Cell autonomous role of PTEN in regulating castration-resistant prostate cancer growth

David J Mulholland, Linh M Tran, Yunfeng Li, Houjian Cai, Ashkan Morim, Shunyou Wang, Seema Plaisier, Isla P Garraway, Jiaoti Huang, Thomas G Graeber, Hong Wu, David J Mulholland, Linh M Tran, Yunfeng Li, Houjian Cai, Ashkan Morim, Shunyou Wang, Seema Plaisier, Isla P Garraway, Jiaoti Huang, Thomas G Graeber, Hong Wu

Abstract

Alteration of the PTEN/PI3K pathway is associated with late-stage and castrate-resistant prostate cancer (CRPC). However, how PTEN loss is involved in CRPC development is not clear. Here, we show that castration-resistant growth is an intrinsic property of Pten null prostate cancer (CaP) cells, independent of cancer development stage. PTEN loss suppresses androgen-responsive gene expressions by modulating androgen receptor (AR) transcription factor activity. Conditional deletion of Ar in the epithelium promotes the proliferation of Pten null cancer cells, at least in part, by downregulating the androgen-responsive gene Fkbp5 and preventing PHLPP-mediated AKT inhibition. Our findings identify PI3K and AR pathway crosstalk as a mechanism of CRPC development, with potentially important implications for CaP etiology and therapy.

Copyright © 2011 Elsevier Inc. All rights reserved.

Figures

Figure 1. Early castration does not prevent…
Figure 1. Early castration does not prevent initiation of Pten-null CaP
(A) Pb-Cre+;PtenL/L mutants were castrated (Cx) at 2, 4 or 6 weeks and aged to 10 weeks. (B) Mutants Cx at 2 weeks were evaluated for carcinoma, PI3K (P-AKT, P-S6) activation coinciding with PTEN loss and invasiveness based on smooth muscle actin (SMA) loss but maintained E-Cadherin expression (arrow). Bar=100 μm (C) Cell proliferation, Ki67+; Cyclin D1+ (Cyc D1) cells, in 2 and 6 week Cx cohorts in comparison to WT Cx controls (**, p<0.01) Bar = 150 μm. Error bars, mean +/− SD. See also Figure S1.
Figure 2. PTEN loss can suppress androgen-responsive…
Figure 2. PTEN loss can suppress androgen-responsive gene expression
(A, B) Expression profile (means +/− s.e.m) of AR activated (A) and suppressed (B) genes in WT and Pten-null murine prostates after castration. (C) Heat map of expression ratios of androgen responsive genes in WT (14-day post castration) and intact Pten-null mutants with respect to intact WT mice. (D) Variation in expression of androgen-responsive genes based on PTEN copy number (CN) in human CaP samples. Left, summary of human samples based on PTEN CN (the numbers inside parentheses indicates the number of metastatic cases); right, a comparative analysis of AR activated (red circles) and suppressed (blue circles) gene expression values in two human CaP datasets. (E) Top, gene expression and NCA derived activities of EGR1, JUN, and AR transcription factors in induced PTEN expression in Pten−/− cells. Bottom, the activity of AR in murine models when PTEN/AKT/mTOR pathway was manipulated genetically or pharmacologically. (F) Expression (means +/− SD) of Ezh2 (left), and AR and EZH2 co-target genes (right) in PIN and cancer (CAN) stages of Pten-null prostate. *, p<0.05; **, p<0.005. See also Figure S2 and Table S1.
Figure 3. Epithelial AR is not required…
Figure 3. Epithelial AR is not required for the initiation of Pten-null CaP
(A) Deletion of epithelial AR in the anterior lobe of Pten-null CaP (Pb-Cre+;PtenL/L;ArL/Y) mutants and the impact on cell proliferation (Ki67+ cells), apoptosis (red arrow) and cancer formation (bar, top = 200 μm; bottom = 50 μm). (B) Cell proliferation (left) and apoptotic indexes (right) in Cre− (C−), Pb-Cre+;ArL/Y (C+;Ar); Pb-Cre+;PtenL/L and (C+;Pt) and Cre+;PtenL/L;ArL/Y (C+;Pt;Ar) mutants. (C) Frequency of invasiveness based on smooth muscle actin (SMA) break down in Pb-Cre+;PtenL/L and Cre+;PtenL/L;ArL/Y mutants during progression. Error bars, means +/− SD. See also Figure S3.
Figure 4. Epithelial AR is not required…
Figure 4. Epithelial AR is not required for transformation by Pten deletion or myristoylated Akt in regeneration assays
(A) Evaluating the impact of cre-mediated deletion of Pten and Ar on histopathology and PI3K signaling. The top panel shows the outline of the experiment and the bottom panels show results of tissues stained as indicated. Bar = 100 μm. (B) Evaluating AR deletion and myristoylated-AKT expression in primary, Pb-Cre+;ArL/Y mutants and the impact on prostate histopathology and PI3K signaling. The top panel shows the outline of the experiment and the bottom panels show results of tissues stained as indicated. Bars, top = 200 μm, bottom = 50 μm. See also Figure S4.
Figure 5. AR down regulates AKT activity…
Figure 5. AR down regulates AKT activity by stimulating FKBP5 and PHLPP-mediated, AKT dephosphorylation
(A) PI3K activation (P-AKT, P-S6) in AR+ regions (arrows) versus AR− regions (arrow heads) in castrate, Pb-Cre+;PtenL/L;ArL/Y mutants. (Bar, low mag = 150 μm, high mag = 75 μm). (B) Effect of AR deletion on FKBP5 expression in Pb-Cre+PtenL/L;ArL/Y mutants (bar = 50 μm). (C, D) Expression of FKBP5, PHLPP and P-AKT in AR+ regions (arrow heads) compared to AR-null regions (arrows) in castrated Pb-Cre+;PtenL/L;ArL/Y mutants at 2 days (C) and 4 weeks (D) after castration (bar = 75 μm). See also Figure S5.
Figure 6. Heterogeneous AR expression in human…
Figure 6. Heterogeneous AR expression in human CaPs correlating with PI3K/AKT signaling and FKBP5 and PHLPP levels
(A) PTEN and AR expression in human CaP (bar = 500 μm). (B) PI3K pathway components (P-AKT, P-S6, P-4EBP1) in Pten−;AR− regions of Pb-Cre+;PtenL/L;ArL/Y mice and in PTEN-negative;AR-low/negative regions of human CaPs. Error bars, means +/− SD. (C) Unsupervised clustering analysis of PTEN, PHLPP, AR, and FKBP5 in human TMA samples (N=91) (left). Chi squared test P-values* (N = 91) were used to quantitate the strength of association between each pair (right, table). Protein level was categorized to high (IHC> 1) and low (< 1) levels). See also Figure S6.
Figure 7. Cooperative effects of AR and…
Figure 7. Cooperative effects of AR and mTOR inhibition in vitro and in vivo
(A) In vitro response of Pten-null;Ar+ mouse (CaP8) and human (LNCaP) CaP cells to AR knockdown (sh-AR) or pharmacological inhibition of AR (MDV3100, 10 μM) with and without rapamycin (R: 1 nM) treatment. (Sc = control sh oligo). (B, D) In vivo response to treatments with castration, MDV3100, rapamycin or their combinations as measured by cell proliferation (Ki67+ cells) and (C, D) tumor burden in Pb-Cre+;PtenL/L and Pb-Cre+;PtenL/L:ArL/Y mutants (C, bar = 2 mm; D, bar = 200 μm; D, inset bar = 75 μm). Error bars = means +/− SD. See also Figure S7.
Figure 8. PTEN loss promotes CRPC development…
Figure 8. PTEN loss promotes CRPC development by two collaborative mechanisms
By regulating EGR1, c-JUN and EZH2 expression and activities, PTEN loss suppresses AR transcription factor activity and output, leading to reduced prostate epithelial differentiation and survival. Collaboratively, PTEN loss activates the PI3K/AKT signaling pathway and reduces the AR-regulated FKBP5-PHLPP negative feedback loop to AKT activation, further enhances AKT activation, leading to androgen/AR-independent prostate epithelial proliferation.

References

    1. Abbas F, Civantos F, Benedetto P, Soloway MS. Small cell carcinoma of the bladder and prostate. Urology. 1995;46:617–630.
    1. Abdulkadir SA, Qu Z, Garabedian E, Song SK, Peters TJ, Svaren J, Carbone JM, Naughton CK, Catalona WJ, Ackerman JJ, et al. Impaired prostate tumorigenesis in Egr1-deficient mice. Nat Med. 2001;7:101–107.
    1. Attard G, Cooper CS, de Bono JS. Steroid hormone receptors in prostate cancer: a hard habit to break? Cancer Cell. 2009a;16:458–462.
    1. Attard G, Reid AH, Olmos D, de Bono JS. Antitumor activity with CYP17 blockade indicates that castration-resistant prostate cancer frequently remains hormone driven. Cancer Res. 2009b;69:4937–4940.
    1. Brognard J, Sierecki E, Gao T, Newton AC. PHLPP and a second isoform, PHLPP2, differentially attenuate the amplitude of Akt signaling by regulating distinct Akt isoforms. Mol Cell. 2007;25:917–931.
    1. Chang CJ, Freeman DJ, Wu H. PTEN regulates Mdm2 expression through the P1 promoter. J Biol Chem. 2004;279:29841–29848.
    1. Chauvin TR, Griswold MD. Androgen-regulated genes in the murine epididymis. Biol Reprod. 2004;71:560–569.
    1. Chen Y, Sawyers CL, Scher HI. Targeting the androgen receptor pathway in prostate cancer. Curr Opin Pharmacol. 2008;8:440–448.
    1. Cunha GR. Role of mesenchymal-epithelial interactions in normal and abnormal development of the mammary gland and prostate. Cancer. 1994;74:1030–1044.
    1. Cunha GR, Chung LW. Stromal-epithelial interactions--I. Induction of prostatic phenotype in urothelium of testicular feminized (Tfm/y) mice. J Steroid Biochem. 1981;14:1317–1324.
    1. Cunha GR, Hayward SW, Wang YZ. Role of stroma in carcinogenesis of the prostate. Differentiation. 2002;70:473–485.
    1. De Gendt K, Swinnen JV, Saunders PT, Schoonjans L, Dewerchin M, Devos A, Tan K, Atanassova N, Claessens F, Lecureuil C, et al. A Sertoli cell-selective knockout of the androgen receptor causes spermatogenic arrest in meiosis. Proc Natl Acad Sci U S A. 2004;101:1327–1332.
    1. Ellwood-Yen K, Graeber TG, Wongvipat J, Iruela-Arispe ML, Zhang J, Matusik R, Thomas GV, Sawyers CL. Myc-driven murine prostate cancer shares molecular features with human prostate tumors. Cancer Cell. 2003;4:223–238.
    1. Gao H, Ouyang X, Banach-Petrosky WA, Shen MM, Abate-Shen C. Emergence of androgen independence at early stages of prostate cancer progression in Nkx3.1; Pten mice. Cancer Res. 2006;66:7929–7933.
    1. Gao T, Furnari F, Newton AC. PHLPP: a phosphatase that directly dephosphorylates Akt, promotes apoptosis, and suppresses tumor growth. Mol Cell. 2005;18:13–24.
    1. Gitenay D, Baron VT. Is EGR1 a potential target for prostate cancer therapy? Future Oncol. 2009;5:993–1003.
    1. Goldstein AS, Huang J, Guo C, Garraway IP, Witte ON. Identification of a cell of origin for human prostate cancer. Science. 2010;329:568–571.
    1. Greenberg NM, DeMayo F, Finegold MJ, Medina D, Tilley WD, Aspinall JO, Cunha GR, Donjacour AA, Matusik RJ, Rosen JM. Prostate cancer in a transgenic mouse. Proc Natl Acad Sci U S A. 1995;92:3439–3443.
    1. Ham WS, Cho NH, Kim WT, Ju HJ, Lee JS, Choi YD. Pathological effects of prostate cancer correlate with neuroendocrine differentiation and PTEN expression after bicalutamide monotherapy. J Urol. 2009;182:1378–1384.
    1. Hill R, Wu H. PTEN, stem cells, and cancer stem cells. J Biol Chem. 2009;284:11755–11759.
    1. Jiao J, Wang S, Qiao R, Vivanco I, Watson PA, Sawyers CL, Wu H. Murine cell lines derived from Pten null prostate cancer show the critical role of PTEN in hormone refractory prostate cancer development. Cancer Res. 2007;67:6083–6091.
    1. Kaarbo M, Mikkelsen OL, Malerod L, Qu S, Lobert VH, Akgul G, Halvorsen T, Maelandsmo GM, Saatcioglu F. PI3K-AKT-mTOR pathway is dominant over androgen receptor signaling in prostate cancer cells. Cell Oncol. 2010;32:11–27.
    1. Kinkade CW, Castillo-Martin M, Puzio-Kuter A, Yan J, Foster TH, Gao H, Sun Y, Ouyang X, Gerald WL, Cordon-Cardo C, Abate-Shen C. Targeting AKT/mTOR and ERK MAPK signaling inhibits hormone-refractory prostate cancer in a preclinical mouse model. J Clin Invest. 2008;118:3051–3064.
    1. Kunderfranco P, Mello-Grand M, Cangemi R, Pellini S, Mensah A, Albertini V, Malek A, Chiorino G, Catapano CV, Carbone GM. ETS transcription factors control transcription of EZH2 and epigenetic silencing of the tumor suppressor gene Nkx3.1 in prostate cancer. PLoS One. 2010;5:e10547.
    1. Lapointe J, Li C, Higgins JP, van de Rijn M, Bair E, Montgomery K, Ferrari M, Egevad L, Rayford W, Bergerheim U, et al. Gene expression profiling identifies clinically relevant subtypes of prostate cancer. Proc Natl Acad Sci U S A. 2004;101:811–816.
    1. Lei Q, Jiao J, Xin L, Chang CJ, Wang S, Gao J, Gleave ME, Witte ON, Liu X, Wu H. NKX3.1 stabilizes p53, inhibits AKT activation, and blocks prostate cancer initiation caused by PTEN loss. Cancer Cell. 2006;9:367–378.
    1. Lin HK, Hu YC, Lee DK, Chang C. Regulation of androgen receptor signaling by PTEN (phosphatase and tensin homolog deleted on chromosome 10) tumor suppressor through distinct mechanisms in prostate cancer cells. Mol Endocrinol. 2004;18:2409–2423.
    1. Lin HK, Hu YC, Yang L, Altuwaijri S, Chen YT, Kang HY, Chang C. Suppression versus induction of androgen receptor functions by the phosphatidylinositol 3-kinase/Akt pathway in prostate cancer LNCaP cells with different passage numbers. J Biol Chem. 2003;278:50902–50907.
    1. Magee JA, Chang LW, Stormo GD, Milbrandt J. Direct, androgen receptor-mediated regulation of the FKBP5 gene via a distal enhancer element. Endocrinology. 2006;147:590–598.
    1. Majumder PK, Febbo PG, Bikoff R, Berger R, Xue Q, McMahon LM, Manola J, Brugarolas J, McDonnell TJ, Golub TR, et al. mTOR inhibition reverses Akt-dependent prostate intraepithelial neoplasia through regulation of apoptotic and HIF-1-dependent pathways. Nat Med. 2004;10:594–601.
    1. Marumoto T, Tashiro A, Friedmann-Morvinski D, Scadeng M, Soda Y, Gage FH, Verma IM. Development of a novel mouse glioma model using lentiviral vectors. Nat Med. 2009;15:110–116.
    1. Messing EM, Manola J, Sarosdy M, Wilding G, Crawford ED, Trump D. Immediate hormonal therapy compared with observation after radical prostatectomy and pelvic lymphadenectomy in men with node-positive prostate cancer. N Engl J Med. 1999;341:1781–1788.
    1. Montgomery RB, Mostaghel EA, Vessella R, Hess DL, Kalhorn TF, Higano CS, True LD, Nelson PS. Maintenance of intratumoral androgens in metastatic prostate cancer: a mechanism for castration-resistant tumor growth. Cancer Res. 2008;68:4447–4454.
    1. Mostaghel EA, Page ST, Lin DW, Fazli L, Coleman IM, True LD, Knudsen B, Hess DL, Nelson CC, Matsumoto AM, et al. Intraprostatic androgens and androgen-regulated gene expression persist after testosterone suppression: therapeutic implications for castration-resistant prostate cancer. Cancer Res. 2007;67:5033–5041.
    1. Mulholland DJ, Xin L, Morim A, Lawson D, Witte O, Wu H. Lin-Sca-1+CD49fhigh stem/progenitors are tumor-initiating cells in the Pten-null prostate cancer model. Cancer Res. 2009;69:8555–8562.
    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 U S A. 2002;99:11890–11895.
    1. Niu Y, Altuwaijri S, Lai KP, Wu CT, Ricke WA, Messing EM, Yao J, Yeh S, Chang C. Androgen receptor is a tumor suppressor and proliferator in prostate cancer. Proc Natl Acad Sci U S A. 2008a;105:12182–12187.
    1. Niu Y, Altuwaijri S, Yeh S, Lai KP, Yu S, Chuang KH, Huang SP, Lardy H, Chang C. Targeting the stromal androgen receptor in primary prostate tumors at earlier stages. Proc Natl Acad Sci U S A. 2008b;105:12188–12193.
    1. Pei H, Li L, Fridley BL, Jenkins GD, Kalari KR, Lingle W, Petersen G, Lou Z, Wang L. FKBP51 affects cancer cell response to chemotherapy by negatively regulating Akt. Cancer Cell. 2009;16:259–266.
    1. Pei H, Lou Z, Wang L. Emerging role of FKBP51 in AKT kinase/protein kinase B signaling. Cell Cycle. 2010;9:6–7.
    1. Plaisier SB, Taschereau R, Wong JA, Graeber TG. Rank-rank hypergeometric overlap: identification of statistically significant overlap between gene-expression signatures. Nucleic Acids Res. 2010;38:e169.
    1. Ratajczak T, Ward BK, Minchin RF. Immunophilin chaperones in steroid receptor signalling. Curr Top Med Chem. 2003;3:1348–1357.
    1. Roudier MP, True LD, Higano CS, Vesselle H, Ellis W, Lange P, Vessella RL. Phenotypic heterogeneity of end-stage prostate carcinoma metastatic to bone. Hum Pathol. 2003;34:646–653.
    1. Sato N, Sadar MD, Bruchovsky N, Saatcioglu F, Rennie PS, Sato S, Lange PH, Gleave ME. Androgenic induction of prostate-specific antigen gene is repressed by protein-protein interaction between the androgen receptor and AP-1/c-Jun in the human prostate cancer cell line LNCaP. J Biol Chem. 1997;272:17485–17494.
    1. Scher HI, Beer TM, Higano CS, Anand A, Taplin ME, Efstathiou E, Rathkopf D, Shelkey J, Yu EY, Alumkal J, et al. Antitumour activity of MDV3100 in castration-resistant prostate cancer: a phase 1–2 study. Lancet. 2010;375:1437–1446.
    1. Sellers WR, Loda M. The EZH2 polycomb transcriptional repressor--a marker or mover of metastatic prostate cancer? Cancer Cell. 2002;2:349–350.
    1. Simanainen U, Allan CM, Lim P, McPherson S, Jimenez M, Zajac JD, Davey RA, Handelsman DJ. Disruption of prostate epithelial androgen receptor impedes prostate lobe-specific growth and function. Endocrinology. 2007;148:2264–2272.
    1. Simanainen U, McNamara K, Gao YR, Handelsman DJ. Androgen sensitivity of prostate epithelium is enhanced by postnatal androgen receptor inactivation. Am J Physiol Endocrinol Metab. 2009;296:E1335–1343.
    1. Stiles B, Groszer M, Wang S, Jiao J, Wu H. PTENless means more. Dev Biol. 2004;273:175–184.
    1. Tanaka H, Kono E, Tran CP, Miyazaki H, Yamashiro J, Shimomura T, Fazli L, Wada R, Huang J, Vessella RL, et al. Monoclonal antibody targeting of N-cadherin inhibits prostate cancer growth, metastasis and castration resistance. Nat Med 2010
    1. Taylor BS, Schultz N, Hieronymus H, Gopalan A, Xiao Y, Carver BS, Arora VK, Kaushik P, Cerami E, Reva B, et al. Integrative genomic profiling of human prostate cancer. Cancer Cell. 2010;18:11–22.
    1. Tetu B, Ro JY, Ayala AG, Johnson DE, Logothetis CJ, Ordonez NG. Small cell carcinoma of the prostate. Part I. A clinicopathologic study of 20 cases. Cancer. 1987;59:1803–1809.
    1. Tran C, Ouk S, Clegg NJ, Chen Y, Watson PA, Arora V, Wongvipat J, Smith-Jones PM, Yoo D, Kwon A, et al. Development of a second-generation antiandrogen for treatment of advanced prostate cancer. Science. 2009;324:787–790.
    1. Tran LM, Hyduke DR, Liao JC. Trimming of mammalian transcriptional networks using network component analysis. BMC Bioinformatics. 2010;11:511.
    1. Varambally S, Dhanasekaran SM, Zhou M, Barrette TR, Kumar-Sinha C, Sanda MG, Ghosh D, Pienta KJ, Sewalt RG, Otte AP, et al. The polycomb group protein EZH2 is involved in progression of prostate cancer. Nature. 2002;419:624–629.
    1. Vivanco I, Sawyers CL. The phosphatidylinositol 3-Kinase AKT pathway in human cancer. Nat Rev Cancer. 2002;2:489–501.
    1. Wang S, Gao J, Lei Q, Rozengurt N, Pritchard C, Jiao J, Thomas GV, Li G, Roy-Burman P, Nelson PS, et al. Prostate-specific deletion of the murine Pten tumor suppressor gene leads to metastatic prostate cancer. Cancer Cell. 2003;4:209–221.
    1. Wang XD, Wang BE, Soriano R, Zha J, Zhang Z, Modrusan Z, Cunha GR, Gao WQ. Expression profiling of the mouse prostate after castration and hormone replacement: implication of H-cadherin in prostate tumorigenesis. Differentiation. 2007;75:219–234.
    1. Wu CT, Altuwaijri S, Ricke WA, Huang SP, Yeh S, Zhang C, Niu Y, Tsai MY, Chang C. Increased prostate cell proliferation and loss of cell differentiation in mice lacking prostate epithelial androgen receptor. Proc Natl Acad Sci U S A. 2007;104:12679–12684.
    1. Wu X, Wu J, Huang J, Powell WC, Zhang J, Matusik RJ, Sangiorgi FO, Maxson RE, Sucov HM, Roy-Burman P. Generation of a prostate epithelial cell-specific Cre transgenic mouse model for tissue-specific gene ablation. Mech Dev. 2001;101:61–69.
    1. Yang SZ, Eltoum IA, Abdulkadir SA. Enhanced EGR1 activity promotes the growth of prostate cancer cells in an androgen-depleted environment. J Cell Biochem. 2006;97:1292–1299.
    1. Yu J, Mani RS, Cao Q, Brenner CJ, Cao X, Wang X, Wu L, Li J, Hu M, Gong Y, et al. An integrated network of androgen receptor, polycomb, and TMPRSS2-ERG gene fusions in prostate cancer progression. Cancer Cell. 2010;17:443–454.
    1. Yuan H, Young CY, Tian Y, Liu Z, Zhang M, Lou H. Suppression of the androgen receptor function by quercetin through protein-protein interactions of Sp1, c-Jun, and the androgen receptor in human prostate cancer cells. Mol Cell Biochem. 2010;339:253–262.
    1. Zhang W, Zhu J, Efferson CL, Ware C, Tammam J, Angagaw M, Laskey J, Bettano KA, Kasibhatla S, Reilly JF, et al. Inhibition of tumor growth progression by antiandrogens and mTOR inhibitor in a Pten-deficient mouse model of prostate cancer. Cancer Res. 2009;69:7466–7472.

Source: PubMed

3
Sottoscrivi