The growth response to androgen receptor signaling in ERα-negative human breast cells is dependent on p21 and mediated by MAPK activation

Joseph P Garay, Bedri Karakas, Abde M Abukhdeir, David P Cosgrove, John P Gustin, Michaela J Higgins, Hiroyuki Konishi, Yuko Konishi, Josh Lauring, Morassa Mohseni, Grace M Wang, Danijela Jelovac, Ashani Weeraratna, Cheryl A Sherman Baust, Patrice J Morin, Antoun Toubaji, Alan Meeker, Angelo M De Marzo, Gloria Lewis, Andrea Subhawong, Pedram Argani, Ben H Park, Joseph P Garay, Bedri Karakas, Abde M Abukhdeir, David P Cosgrove, John P Gustin, Michaela J Higgins, Hiroyuki Konishi, Yuko Konishi, Josh Lauring, Morassa Mohseni, Grace M Wang, Danijela Jelovac, Ashani Weeraratna, Cheryl A Sherman Baust, Patrice J Morin, Antoun Toubaji, Alan Meeker, Angelo M De Marzo, Gloria Lewis, Andrea Subhawong, Pedram Argani, Ben H Park

Abstract

Introduction: Although a high frequency of androgen receptor (AR) expression in human breast cancers has been described, exploiting this knowledge for therapy has been challenging. This is in part because androgens can either inhibit or stimulate cell proliferation in pre-clinical models of breast cancer. In addition, many breast cancers co-express other steroid hormone receptors that can affect AR signaling, further obfuscating the effects of androgens on breast cancer cells.

Methods: To create better-defined models of AR signaling in human breast epithelial cells, we took estrogen receptor (ER)-α-negative and progesterone receptor (PR)-negative human breast epithelial cell lines, both cancerous and non-cancerous, and engineered them to express AR, thus allowing the unambiguous study of AR signaling. We cloned a full-length cDNA of human AR, and expressed this transgene in MCF-10A non-tumorigenic human breast epithelial cells and MDA-MB-231 human breast-cancer cells. We characterized the responses to AR ligand binding using various assays, and used isogenic MCF-10A p21 knock-out cell lines expressing AR to demonstrate the requirement for p21 in mediating the proliferative responses to AR signaling in human breast epithelial cells.

Results: We found that hyperactivation of the mitogen-activated protein kinase (MAPK) pathway from both AR and epidermal growth factor receptor (EGFR) signaling resulted in a growth-inhibitory response, whereas MAPK signaling from either AR or EGFR activation resulted in cellular proliferation. Additionally, p21 gene knock-out studies confirmed that AR signaling/activation of the MAPK pathway is dependent on p21.

Conclusions: These studies present a new model for the analysis of AR signaling in human breast epithelial cells lacking ERα/PR expression, providing an experimental system without the potential confounding effects of ERα/PR crosstalk. Using this system, we provide a mechanistic explanation for previous observations ascribing a dual role for AR signaling in human breast cancer cells. As previous reports have shown that approximately 40% of breast cancers can lack p21 expression, our data also identify potential new caveats for exploiting AR as a target for breast cancer therapy.

Figures

Figure 1
Figure 1
Expression of androgen receptor (AR) in human breast epithelial cells. (A) MCF-10A cells were stably transfected with an AR cDNA, and lysates from single-cell clones were probed for AR expression by western blotting. As a negative control, MCF-10A cells were also transfected with empty vector (10A plus vector) and isolated as single-cell clones. MDA-MB-453 and LNCaP cell lysates served as positive controls for AR expression. All lysates were also probed with GAPDH antibody as a loading control. (B) Androgen Receptor In Breast Epithelium (ARIBE) cells and control cells were transfected with luciferase reporter plasmids containing consensus DNA binding sites and mutated controls for AR as described in Methods. At the time of transfection, cells were treated with 1 nmol/l R1881 or vehicle (ethanol), and analyzed 48 hours later. Firefly luciferase measurements were normalized to Renilla luciferase measurements in all samples. The relative luciferase units (RLU) ratio was calculated as the luciferase expression comparing wild-type ARE/mutant ARE in R1881 versus vehicle-treated control cells (R1881 wtARE/mutARE: ETOH wtARE/mutARE). Error bars represent the standard deviation of three independent experiments. Luciferase expression in each ARIBE clone compared with control cell lines was significant by one-way analysis of variance (ANOVA) (P < 0.05).(C) cDNA was made from RNA of cells treated with 1 nmo/l R1881 or vehicle for 48 hours. Quantitative real-time PCR using SYBR Green was performed on triplicate samples of each cell line using intron-spanning primers for each of three androgen-response genes. All cycle threshold numbers were normalized to a control gene, TATA binding protein (TBP). Ratio is expression in cells treated with drug versus vehicle. Error bars represent the standard deviation of four independent experiments. In both ARIBE lines, induction of all genes after drug treatment was significant by one-way ANOVA compared with control cell lines (P < 0.001).
Figure 2
Figure 2
Activation of the mitogen-activated protein kinase (MAPK) pathway alters the growth response of Androgen Receptor In Breast Epithelium (ARIBE) cells to androgen receptor (AR) ligand. (A) Control cells (MCF-10A and 10A plus vector) and two ARIBE clones were cultured in normal propagation media (with 20 ng/ml epidermal growth factor (EGF)) and treated with vehicle, 10 μmol/l bicalutamide, 1 nmol/l R1881, or a combination of R1881 and bicalutamide. Cells were counted after either 2 or 4 days of treatment, and normalized to values of cells counted on the day of drug addition (day 0). Error bars represent the standard deviation of the mean of three independent cell counts. The growth difference between cells treated with R1881 and R1881 plus bicalutamide was significant by Student's two-tailed t-test (P < 0.001). (B) Cells were cultured under normal propagation conditions except for the absence of EGF from the medium. Cells were treated with vehicle or drugs as above. Cells were counted after either 4 or 8 days of treatment, and normalized to values of cells counted on the day of drug addition (day 0). Error bars represent the standard deviation of the mean of three independent cell counts. The growth difference between cells treated with R1881 and R1881 plus bicalutamide was significant by Student's two-tailed t-test (P < 0.01).
Figure 3
Figure 3
Androgen Receptor In Breast Epithelium (ARIBE) cells stimulate mitogen-activated protein kinase (MAPK) signaling upon androgen receptor (AR) ligand binding. Control cells (MCF-10A and 10A plus vector) and ARIBE cells were cultured under normal propagation conditions in the presence or absence of 20 ng/ml epidermal growth factor (EGF), treated with either vehicle or 1 nmol/l R1881 for 48 hours, and then used to make lysates for western blotting as described in Methods. Blots were probed for phosphorylated extracellular signal-regulated kinase (ERK) (Thr-202/Tyr-204) and total ERK. GAPDH antibody was used as loading control.
Figure 4
Figure 4
MDA-MB-231 cells transfected with androgen receptor (AR) show growth inhibition when treated with androgen. (A) The breast cancer cell line MDA-MB-231 was transfected with an AR cDNA, and lysates from single-cell clones were analyzed for AR protein expression. Two representative clones are shown. MDA-MB-453 and LNCaP lysates served as positive controls for AR expression. Lysates were also probed with GAPDH as a loading control.(B) Two MDA-MB-231 clones transfected with AR (ARc3 and ARc4) and control cells (Vector) were treated with vehicle, 1 μmol/l of the MEK inhibitor U0126, 1 nmol/l of R1881, or a combination of R1881 and U0126. Cells were counted after 4 days of drug treatment, and normalized to counts of vehicle-treated control cells. Error bars represent the standard deviation of the mean of three independent counts. The growth difference between cells treated with R1881 and R1881 plus U0126 was significant by Student's two-tailed t-test (*P < 0.005).
Figure 5
Figure 5
Upregulation of p21 expression upon androgen receptor (AR) ligand binding in the presence of mitogen-activated protein kinase (MAPK)signaling. (A) Control cells (MCF-10A and 10A plus vector) and ARIBE cells were cultured in normal propagation media (with 20 ng/ml EGF) and treated with either vehicle or 1 nmol/l R1881 for 24 hours. Whole-cell lysates were probed for expression of p21 and GAPDH for a loading control. (B) Control cells (MDA-MB-231 and 231 plus vector) and MDA-MB-231 cells expressing AR (ARc3 and ARc4) were cultured in the presence EGF and treated with either vehicle or 1 nmol/l R1881 for 24 hours. Whole-cell lysates were probed for expression of p21 and GAPDH was used as loading control.
Figure 6
Figure 6
p21 knock-down via siRNA abrogates growth inhibition of Androgen Receptor In Breast Epithelium (ARIBE) cells. (A) ARIBE cells were transfected with p21 siRNA constructs, control siRNA, or no siRNA, and incubated for 24 hours. After incubation, whole-cell lysates were probed for expression of p21, and GAPDH was used as a loading control. (B) ARIBE cells transfected for 24 hours with p21 or control siRNA were subsequently treated with vehicle or 1 nmol/l R1881 for 4 days, and cells were counted. The ratio of growth between vehicle and R1881-treated cells was calculated and normalized to untransfected (no siRNA) values to determine the fold increase in growth of siRNA-transfected cells. Error bars represent the standard deviation of the mean of three independent counts. The growth difference between cells transfected with p21 siRNA constructs and mock siRNA was significant by one-way analysis of variance (*P < 0.05).
Figure 7
Figure 7
p21 mediates growth effects of androgen receptor (AR) ligand in MCF-10A cells. (A) MCF-10A p21-/- cells were stably transfected with an AR cDNA, and lysates from single-cell clones were probed for AR expression by western blotting. As a negative control, MCF-10A p21-/- cells were also transfected with empty vector (p21-/- plus vector) and isolated as single-cell clones. Expression of AR was compared with ARIBE cells, which have wild-type p21. MDA-MB-453 and LNCaP cell lysates served as positive controls for AR expression. All lysates were also probed with GAPDH antibody as a loading control. (B) The p21-/- (left) and p21 wild-type (right) cells were cultured in normal propagation media with 20 ng/ml epidermal growth factor (EGF), and treated with 1 nmol/l R1881. Cells were counted after 4 days of treatment, and normalized to values of cells counted on the day of drug addition (day 0). Error bars represent the standard deviation of the mean of three independent cell counts. The growth difference between p21-/- AR-expressing cells and p21-wild-type AR-expressing cells was significant by one-way analysis of variance (ANOVA) (*P < 0.001). (C) Cells were cultured under normal propagation conditions except for the absence of EGF from the medium. Cells were treated with vehicle or drugs as described above. Cells were counted after 8 days of treatment, and normalized to values of cells counted on the day of drug addition (day 0). Error bars represent the standard deviation of the mean of three independent cell counts. The growth difference between p21-/- AR-expressing cells and p21-wild-type AR-expressing cells was significant by one-way ANOVA (*P < 0.01).
Figure 8
Figure 8
p21-/- cells do not stimulate mitogen-activated protein kinase (MAPK) signaling upon androgen receptor (AR) ligand binding. Control cells (MCF-10A p21-/- and p21-/- plus vector) and AR-expressing cells (p21-/- AR-1 and p21-/- AR-4) were cultured under normal propagation conditions in the presence or absence of 20 ng/ml epidermal growth factor (EGF), and treated with either vehicle or 1 nmol/l R1881 for 48 hours. Western blotting was performed on whole-cell lysates. Blots were probed for phosphorylated extracellular signal-regulated kinase (ERK) (Thr-202/Tyr-204) and total ERK. GAPDH antibody was used as a loading control.

References

    1. van de Velde CJ, Verma S, van Nes JG, Masterman C, Pritchard KI. Switching from tamoxifen to aromatase inhibitors for adjuvant endocrine therapy in postmenopausal patients with early breast cancer. Cancer Treat Rev. 2009.
    1. Cauley JA, Lucas FL, Kuller LH, Stone K, Browner W, Cummings SR. Elevated serum estradiol and testosterone concentrations are associated with a high risk for breast cancer. Study of Osteoporotic Fractures Research Group. Ann Intern Med. 1999;130:270–277.
    1. Hankinson SE, Willett WC, Manson JE, Colditz GA, Hunter DJ, Spiegelman D, Barbieri RL, Speizer FE. Plasma sex steroid hormone levels and risk of breast cancer in postmenopausal women. J Natl Cancer Inst. 1998;90:1292–1299. doi: 10.1093/jnci/90.17.1292.
    1. Dimitrakakis C, Bondy C. Androgens and the breast. Breast Cancer Res. 2009;11:212. doi: 10.1186/bcr2413.
    1. Lea OA, Kvinnsland S, Thorsen T. Improved measurement of androgen receptors in human breast cancer. Cancer Res. 1989;49:7162–7167.
    1. Moinfar F, Okcu M, Tsybrovskyy O, Regitnig P, Lax SF, Weybora W, Ratschek M, Tavassoli FA, Denk H. Androgen receptors frequently are expressed in breast carcinomas: potential relevance to new therapeutic strategies. Cancer. 2003;98:703–711. doi: 10.1002/cncr.11532.
    1. Agoff SN, Swanson PE, Linden H, Hawes SE, Lawton TJ. Androgen receptor expression in estrogen receptor-negative breast cancer. Immunohistochemical, clinical, and prognostic associations. Am J Clin Pathol. 2003;120:725–731. doi: 10.1309/42F00D0DJD0J5EDT.
    1. Park S, Koo J, Park HS, Kim JH, Choi SY, Lee JH, Park BW, Lee KS. Expression of androgen receptors in primary breast cancer. Ann Oncol. 2010;21:488–492. doi: 10.1093/annonc/mdp510.
    1. Niemeier LA, Dabbs DJ, Beriwal S, Striebel JM, Bhargava R. Androgen receptor in breast cancer: expression in estrogen receptor-positive tumors and in estrogen receptor-negative tumors with apocrine differentiation. Mod Pathol. 2010;23:205–212. doi: 10.1038/modpathol.2009.159.
    1. Hu R, Dawood S, Holmes MD, Collins LC, Schnitt SJ, Cole K, Marotti JD, Hankinson SE, Colditz GA, Tamimi RM. Androgen receptor expression and breast cancer survival in postmenopausal women. Clin Cancer Res. 2011;17:1867–1874. doi: 10.1158/1078-0432.CCR-10-2021.
    1. Peters AA, Buchanan G, Ricciardelli C, Bianco-Miotto T, Centenera MM, Harris JM, Jindal S, Segara D, Jia L, Moore NL, Henshall SM, Birrell SN, Coetzee GA, Sutherland RL, Butler LM, Tilley WD. Androgen receptor inhibits estrogen receptor-alpha activity and is prognostic in breast cancer. Cancer Res. 2009;69:6131–6140.
    1. Castellano I, Allia E, Accortanzo V, Vandone AM, Chiusa L, Arisio R, Durando A, Donadio M, Bussolati G, Coates AS, Viale G, Sapino A. Androgen receptor expression is a significant prognostic factor in estrogen receptor positive breast cancers. Breast Cancer Res Treat. 2010;124:607–617. doi: 10.1007/s10549-010-0761-y.
    1. Buchanan G, Birrell SN, Peters AA, Bianco-Miotto T, Ramsay K, Cops EJ, Yang M, Harris JM, Simila HA, Moore NL, Bentel JM, Ricciardelli C, Horsfall DJ, Butler LM, Tilley WD. Decreased androgen receptor levels and receptor function in breast cancer contribute to the failure of response to medroxyprogesterone acetate. Cancer Res. 2005;65:8487–8496. doi: 10.1158/0008-5472.CAN-04-3077.
    1. Lonning PE. Additive endocrine therapy for advanced breast cancer - back to the future. Acta Oncol. 2009;48:1092–1101. doi: 10.3109/02841860903117816.
    1. Gucalp A, Traina TA. Triple-negative breast cancer: role of the androgen receptor. Cancer J. 2010;16:62–65. doi: 10.1097/PPO.0b013e3181ce4ae1.
    1. Lehmann BD, Bauer JA, Chen X, Sanders ME, Chakravarthy AB, Shyr Y, Pietenpol JA. Identification of human triple-negative breast cancer subtypes and preclinical models for selection of targeted therapies. J Clin Invest. 2011;121:2750–2767. doi: 10.1172/JCI45014.
    1. Robinson JL, Macarthur S, Ross-Innes CS, Tilley WD, Neal DE, Mills IG, Carroll JS. Androgen receptor driven transcription in molecular apocrine breast cancer is mediated by FoxA1. Embo J. 2011;30:3019–3027. doi: 10.1038/emboj.2011.216.
    1. Naderi A, Chia KM, Liu J. Synergy between inhibitors of androgen receptor and MEK has therapeutic implications in estrogen receptor-negative breast cancer. Breast Cancer Res. 2011;13:R36. doi: 10.1186/bcr2858.
    1. Nahleh Z. Androgen receptor as a target for the treatment of hormone receptor-negative breast cancer: an unchartered territory. Future Oncol. 2008;4:15–21. doi: 10.2217/14796694.4.1.15.
    1. Ni M, Chen Y, Lim E, Wimberly H, Bailey ST, Imai Y, Rimm DL, Liu XS, Brown M. Targeting androgen receptor in estrogen receptor-negative breast cancer. Cancer Cell. 2011;20:119–131. doi: 10.1016/j.ccr.2011.05.026.
    1. Kennedy BJ. Fluoxymesterone therapy in advanced breast cancer. N Engl J Med. 1958;259:673–675. doi: 10.1056/NEJM195810022591404.
    1. Birrell SN, Bentel JM, Hickey TE, Ricciardelli C, Weger MA, Horsfall DJ, Tilley WD. Androgens induce divergent proliferative responses in human breast cancer cell lines. J Steroid Biochem Mol Biol. 1995;52:459–467. doi: 10.1016/0960-0760(95)00005-K.
    1. Yoon DS, Wersto RP, Zhou W, Chrest FJ, Garrett ES, Kwon TK, Gabrielson E. Variable levels of chromosomal instability and mitotic spindle checkpoint defects in breast cancer. Am J Pathol. 2002;161:391–397. doi: 10.1016/S0002-9440(10)64194-6.
    1. De Amicis F, Thirugnansampanthan J, Cui Y, Selever J, Beyer A, Parra I, Weigel NL, Herynk MH, Tsimelzon A, Lewis MT, Chamness GC, Hilsenbeck SG, Ando S, Fuqua SA. Androgen receptor overexpression induces tamoxifen resistance in human breast cancer cells. Breast Cancer Res Treat. 2010;121:1–11. doi: 10.1007/s10549-009-0436-8.
    1. Isola JJ. Immunohistochemical demonstration of androgen receptor in breast cancer and its relationship to other prognostic factors. J Pathol. 1993;170:31–35. doi: 10.1002/path.1711700106.
    1. Doane AS, Danso M, Lal P, Donaton M, Zhang L, Hudis C, Gerald WL. An estrogen receptor-negative breast cancer subset characterized by a hormonally regulated transcriptional program and response to androgen. Oncogene. 2006;25:3994–4008. doi: 10.1038/sj.onc.1209415.
    1. Kuenen-Boumeester V, Van der Kwast TH, van Putten WL, Claassen C, van Ooijen B, Henzen-Logmans SC. Immunohistochemical determination of androgen receptors in relation to oestrogen and progesterone receptors in female breast cancer. Int J Cancer. 1992;52:581–584. doi: 10.1002/ijc.2910520415.
    1. Riva C, Dainese E, Caprara G, Rocca PC, Massarelli G, Tot T, Capella C, Eusebi V. Immunohistochemical study of androgen receptors in breast carcinoma. Evidence of their frequent expression in lobular carcinoma. Virchows Arch. 2005;447:695–700. doi: 10.1007/s00428-005-0003-6.
    1. Panet-Raymond V, Gottlieb B, Beitel LK, Pinsky L, Trifiro MA. Interactions between androgen and estrogen receptors and the effects on their transactivational properties. Mol Cell Endocrinol. 2000;167:139–150. doi: 10.1016/S0303-7207(00)00279-3.
    1. Bentel JM, Birrell SN, Pickering MA, Holds DJ, Horsfall DJ, Tilley WD. Androgen receptor agonist activity of the synthetic progestin, medroxyprogesterone acetate, in human breast cancer cells. Mol Cell Endocrinol. 1999;154:11–20. doi: 10.1016/S0303-7207(99)00109-4.
    1. Bhattacharyya RS, Krishnan AV, Swami S, Feldman D. Fulvestrant (ICI 182,780) down-regulates androgen receptor expression and diminishes androgenic responses in LNCaP human prostate cancer cells. Mol Cancer Ther. 2006;5:1539–1549.
    1. Wasielewski M, Elstrodt F, Klijn JG, Berns EM, Schutte M. Thirteen new p53 gene mutants identified among 41 human breast cancer cell lines. Breast Cancer Res Treat. 2006;99:97–101. doi: 10.1007/s10549-006-9186-z.
    1. She QB, Chandarlapaty S, Ye Q, Lobo J, Haskell KM, Leander KR, DeFeo-Jones D, Huber HE, Rosen N. Breast tumor cells with PI3K mutation or HER2 amplification are selectively addicted to Akt signaling. PLoS ONE. 2008;3:e3065. doi: 10.1371/journal.pone.0003065.
    1. Abukhdeir AM, Blair BG, Brenner K, Karakas B, Konishi H, Lim J, Sahasranaman V, Huang Y, Keen J, Davidson N, Vitolo MI, Bachman KE, Park BH. Physiologic estrogen receptor alpha signaling in non-tumorigenic human mammary epithelial cells. Breast Cancer Res Treat. 2006;99:23–33. doi: 10.1007/s10549-006-9177-0.
    1. Soule HD, Maloney TM, Wolman SR, Peterson WD, Brenz R, McGrath CM, Russo J, Pauley RJ, Jones RF, Brooks SC. Isolation and characterization of a spontaneously immortalized human breast epithelial cell line, MCF-10. Cancer Res. 1990;50:6075–6086.
    1. Bachman KE, Blair BG, Brenner K, Bardelli A, Arena S, Zhou S, Hicks J, De Marzo AM, Argani P, Park BH. p21(WAF1/CIP1) Mediates the Growth Response to TGF-beta in Human Epithelial Cells. Cancer Biol Ther. 2004;3:221–225.
    1. Favata MF, Horiuchi KY, Manos EJ, Daulerio AJ, Stradley DA, Feeser WS, Van Dyk DE, Pitts WJ, Earl RA, Hobbs F, Copeland RA, Magolda RL, Scherle PA, Trzaskos JM. Identification of a novel inhibitor of mitogen-activated protein kinase kinase. J Biol Chem. 1998;273:18623–18632. doi: 10.1074/jbc.273.29.18623.
    1. ImageJ.
    1. Gonzalez-Angulo AM, Stemke-Hale K, Palla SL, Carey M, Agarwal R, Meric-Berstam F, Traina TA, Hudis C, Hortobagyi GN, Gerald WL, Mills GB, Hennessy BT. Androgen receptor levels and association with PIK3CA mutations and prognosis in breast cancer. Clin Cancer Res. 2009;15:2472–2478. doi: 10.1158/1078-0432.CCR-08-1763.
    1. Linja MJ, Savinainen KJ, Saramaki OR, Tammela TL, Vessella RL, Visakorpi T. Amplification and overexpression of androgen receptor gene in hormone-refractory prostate cancer. Cancer Res. 2001;61:3550–3555.
    1. Albertson DG. Conflicting evidence on the frequency of ESR1 amplification in breast cancer. Nat Genet. 2008;40:821–822. doi: 10.1038/ng0708-821.
    1. Liu W, Xie CC, Zhu Y, Li T, Sun J, Cheng Y, Ewing CM, Dalrymple S, Turner AR, Isaacs JT, Chang BL, Zheng SL, Isaacs WB, Xu J. Homozygous deletions and recurrent amplifications implicate new genes involved in prostate cancer. Neoplasia. 2008;10:897–907.
    1. Magklara A, Grass L, Diamandis EP. Differential steroid hormone regulation of human glandular kallikrein (hK2) and prostate-specific antigen (PSA) in breast cancer cell lines. Breast Cancer Res Treat. 2000;59:263–270. doi: 10.1023/A:1006304518750.
    1. Vendola K, Zhou J, Wang J, Bondy CA. Androgens promote insulin-like growth factor-I and insulin-like growth factor-I receptor gene expression in the primate ovary. Hum Reprod. 1999;14:2328–2332. doi: 10.1093/humrep/14.9.2328.
    1. Yanase T, Fan W. Modification of androgen receptor function by IGF-1 signaling implications in the mechanism of refractory prostate carcinoma. Vitam Horm. 2009;80:649–666.
    1. Lu S, Liu M, Epner DE, Tsai SY, Tsai MJ. Androgen regulation of the cyclin-dependent kinase inhibitor p21 gene through an androgen response element in the proximal promoter. Mol Endocrinol. 1999;13:376–384. doi: 10.1210/me.13.3.376.
    1. Nelson PS, Clegg N, Arnold H, Ferguson C, Bonham M, White J, Hood L, Lin B. The program of androgen-responsive genes in neoplastic prostate epithelium. Proc Natl Acad Sci USA. 2002;99:11890–11895. doi: 10.1073/pnas.182376299.
    1. Bolton EC, So AY, Chaivorapol C, Haqq CM, Li H, Yamamoto KR. Cell- and gene-specific regulation of primary target genes by the androgen receptor. Genes Dev. 2007;21:2005–2017. doi: 10.1101/gad.1564207.
    1. von Bueren AO, Ma R, Schlumpf M, Lichtensteiger W. Salbutamol exhibits androgenic activity in vitro. Br J Sports Med. 2007;41:874–878. doi: 10.1136/bjsm.2007.035162. discussion 878.
    1. Blankvoort BM, de Groene EM, van Meeteren-Kreikamp AP, Witkamp RF, Rodenburg RJ, Aarts JM. Development of an androgen reporter gene assay (AR-LUX) utilizing a human cell line with an endogenously regulated androgen receptor. Anal Biochem. 2001;298:93–102. doi: 10.1006/abio.2001.5352.
    1. Ma R, Cotton B, Lichtensteiger W, Schlumpf M. UV filters with antagonistic action at androgen receptors in the MDA-kb2 cell transcriptional-activation assay. Toxicol Sci. 2003;74:43–50. doi: 10.1093/toxsci/kfg102.
    1. Chou JL, Fan Z, DeBlasio T, Koff A, Rosen N, Mendelsohn J. Constitutive overexpression of cyclin D1 in human breast epithelial cells does not prevent G1 arrest induced by deprivation of epidermal growth factor. Breast Cancer Res Treat. 1999;55:267–283. doi: 10.1023/A:1006217413089.
    1. Zhu X, Li H, Liu JP, Funder JW. Androgen stimulates mitogen-activated protein kinase in human breast cancer cells. Mol Cell Endocrinol. 1999;152:199–206. doi: 10.1016/S0303-7207(99)00031-3.
    1. Davidson NE, Gelmann EP, Lippman ME, Dickson RB. Epidermal growth factor receptor gene expression in estrogen receptor-positive and negative human breast cancer cell lines. Mol Endocrinol. 1987;1:216–223. doi: 10.1210/mend-1-3-216.
    1. Fitzpatrick SL, LaChance MP, Schultz GS. Characterization of epidermal growth factor receptor and action on human breast cancer cells in culture. Cancer Res. 1984;44:3442–3447.
    1. Ho-Kim MA, Tremblay RR, Dube JY. Binding of methyltrienolone to glucocorticoid receptors in rat muscle cytosol. Endocrinology. 1981;109:1418–1423. doi: 10.1210/endo-109-5-1418.
    1. Sarkisian CJ, Keister BA, Stairs DB, Boxer RB, Moody SE, Chodosh LA. Dose-dependent oncogene-induced senescence in vivo and its evasion during mammary tumorigenesis. Nat Cell Biol. 2007;9:493–505. doi: 10.1038/ncb1567.
    1. el-Deiry WS, Harper JW, O'Connor PM, Velculescu VE, Canman CE, Jackman J, Pietenpol JA, Burrell M, Hill DE, Wang Y. et al.WAF1/CIP1 is induced in p53-mediated G1 arrest and apoptosis. Cancer Res. 1994;54:1169–1174.
    1. el-Deiry WS, Tokino T, Velculescu VE, Levy DB, Parsons R, Trent JM, Lin D, Mercer WE, Kinzler KW, Vogelstein B. WAF1, a potential mediator of p53 tumor suppression. Cell. 1993;75:817–825. doi: 10.1016/0092-8674(93)90500-P.
    1. Sherr CJ, Roberts JM. Living with or without cyclins and cyclin-dependent kinases. Genes Dev. 2004;18:2699–2711. doi: 10.1101/gad.1256504.
    1. Karakas B, Weeraratna AT, Abukhdeir AM, Konishi H, Gustin JP, Vitolo MI, Bachman KE, Park BH. p21 Gene knock Down Does Not Identify Genetic Effectors Seen with Gene Knock Out. Cancer Biol Ther. 2007;6:1025–1030. doi: 10.4161/cbt.6.7.4202.
    1. Abukhdeir AM, Park BH. P21 and p27: roles in carcinogenesis and drug resistance. Expert Rev Mol Med. 2008;10:e19.
    1. Strickfaden SC, Winters MJ, Ben-Ari G, Lamson RE, Tyers M, Pryciak PM. A mechanism for cell-cycle regulation of MAP kinase signaling in a yeast differentiation pathway. Cell. 2007;128:519–531. doi: 10.1016/j.cell.2006.12.032.
    1. Higgins MJ, Beaver JA, Wong HY, Gustin JP, Lauring JD, Garay JP, Konishi H, Mohseni M, Wang GM, Cidado J, Jelovac D, Cosgrove DP, Tamaki A, Abukhdeir AM, Park BH. PIK3CA mutations and EGFR overexpression predict for lithium sensitivity in human breast epithelial cells. Cancer Biol Ther. 2011;11:35–44.

Source: PubMed

3
Abonnieren