of hormonal therapies for this disease. In contrast, androgens predominantly have a growth inhibitory role in the breast, consistent with an emerging view of the AR playing a tumor-suppressive role in breast tumorigenesis.

Type
Chapter
Information
Molecular Oncology
Causes of Cancer and Targets for Treatment
, pp. 378 - 391
Publisher: Cambridge University Press
Print publication year: 2013

Access options

Get access to the full version of this content by using one of the access options below. (Log in options will check for institutional or personal access. Content may require purchase if you do not have access.)

References

Wilhelm, D, Koopman, P. The makings of maleness: towards an integrated view of male sexual development. Nature Reviews Genetics 2006;7:620–31.CrossRef
Labrie, F, Luu-The, V, Labrie, C, et al. Endocrine and intracrine sources of androgens in women: inhibition of breast cancer and other roles of androgens and their precursor dehydroepiandrosterone. Endocrine Reviews 2003;24:152–82.CrossRef
Bolton, EC, So, AY, Chaivorapol C, et al. Cell- and gene-specific regulation of primary target genes by the androgen receptor. Genes and Development 2007;21:2005–17.CrossRef
Evans, RM. The steroid and thyroid hormone receptor superfamily. Science 1988;240:889–95.CrossRef
Nuclear Receptors Nomenclature Committee. A unified nomenclature system for the nuclear receptor superfamily. Cell 1999;97:161–3.CrossRef
Lubahn, DB, Brown, TR, Simental, JA, et al. Sequence of the intron/exon junctions of the coding region of the human androgen receptor gene and identification of a point mutation in a family with complete androgen sensitivity. Proceedings of the National Academy of Sciences USA 1989;86:9534–38.CrossRef
Tilley, WD, Marcelli, M, Wilson, JD, McPhaul, MJ. Characterization and expression of a cDNA encoding the human androgen receptor. Proceedings of the National Academy of Sciences USA 1989;86:327–31.CrossRef
Matias, PM, Donner, P, Coelho, R, et al. Structural evidence for ligand specificity in the binding domain of the human androgen receptor. Implications for pathogenic gene mutations. Journal of Biological Chemistry 2000;275:26 164–71.CrossRefGoogle ScholarPubMed
Sack, JS, Kish, KF, Wang, C, et al. Crystallographic structures of the ligand-binding domains of the androgen receptor and its T877A mutant complexed with the natural agonist dihydrotestosterone. Proceedings of the National Academy of Sciences USA 2001;98:4904–9.CrossRef
Bohl, CE, Wu, Z, Miller, DD, Bell, CE, Dalton, JT.Crystal structure of the T877A human androgen receptor ligand-binding domain complexed to cyproterone acetate provides insight for ligand-induced conformational changes and structure-based drug design. Journal of Biological Chemistry 2007;282:13 648–55.CrossRefGoogle ScholarPubMed
Glass, CK, Rosenfeld, MG. The coregulator exchange in transcriptional functions of nuclear receptors. Genes and Development 2000;14:121–41.
Heery, DM, Kalkhoven, E, Hoare, S, Parker, MG. A signature motif in transcriptional co-activators mediates binding to nuclear receptors. Nature 1997;387:733–6.CrossRef
He, B, Wilson, EM. Electrostatic modulation in steroid receptor recruitment of LXXLL and FXXLF motifs. Molecular and Cellular Biology 2003;23:2135–50.CrossRef
Langley, E, Kemppainen, JA, Wilson, EM.Intermolecular NH2-/carboxyl-terminal interactions in androgen receptor dimerization revealed by mutations that cause androgen insensitivity. Journal of Biological Chemistry 1998;273:92–101.CrossRefGoogle ScholarPubMed
Berrevoets, CA, Doesburg, P, Steketee, K, Trapman, J, Brinkmann, AO. Functional interactions of the AF-2 activation domain core region of the human androgen receptor with the amino-terminal domain and with the transcriptional coactivator TIF2 (transcriptional intermediary factor2). Molecular Endocrinology 1998;12:1172–83.CrossRef
Ikonen, T, Palvimo, JJ, Janne, OA.Interaction between the amino- and carboxyl-terminal regions of the rat androgen receptor modulates transcriptional activity and is influenced by nuclear receptor coactivators. Journal of Biological Chemistry 1997;272:29 821–8.CrossRefGoogle ScholarPubMed
He, B, Bowen, NT, Minges, JT, Wilson, EM.Androgen-induced NH2- and COOH-terminal interaction inhibits p160 coactivator recruitment by activation function 2. Journal of Biological Chemistry 2001;276:42 293–301.CrossRefGoogle ScholarPubMed
Schaufele, F, Carbonell, X, Guerbadot, M, et al. The structural basis of androgen receptor activation: intramolecular and intermolecular amino-carboxy interactions. Proceedings of the National Academy of Sciences USA 2005;102:9802–7.CrossRef
van Royen, ME, Cunha, SM, Brink, MC, et al. Compartmentalization of androgen receptor protein-protein interactions in living cells. Journal of Cell Biology 2007;177:63–72.CrossRefGoogle ScholarPubMed
Jenster, G, van der Korput, HA, Trapman, J, Brinkmann, AO.Identification of two transcription activation units in the N-terminal domain of the human androgen receptor. Journal of Biological Chemistry 1995;270:7341–6.CrossRefGoogle ScholarPubMed
He, B, Gampe, RT, Kole, AJ, et al. Structural basis for androgen receptor interdomain and coactivator interactions suggests a transition in nuclear receptor activation function dominance. Molecular Cell 2004;16:425–38.CrossRef
He, B, Kemppainen, JA, Wilson, EM.FXXLF and WXXLF sequences mediate the NH2-terminal interaction with the ligand binding domain of the androgen receptor. Journal of Biological Chemistry 2000;275:22 986–94.CrossRefGoogle ScholarPubMed
Baumann, CT, Lim, CS, Hager, GL. Intracellular localization and trafficking of steroid receptors. Cell Biochemistry and Biophysics 1999;31:119–27.CrossRef
Jenster, G, Trapman, J, Brinkmann, AO. Nuclear import of the human androgen receptor. Biochemical Journal 1993;293:761–8.CrossRef
Jenster, G, van der Korput, HA, van Vroonhoven, C, et al. Domains of the human androgen receptor involved in steroid binding, transcriptional activation, and subcellular localization. Molecular Endocrinology 1991;5:1396–404.CrossRef
Zou, J, Guo, Y, Guettouche, T, Smith, DF, Voellmy, R. Repression of heat shock transcription factor HSF1 activation by HSP90 (HSP90 complex) that forms a stress-sensitive complex with HSF1. Cell 1998;94:471–80.CrossRef
Fu, M, Rao, M, Wang, C, et al. Acetylation of androgen receptor enhances coactivator binding and promotes prostate cancer cell growth. Molecular and Cellular Biology 2003;23:8563–75.CrossRef
Tanner, T, Claessens, F, Haelens, A. The hinge region of the androgen receptor plays a role in proteasome-mediated transcriptional activation. Annals of the New York Academy of Science 2004;1030:587–92.CrossRef
Zhou, ZX, Kemppainen, JA, Wilson, EM. Identification of three proline-directed phosphorylation sites in the human androgen receptor. Molecular Endocrinology 1995;9:605–15.
Faus, H and Haendler, B. Post translational modifications of steroid receptors. Biomedicine and Pharmacotherapy 2006; 60:520–8.CrossRef
Glass, CK. Differential recognition of target genes by nuclear receptor monomers, dimers, and heterodimers. Endocrine Reviews 1994;15:391–407.
Zilliacus, J, Dahlman-Wright, K, Carlstedt-Duke, J, Gustafsson, JA.Zinc coordination scheme for the C-terminal zinc binding site of nuclear hormone receptors. Journal of Steroid Biochemistry and Molecular Biology 1992;42:131–9.CrossRefGoogle ScholarPubMed
Freedman, LP, Luisi, BF, Korszun, ZR, et al. The function and structure of the metal coordination sites within the glucocorticoid receptor DNA binding domain. Nature 1988;334:543–6.CrossRef
Luisi, BF, Xu, WX, Otwinowski, Z, et al. Crystallographic analysis of the interaction of the glucocorticoid receptor with DNA. Nature 1991;352:497–505.CrossRef
Schoenmakers, E, Alen, P, Verrijdt, G, et al. Differential DNA binding by the androgen and glucocorticoid receptors involves the second Zn-finger and a C-terminal extension of the DNA-binding domains. Biochemical Journal 1999;341:515–21.CrossRef
Umesono, K, Evans, RM. Determinants of target gene specificity for steroid/thyroid hormone receptors. Cell 1989;57:1139–46.CrossRef
Dahlman-Wright, K, Wright, A, Gustafsson, JA, Carlstedt-Duke, J.Interaction of the glucocorticoid receptor DNA-binding domain with DNA as a dimer is mediated by a short segment of five amino acids. Journal of Biological Chemistry 1991;266:3107–12.Google ScholarPubMed
Remerowski, ML, Kellenbach, E, Boelens, R, et al. 1H NMR studies of DNA recognition by the glucocorticoid receptor: complex of the DNA binding domain with a half-site response element. Biochemistry 1991;30:11 620–4.
Nazareth, LV, Stenoien, DL, Bingman, WE, 3rd, et al. A C619Y mutation in the human androgen receptor causes inactivation and mislocalization of the receptor with concomitant sequestration of SRC-1 (steroid receptor coactivator 1). Molecular Endocrinology 1999;13:2065–75.CrossRef
Schoenmakers, E, Verrijdt, G, Peeters, B, et al. Differences in DNA binding characteristics of the androgen and glucocorticoid receptors can determine hormone-specific responses. Journal of Biological Chemistry 2000;275:12 290–7.CrossRefGoogle ScholarPubMed
Haelens, A, Verrijdt, G, Callewaert, L, et al. Androgen-receptor-specific DNA binding to an element in the first exon of the human secretory component gene. Biochemical Journal 2001;353:611–20.CrossRef
Beato, M. Gene regulation by steroid hormones. Cell 1989;56():335–44.
Roche, PJ, Hoare, SA, Parker, MG. A consensus DNA-binding site for the androgen receptor. Molecular Endocrinology 1992;6:2229–35.
Claessens, F, Celis, L, Peeters, B, et al. Functional characterization of an androgen response element in the first intron of the C3(1) gene of prostatic binding protein. Biochemical and Biophysical Research Communications 1989;164:833–40.CrossRef
Claessens, F, Alen, P, Devos, A, et al. The androgen-specific probasin response element 2 interacts differentially with androgen and glucocorticoid receptors. Journal of Biological Chemistry 1996;271:19 013–6.CrossRefGoogle ScholarPubMed
Rennie, PS, Bruchovsky, N, Leco, KJ, et al. Characterization of two cis-acting DNA elements involved in the androgen regulation of the probasin gene. Molecular Endocrinology 1993;7:23–36.
Verrijdt, G, Schoenmakers, E, Alen, P, et al. Androgen specificity of a response unit upstream of the human secretory component gene is mediated by differential receptor binding to an essential androgen response element. Molecular Endocrinology 1999;13:1558–70.CrossRef
Kasper, S, Rennie, PS, Bruchovsky, N, et al. Cooperative binding of androgen receptors to two DNA sequences is required for androgen induction of the probasin gene. Journal of Biological Chemistry 1994;269:31 763–9.Google ScholarPubMed
Verrijdt, G, Haelens, A, Claessens, F. Selective DNA recognition by the androgen receptor as a mechanism for hormone-specific regulation of gene expression. Molecular Genetics and Metabolism 2003;78:175–85.CrossRef
Haelens, A, Verrijdt, G, Callewaert, L, et al. DNA recognition by the androgen receptor: evidence for an alternative DNA-dependent dimerization, and an active role of sequences flanking the response element on transactivation. Biochemical Journal 2003;369:141–51.CrossRef
Centenera, MM, Harris, JM, Tilley, WD, Butler, LM. The contribution of different androgen receptor domains to receptor dimerization and signaling. Molecular Endocrinology 2008;22:2373–82.CrossRef
Chamberlain, NL, Whitacre, DC, Miesfeld, RL.Delineation of two distinct type 1 activation functions in the androgen receptor amino-terminal domain. Journal of Biological Chemistry 1996;271:26 772–8.CrossRefGoogle ScholarPubMed
Edwards, A, Hammond, HA, Jin, L, Caskey, CT, Chakraborty, R. Genetic variation at five trimeric and tetrameric tandem repeat loci in four human population groups. Genomics 1992;12:241–53.CrossRef
Sartor, O, Zheng, Q, Eastham, JA. Androgen receptor gene CAG repeat length varies in a race-specific fashion in men without prostate cancer. Urology 1999;53:378–80.CrossRef
Beilin, J, Ball, EM, Favaloro, JM, Zajac, JD.Effect of the androgen receptor CAG repeat polymorphism on transcriptional activity: specificity in prostate and non-prostate cell lines. Journal of Molecular Endocrinology 2000;25:85–96.CrossRefGoogle ScholarPubMed
Kazemi-Esfarjani, P, Trifiro, MA, Pinsky, L. Evidence for a repressive function of the long polyglutamine tract in the human androgen receptor: possible pathogenetic relevance for the (CAG)n-expanded neuronopathies. Human Molecular Genetics 1995;4:523–7.CrossRef
Tut, TG, Ghadessy, FJ, Trifiro, MA, Pinsky, L, Yong, EL.Long polyglutamine tracts in the androgen receptor are associated with reduced trans-activation, impaired sperm production, and male infertility. Journal of Clinical Endocrinology and Metabolism 1997;82:3777–82.Google ScholarPubMed
Buchanan, G, Yang, M, Cheong, A, et al. Structural and functional consequences of glutamine tract variation in the androgen receptor. Human Molecular Genetics 2004;13:1677–92.CrossRef
La Spada, AR, Wilson, EM, Lubahn, DB, Harding, AE, Fischbeck, KH. Androgen receptor gene mutations in X-linked spinal and bulbar muscular atrophy. Nature 1991;352:77–9.CrossRef
MacLean, HE, Warne, GL, Zajac, JD.Spinal and bulbar muscular atrophy: androgen receptor dysfunction caused by a trinucleotide repeat expansion. Journal of the Neurological Sciences 1996;135:149–57.CrossRefGoogle ScholarPubMed
Giovannucci, E, Stampfer, MJ, Krithivas, K, et al. The CAG repeat within the androgen receptor gene and its relationship to prostate cancer. Proceedings of the National Academy of Sciences USA 1997;94:3320–3.CrossRef
Hakimi, JM, Schoenberg, MP, Rondinelli, RH, Piantadosi, S, Barrack, ER. Androgen receptor variants with short glutamine or glycine repeats may identify unique subpopulations of men with prostate cancer. Clinical Cancer Research 1997;3:1599–608.
Hsing, AW, Gao, YT, Wu, G, et al. Polymorphic CAG and GGN repeat lengths in the androgen receptor gene and prostate cancer risk: a population-based case-control study in China. Cancer Research 2000;60:5111–16.
Price, DK, Chau, CH, Till, C, et al. Androgen receptor CAG repeat length and association with prostate cancer risk: results from the prostate cancer prevention trial. Journal of Urology 2010;184:2297–302.CrossRefGoogle ScholarPubMed
Lindström, S, Ma, J, Altshuler, D, et al. A large study of androgen receptor germline variants and their relation to sex hormone levels and prostate cancer risk. Results from the National Cancer Institute Breast and Prostate Cancer Cohort Consortium. Journal of Clinical Endocrinology and Metabolism 2010;95:E121–7.CrossRefGoogle ScholarPubMed
Smith, DF, Toft, DO. Steroid receptors and their associated proteins. Molecular Endocrinology 1993;7:4–11.
Vanaja, DK, Mitchell, SH, Toft, DO, Young, CY. Effect of geldanamycin on androgen receptor function and stability. Cell Stress Chaperones 2002;7:55–64.2.0.CO;2>CrossRef
Pratt, WB, Toft, DO. Steroid receptor interactions with heat shock protein and immunophilin chaperones. Endocrine Reviews 1997;18:306–60.
Claessens, F, Verrijdt, G, Schoenmakers, E, et al. Selective DNA binding by the androgen receptor as a mechanism for hormone-specific gene regulation. Journal of Steroid Biochemistry and Molecular Biology 2001;76:23–30.CrossRefGoogle ScholarPubMed
Rachez, C, Freedman, LP. Mediator complexes and transcription. Current Opinion in Cell Biology 2001;13:274–80.CrossRef
Chmelar, R, Buchanan, G, Need, EF, et al. Androgen receptor coregulators and their involvement in the development and progression of prostate cancer. International Journal of Cancer 2006;120: 719–33.CrossRefGoogle Scholar
Wang, Q, Li, W, Liu, XS, et al. A hierarchical network of transcription factors governs androgen receptor-dependent prostate cancer growth. Molecular Cell 2007;27:380–92.CrossRef
Massie, CE, Adryan, B, Barbosa-Morais, NL, et al. New androgen receptor genomic targets show an interaction with the ETS1 transcription factor. EMBO Reports 2007;8:871–8.CrossRef
Lupien, M, Brown, M. Cistromics of hormone-dependent cancer. Endocrine-Related Cancer 2009;16:381–9.CrossRef
Foradori, CD, Weiser, MJ, Handa, RJ. Non-genomic actions of androgens. Frontiers in Neuroendocrinology 2008;29:169–81.CrossRef
Heinlein, CA, Chang, C. The roles of androgen receptors and androgen-binding proteins in nongenomic androgen actions. Molecular Endocrinology 2002;16:2181–7.CrossRef
Baron, S, Manim, M, Beaudoin, C, et al. Androgen receptor mediates non-genomic activation of phosphatidylinositol 3-OH kinase in androgen-sensitive epithelial cells. Journal of Biological Chemistry 2004;9:14 579–86.CrossRefGoogle Scholar
Migliaccio, A, Castoria, G, Di Domenico, M, et al. Steroid-induced androgen receptor-oestradiol receptor beta-Src complex triggers prostate cancer cell proliferation. EMBO Journal 2000;19:5406–17.CrossRef
Walters, KA, Simanainen, U, Handelsman, DJ. Molecular insights into androgen actions in male and female reproductive function from androgen receptor knockout models. Human Reproduction Update 2010;16: 543–58.CrossRef
Yeh, S, Tsai, MY, Xu, Q, et al. Generation and characterization of androgen receptor knockout (ARKO) mice: an in vivo model for the study of androgen functions in selective tissues, Proceedings of the National Academy of Sciences USA 2002;99:13 498–503.
Sato, T, Kawano, H, Kato, S.Study of androgen action in bone by analysis of androgen-receptor deficient mice. Journal of Bone and Mineral Metabolism 2002;20:326–30.CrossRefGoogle ScholarPubMed
Notini, AJ, Davey, RA, McManus, JF, et al. Genomic actions of the androgen receptor are required for normal male sexual differentiation in a mouse model. Journal of Molecular Endocrinology 2005;35:547–55.CrossRefGoogle ScholarPubMed
Holdcraft, RW, Braun, RE. Androgen receptor function is required in Sertoli cells for the terminal differentiation of haploid spermatids. Development 2004;131:459–67.CrossRef
Matsumoto, T, Takeyama, K, Sato, T, Kato, S.Androgen receptor functions from reverse genetic models. Journal of Steroid Biochemistry and Molecular Biology 2003;85:95–9.CrossRefGoogle ScholarPubMed
Sato, T, Matsumoto, T, Kawano, H, et al. Brain masculinization requires androgen receptor function Proceedings of the National Academy of Sciences USA 2004;101:1673–8.
Yeh, S, Hu, YC, Wang, PH, et al. Abnormal mammary gland development and growth retardation in female mice and MCF7 breast cancer cells lacking androgen receptor. Journal of Experimental Medicine 2003;198:1899–908.CrossRefGoogle ScholarPubMed
Hu, YC, Wang, PH, Yeh, S, et al. Subfertility and defective folliculogenesis in female mice lacking androgen receptor. Proceedings of the National Academy of Sciences USA 2004;101:11 209–14.
Shiina, H, Matsumoto, T, Sato, T, et al. Premature ovarian failure in androgen receptor-deficient mice. Proceedings of the National Academy of Sciences USA 2006;103:224–9.CrossRef
Walters, KA, Allan, CM, Jimenez, M, et al. Female mice haploinsufficient for an inactivated androgen receptor (AR) exhibit age-dependent defects that resemble the AR null phenotype of dysfunctional late follicle development, ovulation, and fertility. Endocrinology 2007;148:3674–84.CrossRef
Walters, KA, McTavish, KJ, Seneviratne, MG, et al. Subfertile female androgen receptor knockout mice exhibit defects in neuroendocrine signaling, intraovarian function, and uterine development but not uterine function. Endocrinology 2009;150:3274–82.CrossRef
Simanainen, U, Allan, CM, Lim, P, et al. Disruption of prostate epithelial androgen receptor impedes prostate lobe-specific growth and function. Endocrinology 2007;148:2264–72.CrossRef
Simanainen, U, McNamara, K, Gao, YR, Handelsman, DJ.Androgen sensitivity of prostate epithelium is enhanced by postnatal androgen receptor inactivation. American Journal of Physiology, Endocrinology and Metabolism 2009;296: E1335–43.CrossRefGoogle ScholarPubMed
Wu, CT, Altuwaijri, S, Ricke, WA, et al. Increased prostate cell proliferation and loss of cell differentiation in mice lacking prostate epithelial androgen receptor. Proceedings of the National Academy of Sciences USA 2007; 104:12 679–84.
Sen, A; Hammes, SR. Granulosa cell-specific androgen receptors are critical regulators of ovarian development and function. Molecular Endocrinology 2010;24:1393–403.CrossRef
Peters, AA, Buchanan, G, Ricciardelli, C, et al. Androgen receptor inhibits estrogen receptor-alpha activity and is prognostic in breast cancer. Cancer Research 2009;69:6131–40.CrossRef
Dimitrakakis, C, Zhou, J, Wang, J, et al. A physiologic role for testosterone in limiting estrogenic stimulation of the breast. Menopause 2003;10:292–8.CrossRef
Ando, S, De Amicis, F, Rago, V, et al. Breast cancer: from estrogen to androgen receptor. Molecular and Cellular Endocrinology 2002;193:121–8.CrossRef
Greenberg, NM, DeMayo, F, Finegold, MJ, et al. Prostate cancer in a transgenic mouse, Proceedings of the National Academy of Sciences USA 1995;92:3439–43.
Gingrich, JR, Barrios, RJ, Morton, RA, et al. Metastatic prostate cancer in a transgenic mouse. Cancer Research 1996;56:4096–102.
Niu, Y, Altuwaijri, S, Yeh, S, et al. Targeting the stromal androgen receptor in primary prostate tumors at earlier stages. Proceedings of the National Academy of Sciences USA 2008;105:12 188–93.
Stanbrough, M, Leav, I, Kwan, PW, Bubley, GJ, Balk, SP. Prostatic intraepithelial neoplasia in mice expressing an androgen receptor transgene in prostate epithelium. Proceedings of the National Academy of Sciences USA 2001;98:10 823–28.
Albertelli, MA, Scheller, A, Brogley, M, Robins, DM. Replacing the mouse androgen receptor with human alleles demonstrates glutamine tract length-dependent effects on physiology and tumorigenesis in mice. Molecular Endocrinology 2006;20:1248–60.CrossRef
Albertelli, MA, O’Mahony, OA, Brogley, M, et al. Glutamine tract length of human androgen receptors affects hormone-dependent and -independent prostate cancer in mice. Human Molecular Genetics 2008;17:98–110.CrossRef
Han, G, Foster, BA, Mistry, S, et al. Hormone status selects for spontaneous somatic androgen receptor variants that demonstrate specific ligand and cofactor dependent activities in autochthonous prostate cancer. Journal of Biological Chemistry 2001;276:11 204–13.CrossRefGoogle ScholarPubMed
Han, G, Buchanan, G, Ittmann, M, et al. Mutation of the androgen receptor causes oncogenic transformation of the prostate. Proceedings of the National Academy of Sciences USA 2005;102:1151–6.CrossRef
O’Mahony, OA, Steinkamp, MP, Albertelli, MA et al. Profiling human androgen receptor mutations reveals treatment effects in a mouse model of prostate cancer. Molecular Cancer Research 2008;6:1691–701.
Gottlieb, B, Lehvaslaiho, H, Beitel, LK, et al. The Androgen Receptor Gene Mutations Database. Nucleic Acids Research 1998;26:234–8.CrossRef
Brinkmann, AO. Molecular basis of androgen insensitivity. Molecular and Cellular Endocrinology 2001;179:105–9.CrossRef
Gottlieb, B, Beitel, LK, Wu, JH, Trifiro M. The androgen receptor gene mutations database (ARDB): 2004 update. Human Mutation 2004;23:527–33.CrossRef
Taplin, ME, Bubley, GJ, Shuster, TD, et al. Mutation of the androgen-receptor gene in metastatic androgen-independent prostate cancer. New England Journal of Medicine 1995;332: 1393–8.CrossRefGoogle ScholarPubMed
Steinkamp, MP, O’Mahony, OA, Brogley, M, et al. Treatment-dependent androgen receptor mutations in prostate cancer exploit multiple mechanisms to evade therapy. Cancer Research 2009;69:4434–42.CrossRef
Jiang, Y, Palma, JF, Agus, DB, Wang, Y, Gross, ME. Detection of androgen receptor mutations in circulating tumor cells in castration-resistant prostate cancer. Clinical Chemistry 2010;56:1492–5.CrossRef
Marcelli, M, Ittmann, M, Mariani, S, et al. Androgen receptor mutations in prostate cancer. Cancer Research 2000;60:944–9.
Suzuki, H, Sato, N, Watabe, Y, et al. Androgen receptor gene mutations in human prostate cancer. Journal of Steroid Biochemistry and Molecular Biology 1993;46:759–65.CrossRefGoogle ScholarPubMed
Culig, Z, Hobisch, A, Cronauer, MV, et al. Mutant androgen receptor detected in an advanced-stage prostatic carcinoma is activated by adrenal androgens and progesterone. Molecular Endocrinology 1993;7:1541–50.
Wallen, MJ, Linja, M, Kaartinen, K, Schleutker, J, Visakorpi, T.Androgen receptor gene mutations in hormone-refractory prostate cancer. Journal of Pathology 1999;189:559–63.3.0.CO;2-Y>CrossRefGoogle ScholarPubMed
Castagnaro, M, Yandell, DW, Dockhorn-Dworniczak, B, Wolfe, HJ, Poremba, C. Androgen receptor gene mutations and p53 gene analysis in advanced prostate cancer. Verhandlung der Deutschen Gesellschaft für Pathologie 1993;77:119–23.
Tilley, WD, Buchanan, G, Hickey, TE, Bentel, JM. Mutations in the androgen receptor gene are associated with progression of human prostate cancer to androgen independence. Clinical Cancer Research 1996;2:277–85.
Taplin, ME, Bubley, GJ, Ko, YJ, et al. Selection for androgen receptor mutations in prostate cancers treated with androgen antagonist. Cancer Research 1999;59:2511–15.
Watanabe, M, Ushijima, T, Shiraishi, T, et al. Genetic alterations of androgen receptor gene in Japanese human prostate cancer. Japanese Journal of Clinical Oncology 1997;27:389–93.CrossRefGoogle ScholarPubMed
Elo, JP, Kvist, L, Leinonen, K, et al. Mutated human androgen receptor gene detected in a prostatic cancer patient is also activated by estradiol. Journal of Clinical Endocrinology and Metabolism 1995;80:3494–500.CrossRefGoogle Scholar
Suzuki, H, Akakura, K, Komiya, A, et al. Codon 877 mutation in the androgen receptor gene in advanced prostate cancer: relation to antiandrogen withdrawal syndrome. Prostate 1996;29: 153–8.3.0.CO;2-5>CrossRef
Gaddipati, JP, McLeod, DG, Heidenberg, HB, et al. Frequent detection of codon 877 mutation in the androgen receptor gene in advanced prostate cancers. Cancer Research 1994;54:2861–4.
Newmark, JR, Hardy, DO, Tonb, DC, et al. Androgen receptor gene mutations in human prostate cancer. Proceedings of the National Academy of Sciences USA 1992;89:6319–23.CrossRef
Buchanan, G, Irvine, RA, Coetzee, GA, Tilley, WD. Contribution of the androgen receptor to prostate cancer predisposition and progression. Cancer Metastasis Reviews 2001;20:207–23.CrossRef
Shi, XB, Ma, AH, Xia, L, Kung, HJ, Vere, White RW. Functional analysis of 44 mutant androgen receptors from human prostate cancer. Cancer Research 2002;62: 1496–502.
Buchanan, G, Greenberg, NM, Scher, HI, et al. Collocation of androgen receptor gene mutations in prostate cancer. Clinical Cancer Research 2001;7:1273–81.
Scher, HI, Buchanan, G, Gerald, W, Butler, LM, Tilley, WD. Targeting the androgen receptor: improving outcomes for castration-resistant prostate cancer. Endocrine-Related Cancer 2004;11:459–76.CrossRef
Bergerat, JP, Ceraline, J. Pleiotropic functional properties of androgen receptor mutants in prostate cancer. Human Mutation 2009;30:145–57.CrossRef
Buchanan, G, Birrell, SN, Peters, AA et al. Decreased androgen receptor levels and receptor function in breast cancer contribute to the failure of response to medroxyprogesterone acetate. Cancer Research 2005;65:8487–96.CrossRef
Birrell, SN, Butler, LM, Harris, JM, Buchanan, G, Tilley, WD. Disruption of androgen receptor signaling by synthetic progestins may increase risk of developing breast cancer. FASEB Journal 2007;21:2285–93.CrossRef
Birrell, SN, Roder, DM, Horsfall, DJ, Bentel, JM, Tilley, WD.Medroxyprogesterone acetate therapy in advanced breast cancer: the predictive value of androgen receptor expression. Journal of Clinical Oncology 1995;13:1572–7.CrossRefGoogle ScholarPubMed
Hu, SY, Liu, T, Liu, Z, et al. Identification of a novel germline missense mutation of the androgen receptor in African American men with familial prostate cancer. Asian Journal of Andrology 2010;123:336–43.CrossRefGoogle Scholar
Mononen, N, Syrjakoski, K, Matikainen, M, et al. Two percent of Finnish prostate cancer patients have a germ-line mutation in the hormone-binding domain of the androgen receptor gene. Cancer Research 2000;60:6479–81.
Koivisto, PA, Hyytinen, ER, Matikainen, M, et al. Germline mutation analysis of the androgen receptor gene in Finnish patients with prostate cancer. Journal of Urology 2004;171:431–3.CrossRefGoogle ScholarPubMed
Wooster, R, Mangion, J, Eeles, R, et al. A germline mutation in the androgen receptor gene in two brothers with breast cancer and Reifenstein syndrome. Nature Genetics 1992;2:132–4.CrossRef
Lobaccaro, JM, Lumbroso, S, Belon, C, et al. Androgen receptor gene mutation in male breast cancer. Human Molecular Genetics 1993;2:1799–802.CrossRef
Buchanan, G, Yang, M, Harris, JM, et al. Mutations at the boundary of the hinge and ligand binding domain of the androgen receptor confer increased transactivation function. Molecular Endocrinology 2001;15:46–56.CrossRef
Estebanez-Perpina, E, Arnold, LA, Nguyen, P, et al. A surface on the androgen receptor that allosterically regulates coactivator binding. Proceedings of the National Academy of Sciences USA 2007;104:16 074–9.
Veldscholte, J, Ris-Stalpers, C, Kuiper, GG, et al. A mutation in the ligand binding domain of the androgen receptor of human LNCaP cells affects steroid binding characteristics and response to anti-androgens. Biochemical and Biophysical Research Communications 1990;173:534–40.CrossRef
Berrevoets, CA, Veldscholte, J, Mulder, E.Effects of antiandrogens on transformation and transcription activation of wild-type and mutated (LNCaP) androgen receptors. Journal of Steroid Biochemistry and Molecular Biology 1993;46:731–6.CrossRefGoogle ScholarPubMed
Veldscholte, J, Berrevoets, CA, Ris-Stalpers, C, et al. The androgen receptor in LNCaP cells contains a mutation in the ligand binding domain which affects steroid binding characteristics and response to antiandrogens. Journal of Steroid Biochemistry and Molecular Biology 1992;41:665–9.CrossRefGoogle ScholarPubMed
Taplin, ME, Rajeshkumar, B, Halabi, S, et al. Androgen receptor mutations in androgen-independent prostate cancer: Cancer and Leukemia Group B Study 9663. Journal of Clinical Oncology 2003;21:2673–8.CrossRefGoogle Scholar
Ceraline, J, Cruchant, MD, Erdmann, E, et al. Constitutive activation of the androgen receptor by a point mutation in the hinge region: a new mechanism for androgen-independent growth in prostate cancer. International Journal of Cancer 2004;108:152–7.CrossRefGoogle ScholarPubMed
He, B, Gampe, RT, Hnat, AT, et al. Probing the functional link between androgen receptor coactivator and ligand-binding sites in prostate cancer and androgen insensitivity. Journal of Biological Chemistry 2006;281:6648–63.CrossRefGoogle ScholarPubMed
Askew, EB, Gampe, RT, Stanley, TB, Faggart, JL, Wilson, EM.Modulation of androgen receptor activation function 2 by testosterone and dihydrotesto-sterone. Journal of Biological Chemistry 2007;282: 25 801–16.CrossRefGoogle Scholar
Duff, J, McEwan, IJ. Mutation of histidine 874 in the androgen receptor ligand-binding domain leads to promiscuous ligand activation and altered p160 coactivator interactions. Molecular Endocrinology 2005;19:2943–54.CrossRef
McDonald, S, Brive, L, Agus, DB, Scher, HI, Ely, KR. Ligand responsiveness in human prostate cancer: structural analysis of mutant androgen receptors from LNCaP and CWR22 tumors. Cancer Research 2000;60:2317–22.
Matias, PM, Carrondo, MA, Coelho, R, et al. Structural basis for the glucocorticoid response in a mutant human androgen receptor (AR(ccr)) derived from an androgen-independent prostate cancer. Journal of Medicinal Chemistry 2002;45:1439–46.CrossRefGoogle Scholar
Brooke, GN, Parker, MG, Bevan, CL. Mechanisms of androgen receptor activation in advanced prostate cancer: differential co-activator recruitment and gene expression. Oncogene 2008;27: 2941–50.CrossRef
Krishnan, AV, Zhao, XY, Swami, S, et al. A glucocorticoid-responsive mutant androgen receptor exhibits unique ligand specificity: therapeutic implications for androgen-independent prostate cancer. Endocrinology 2002;143:1889–900.CrossRef
van de Wijngaart, DJ, Molier, M, Lusher, SJ, et al. Systematic structure-function analysis of androgen receptor Leu701 mutants explains the properties of the prostate cancer mutant L701H. Journal of Biological Chemistry 2010; 285: 5097–105.CrossRefGoogle ScholarPubMed
Yeh, SH, Chiu, CM, Chen, CL, et al. Somatic mutations at the trinucleotide repeats of androgen receptor gene in male hepatocellular carcinoma. International Journal of Cancer 2007;120:1610–17.CrossRefGoogle ScholarPubMed
Monge, A, Jagla, M, Lapouge, G, et al. Unfaithfulness and promiscuity of a mutant androgen receptor in a hormone-refractory prostate cancer. Cellular and Molecular Life Sciences 2006;63: 487–97.CrossRef
Shi, XB, Xue, L, Tepper, CG. The oncogenic potential of a prostate cancer-derived androgen receptor mutant, Prostate 2007;67:591–602.
Haelens, A, Tanner, T, Denayer, S, Callewaert, L, Claessens, F. The hinge region regulates DNA binding, nuclear translocation, and transactivation of the androgen receptor. Cancer Research 2007;67:4514–23.CrossRef
Aarnisalo, P, Santti, H, Poukka, H, Palvimo, JJ, Janne, OA. Transcription activating and repressing functions of the androgen receptor are differentially influenced by mutations in the deoxyribonucleic acid-binding domain. Endocrinology 1999;140:3097–105.CrossRef
Poujol, N, Lobaccaro, JM, Chiche, L, Lumbroso, S, Sultan, C. Functional and structural analysis of R607Q and R608K androgen receptor substitutions associated with male breast cancer. Molecular and Cellular Endocrinology 1997;130:43–51CrossRef
Need, EF, Scher, HI, Peters, AA, et al. A novel androgen receptor amino terminal region reveals two classes of amino/carboxyl interaction-deficient variants with divergent capacity to activate responsive sites in chromatin. Endocrinology 2009;150:2674–82.CrossRef
Li, W, Cavasotto, CN, Cardozo, T, et al. Androgen receptor mutations identified in prostate cancer and androgen insensitivity syndrome display aberrant ART-27 coactivator function. Molecular Endocrinology 2005;19:2273–82.CrossRef
Garolla, A, Ferlin, A, Vinanzi, C, et al. Molecular analysis of the androgen receptor gene in testicular cancer. Endocrine-Related Cancer 2005;12:645–55.CrossRef
Tepper, CG, Boucher, DL, Ryan, PE, et al. Characterization of a novel androgen receptor mutation in a relapsed CWR22 prostate cancer xenograft and cell line. Cancer Research 2002;62: 6606–14.
Dehm, SM, Schmidt, LJ, Heemers, HV, Vessella, RL, Tindall, DJ. Splicing of a novel androgen receptor exon generates a constitutively active androgen receptor that mediates prostate cancer therapy resistance. Cancer Research 2008;68:5469–77.CrossRef
Marcias, G, Erdmann, E, Lapouge, G, et al. Identification of novel truncated androgen receptor (AR) mutants including unreported pre-mRNA splicing variants in the 22Rv1 hormone-refractory prostate cancer (PCa) cell line. Human Mutation 2010;31:74–80.CrossRef
Hu, R, Dunn, TA, Wei, S, et al. Ligand-independent androgen receptor variants derived from splicing of cryptic exons signify hormone-refractory prostate cancer. Cancer Research 2009;69:16–22.CrossRef
Watson, PA, Chen, YF, Balbas, MD, et al. Constitutively active androgen receptor splice variants expressed in castration-resistant prostate cancer require full-length androgen receptor. Proceedings of the National Academy of Sciences USA 2010;107:16 759–65.
Sun, S, Sprenger, CC, Vessella, RL, et al. Castration resistance in human prostate cancer is conferred by a frequently occurring androgen receptor splice variant. Journal of Clinical Investigation 2010;120:2715–30.CrossRefGoogle ScholarPubMed
Jagla, M, Feve, M, Kessler, P, et al. A splicing variant of the androgen receptor detected in a metastatic prostate cancer exhibits exclusively cytoplasmic actions. Endocrinology 2007;148: 4334–43.CrossRef
Lapouge, G, Marcias, G, Erdmann, E, et al. Specific properties of a C-terminal truncated androgen receptor detected in hormone refractory prostate cancer. Advances in Experimental Medicine and Biology 2008;617:529–34.CrossRef
Lapouge, G, Erdmann, E, Marcias, G, et al. Unexpected paracrine action of prostate cancer cells harboring a new class of androgen receptor mutation–a new paradigm for cooperation among prostate tumor cells. International Journal of Cancer 2007;121:1238–44.CrossRefGoogle ScholarPubMed

Save book to Kindle

To save this book to your Kindle, first ensure coreplatform@cambridge.org is added to your Approved Personal Document E-mail List under your Personal Document Settings on the Manage Your Content and Devices page of your Amazon account. Then enter the ‘name’ part of your Kindle email address below. Find out more about saving to your Kindle.

Note you can select to save to either the @free.kindle.com or @kindle.com variations. ‘@free.kindle.com’ emails are free but can only be saved to your device when it is connected to wi-fi. ‘@kindle.com’ emails can be delivered even when you are not connected to wi-fi, but note that service fees apply.

Find out more about the Kindle Personal Document Service.

  • Androgens and the androgen receptor (AR)
    • By Nicole L. Moore, Dame Roma Mitchell Cancer Research Laboratories, Adelaide University/Hanson Institute, Adelaide, South Australia, Margaret M. Centenera, Dame Roma Mitchell Cancer Research Laboratories, Adelaide University/Hanson Institute, Adelaide, South Australia, Lisa M. Butler, Dame Roma Mitchell Cancer Research Laboratories, Adelaide University/Hanson Institute, Adelaide, South Australia, Theresa E. Hickey, Dame Roma Mitchell Cancer Research Laboratories, Adelaide University/Hanson Institute, Adelaide, South Australia, Wayne D. Tilley, Dame Roma Mitchell Cancer Research Laboratories, Adelaide University/Hanson Institute, Adelaide, South Australia
  • Edited by Edward P. Gelmann, Columbia University, New York, Charles L. Sawyers, Memorial Sloan-Kettering Cancer Center, New York, Frank J. Rauscher, III
  • Book: Molecular Oncology
  • Online publication: 05 February 2015
  • Chapter DOI: https://doi.org/10.1017/CBO9781139046947.033
Available formats
×

Save book to Dropbox

To save content items to your account, please confirm that you agree to abide by our usage policies. If this is the first time you use this feature, you will be asked to authorise Cambridge Core to connect with your account. Find out more about saving content to Dropbox.

  • Androgens and the androgen receptor (AR)
    • By Nicole L. Moore, Dame Roma Mitchell Cancer Research Laboratories, Adelaide University/Hanson Institute, Adelaide, South Australia, Margaret M. Centenera, Dame Roma Mitchell Cancer Research Laboratories, Adelaide University/Hanson Institute, Adelaide, South Australia, Lisa M. Butler, Dame Roma Mitchell Cancer Research Laboratories, Adelaide University/Hanson Institute, Adelaide, South Australia, Theresa E. Hickey, Dame Roma Mitchell Cancer Research Laboratories, Adelaide University/Hanson Institute, Adelaide, South Australia, Wayne D. Tilley, Dame Roma Mitchell Cancer Research Laboratories, Adelaide University/Hanson Institute, Adelaide, South Australia
  • Edited by Edward P. Gelmann, Columbia University, New York, Charles L. Sawyers, Memorial Sloan-Kettering Cancer Center, New York, Frank J. Rauscher, III
  • Book: Molecular Oncology
  • Online publication: 05 February 2015
  • Chapter DOI: https://doi.org/10.1017/CBO9781139046947.033
Available formats
×

Save book to Google Drive

To save content items to your account, please confirm that you agree to abide by our usage policies. If this is the first time you use this feature, you will be asked to authorise Cambridge Core to connect with your account. Find out more about saving content to Google Drive.

  • Androgens and the androgen receptor (AR)
    • By Nicole L. Moore, Dame Roma Mitchell Cancer Research Laboratories, Adelaide University/Hanson Institute, Adelaide, South Australia, Margaret M. Centenera, Dame Roma Mitchell Cancer Research Laboratories, Adelaide University/Hanson Institute, Adelaide, South Australia, Lisa M. Butler, Dame Roma Mitchell Cancer Research Laboratories, Adelaide University/Hanson Institute, Adelaide, South Australia, Theresa E. Hickey, Dame Roma Mitchell Cancer Research Laboratories, Adelaide University/Hanson Institute, Adelaide, South Australia, Wayne D. Tilley, Dame Roma Mitchell Cancer Research Laboratories, Adelaide University/Hanson Institute, Adelaide, South Australia
  • Edited by Edward P. Gelmann, Columbia University, New York, Charles L. Sawyers, Memorial Sloan-Kettering Cancer Center, New York, Frank J. Rauscher, III
  • Book: Molecular Oncology
  • Online publication: 05 February 2015
  • Chapter DOI: https://doi.org/10.1017/CBO9781139046947.033
Available formats
×