|
|
Mechanisms navigating the TGF-β pathway in prostate cancer |
Zheng Cao1,2,3,4, Natasha Kyprianou1,2,3,4
|
1. Department of Toxicology, University of Kentucky College of Medicine, Lexington, KY, USA;
2. Department of Urology, University of Kentucky College of Medicine, Lexington, KY, USA;
3. Department of Pathology, University of Kentucky College of Medicine, Lexington, KY, USA;
4. Department of Molecular and Cellular Biochemistry, University of Kentucky College of Medicine, Lexington, KY, USA |
|
|
Abstract Few pharmacotherapies are currently available to treat castration resistant prostate cancer (CRPC), with low impact on patient survival. Transforming growth factor-β (TGF-b) is a multi-functional peptide with opposite roles in prostate tumorigenesis as an inhibitor in normal growth and early stage disease and a promoter in advanced prostate cancer. Dysregulated TGF-β signaling leads to a cascade of events contributing to oncogenesis, including upregulated proliferation, decreased apoptosis, epithelial-to-mesenchymal transition (EMT) and evasion of immune surveillance. TGF-β signaling pathway presents an appropriate venue for establishing a therapeutic targeting platform in CRPC. Exploitation of TGF-β effectors and their cross talk with the androgen axis pathway will provide new insights into mechanisms of resistance of the current antiandrogen therapeutic strategies and lead to generation of new effective treatment modalities for CRPC. Points of functional convergence of TGF-β with key oncogenic pathways, including mitogen-activated protein kinase (MAPK) and androgen receptor (AR), are discussed as navigated within the EMT landscape in the tumor microenvironment. In this context the emerging anti-TGF-β pharmacotherapies for prostate cancer treatment are considered. Targeting the functional cross-talk between the TGF-β signaling effectors with the androgen axis supports the development of novel therapeutic strategies for treating CRPC with high specificity and efficacy in a personalized-medicine approach.
|
Received: 15 July 2014
Published: 06 February 2015
|
Fund:This work was supported by an NIH grant (RO1 DK 083761). |
Corresponding Authors:
Natasha Kyprianou
E-mail: nkypr2@uky.edu
|
|
|
[1] |
Siegel R, Naishadham D, Jemal A. Cancer statistics, 2013. CA Cancer J Clin 2013;63:11-30.
|
[2] |
Scardino PT. Early detection of prostate cancer. Urol Clin North Am 1989;16:635-55.
|
[3] |
HHS. U.S. Department of health and human services (HHS), a profile of older Americans. 2011.
|
[4] |
Sridhar SS, Freedland SJ, Gleave ME, Higano C, Mulders P, Parker C, et al. Castration-resistant prostate cancer: from new pathophysiology to new treatment. Eur Urol 2014;65: 289-99.
|
[5] |
Kageyama Y, Hyochi N, Kihara K, Sugiyama H. The androgen receptor as putative therapeutic target in hormonerefractory prostate cancer. Recent Pat Anticancer Drug Discov 2007;2:203-11.
|
[6] |
Jones E, Pu H, Kyprianou N. Targeting TGF-beta in prostate cancer: therapeutic possibilities during tumor progression. Expert Opin Ther Targets 2009;13:227-34.
|
[7] |
Lorente D, De Bono JS. Molecular alterations and emerging targets in castration resistant prostate cancer. Eur J Cancer 2014;50:753-64.
|
[8] |
Herpin A, Lelong C, Favrel P. Transforming growth factorbeta-related proteins: an ancestral and widespread superfamily of cytokines in metazoans. Dev Comp Immunol 2004; 28:461-85.
|
[9] |
Massague J. TGFbeta signalling in context. Nat Rev Mol Cell Biol 2012;13:616-30.
|
[10] |
Kulkarni AB, Karlsson S. Transforming growth factor-beta 1 knockout mice. A mutation in one cytokine gene causes a dramatic inflammatory disease. Am J Pathol 1993;143:3-9.
|
[11] |
Proetzel G, Pawlowski SA, Wiles MV, Yin M, Boivin GP, Howles PN, et al. Transforming growth factor-beta 3 is required for secondary palate fusion. Nat Genet 1995;11:409-14.
|
[12] |
Sanford LP, Ormsby I, Gittenberger-de Groot AC, Sariola H, Friedman R, Boivin GP, et al. TGFbeta2 knockout mice have multiple developmental defects that are non-overlapping with other TGFbeta knockout phenotypes. Development 1997;124:2659-70.
|
[13] |
Annes JP, Munger JS, Rifkin DB. Making sense of latent TGFbeta activation. J Cell Sci 2003;116:217-24.
|
[14] |
Guo Y, Kyprianou N. Overexpression of transforming growth factor (TGF) beta1 type II receptor restores TGF-beta1 sensitivity and signaling in human prostate cancer cells. Cell Growth Differ 1998;9:185-93.
|
[15] |
Massague J, Blain SW, Lo RS. TGFbeta signaling in growth control, cancer, and heritable disorders. Cell 2000;103: 295-309.
|
[16] |
Jang CW, Chen CH, Chen CC, Chen JY, Su YH, Chen RH. TGFbeta induces apoptosis through smad-mediated expression of DAP-kinase. Nat Cell Biol 2002;4:51-8.
|
[17] |
Azar R, Alard A, Susini C, Bousquet C, Pyronnet S. 4E-BP1 is a target of Smad4 essential for TGFbeta-mediated inhibition of cell proliferation. EMBO J 2009;28:3514-22.
|
[18] |
Hussey GS, Chaudhury A, Dawson AE, Lindner DJ, Knudsen CR, Wilce MC, et al. Identification of an mRNP complex regulating tumorigenesis at the translational elongation step. Mol Cell 2011;41:419-31.
|
[19] |
Kavsak P, Rasmussen RK, Causing CG, Bonni S, Zhu H, Thomsen GH, et al. Smad7 binds to Smurf2 to form an E3 ubiquitin ligase that targets the TGF beta receptor for degradation. Mol Cell 2000;6:1365-75.
|
[20] |
Zhu B, Kyprianou N. Transforming growth factor beta and prostate cancer. Cancer Treat Res 2005;126:157-73.
|
[21] |
Collazo J, Zhu B, Larkin S, Martin SK, Pu H, Horbinski C, et al. Cofilin drives cell-invasive and metastatic responses to TGFbeta in prostate cancer. Cancer Res 2014;74:2362-73.
|
[22] |
Zhu B, Fukada K, Zhu H, Kyprianou N. Prohibitin and cofilin are intracellular effectors of transforming growth factor beta signaling in human prostate cancer cells. Cancer Res 2006; 66:8640-7.
|
[23] |
Bacman D, Merkel S, Croner R, Papadopoulos T, Brueckl W, Dimmler A. TGF-beta receptor 2 downregulation in tumourassociated stroma worsens prognosis and high-grade tumours show more tumour-associated macrophages and lower TGF-beta1 expression in colon carcinoma: a retrospective study. BMC Cancer 2007;7:156.
|
[24] |
Dong M, How T, Kirkbride KC, Gordon KJ, Lee JD, Hempel N, et al. The type III TGF-beta receptor suppresses breast cancer progression. J Clin Invest 2007;117:206-17.
|
[25] |
Guo Y, Jacobs SC, Kyprianou N. Down-regulation of protein and mRNA expression for transforming growth factor-beta (TGF-beta1) type I and type II receptors in human prostate cancer. Int J Cancer 1997;71:573-9.
|
[26] |
Guo Y, Kyprianou N. Restoration of transforming growth factor beta signaling pathway in human prostate cancer cells suppresses tumorigenicity via induction of caspase-1-mediated apoptosis. Cancer Res 1999;59:1366-71.
|
[27] |
Pu H, Collazo J, Jones E, Gayheart D, Sakamoto S, Vogt A, et al. Dysfunctional transforming growth factor-beta receptor II accelerates prostate tumorigenesis in the TRAMP mouse model. Cancer Res 2009;69:7366-74.
|
[28] |
Teixeira AL, Gomes M, Nogueira A, Azevedo AS, Assis J, Dias F, et al. Improvement of a predictive model of castration-resistant prostate cancer: functional genetic variants in TGFbeta1 signaling pathway modulation. PLoS One 2013;8:e72419.
|
[29] |
Gatza CE, Oh SY, Blobe GC. Roles for the type III TGF-beta receptor in human cancer. Cell Signal 2010;22:1163-74.
|
[30] |
Sharifi N, Hurt EM, Kawasaki BT, Farrar WL. TGFBR3 loss and consequences in prostate cancer. Prostate 2007;67: 301-11.
|
[31] |
Turley RS, Finger EC, Hempel N, How T, Fields TA, Blobe GC. The type III transforming growth factor-beta receptor as a novel tumor suppressor gene in prostate cancer. Cancer Res 2007;67:1090-8.
|
[32] |
Bandyopadhyay A, Wang L, López-Casillas F, Mendoza V, Yeh IT, Sun L. Systemic administration of a soluble betaglycan suppresses tumor growth, angiogenesis, and matrix metalloproteinase-9 expression in a human xenograft model of prostate cancer. Prostate 2005;63:81-90.
|
[33] |
Yu N, Kozlowski JM, Park II, Chen L, Zhang Q, Xu D, et al. Overexpression of transforming growth factor beta1 in malignant prostate cells is partly caused by a runaway of TGFbeta1 auto-induction mediated through a defective recruitment of protein phosphatase 2A by TGF-beta type I receptor. Urology 2010;76:1519.e8-1519.e13.
|
[34] |
Zhang Q, Chen L, Helfand BT, Jang TL, Sharma V, Kozlowski J, et al. TGF-beta regulates DNA methyltransferase expression in prostate cancer, correlates with aggressive capabilities, and predicts disease recurrence. PLoS One 2011;6:e25168.
|
[35] |
Mu Y, Sundar R, Thakur N, Ekman M, Gudey SK, Yakymovych M, et al. TRAF6 ubiquitinates TGFbeta type I receptor to promote its cleavage and nuclear translocation in cancer. Nat Commun 2011;2:330.
|
[36] |
Wang W, Mouneimne G, Sidani M, Wyckoff J, Chen X, Makris A, et al. The activity status of cofilin is directly related to invasion, intravasation, and metastasis of mammary tumors. J Cell Biol 2006;173:395-404.
|
[37] |
Zhu ML, Kyprianou N. Androgen receptor and growth factor signaling cross-talk in prostate cancer cells. Endocr Relat Cancer 2008;15:841-9.
|
[38] |
Bruckheimer EM, Kyprianou N. Dihydrotestosterone enhances transforming growth factor-beta-induced apoptosis in hormone-sensitive prostate cancer cells. Endocrinology 2001;142:2419-26.
|
[39] |
Bruckheimer EM, Kyprianou N. Bcl-2 antagonizes the combined apoptotic effect of transforming growth factor-beta and dihydrotestosterone in prostate cancer cells. Prostate 2002;53:133-42.
|
[40] |
Wang H, Song K, Sponseller TL, Danielpour D. Novel function of androgen receptor-associated protein 55/Hic-5 as a negative regulator of Smad3 signaling. J Biol Chem 2005;280: 5154-62.
|
[41] |
Yoon G, Kim JY, Choi YK, Won YS, Lim IK. Direct activation of TGF-beta1 transcription by androgen and androgen receptor complex in Huh7 human hepatoma cells and its tumor in nude mice. J Cell Biochem 2006;97:393-411.
|
[42] |
Hayes SA, Zarnegar M, Sharma M, Yang F, Peehl DM, ten Dijke P, et al. SMAD3 represses androgen receptor-mediated transcription. Cancer Res 2001;61:2112-8.
|
[43] |
Song K, Wang H, Krebs TL, Wang B, Kelley TJ, Danielpour D. DHT selectively reverses Smad3-mediated/TGF-betainduced responses through transcriptional down-regulation of Smad3 in prostate epithelial cells. Mol Endocrinol 2010; 24:2019-29.
|
[44] |
Qi W, Gao S, Chu J, Zhou L, Wang Z. Negative androgenresponse elements mediate androgen-dependent transcriptional inhibition of TGF-beta1 and CDK2 promoters in the prostate gland. J Androl 2012;33:27-36.
|
[45] |
Yu S, Xia S, Yang D, Wang K, Yeh S, Gao Z, et al. Androgen receptor in human prostate cancer-associated fibroblasts promotes prostate cancer epithelial cell growth and invasion. Med Oncol 2013;30:674.
|
[46] |
Mishra S, Deng JJ, Gowda PS, Rao MK, Lin CL, Chen CL, et al. Androgen receptor and microRNA-21 axis downregulates transforming growth factor beta receptor II (TGFBR2) expression in prostate cancer. Oncogene 2014;33:4097-106.
|
[47] |
Ao M, Williams K, Bhowmick NA, Hayward SW. Transforming growth factor-beta promotes invasion in tumorigenic but not in nontumorigenic human prostatic epithelial cells. Cancer Res 2006;66:8007-16.
|
[48] |
Roberts AB, Tian F, Byfield SD, Stuelten C, Ooshima A, Saika S, et al. Smad3 is key to TGF-beta-mediated epithelialto-mesenchymal transition, fibrosis, tumor suppression and metastasis. Cytokine Growth Factor Rev 2006;17(1-2): 19-27.
|
[49] |
Zhang X, Fournier MV, Ware JL, Bissell MJ, Yacoub A, Zehner ZE. Inhibition of vimentin or beta1 integrin reverts morphology of prostate tumor cells grown in laminin-rich extracellular matrix gels and reduces tumor growth in vivo. Mol Cancer Ther 2009;8:499-508.
|
[50] |
Zhang Q, Helfand BT, Jang TL, Zhu LJ, Chen L, Yang XJ, et al. Nuclear factor-kappaB-mediated transforming growth factor-beta-induced expression of vimentin is an independent predictor of biochemical recurrence after radical prostatectomy. Clin Cancer Res 2009;15:3557-67.
|
[51] |
Slabáková E, Pernicová Z, Slaví?ková E, Staršíchová A, Kozubík A, Sou?ek K. TGF-beta1-induced EMT of nontransformed prostate hyperplasia cells is characterized by early induction of SNAI2/Slug. Prostate 2011;71:1332-43.
|
[52] |
Mani SA, Guo W, Liao MJ, Eaton EN, Ayyanan A, Zhou AY, et al. The epithelial-mesenchymal transition generates cells with properties of stem cells. Cell 2008;133:704-15.
|
[53] |
Konrad L, Scheiber JA, Schwarz L, Schrader AJ, Hofmann R. TGF-beta1 and TGF-beta2 strongly enhance the secretion of plasminogen activator inhibitor-1 and matrix metalloproteinase-9 of the human prostate cancer cell line PC-3. Regul Pept 2009;155(1-3):28-32.
|
[54] |
Ha B, Ko H, Kim B, Sohn EJ, Jung JH, Kim JS, et al. Regulation of crosstalk between epithelial to mesenchymal transition molecules and MMP-9 mediates the antimetastatic activity of anethole in DU145 prostate cancer cells. J Nat Prod 2014;77: 63-9.
|
[55] |
Lin TH, Lee SO, Niu Y, Xu D, Liang L, Li L, et al. Differential androgen deprivation therapies with anti-androgens casodex/ bicalutamide or MDV3100/Enzalutamide versus antiandrogen receptor ASC-J9(R) Lead to promotion versus suppression of prostate cancer metastasis. J Biol Chem 2013; 288:19359-69.
|
[56] |
Wang X, Lee SO, Xia S, Jiang Q, Luo J, Li L, et al. Endothelial cells enhance prostate cancer metastasis via IL-6->androgen receptor->TGF-beta->MMP-9 signals. Mol Cancer Ther 2013; 12:1026-37.
|
[57] |
Morimoto K, Tanaka T, Nitta Y, Ohnishi K, Kawashima H, Nakatani T. NEDD9 crucially regulates TGF-beta-triggered epithelial-mesenchymal transition and cell invasion in prostate cancer cells: involvement in cancer progressiveness. Prostate 2014;74:901-10.
|
[58] |
Shiota M, Zardan A, Takeuchi A, Kumano M, Beraldi E, Naito S, et al. Clusterin mediates TGF-beta-induced epithelial-mesenchymal transition and metastasis via twist1 in prostate cancer cells. Cancer Res 2012;72:5261-72.
|
[59] |
Barron DA, Rowley DR. The reactive stroma microenvironment and prostate cancer progression. Endocr Relat Cancer 2012;19:R187-204.
|
[60] |
Ting HJ, Deep G, Jain AK, Cimic A, Sirintrapun J, Romero LM, et al. Silibinin prevents prostate cancer cell-mediated differentiation of naive fibroblasts into cancer-associated fibroblast phenotype by targeting TGF beta2. Mol Carcinog 2014. http://dx.doi.org/10.1002/mc.22135 [Epub ahead of print].
|
[61] |
Epstein JI. Diagnosis and reporting of limited adenocarcinoma of the prostate on needle biopsy. Mod Pathol 2004;17: 307-15.
|
[62] |
Mirzoeva S, Franzen CA, Pelling JC. Apigenin inhibits TGFbeta-induced VEGF expression in human prostate carcinoma cells via a Smad2/3-and Src-dependent mechanism. Mol Carcinog 2014;53:598-609.
|
[63] |
Bhowmick NA, Chytil A, Plieth D, Gorska AE, Dumont N, Shappell S, et al. TGF-beta signaling in fibroblasts modulates the oncogenic potential of adjacent epithelia. Science 2004; 303:848-51.
|
[64] |
Bartholin L, Cyprian FS, Vincent D, Garcia CN, Martel S, Horvat B, et al. Generation of mice with conditionally activated transforming growth factor beta signaling through the TbetaRI/ALK5 receptor. Genesis 2008;46:724-31.
|
[65] |
Marie JC, Liggitt D, Rudensky AY. Cellular mechanisms of fatal early-onset autoimmunity in mice with the T cellspecific targeting of transforming growth factor-beta receptor. Immunity 2006;25:441-54.
|
[66] |
Donkor MK, Sarkar A, Savage PA, Franklin RA, Johnson LK, Jungbluth AA, et al. T cell surveillance of oncogene-induced prostate cancer is impeded by T cell-derived TGF-beta1 cytokine. Immunity 2011;35:123-34.
|
[67] |
Fontenot JD, Gavin MA, Rudensky AY. Foxp3 programs the development and function of CD4+CD25+ regulatory T cells. Nat Immunol 2003;4:330-6.
|
[68] |
Chen W, Jin W, Hardegen N, Lei KJ, Li L, Marinos N, et al. Conversion of peripheral CD4+CD25- naive T cells to CD4+CD25+ regulatory T cells by TGF-beta induction of transcription factor Foxp3. J Exp Med 2003;198:1875-86.
|
[69] |
Zhang Q, Jang TL, Yang X, Park I, Meyer RE, Kundu S, et al. Infiltration of tumor-reactive transforming growth factorbeta insensitive CD8+ T cells into the tumor parenchyma is associated with apoptosis and rejection of tumor cells. Prostate 2006;66:235-47.
|
[70] |
Hensley PJ, Kyprianou N. Modeling prostate cancer in mice: limitations and opportunities. J Androl 2012;33:133-44.
|
[71] |
Li X, Sterling JA, Fan KH, Vessella RL, Shyr Y, Hayward SW, et al. Loss of TGF-beta responsiveness in prostate stromal cells alters chemokine levels and facilitates the development of mixed osteoblastic/osteolytic bone lesions. Mol Cancer Res 2012;10:494-503.
|
[72] |
Thompson TC, Truong LD, Timme TL, Kadmon D, McCune BK, Flanders KC, et al. Transgenic models for the study of prostate cancer. Cancer 1993;71(3 Suppl):1165-71.
|
[73] |
Ding Z, Wu CJ, Chu GC, Xiao Y, Ho D, Zhang J, et al. SMAD4-dependent barrier constrains prostate cancer growth and metastatic progression. Nature 2011;470:269-73.
|
[74] |
Flavell RA, Sanjabi S, Wrzesinski SH, Licona-Limón P. The polarization of immune cells in the tumour environment by TGFbeta. Nat Rev Immunol 2010;10:554-67.
|
[75] |
Calve-Aller E. First human dose escalation study in patients with metastatic malignancies to determine safety and pharmacokinetics of LY2157299, a small molecule inhibitor of the transforming growth factor-b receptor I kinase. ASCO Annu Meet J Clin Oncol 2008;26:14554. Abstr.
|
[76] |
Trachtman H, Fervenza FC, Gipson DS, Heering P, Jayne DR, Peters H, et al. A phase 1, single-dose study of fresolimumab, an anti-TGF-beta antibody, in treatment-resistant primary focal segmental glomerulosclerosis. Kidney Int 2011;79: 1236-43.
|
[77] |
Katragadda L, Carter BZ, Borthakur G. XIAP antisense therapy with AEG 35156 in acute myeloid leukemia. Expert Opin Investig Drugs 2013;22:663-70.
|
[78] |
Muraoka RS, Dumont N, Ritter CA, Dugger TC, Brantley DM, Chen J, et al. Blockade of TGF-beta inhibits mammary tumor cell viability, migration, and metastases. J Clin Invest 2002; 109:1551-9.
|
[1] |
Zhao Pei,Zhu Yezi,Cheng Liang,Luo Jun. Detection of androgen receptor (AR) and AR-V7 in small cell prostate carcinoma: Diagnostic and therapeutic implications[J]. Asian Journal of Urology, 2019, 6(1): 109-113. |
[2] |
M. Armstrong Cameron,C. Gao Allen. Current strategies for targeting the activity of androgen receptor variants[J]. Asian Journal of Urology, 2019, 6(1): 42-49. |
[3] |
Xu Lingfan,Chen Junyi,Liu Weipeng,Liang Chaozhao,Hu Hailiang,Huang Jiaoti. Targeting androgen receptor-independent pathways in therapy-resistant prostate cancer[J]. Asian Journal of Urology, 2019, 6(1): 91-98. |
[4] |
Jin Xu, Yun Qiu. Role of androgen receptor splice variants in prostate cancer metastasis[J]. Asian Journal of Urology, 2016, 3(4): 177-184. |
[5] |
Takashi Kobayashi. Understanding the biology of urothelial cancer metastasis[J]. Asian Journal of Urology, 2016, 3(4): 211-222. |
[6] |
Jun Luo. Non-invasive actionable biomarkers for metastatic prostate cancer[J]. Asian Journal of Urology, 2016, 3(4): 170-176. |
[7] |
Styliani Karanika, Theodoros Karantanos, Jianhua Yin, Likun Li, Timothy C. Thompson. Novel anti-androgen receptor signaling agents: understanding the mechanisms of resistance[J]. Asian Journal of Urology, 2014, 1(1): 28-37. |
|
|
|
|