全部 标题 作者
关键词 摘要

OALib Journal期刊
ISSN: 2333-9721
费用:99美元

查看量下载量

相关文章

更多...

Dissecting Major Signaling Pathways throughout the Development of Prostate Cancer

DOI: 10.1155/2013/920612

Full-Text   Cite this paper   Add to My Lib

Abstract:

Prostate cancer (PCa) is one of the most common malignancies found in males. The development of PCa involves several mutations in prostate epithelial cells, usually linked to developmental changes, such as enhanced resistance to apoptotic death, constitutive proliferation, and, in some cases, to differentiation into an androgen deprivation-resistant phenotype, leading to the appearance of castration-resistant PCa (CRPCa), which leads to a poor prognosis in patients. In this review, we summarize recent findings concerning the main deregulations into signaling pathways that will lead to the development of PCa and/or CRPCa. Key mutations in some pathway molecules are often linked to a higher prevalence of PCa, by directly affecting the respective cascade and, in some cases, by deregulating a cross-talk node or junction along the pathways. We also discuss the possible environmental and nonenvironmental inducers for these mutations, as well as the potential therapeutic strategies targeting these signaling pathways. A better understanding of how some risk factors induce deregulation of these signaling pathways, as well as how these deregulated pathways affect the development of PCa and CRPCa, will further help in the development of new treatments and prevention strategies for this disease. 1. Introduction The long-term ineffectiveness of current treatments for prostate cancer (PCa) has spurred an increasing interest in understanding the molecular mechanisms that underlie PCa tumorigenesis [1]. Currently, PCa is considered the most common nonmelanoma neoplasia among men [2–4]. According to the current trends in population growth, the incidence of PCa will exceed 1.7 million new cases by 2030 [5]. In the United States, nearly 2.8 million men are potentially living with this condition, and approximately 240,000 new cases were diagnosed in 2012 [3]. PCa predominately affects elderly men with higher incidence [6], and it is more prevalent in Western countries [7], where the average life expectation is over 75 years old. In developing countries like Brazil, PCa has recently surpassed the population incidence of breast cancer, and it has become the most common tumor malignancy, with approximately 50,000 new cases occurring per year [4, 5]. Yet, there is a considerable heterogeneity in the mortality rates and incidence among different countries, probably due to the variable penetrance of some risk factors such as age, race, genetics (family history), diet and environmental factors [8], and also behavioral factors, like frequent consumption of dairy products and meat

References

[1]  M. Verras and Z. Sun, “Roles and regulation of Wnt signaling and β-catenin in prostate cancer,” Cancer Letters, vol. 237, no. 1, pp. 22–32, 2006.
[2]  J. K. Mullins and S. Loeb, “Environmental exposures and prostate cancer,” Urologic Oncology, vol. 30, pp. 216–219, 2012.
[3]  R. Siegel, C. DeSantis, K. Virgo et al., “Cancer treatment and survivorship statistics, 2012,” CA: A Cancer Journal for Clinicians, vol. 62, pp. 220–241, 2012.
[4]  F. C. Maluf, O. Smaletz, and D. Herchenhorn, “Castration-resistant prostate cancer: systemic therapy in 2012,” Clinics, vol. 67, pp. 389–394, 2012.
[5]  M. M. Center, A. Jemal, J. Lortet-Tieulent et al., “International variation in prostate cancer incidence and mortality rates,” European Urology, vol. 61, pp. 1079–1092, 2012.
[6]  G. Arcangeli, B. Saracino, S. Gomellini et al., “A Prospective phase III randomized trial of hypofractionation versus conventional fractionation in patients with high-risk prostate cancer,” International Journal of Radiation Oncology Biology Physics, vol. 78, no. 1, pp. 11–18, 2010.
[7]  H. G. Sim and C. W. S. Cheng, “Changing demography of prostate cancer in Asia,” European Journal of Cancer, vol. 41, no. 6, pp. 834–845, 2005.
[8]  S. H. Reuben, “Reducing environmental cancer risk: what we can do now,” The President's Cancer Panel, pp. 1–240, 2010.
[9]  M. F. Leitzmann and S. Rohrmann, “Risk factors for the onset of prostatic cancer: age, location, and behavioral correlates,” Clinical Epidemiology, vol. 4, pp. 1–11, 2012.
[10]  B. G. Timms, “Prostate development: a historical perspective,” Differentiation, vol. 76, no. 6, pp. 565–577, 2008.
[11]  A. R. Kristal, K. B. Arnold, M. L. Neuhouser et al., “Diet, supplement use, and prostate cancer risk: results from the prostate cancer prevention trial,” American Journal of Epidemiology, vol. 172, no. 5, pp. 566–577, 2010.
[12]  L. J. Su, L. Arab, S. E. Steck et al., “Obesity and prostate cancer aggressiveness among African and Caucasian Americans in a population-based study,” Cancer Epidemiology Biomarkers and Prevention, vol. 20, no. 5, pp. 844–853, 2011.
[13]  S. Koifman and R. J. Koifman, “Environment and cancer in Brazil: an overview from a public health perspective,” Mutation Research, vol. 544, no. 2-3, pp. 305–311, 2003.
[14]  R. Sichieri, J. E. Everhart, and G. A. Mendonca, “Diet and mortality from common cancers in Brazil: an ecological study,” Cadernos de Saúde Pública, vol. 12, pp. 53–59, 1996.
[15]  P. Cocco, “On the rumors about the silent spring. Review of the scientific evidence linking occupational and environmental pesticide exposure to endocrine disruption health effects,” Cadernos de Saúde Pública, vol. 18, no. 2, pp. 379–402, 2002.
[16]  H. J. van der Rhee, E. de Vries, and J. W. W. Coebergh, “Does sunlight prevent cancer? A systematic review,” European Journal of Cancer, vol. 42, no. 14, pp. 2222–2232, 2006.
[17]  S. J. Freedland, W. B. Isaacs, E. A. Platz et al., “Prostate size and risk of high-grade, advanced prostate cancer and biochemical progression after radical prostatectomy: a search database study,” Journal of Clinical Oncology, vol. 23, no. 30, pp. 7546–7554, 2005.
[18]  H. T. Lynch and J. F. Lynch, “The Lynch syndrome: melding natural history and molecular genetics to genetic counseling and cancer control,” Cancer Control, vol. 3, no. 1, pp. 13–19, 1996.
[19]  J. K. Jin, F. Dayyani, and G. E. Gallick, “Steps in prostate cancer progression that lead to bone metastasis,” International Journal of Cancer, vol. 128, no. 11, pp. 2545–2561, 2011.
[20]  P. E. Lonergan and D. J. Tindall, “Androgen receptor signaling in prostate cancer development and progression,” Journal of Carcinogenesis, vol. 10, article 20, 2011.
[21]  S. A. Boorjian, J. A. Eastham, M. Graefen et al., “A critical analysis of the long-term impact of radical prostatectomy on cancer control and function outcomes,” European Urology, vol. 61, pp. 664–675, 2012.
[22]  Z. Culig, A. Hobisch, M. V. Cronauer et al., “Androgen receptor activation in prostatic tumor cell lines by insulin- like growth factor-I, keratinocyte growth factor, and epidermal growth factor,” Cancer Research, vol. 54, no. 20, pp. 5474–5478, 1994.
[23]  A. Wolk, C. S. Mantzoros, S. O. Andersson et al., “Insulin-like growth factor 1 and prostate cancer risk: a population-based, case-control study,” Journal of the National Cancer Institute, vol. 90, no. 12, pp. 911–915, 1998.
[24]  D. Gioeli, S. B. Ficarro, J. J. Kwiek et al., “Androgen receptor phosphorylation. Regulation and identification of the phosphorylation sites,” Journal of Biological Chemistry, vol. 277, no. 32, pp. 29304–29314, 2002.
[25]  Z. Guo, B. Dai, T. Jiang et al., “Regulation of androgen receptor activity by tyrosine phosphorylation,” Cancer Cell, vol. 10, no. 4, pp. 309–319, 2006.
[26]  E. Hernes, Foss?, A. Berner, B. Otnes, and J. M. Nesland, “Expression of the epidermal growth factor receptor family in prostate carcinoma before and during androgen-independence,” British Journal of Cancer, vol. 90, no. 2, pp. 449–454, 2004.
[27]  S. M. Green, E. A. Mostaghel, and P. S. Nelson, “Androgen action and metabolism in prostate cancer,” Molecular and Cellular Endocrinology, vol. 360, pp. 3–13, 2012.
[28]  S. Q. Yu, K. P. Lai, S. J. Xia, H. C. Chang, C. Chang, and S. Yeh, “The diverse and contrasting effects of using human prostate cancer cell lines to study androgen receptor roles in prostate cancer,” Asian Journal of Andrology, vol. 11, no. 1, pp. 39–48, 2009.
[29]  C. GIampietri, S. Petrungaro, F. Padula et al., “Autophagy modulators sensitize prostate epithelial cancer cell lines to TNF-alpha-dependent apoptosis,” Apoptosis, vol. 17, no. 11, pp. 1210–1222, 2012.
[30]  X. Huang, F. Yuan, M. Liang et al., “M-HIFU inhibits tumor growth, suppresses STAT3 activity and enhances tumor specific immunity in a transplant tumor model of prostate cancer,” PLoS ONE, vol. 7, Article ID e41632, 2012.
[31]  A. Vajda, L. Marignol, C. Barrett et al., “Gene expression analysis in prostate cancer: the importance of the endogenous control,” Prostate, vol. 73, pp. 382–390, 2012.
[32]  C. E. Lin, S. U. Chen, C. C. Lin et al., “Lysophosphatidic acid enhances vascular endothelial growth factor-C expression in human prostate cancer PC-3 cells,” PLoS ONE, vol. 7, Article ID e41096, 2012.
[33]  P. Saraon, N. Musrap, D. Cretu et al., “Proteomic profiling of androgen-independent prostate cancer cell lines reveals a role for protein S during the development of high grade and castration-resistant prostate cancer,” Journal of Biological Chemistry, vol. 287, pp. 34019–34031, 2012.
[34]  B. S. Carver, C. Chapinski, J. Wongvipat et al., “Reciprocal feedback regulation of PI3K and androgen receptor signaling in PTEN-deficient prostate cancer,” Cancer Cell, vol. 19, no. 5, pp. 575–586, 2011.
[35]  J. A. Ewald, J. A. Desotelle, D. R. Church et al., “Androgen deprivation induces senescence characteristics in prostate cancer cells in vitro and in vivo,” Prostate, vol. 73, pp. 337–345, 2012.
[36]  P. J. Maxwell, J. Coulter, S. M. Walker et al., “Potentiation of inflammatory CXCL8 signalling sustains cell survival in PTEN-deficient prostate carcinoma,” European Urology, 2012.
[37]  R. S. Jackson II, W. Placzek, A. Fernandez et al., “Sabutoclax, a Mcl-1 antagonist, inhibits tumorigenesis in transgenic mouse and human xenograft models of prostate cancer,” Neoplasia, vol. 14, pp. 656–665, 2012.
[38]  H. Wang, Y. Xu, Z. Fang, S. Chen, S. P. Balk, and X. Yuan, “Doxycycline regulated induction of AKT in murine prostate drives proliferation independently of p27 cyclin dependent kinase inhibitor downregulation,” PLoS ONE, vol. 7, Article ID e41330, 2012.
[39]  M. A. Reynolds, “Molecular alterations in prostate cancer,” Cancer Letters, vol. 271, no. 1, pp. 13–24, 2008.
[40]  M. E. Grossmann, H. Huang, and D. J. Tindall, “Androgen receptor signaling in androgen-refractory prostate cancer,” Journal of the National Cancer Institute, vol. 93, no. 22, pp. 1687–1697, 2001.
[41]  S. C. Chan, Y. Li, and S. M. Dehm, “Androgen receptor splice variants activate androgen receptor target genes and support aberrant prostate cancer cell growth independent of canonical androgen receptor nuclear localization signal,” Journal of Biological Chemistry, vol. 287, pp. 19736–19749, 2012.
[42]  G. Castoria, L. D'Amato, A. Ciociola et al., “Androgen-induced cell migration: role of androgen receptor/filamin A association,” PLoS ONE, vol. 6, Article ID e17218, 2012.
[43]  K. K. Waltering, A. Urbanucci, and T. Visakorpi, “Androgen receptor (AR) aberrations in castration-resistant prostate cancer,” Molecular and Cellular Endocrinology, vol. 360, pp. 38–43, 2012.
[44]  H. Wang, C. Zhang, A. Rorick et al., “CCI-779 inhibits cell-cycle G2-M progression and invasion of castration-resistant prostate cancer via attenuation of UBE2C transcription and mRNA stability,” Cancer Research, vol. 71, no. 14, pp. 4866–4876, 2011.
[45]  A. Urbanucci, B. Sahu, J. Seppala et al., “Overexpression of androgen receptor enhances the binding of the receptor to the chromatin in prostate cancer,” Oncogene, vol. 31, pp. 2153–2163, 2012.
[46]  M. Shiota, A. Yokomizo, and S. Naito, “Increased androgen receptor transcription: a cause of castration-resistant prostate cancer and a possible therapeutic target,” Journal of Molecular Endocrinology, vol. 47, no. 1, pp. R25–R41, 2011.
[47]  M. Shiota, A. Yokomizo, and S. Naito, “Oxidative stress and androgen receptor signaling in the development and progression of castration-resistant prostate cancer,” Free Radical Biology and Medicine, vol. 51, pp. 1320–1328, 2011.
[48]  Y. Zuo and J. K. Cheng, “Small ubiquitin-like modifier protein-specific protease 1 and prostate cancer,” Asian Journal of Andrology, vol. 11, no. 1, pp. 36–38, 2009.
[49]  T. Bawa-Khalfe, J. Cheng, Z. Wang, and E. T. H. Yeh, “Induction of the SUMO-specific protease 1 transcription by the androgen receptor in prostate cancer cells,” Journal of Biological Chemistry, vol. 282, no. 52, pp. 37341–37349, 2007.
[50]  N. D. Perkins, “Integrating cell-signalling pathways with NF-kappaB and IKK function,” Nature Reviews Molecular Cell Biology, vol. 8, pp. 49–62, 2007.
[51]  C. Cai, F. N. Jiang, Y. X. Liang et al., “Classical and alternative nuclear factor-kappaB pathways: a comparison among normal prostate, benign prostate hyperplasia and prostate cancer,” Pathology and Oncology Research, vol. 17, pp. 873–878, 2011.
[52]  V. Tergaonkar, “NFκB pathway: a good signaling paradigm and therapeutic target,” International Journal of Biochemistry and Cell Biology, vol. 38, no. 10, pp. 1647–1653, 2006.
[53]  B. Razani, A. D. Reichardt, and G. Cheng, “Non-canonical NF-kappaB signaling activation and regulation: principles and perspectives,” Immunological Reviews, vol. 244, pp. 44–54, 2011.
[54]  R. C. Rickert, J. Jellusova, and A. V. Miletic, “Signaling by the tumor necrosis factor receptor superfamily in B-cell biology and disease,” Immunological Reviews, vol. 244, pp. 115–133, 2011.
[55]  L. Gu, A. Dagvadorj, J. Lutz et al., “Transcription factor Stat3 stimulates metastatic behavior of human prostate cancer cells in vivo, whereas Stat5b has a preferential role in the promotion of prostate cancer cell viability and tumor growth,” American Journal of Pathology, vol. 176, no. 4, pp. 1959–1972, 2010.
[56]  J. Chen, K. V. Giridhar, L. Zhang, S. Xu, and Q. Jane Wang, “A protein kinase C/protein kinase D pathway protects LNCaP prostate cancer cells from phorbol ester-induced apoptosis by promoting ERK1/2 and NF-κB activities,” Carcinogenesis, vol. 32, no. 8, pp. 1198–1206, 2011.
[57]  A. Hsu, R. S. Bruno, C. V. L?hr et al., “Dietary soy and tea mitigate chronic inflammation and prostate cancer via NFκB pathway in the Noble rat model,” Journal of Nutritional Biochemistry, vol. 22, no. 5, pp. 502–510, 2011.
[58]  R. Benelli, R. Vene, M. Ciarlo, S. Carlone, O. Barbieri, and N. Ferrari, “The AKT/NF-kappaB inhibitor xanthohumol is a potent anti-lymphocytic leukemia drug overcoming chemoresistance and cell infiltration,” Biochemical Pharmacology, vol. 83, pp. 1634–1642, 2012.
[59]  G. Jain, C. Voogdt, A. Tobias et al., “IkappaB kinases modulate the activity of the androgen receptor in prostate carcinoma cell lines,” Neoplasia, vol. 14, pp. 178–189, 2012.
[60]  Y. Fang, H. Sun, J. Zhai et al., “Antitumor activity of NF-kB decoy oligodeoxynucleotides in a prostate cancer cell line,” Asian Pacific Journal of Cancer Prevention, vol. 12, pp. 2721–2726, 2012.
[61]  W. Xiao, D. R. Hodge, L. Wang, X. Yang, X. Zhang, and W. L. Farrar, “Co-operative functions between nuclear factors NFκB and CCAT/enhancer-binding protein-β (C/EBP-β) regulate the IL-6 promoter in autocrine human prostate cancer cells,” Prostate, vol. 61, no. 4, pp. 354–370, 2004.
[62]  S. Shukla, G. T. MacLennan, P. Fu et al., “Nuclear factor-κB/p65 (Rel A) is constitutively activated in human prostate adenocarcinoma and correlates with disease progression,” Neoplasia, vol. 6, no. 4, pp. 390–400, 2004.
[63]  C. D. Chen and C. L. Sawyers, “NF-κB activates prostate-specific antigen expression and is upregulated in androgen-independent prostate cancer,” Molecular and Cellular Biology, vol. 22, no. 8, pp. 2862–2870, 2002.
[64]  M. Paz De Miguel, M. Royuela, F. R. Bethencourt, L. Santamaría, B. Fraile, and R. Paniagua, “Immunoexpression of tumour necrosis factor-α and its receptors 1 and 2 correlates with proliferation/apoptosis equilibrium in normal, hyperplasic and carcinomatous human prostrate,” Cytokine, vol. 12, no. 5, pp. 535–538, 2000.
[65]  G. Rodriguez-Berriguete, B. Fraile, R. Paniagua, P. Aller, and M. Royuela, “Expression of NF-kappaB-related proteins and their modulation during TNF-alpha-provoked apoptosis in prostate cancer cells,” Prostate, vol. 72, pp. 40–50, 2012.
[66]  Y. Bouraoui, M. Ricote, I. García-Tu?ón et al., “Pro-inflammatory cytokines and prostate-specific antigen in hyperplasia and human prostate cancer,” Cancer Detection and Prevention, vol. 32, no. 1, pp. 23–32, 2008.
[67]  S. Srinivasan, R. Kumar, S. Koduru, A. Chandramouli, and C. Damodaran, “Inhibiting TNF-mediated signaling: a novel therapeutic paradigm for androgen independent prostate cancer,” Apoptosis, vol. 15, no. 2, pp. 153–161, 2010.
[68]  G. Rodríguez-Berriguete, B. Fraile, F. R. de Bethencourt et al., “Role of IAPs in prostate cancer progression: immunohistochemical study in normal and pathological (benign hyperplastic, prostatic intraepithelial neoplasia and cancer) human prostate,” BMC Cancer, vol. 10, article 18, 2010.
[69]  C. D. Chen, D. S. Welsbie, C. Tran et al., “Molecular determinants of resistance to antiandrogen therapy,” Nature Medicine, vol. 10, no. 1, pp. 33–39, 2004.
[70]  L. Zhang, S. Altuwaijri, F. Deng et al., “NF-κB regulates androgen receptor expression and prostate cancer growth,” American Journal of Pathology, vol. 175, no. 2, pp. 489–499, 2009.
[71]  G. Jain, M. V. Cronauer, M. Schrader, P. Moller, and R. B. Marienfeld, “NF-kappaB signaling in prostate cancer: a promising therapeutic target?” World Journal of Urology, vol. 30, pp. 303–310, 2012.
[72]  S. I. Grivennikov and M. Karin, “Dangerous liaisons: STAT3 and NF-κB collaboration and crosstalk in cancer,” Cytokine and Growth Factor Reviews, vol. 21, no. 1, pp. 11–19, 2010.
[73]  G. Schneider and O. H. Kramer, “NFkappaB/p53 crosstalk-a promising new therapeutic target,” Biochimica et Biophysica Acta, vol. 1815, pp. 90–103, 2011.
[74]  A. Oeckinghaus, M. S. Hayden, and S. Ghosh, “Crosstalk in NF-kappaB signaling pathways,” Nature Immunology, vol. 12, pp. 695–708, 2011.
[75]  I. Vivanco and C. L. Sawyers, “The phosphatidylinositol 3-kinase-AKT pathway in human cancer,” Nature Reviews Cancer, vol. 2, no. 7, pp. 489–501, 2002.
[76]  A. G. Bader, S. Kang, L. Zhao, and P. K. Vogt, “Oncogenic PI3K deregulates transcription and translation,” Nature Reviews Cancer, vol. 5, no. 12, pp. 921–929, 2005.
[77]  J. A. Engelman, J. Luo, and L. C. Cantley, “The evolution of phosphatidylinositol 3-kinases as regulators of growth and metabolism,” Nature Reviews Genetics, vol. 7, no. 8, pp. 606–619, 2006.
[78]  D. A. Guertin and D. M. Sabatini, “Defining the role of mTOR in cancer,” Cancer Cell, vol. 12, no. 1, pp. 9–22, 2007.
[79]  L. Bozulic and B. A. Hemmings, “PIKKing on PKB: regulation of PKB activity by phosphorylation,” Current Opinion in Cell Biology, vol. 21, no. 2, pp. 256–261, 2009.
[80]  M. P. Scheid and J. R. Woodgett, “PKB/AKT: functional insights from genetic models,” Nature Reviews Molecular Cell Biology, vol. 2, no. 10, pp. 760–768, 2001.
[81]  B. D. Manning and L. C. Cantley, “AKT/PKB signaling: navigating downstream,” Cell, vol. 129, no. 7, pp. 1261–1274, 2007.
[82]  L. C. Cantley, “The phosphoinositide 3-kinase pathway,” Science, vol. 296, no. 5573, pp. 1655–1657, 2002.
[83]  T. M. Morgan, T. D. Koreckij, and E. Corey, “Targeted therapy for advanced prostate cancer: inhibition of the PI3K/Akt/mTOR pathway,” Current Cancer Drug Targets, vol. 9, no. 2, pp. 237–249, 2009.
[84]  A. Di Cristofano, B. Pesce, C. Cordon-Cardo, and P. P. Pandolfi, “Pten is essential for embryonic development and tumour suppression,” Nature Genetics, vol. 19, no. 4, pp. 348–355, 1998.
[85]  J. L. Boormans, H. Korsten, A. C. J. Ziel-Van Der Made, G. J. L. H. Van Leenders, P. C. M. S. Verhagen, and J. Trapman, “E17K substitution in AKT1 in prostate cancer,” British Journal of Cancer, vol. 102, no. 10, pp. 1491–1494, 2010.
[86]  L. C. Trotman, M. Niki, Z. A. Dotan et al., “Pten dose dictates cancer progression in the prostate,” PLoS Biology, vol. 1, no. 3, article E59, 2003.
[87]  Z. Chen, L. C. Trotman, D. Shaffer et al., “Crucial role of p53-dependent cellular senescence in suppression of Pten-deficient tumorigenesis,” Nature, vol. 436, no. 7051, pp. 725–730, 2005.
[88]  A. Di Cristofano, M. De Acetis, A. Koff, C. Cordon-Cardo, and P. P Pandolfi, “Pten and p27KIP1 cooperate in prostate cancer tumor suppression in the mouse,” Nature Genetics, vol. 27, no. 2, pp. 222–224, 2001.
[89]  L. Poliseno, L. Salmena, L. Riccardi et al., “Identification of the miR-106b~25 microRNA cluster as a proto-oncogenic PTEN-targeting intron that cooperates with its host gene MCM7 in transformation,” Science Signaling, vol. 3, no. 117, article ra29, 2010.
[90]  M. S. Song, L. Salmena, A. Carracedo et al., “The deubiquitinylation and localization of PTEN are regulated by a HAUSP-PML network,” Nature, vol. 455, no. 7214, pp. 813–817, 2008.
[91]  M. S. Song, L. Salmena, and P. P. Pandolfi, “The functions and regulation of the PTEN tumour suppressor,” Nature Reviews Molecular Cell Biology, vol. 13, pp. 283–296, 2012.
[92]  A. Almasan and A. Ashkenazi, “Apo2L/TRAIL: apoptosis signaling, biology, and potential for cancer therapy,” Cytokine and Growth Factor Reviews, vol. 14, no. 3-4, pp. 337–348, 2003.
[93]  J. Xu, J. Y. Zhou, W. Z. Wei, and G. S. Wu, “Activation of the Akt survival pathway contributes to TRAIL resistance in cancer cells,” PLoS ONE, vol. 5, no. 4, Article ID e10226, 2010.
[94]  N. Tzenaki, M. Andreou, K. Stratigi et al., “High levels of p110delta PI3K expression in solid tumor cells suppress PTEN activity, generating cellular sensitivity to p110delta inhibitors through PTEN activation,” FASEB Journal, vol. 26, pp. 2498–2508, 2012.
[95]  S. H. Lee, G. Poulogiannis, S. Pyne et al., “A constitutively activated form of the p110β isoform of PI3-kinase induces prostatic intraepithelial neoplasia in mice,” Proceedings of the National Academy of Sciences of the United States of America, vol. 107, no. 24, pp. 11002–11007, 2010.
[96]  B. Cinar, P. K. Fang, M. Lutchman et al., “The pro-apoptotic kinase Mst1 and its caspase cleavage products are direct inhibitors of Akt1,” EMBO Journal, vol. 26, no. 21, pp. 4523–4534, 2007.
[97]  B. Cinar, F. K. Collak, D. Lopez et al., “MST1 is a multifunctional caspase-independent inhibitor of androgenic signaling,” Cancer Research, vol. 71, no. 12, pp. 4303–4313, 2011.
[98]  Z. Yuan, D. Kim, S. Shu et al., “Phosphoinositide 3-kinase/Akt inhibits MST1-mediated pro-apoptotic signaling through phosphorylation of threonine 120,” Journal of Biological Chemistry, vol. 285, no. 6, pp. 3815–3824, 2010.
[99]  K. Mahajan, D. Coppola, S. Challa et al., “Ack1 mediated AKT/PKB tyrosine 176 phosphorylation regulates its activation,” PloS ONE, vol. 5, no. 3, article e9646, 2010.
[100]  K. Nacerddine, J. B. Beaudry, V. Ginjala et al., “Akt-mediated phosphorylation of Bmi1 modulates its oncogenic potential, E3 ligase activity, and DNA damage repair activity in mouse prostate cancer,” Journal of Clinical Investigation, vol. 122, pp. 1920–1932, 2012.
[101]  M. Gao, R. Patel, I. Ahmad et al., “SPRY2 loss enhances ErbB trafficking and PI3K/AKT signalling to drive human and mouse prostate carcinogenesis,” EMBO Molecular Medicine, vol. 4, pp. 776–790, 2012.
[102]  V. M. Adhami, I. A. Siddiqui, S. Sarfaraz et al., “Effective prostate cancer chemopreventive intervention with green tea polyphenols in the TRAMP model depends on Thestage of the disease,” Clinical Cancer Research, vol. 15, no. 6, pp. 1947–1953, 2009.
[103]  V. M. Adhami, I. A. Siddiqui, N. Ahmad, S. Gupta, and H. Mukhtar, “Oral consumption of green tea polyphenols inhibits insulin-like growth factor-I-induced signaling in an autochthonous mouse model of prostate cancer,” Cancer Research, vol. 64, no. 23, pp. 8715–8722, 2004.
[104]  H. J. Kung and C. P. Evans, “Oncogenic activation of androgen receptor,” Urologic Oncology, vol. 27, no. 1, pp. 48–52, 2009.
[105]  N. J. Clegg, S. S. Couto, J. Wongvipat et al., “MYC cooperates with AKT in prostate tumorigenesis and alters sensitivity to mTOR inhibitors,” PLoS ONE, vol. 6, no. 3, Article ID e17449, 2011.
[106]  D. N. Gunadharini, P. Elumalai, R. Arunkumar, K. Senthilkumar, and J. Arunakaran, “Induction of apoptosis and inhibition of PI3K/Akt pathway in PC-3 and LNCaP prostate cancer cells by ethanolic neem leaf extract,” Journal of Ethnopharmacology, vol. 134, no. 3, pp. 644–650, 2011.
[107]  K. R. Park, D. Nam, H. M. Yun et al., “beta-Caryophyllene oxide inhibits growth and induces apoptosis through the suppression of PI3K/AKT/mTOR/S6K1 pathways and ROS-mediated MAPKs activation,” Cancer Letters, vol. 312, pp. 178–188, 2011.
[108]  V. M. Adhami, D. N. Syed, N. Khan, and H. Mukhtar, “Dietary flavonoid fisetin: a novel dual inhibitor of PI3K/Akt and mTOR for prostate cancer management,” Biochemical Pharmacology, vol. 84, pp. 1277–1281, 2012.
[109]  Z. Wang, Y. Zhang, S. Banerjee, Y. Li, and F. H. Sarkar, “Notch-1 down-regulation by curcumin is associated with the inhibition of cell growth and the induction of apoptosis in pancreatic cancer cells,” Cancer, vol. 106, no. 11, pp. 2503–2513, 2006.
[110]  M. J. Ryu, M. Cho, J. Y. Song et al., “Natural derivatives of curcumin attenuate the Wnt/β-catenin pathway through down-regulation of the transcriptional coactivator p300,” Biochemical and Biophysical Research Communications, vol. 377, no. 4, pp. 1304–1308, 2008.
[111]  S. Yu, G. Shen, O. K. Tin, J. H. Kim, and A. N. T. Kong, “Curcumin inhibits Akt/mammalian target of rapamycin signaling through protein phosphatase-dependent mechanism,” Molecular Cancer Therapeutics, vol. 7, no. 9, pp. 2609–2620, 2008.
[112]  A. ?lusarz, N. S. Shenouda, M. S. Sakla et al., “Common botanical compounds inhibit the hedgehog signaling pathway in prostate cancer,” Cancer Research, vol. 70, no. 8, pp. 3382–3390, 2010.
[113]  O. W. Rokhlin, N. V. Guseva, A. F. Taghiyev, R. A. Glover, and M. B. Cohen, “KN-93 inhibits androgen receptor activity and induces cell death irrespective of p53 and Akt status in prostate cancer,” Cancer Biology and Therapy, vol. 9, no. 3, pp. 224–235, 2010.
[114]  J. J. Wallin, K. A. Edgar, J. Guan et al., “GDC-0980 is a novel class I PI3K/mTOR kinase inhibitor with robust activity in cancer models driven by the PI3K pathway,” Molecular Cancer Therapeutics, vol. 10, pp. 2426–2436, 2011.
[115]  S. M. Manohar, A. A. Padgaonkar, A. J. Badhwar et al., “A novel inhibitor of hypoxia-inducible factor-1alpha P3155 also modulates PI3K pathway and inhibits growth of prostate cancer cells,” BMC Cancer, vol. 11, article 338, 2011.
[116]  L. Lu, D. Tang, L. Wang et al., “Gambogic acid inhibits TNF-alpha-induced invasion of human prostate cancer PC3 cells in vitro through PI3K/Akt and NF-kappaB signaling pathways,” Acta Pharmacologica Sinica, vol. 33, pp. 531–541, 2012.
[117]  S. L. Burgio, F. Fabbri, I. J. Seymour, W. Zoli, D. Amadori, and U. De Giorgi, “Perspectives on mTOR inhibitors for castration-refractory prostate cancer,” Current Cancer Drug Targets, vol. 12, pp. 940–949, 2012.
[118]  E. C. Nelson, C. P. Evans, P. C. Mack, R. W. Devere-White, and P. N. Lara Jr., “Inhibition of Akt pathways in the treatment of prostate cancer,” Prostate Cancer and Prostatic Diseases, vol. 10, no. 4, pp. 331–339, 2007.
[119]  C. A. Granville, R. M. Memmott, J. J. Gills, and P. A. Dennis, “Handicapping the race to develop inhibitors of the phosphoinositide 3-kinase/Akt/mammalian target of rapamycin pathway,” Clinical Cancer Research, vol. 12, no. 3, pp. 679–689, 2006.
[120]  S. B. Kondapaka, S. S. Singh, G. P. Dasmahapatra, E. A. Sausville, and K. K. Roy, “Perifosine, a novel alkylphospholipid, inhibits protein kinase B activation,” Molecular Cancer Therapeutics, vol. 2, no. 11, pp. 1093–1103, 2003.
[121]  C. G. Tepper, R. L. Vinall, C. B. Wee et al., “GCP-mediated growth inhibition and apoptosis of prostate cancer cells via androgen receptor-dependent and -independent mechanisms,” Prostate, vol. 67, no. 5, pp. 521–535, 2007.
[122]  P. B. Makhov, K. Golovine, A. Kutikov et al., “Modulation of Akt/mTOR signaling overcomes sunitinib resistance in renal and prostate cancer cells,” Molecular Cancer Therapeutics, vol. 11, pp. 1510–1517, 2012.
[123]  H. Kiu and S. E. Nicholson, “Biology and significance of the JAK/STAT signalling pathways,” Growth Factors, vol. 30, pp. 88–106, 2012.
[124]  D. A. Harrison, “The Jak/STAT pathway,” Cold Spring Harbor Perspectives in Biology, vol. 4, 2012.
[125]  W. X. Li, “Canonical and non-canonical JAK-STAT signaling,” Trends in Cell Biology, vol. 18, no. 11, pp. 545–551, 2008.
[126]  D. Hebenstreit, J. Horejs-Hoeck, and A. Duschl, “JAK/STAT-dependent gene regulation by cytokines,” Drug News and Perspectives, vol. 18, no. 4, pp. 243–249, 2005.
[127]  J. J. O'Shea, H. Park, M. Pesu, D. Borie, and P. Changelian, “New strategies for immunosuppression: interfering with cytokines by targeting the Jak/Stat pathway,” Current Opinion in Rheumatology, vol. 17, no. 3, pp. 305–311, 2005.
[128]  P. Igaz, S. Tóth, and A. Falus, “Biological and clinical significance of the JAK-STAT pathway; lessons from knockout mice,” Inflammation Research, vol. 50, no. 9, pp. 435–441, 2001.
[129]  J. J. O'Shea, M. Gadina, and R. D. Schreiber, “Cytokine signaling in 2002: new surprises in the Jak/Stat pathway,” Cell, vol. 109, no. 2, pp. S121–S131, 2002.
[130]  J. E. Darnell Jr., “STATs and gene regulation,” Science, vol. 277, no. 5332, pp. 1630–1635, 1997.
[131]  J. N. Ihle, W. Thierfelder, S. Teglund et al., “Signaling by the cytokine receptor superfamily,” Annals of the New York Academy of Sciences, vol. 865, pp. 1–9, 1998.
[132]  K. Shuai, C. M. Horvath, L. H. T. Huang, S. A. Qureshi, D. Cowburn, and J. E. Darnell Jr., “Interferon activation of the transcription factor Stat91 involves dimerization through SH2-phosphotyrosyl peptide interactions,” Cell, vol. 76, no. 5, pp. 821–828, 1994.
[133]  S. Gupta, T. Barrett, A. J. Whitmarsh et al., “Selective interaction of JNK protein kinase isoforms with transcription factors,” EMBO Journal, vol. 15, no. 11, pp. 2760–2770, 1996.
[134]  K. Mowen and M. David, “Role of the STAT1-SH2 domain and STAT2 in the activation and nuclear translocation of STAT1,” Journal of Biological Chemistry, vol. 273, no. 46, pp. 30073–30076, 1998.
[135]  J. S. Rawlings, K. M. Rosler, and D. A. Harrison, “The JAK/STAT signaling pathway,” Journal of Cell Science, vol. 117, no. 8, pp. 1281–1283, 2004.
[136]  J. Bromberg, “Stat proteins and oncogenesis,” Journal of Clinical Investigation, vol. 109, no. 9, pp. 1139–1142, 2002.
[137]  H. Yu and R. Jove, “The stats of cancer—new molecular targets come of age,” Nature Reviews Cancer, vol. 4, no. 2, pp. 97–105, 2004.
[138]  A. Tutt and A. Ashworth, “The relationship between the roles of BRCA genes in DNA repair and cancer predisposition,” Trends in Molecular Medicine, vol. 8, no. 12, pp. 571–576, 2002.
[139]  H. Wallerand, G. Robert, J. C. Bernhard, A. Ravaud, and J. J. Patard, “Tyrosine-kinase inhibitors in the treatment of muscle invasive bladder cancer and hormone refractory prostate cancer,” Archivos Espanoles de Urologia, vol. 63, no. 9, pp. 773–787, 2010.
[140]  E. M. Kwon, S. K. Holt, R. Fu et al., “Androgen metabolism and JAK/STAT pathway genes and prostate cancer risk,” Cancer Epidemiology, vol. 36, pp. 347–353, 2012.
[141]  D. A. Frank, “STAT3 as a central mediator of neoplastic cellular transformation,” Cancer Letters, vol. 251, no. 2, pp. 199–210, 2007.
[142]  B. Gao, X. Shen, G. Kunos et al., “Constitutive activation of JAK-STAT3 signaling by BRCA1 in human prostate cancer cells,” FEBS Letters, vol. 488, no. 3, pp. 179–184, 2001.
[143]  A. R. Venkitaraman, “Cancer susceptibility and the functions of BRCA1 and BRCA2,” Cell, vol. 108, no. 2, pp. 171–182, 2002.
[144]  E. A. Ostrander and M. S. Udler, “The role of the BRCA2 gene in susceptibility to prostate cancer revisited,” Cancer Epidemiology Biomarkers and Prevention, vol. 17, no. 8, pp. 1843–1848, 2008.
[145]  S. Panchal, O. Shachar, F. O'Malley et al., “Breast cancer in a BRCA2 mutation carrier with a history of prostate cancer,” Nature Reviews Clinical Oncology, vol. 6, no. 10, pp. 604–607, 2009.
[146]  N. Rahman and M. R. Stratton, “The genetics of breast cancer susceptibility,” Annual Review of Genetics, vol. 32, pp. 95–121, 1998.
[147]  P. Kerr and A. Ashworth, “New complexities for BRCA1 and BRCA2,” Current Biology, vol. 11, no. 16, pp. R668–R676, 2001.
[148]  M. Sakaguchi, M. Oka, T. Iwasaki, Y. Fukami, and C. Nishigori, “Role and regulation of STAT3 phosphorylation at Ser727 in melanocytes and melanoma cells,” Journal of Investigative Dermatology, vol. 132, pp. 1877–1885, 2012.
[149]  J. Cao, P. Kozarekar, M. Pavlaki, C. Chiarelli, W. F. Bahou, and S. Zucker, “Distinct roles for the catalytic and hemopexin domains of membrane type 1-matrix metalloproteinase in substrate degradation and cell migration,” Journal of Biological Chemistry, vol. 279, no. 14, pp. 14129–14139, 2004.
[150]  V. I. Romanov, T. Whyard, H. L. Adler, W. C. Waltzer, and S. Zucker, “Prostate cancer cell adhesion to bone marrow endothelium: the role of prostate-specific antigen,” Cancer Research, vol. 64, no. 6, pp. 2083–2089, 2004.
[151]  H. L. Nguyen, S. Zucker, K. Zarrabi, P. Kadam, C. Schmidt, and J. Cao, “Oxidative stress and prostate cancer progression are elicited by membrane-type 1 matrix metalloproteinase,” Molecular Cancer Research, vol. 9, pp. 1305–1318, 2011.
[152]  D. Kesanakurti, C. Chetty, D. H. Dinh, M. Gujrati, and J. S. Rao, “Role of MMP-2 in the regulation of IL-6/Stat3 survival signaling via interaction with alpha5beta1 integrin in glioma,” Oncogene, vol. 32, pp. 327–340, 2012.
[153]  R. Dhir, Z. Ni, W. Lou, F. DeMiguel, J. R. Grandis, and A. C. Gao, “Stat3 activation in prostatic carcinomas,” Prostate, vol. 51, no. 4, pp. 241–246, 2002.
[154]  X. Liu, Z. He, C. H. Li, G. Huang, C. Ding, and H. Liu, “Correlation analysis of JAK-STAT pathway components on prognosis of patients with prostate cancer,” Pathology and Oncology Research, vol. 18, pp. 17–23, 2012.
[155]  R. Aalinkeel, Z. Hu, B. B. Nair et al., “Genomic analysis highlights the role of the JAK-STAT signaling in the anti-proliferative effects of dietary—flavonoid “ashwagandha” in prostate cancer cells,” Evidence-Based Complementary and Alternative Medicine, vol. 7, no. 2, pp. 177–187, 2010.
[156]  K. Ishihara and T. Hirano, “IL-6 in autoimmune disease and chronic inflammatory proliferative disease,” Cytokine and Growth Factor Reviews, vol. 13, no. 4-5, pp. 357–368, 2002.
[157]  T. Kishimoto, “Interleukin-6: from basic science to medicine—40 years in immunology,” Annual Review of Immunology, vol. 23, pp. 1–21, 2005.
[158]  A. Hobisch, H. Rogatsch, A. Hittmair, et al., “Immunohistochemical localization of interleukin-6 and its receptor in benign, premalignant and malignant prostate tissue,” Journal of Pathology, vol. 191, pp. 239–244, 2000.
[159]  P. C. Smith, A. Hobisch, D. L. Lin, Z. Culig, and E. T. Keller, “Interleukin-6 and prostate cancer progression,” Cytokine and Growth Factor Reviews, vol. 12, no. 1, pp. 33–40, 2001.
[160]  J. R. Stark, H. Li, P. Kraft et al., “Circulating prediagnostic interleukin-6 and C-reactive protein and prostate cancer incidence and mortality,” International Journal of Cancer, vol. 124, no. 11, pp. 2683–2689, 2009.
[161]  J. Karkera, H. Steiner, W. Li et al., “The anti-interleukin-6 antibody siltuximab down-regulates genes implicated in tumorigenesis in prostate cancer patients from a phase I study,” Prostate, vol. 71, pp. 1455–1465, 2011.
[162]  D. A. Twillie, M. A. Eisenberger, M. A. Carducci, W. S. Hseih, W. Y. Kim, and J. W. Simons, “Interleukin-6: a candidate mediator of human prostate cancer morbidity,” Urology, vol. 45, no. 3, pp. 542–549, 1995.
[163]  M. Okamoto, C. Lee, and R. Oyasu, “Interleukin-6 as a paracrine and autocrine growth factor in human prostatic carcinoma cells in vitro,” Cancer Research, vol. 57, no. 1, pp. 141–146, 1997.
[164]  I. T. Cavarretta, H. Neuwirt, G. Untergasser et al., “The antiapoptotic effect of IL-6 autocrine loop in a cellular model of advanced prostate cancer is mediated by Mcl-1,” Oncogene, vol. 26, no. 20, pp. 2822–2832, 2007.
[165]  T. D. Chung, J. J. Yu, M. T. Spiotto, M. Bartkowski, and J. W. Simons, “Characterization of the role of IL-6 in the progression of prostate cancer,” Prostate, vol. 38, pp. 199–207, 1999.
[166]  W. Lou, Z. Ni, K. Dyer, D. J. Tweardy, and A. C. Gao, “Interleukin-6 induces prostate cancer cell growth accompanied by activation of stat3 signaling pathway,” Prostate, vol. 42, pp. 239–242, 2000.
[167]  I. Sakai, H. Miyake, T. Terakawa, and M. Fujisawa, “Inhibition of tumor growth and sensitization to chemotherapy by RNA interference targeting interleukin-6 in the androgen-independent human prostate cancer PC3 model,” Cancer Science, vol. 102, no. 4, pp. 769–775, 2011.
[168]  L. R. Dearth and J. DeWille, “An AU-rich element in the untranslated region of the C/EBPδ mRNA is important for protein binding during G0 growth arrest,” Biochemical and Biophysical Research Communications, vol. 304, no. 2, pp. 344–350, 2003.
[169]  L. R. Dearth and J. DeWille, “Posttranscriptional and posttranslational regulation of C/EBPδ in Go growth-arrested mammary epithelial cells,” Journal of Biological Chemistry, vol. 278, no. 13, pp. 11246–11255, 2003.
[170]  D. P. Ramji and P. Foka, “CCAAT/enhancer-binding proteins: structure, function and regulation,” Biochemical Journal, vol. 365, no. 3, pp. 561–575, 2002.
[171]  D. C. Sanford and J. W. DeWille, “C/EBPδ is a downstream mediator of IL-6 induced growth inhibition of prostate cancer cells,” Prostate, vol. 63, no. 2, pp. 143–154, 2005.
[172]  E. LaTulippe, J. Satagopan, A. Smith et al., “Comprehensive gene expression analysis of prostate cancer reveals distinct transcriptional programs associated with metastatic disease,” Cancer Research, vol. 62, no. 15, pp. 4499–4506, 2002.
[173]  B. Binétruy, L. Heasley, F. Bost, L. Caron, and M. Aouadi, “Concise review: regulation of embryonic stem cell lineage commitment by mitogen-activated protein kinases,” Stem Cells, vol. 25, no. 5, pp. 1090–1095, 2007.
[174]  A. S. Dhillon, S. Hagan, O. Rath, and W. Kolch, “MAP kinase signalling pathways in cancer,” Oncogene, vol. 26, no. 22, pp. 3279–3290, 2007.
[175]  E. F. Wagner and A. R. Nebreda, “Signal integration by JNK and p38 MAPK pathways in cancer development,” Nature Reviews Cancer, vol. 9, no. 8, pp. 537–549, 2009.
[176]  Y. T. Ip and R. J. Davis, “Signal transduction by the c-Jun N-terminal kinase (JNK)—from inflammation to development,” Current Opinion in Cell Biology, vol. 10, no. 2, pp. 205–219, 1998.
[177]  T. H. Holmstr?m, I. Schmitz, T. S. S?derstr?m et al., “MAPK/ERK signaling in activated T cells inhibits CD95/Fas-mediated apoptosis downstream of DISC assembly,” EMBO Journal, vol. 19, no. 20, pp. 5418–5428, 2000.
[178]  K. Gupta, S. Kshirsagar, W. Li et al., “VEGF prevents apoptosis of human microvascular endothelial cells via opposing effects on MAPK/ERK and SAPK/JNK signaling,” Experimental Cell Research, vol. 247, no. 2, pp. 495–504, 1999.
[179]  A. Ogata, D. Chauhan, G. Teoh et al., “IL-6 triggers cell growth via the ras-dependent mitogen-activated protein kinase cascade,” Journal of Immunology, vol. 159, no. 5, pp. 2212–2221, 1997.
[180]  V. Flati, E. Pasini, G. D'Antona, S. Speca, E. Toniato, and S. Martinotti, “Intracellular mechanisms of metabolism regulation: the role of signaling via the mammalian target of rapamycin pathway and other routes,” American Journal of Cardiology, vol. 101, no. 11, pp. S16–S21, 2008.
[181]  S. Davis, P. Vanhoutte, C. Pagès, J. Caboche, and S. Laroche, “The MAPK/ERK cascade targets both Elk-1 and cAMP response element- binding protein to control long-term potentiation-dependent gene expression in the dentate gyrus in vivo,” Journal of Neuroscience, vol. 20, no. 12, pp. 4563–4572, 2000.
[182]  J. Guicheux, J. Lemonnier, C. Ghayor, A. Suzuki, G. Palmer, and J. Caverzasio, “Activation of p38 mitogen-activated protein kinase and c-Jun-NH2-terminal kinase by BMP-2 and their implication in the stimulation of osteoblastic cell differentiation,” Journal of Bone and Mineral Research, vol. 18, no. 11, pp. 2060–2068, 2003.
[183]  C. Huang, K. Jacobson, and M. D. Schaller, “MAP kinases and cell migration,” Journal of Cell Science, vol. 117, no. 20, pp. 4619–4628, 2004.
[184]  M. A. Lemmon and J. Schlessinger, “Cell signaling by receptor tyrosine kinases,” Cell, vol. 141, no. 7, pp. 1117–1134, 2010.
[185]  L. A. Fecher, R. K. Amaravadi, and K. T. Flaherty, “The MAPK pathway in melanoma,” Current Opinion in Oncology, vol. 20, no. 2, pp. 183–189, 2008.
[186]  Y. Fleming, C. G. Armstrong, N. Morrice, A. Paterson, M. Goedert, and P. Cohen, “Synergistic activation of stress-activated protein kinase 1/c-Jun N-terminal kinase (SAPK1/JNK) isoforms by mitogen-activated protein kinase kinase 4 (MKK4) and MKK7,” Biochemical Journal, vol. 352, no. 1, pp. 145–154, 2000.
[187]  W. Haeusgen, T. Herdegen, and V. Waetzig, “The bottleneck of JNK signaling: molecular and functional characteristics of MKK4 and MKK7,” European Journal of Cell Biology, vol. 90, no. 6-7, pp. 536–544, 2011.
[188]  W. Xin, K. J. Yun, F. Ricci et al., “MAP2K4/MKK4 expression in pancreatic cancer: genetic validation of immunohistochemistry and relationship to disease course,” Clinical Cancer Research, vol. 10, no. 24, pp. 8516–8520, 2004.
[189]  S. D. Yamada, J. A. Hickson, Y. Hrobowski et al., “Mitogen-activated protein kinase kinase 4 (MKK4) acts as a metastasis suppressor gene in human ovarian carcinoma,” Cancer Research, vol. 62, no. 22, pp. 6717–6723, 2002.
[190]  R. Chiu, W. J. Boyle, J. Meek, T. Smeal, T. Hunter, and M. Karin, “The c-Fos protein interacts with c-Jun/AP-1 to stimulate transcription of AP-1 responsive genes,” Cell, vol. 54, no. 4, pp. 541–552, 1988.
[191]  C. Jonat, H. J. Rahmsdorf, K. K. Park et al., “Antitumor promotion and antiinflammation: down-modulation of AP-1 (Fos/Jun) activity by glucocorticoid hormone,” Cell, vol. 62, no. 6, pp. 1189–1204, 1990.
[192]  C. W. Kinkade, M. Castillo-Martin, A. Puzio-Kuter et al., “Targeting AKT/mTOR and ERK MAPK signaling inhibits hormone-refractory prostate cancer in a preclinical mouse model,” Journal of Clinical Investigation, vol. 118, no. 9, pp. 3051–3064, 2008.
[193]  M. T. Abreu-Martin, A. Chari, A. A. Palladino, N. A. Craft, and C. L. Sawyers, “Mitogen-activated protein kinase kinase kinase 1 activates androgen receptor-dependent transcription and apoptosis in prostate cancer,” Molecular and Cellular Biology, vol. 19, no. 7, pp. 5143–5154, 1999.
[194]  D. Gioeli, J. W. Mandell, G. R. Petroni, H. F. Frierson, and M. J. Weber, “Activation of mitogen-activated protein kinase associated with prostate cancer progression,” Cancer Research, vol. 59, no. 2, pp. 279–284, 1999.
[195]  A. M. Carey, R. Pramanik, L. J. Nicholson et al., “Ras-MEK-ERK signaling cascade regulates androgen receptor element-inducible gene transcription and DNA synthesis in prostate cancer cells,” International Journal of Cancer, vol. 121, no. 3, pp. 520–527, 2007.
[196]  T. J. Dorkin, M. C. Robinson, C. Marsh, A. Bjartell, D. E. Neal, and H. Y. Leung, “FGF8 over-expression in prostate cancer is associated with decreased patient survival and persists in androgen independent disease,” Oncogene, vol. 18, no. 17, pp. 2755–2761, 1999.
[197]  H. Steiner, S. Godoy-Tundidor, H. Rogatsch et al., “Accelerated in vivo growth of prostate tumors that up-regulate interleukin-6 is associated with reduced retinoblastoma protein expression and activation of the mitogen-activated protein kinase pathway,” American Journal of Pathology, vol. 162, no. 2, pp. 655–663, 2003.
[198]  R. E. Bakin, D. Gioeli, E. A. Bissonette, and M. J. Weber, “Attenuation of Ras signaling restores androgen sensitivity to hormone-refractory C4-2 prostate cancer cells,” Cancer Research, vol. 63, no. 8, pp. 1975–1980, 2003.
[199]  J. H. Jeong, Z. Wang, A. S. Guimaraes et al., “BRAF activation initiates but does not maintain invasive prostate adenocarcinoma,” PLoS ONE, vol. 3, no. 12, Article ID e3949, 2008.
[200]  H. B. Pearson, T. J. Phesse, and A. R. Clarke, “K-ras and Wnt signaling synergize to accelerate prostate tumorigenesis in the mouse,” Cancer Research, vol. 69, no. 1, pp. 94–101, 2009.
[201]  N. Palanisamy, B. Ateeq, S. Kalyana-Sundaram et al., “Rearrangements of the RAF kinase pathway in prostate cancer, gastric cancer and melanoma,” Nature Medicine, vol. 16, no. 7, pp. 793–798, 2010.
[202]  L. Tie, N. Lu, X. Y. Pan et al., “Hypoxia-induced up-regulation of aquaporin-1 protein in prostate cancer cells in a p38-dependent manner,” Cellular Physiology and Biochemistry, vol. 29, pp. 269–280, 2012.
[203]  Y. Limami, A. Pinon, D. Y. Leger et al., “The P2Y2/Src/p38/COX-2 pathway is involved in the resistance to ursolic acid-induced apoptosis in colorectal and prostate cancer cells,” Biochimie, vol. 94, pp. 1754–1763, 2012.
[204]  M. Katoh, “FGFR2 abnormalities underlie a spectrum of bone, skin, and cancer pathologies,” Journal of Investigative Dermatology, vol. 129, no. 8, pp. 1861–1867, 2009.
[205]  P. C. Marker, A. A. Donjacour, R. Dahiya, and G. R. Cunha, “Hormonal, cellular, and molecular control of prostatic development,” Developmental Biology, vol. 253, no. 2, pp. 165–174, 2003.
[206]  A. A. Donjacour, A. A. Thomson, and G. R. Cunha, “FGF-10 plays an essential role in the growth of the fetal prostate,” Developmental Biology, vol. 261, no. 1, pp. 39–54, 2003.
[207]  Y. Lin, G. Liu, Y. Zhang et al., “Fibroblast growth factor receptor 2 tyrosine kinase is required for prostatic morphogenesis and the acquisition of strict androgen dependency for adult tissue homeostasis,” Development, vol. 134, no. 4, pp. 723–734, 2007.
[208]  Y. Zhang, J. Zhang, Y. Lin et al., “Role of epithelial cell fibroblast growth factor receptor substrate 2α in prostate development, regeneration and tumorigenesis,” Development, vol. 135, no. 4, pp. 775–784, 2008.
[209]  S. L. Kuslak and P. C. Marker, “Fibroblast growth factor receptor signaling through MEK-ERK is required for prostate bud induction,” Differentiation, vol. 75, no. 7, pp. 638–651, 2007.
[210]  M. J. Hour, S. C. Tsai, H. C. Wu et al., “Antitumor effects of the novel quinazolinone MJ-33: inhibition of metastasis through the MAPK, AKT, NF-kappaB and AP-1 signaling pathways in DU145 human prostate cancer cells,” International Journal of Oncology, 2012.
[211]  T. Shimizu, A. W. Tolcher, K. P. Papadopoulos et al., “The clinical effect of the dual-targeting strategy involving PI3K/AKT/mTOR and RAS/MEK/ERK pathways in patients with advanced cancer,” Clinical Cancer Research, vol. 18, pp. 2316–2325, 2012.
[212]  G. C. Blobe, W. P. Schiemann, and H. F. Lodish, “Role of transforming growth factor β in human disease,” New England Journal of Medicine, vol. 342, no. 18, pp. 1350–1358, 2000.
[213]  J. Massagué, S. W. Blain, and R. S. Lo, “TGFβ signaling in growth control, cancer, and heritable disorders,” Cell, vol. 103, no. 2, pp. 295–309, 2000.
[214]  B. Schmierer and C. S. Hill, “TGFβ-SMAD signal transduction: molecular specificity and functional flexibility,” Nature Reviews Molecular Cell Biology, vol. 8, no. 12, pp. 970–982, 2007.
[215]  J. Massague, “TGFbeta in cancer,” Cell, vol. 134, pp. 215–230, 2008.
[216]  Y. Shi and J. Massagué, “Mechanisms of TGF-β signaling from cell membrane to the nucleus,” Cell, vol. 113, no. 6, pp. 685–700, 2003.
[217]  F. L. Miles, N. S. Tung, A. A. Aguiar, S. Kurtoglu, and R. A. Sikes, “Increased TGF-beta1-mediated suppression of growth and motility in castrate-resistant prostate cancer cells is consistent with Smad2/3 signaling,” Prostate, vol. 72, pp. 1339–1350, 2012.
[218]  M. K. Donkor, A. Sarkar, and M. O. Li, “Tgf-beta1 produced by activated CD4+ T cells antagonizes T cell surveillance of tumor development,” Oncoimmunology, vol. 1, pp. 162–171, 2012.
[219]  X. Wan, Z. G. Li, J. M. Yingling et al., “Effect of transforming growth factor beta (TGF-beta) receptor I kinase inhibitor on prostate cancer bone growth,” Bone, vol. 50, pp. 695–703, 2012.
[220]  P. Wikstrom, P. Stattin, I. Franck-Lissbrant, J. E. Damber, and A. Bergh, “Transforming growth factor beta1 is associated with angiogenesis, metastasis, and poor clinical outcome in prostate cancer,” Prostate, vol. 37, pp. 19–29, 1998.
[221]  H. L. Adler, M. A. McCurdy, M. W. Kattan, T. L. Timme, P. T. Scardino, and T. C. Thompson, “Elevated levels of circulating interleukin-6 and transforming growth factor-β1 in patients with metastatic prostatic carcinoma,” Journal of Urology, vol. 161, no. 1, pp. 182–187, 1999.
[222]  S. F. Shariat, M. Shalev, A. Menesses-Diaz et al., “Preoperative plasma levels of transforming growth factor beta1 (TGF-β1 strongly predict progression in patients undergoing radical prostatectomy,” Journal of Clinical Oncology, vol. 19, no. 11, pp. 2856–2864, 2001.
[223]  R. Derynck, R. J. Akhurst, and A. Balmain, “TGF-β signaling in tumor suppression and cancer progression,” Nature Genetics, vol. 29, no. 2, pp. 117–129, 2001.
[224]  L. M. Wakefield and A. B. Roberts, “TGF-β signaling: positive and negative effects on tumorigenesis,” Current Opinion in Genetics and Development, vol. 12, no. 1, pp. 22–29, 2002.
[225]  J. Yang, R. Wahdan-Alaswad, and D. Danielpour, “Critical role of smad2 in tumor suppression and transforming growth factor-β-Lnduced apoptosis of prostate epithelial cells,” Cancer Research, vol. 69, no. 6, pp. 2185–2190, 2009.
[226]  L. Ye, H. Kynaston, and W. G. Jiang, “Bone morphogenetic protein-10 suppresses the growth and aggressiveness of prostate cancer cells through a Smad independent pathway,” Journal of Urology, vol. 181, no. 6, pp. 2749–2759, 2009.
[227]  B. T. Vo and S. A. Khan, “Expression of nodal and nodal receptors in prostate stem cells and prostate cancer cells: autocrine effects on cell proliferation and migration,” Prostate, vol. 71, no. 10, pp. 1084–1096, 2011.
[228]  H. Y. Kang, H. Y. Huang, C. Y. Hsieh et al., “Activin A enhances prostate cancer cell migration through activation of androgen receptor and is overexpressed in metastatic prostate cancer,” Journal of Bone and Mineral Research, vol. 24, no. 7, pp. 1180–1193, 2009.
[229]  J. L. Whyte, A. A. Smith, and J. A. Helms, “Wnt signaling and injury repair,” Cold Spring Harbor Perspectives in Biology, vol. 4, Article ID a008078, 2012.
[230]  S. Angers and R. T. Moon, “Proximal events in Wnt signal transduction,” Nature Reviews Molecular Cell Biology, vol. 10, no. 7, pp. 468–477, 2009.
[231]  S. Thiele, M. Rauner, C. Goettsch et al., “Expression profile of WNT molecules in prostate cancer and its regulation by aminobisphosphonates,” Journal of Cellular Biochemistry, vol. 112, no. 6, pp. 1593–1600, 2011.
[232]  W. Lu, H. N. Tinsley, A. Keeton, Z. Qu, G. A. Piazza, and Y. Li, “Suppression of Wnt/β-catenin signaling inhibits prostate cancer cell proliferation,” European Journal of Pharmacology, vol. 602, no. 1, pp. 8–14, 2009.
[233]  C. L. Hall, S. Kang, O. A. MacDougald, and E. T. Keller, “Role of Wnts in prostate cancer bone metastases,” Journal of Cellular Biochemistry, vol. 97, no. 4, pp. 661–672, 2006.
[234]  D. Kimelman and W. Xu, “β-Catenin destruction complex: insights and questions from a structural perspective,” Oncogene, vol. 25, no. 57, pp. 7482–7491, 2006.
[235]  J. Huelsken and J. Behrens, “The Wnt signalling pathway,” Journal of Cell Science, vol. 115, no. 21, pp. 3977–3978, 2002.
[236]  H. C. Whitaker, J. Girling, A. Y. Warren, H. Leung, I. G. Mills, and D. E. Neal, “Alterations in β-catenin expression and localization in prostate cancer,” Prostate, vol. 68, no. 11, pp. 1196–1205, 2008.
[237]  S. Majid, S. Saini, and R. Dahiya, “Wnt signaling pathways in urological cancers: past decades and still growing,” Molecular Cancer, vol. 11, article 7, 2012.
[238]  S. Barolo, “Transgenic Wnt/TCF pathway reporters: all you need is Lef?” Oncogene, vol. 25, no. 57, pp. 7505–7511, 2006.
[239]  C. Mosimann, G. Hausmann, and K. Basler, “β-Catenin hits chromatin: regulation of Wnt target gene activation,” Nature Reviews Molecular Cell Biology, vol. 10, no. 4, pp. 276–286, 2009.
[240]  J. Zhao, W. Yue, M. J. Zhu, N. Sreejayan, and M. Du, “AMP-activated protein kinase (AMPK) cross-talks with canonical Wnt signaling via phosphorylation of beta-catenin at Ser 552,” Biochemical and Biophysical Research Communications, vol. 395, no. 1, pp. 146–151, 2010.
[241]  W. J. Gullick, “Prevalence of aberrant expression of the epidermal growth factor receptor in human cancers,” British Medical Bulletin, vol. 47, no. 1, pp. 87–98, 1991.
[242]  K. K. Guturi, T. Mandal, A. Chatterjee et al., “Mechanism of beta-catenin-mediated transcriptional regulation of epidermal growth factor receptor expression in glycogen synthase kinase 3 beta-inactivated prostate cancer cells,” Journal of Biological Chemistry, vol. 287, pp. 18287–18296, 2012.
[243]  P. C. Marker, “Does prostate cancer co-opt the developmental program?” Differentiation, vol. 76, no. 6, pp. 736–744, 2008.
[244]  N. S. Fearnhead, M. P. Britton, and W. F. Bodmer, “The ABC of APC,” Human Molecular Genetics, vol. 10, no. 7, pp. 721–733, 2001.
[245]  D. R. Chesire, C. M. Ewing, J. Sauvageot, G. S. Bova, and W. B. Isaacs, “Detection and analysis of beta-catenin mutations in prostate cancer,” Prostate, vol. 45, pp. 323–334, 2000.
[246]  L. Richiardi, V. Fiano, L. Vizzini et al., “Promoter methylation in APC, RUNX3, and GSTP1 and mortality in prostate cancer patients,” Journal of Clinical Oncology, vol. 27, no. 19, pp. 3161–3168, 2009.
[247]  L. E. Pascal, R. Z. N. Vêncio, L. S. Page et al., “Gene expression relationship between prostate cancer cells of Gleason 3, 4 and normal epithelial cells as revealed by cell type-specific transcriptomes,” BMC Cancer, vol. 9, article 452, 2009.
[248]  C. M. Koh, C. J. Bieberich, C. V. Dang, W. G. Nelson, S. Yegnasubramanian, and A. M. De Marzo, “MYC and prostate cancer,” Genes and Cancer, vol. 1, no. 6, pp. 617–628, 2010.
[249]  J. P. Alao, “The regulation of cyclin D1 degradation: roles in cancer development and the potential for therapeutic invention,” Molecular Cancer, vol. 6, article 24, 2007.
[250]  T. Grigoryan, P. Wend, A. Klaus, and W. Birchmeier, “Deciphering the function of canonical Wnt signals in development and disease: conditional loss- and gain-of-function mutations of β-catenin in mice,” Genes and Development, vol. 22, no. 17, pp. 2308–2341, 2008.
[251]  C. L. Hall, A. Bafico, J. Dai, S. A. Aaronson, and E. T. Keller, “Prostate cancer cells promote osteoblastic bone metastases through Wnts,” Cancer Research, vol. 65, no. 17, pp. 7554–7560, 2005.
[252]  K. J. Bruxvoort, H. M. Charbonneau, T. A. Giambernardi et al., “Inactivation of Apc in the mouse prostate causes prostate carcinoma,” Cancer Research, vol. 67, no. 6, pp. 2490–2496, 2007.
[253]  P. Uysal-Onganer, Y. Kawano, M. Caro et al., “Wnt-11 promotes neuroendocrine-like differentiation, survival and migration of prostate cancer cells,” Molecular Cancer, vol. 9, article 55, 2010.
[254]  S. Gupta, K. Iljin, H. Sara et al., “FZD4 as a mediator of ERG oncogene-induced WNT signaling and epithelial-to-mesenchymal transition in human prostate cancer cells,” Cancer Research, vol. 70, no. 17, pp. 6735–6745, 2010.
[255]  M. A. Liss, M. Schlicht, A. Kahler et al., “Characterization of soy-based changes in Wnt-frizzled signaling in prostate cancer,” Cancer Genomics and Proteomics, vol. 7, no. 5, pp. 245–252, 2010.
[256]  D. R. Chesire and W. B. Isaacs, “β-Catenin signaling in prostate cancer: an early perspective,” Endocrine-Related Cancer, vol. 10, no. 4, pp. 537–560, 2003.
[257]  B. He, L. You, K. Uematsu et al., “A monoclonal antibody against Wnt-1 induces apoptosis in human cancer cells,” Neoplasia, vol. 6, no. 1, pp. 7–14, 2004.
[258]  G. W. Yardy and S. F. Brewster, “Wnt signalling and prostate cancer,” Prostate Cancer and Prostatic Diseases, vol. 8, no. 2, pp. 119–126, 2005.
[259]  S. Le Guellec, I. Soubeyran, P. Rochaix et al., “CTNNB1 mutation analysis is a useful tool for the diagnosis of desmoid tumors: a study of 260 desmoid tumors and 191 potential morphologic mimics,” Modern Pathology, vol. 25, pp. 1551–1558, 2012.
[260]  K. Willert, J. D. Brown, E. Danenberg et al., “Wnt proteins are lipid-modified and can act as stem cell growth factors,” Nature, vol. 423, no. 6938, pp. 448–452, 2003.
[261]  M. Sharma, W. W. Chuang, and Z. Sun, “Phosphatidylinositol 3-kinase/Akt stimulates androgen pathway through GSK3β inhibition and nuclear β-catenin accumulation,” Journal of Biological Chemistry, vol. 277, no. 34, pp. 30935–30941, 2002.
[262]  P. Polakis, “The many ways of Wnt in cancer,” Current Opinion in Genetics and Development, vol. 17, no. 1, pp. 45–51, 2007.
[263]  P. Wend, J. D. Holland, U. Ziebold, and W. Birchmeier, “Wnt signaling in stem and cancer stem cells,” Seminars in Cell and Developmental Biology, vol. 21, no. 8, pp. 855–863, 2010.
[264]  T. Reya, S. J. Morrison, M. F. Clarke, and I. L. Weissman, “Stem cells, cancer, and cancer stem cells,” Nature, vol. 414, no. 6859, pp. 105–111, 2001.
[265]  M. D. Castellone, H. Teramoto, B. O. Williams, K. M. Druey, and J. S. Gutkind, “Medicine: prostaglandin E2 promotes colon cancer cell growth through a Gs-axin-β-catenin signaling axis,” Science, vol. 310, no. 5753, pp. 1504–1510, 2005.
[266]  M. Lepourcelet, Y. N. P. Chen, D. S. France et al., “Small-molecule antagonists of the oncogenic Tcf/β-catenin protein complex,” Cancer Cell, vol. 5, no. 1, pp. 91–102, 2004.
[267]  J. P. Rey and D. L. Ellies, “Wnt modulators in the biotech pipeline,” Developmental Dynamics, vol. 239, no. 1, pp. 102–114, 2010.

Full-Text

comments powered by Disqus

Contact Us

service@oalib.com

QQ:3279437679

WhatsApp +8615387084133

WeChat 1538708413