Cell Plasticity and Prostate Cancer: The Role of Epithelial–Mesenchymal Transition in Tumor Progression, Invasion, Metastasis and Cancer Therapy Resistance
Abstract
:Simple Summary
Abstract
1. Introduction
2. Molecular Events Governing EMT in Cancer Progression
2.1. EMT and Signaling Pathways: The Crosstalk with Prostate Cancer
Transforming Growth Factor-β (TGF-β): A Signaling Master of EMT
2.2. TGF-β Signaling and Androgen Axis in Prostate Cancer
2.3. Androgen and Estrogen Signaling in EMT
2.4. Src Signaling
2.5. Wnt, Hedgehog, Notch Signaling, and EGFR Signaling
2.6. Novel Transcription Factors Regulating EMT in PCa
2.7. Endothelin Axis and EMT in Prostate Cancer
3. Epigenetic Changes and EMT in Prostate Cancer
4. EMT and Therapy Resistance in Prostate Cancer
4.1. Androgen Deprivation Therapy
4.2. Chemotherapy
4.3. Radiation Therapy
4.4. The Role of Tumor Microenviroment (TME)
5. Conclusions and Future Directions
Author Contributions
Funding
Conflicts of Interest
References
- Bray, F.; Ferlay, J.; Soerjomataram, I.; Siegel, R.L.; Torre, L.A.; Jemal, A. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J. Clin. 2018, 68, 394–424. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Torre, L.A.; Siegel, R.L.; Ward, E.M.; Jemal, A. Global Cancer Incidence and Mortality Rates and Trends—An Update. Cancer Epidemiol. Biomark. Prev. 2016, 25, 16–27. [Google Scholar] [CrossRef] [Green Version]
- Sartor, O. Why is prostate cancer incidence rising in young men? Cancer 2020, 126, 17–18. [Google Scholar] [CrossRef] [Green Version]
- Ferlay, J.; Soerjomataram, I.; Dikshit, R.; Eser, S.; Mathers, C.; Rebelo, M.; Parkin, D.M.; Forman, D.; Bray, F. Cancer incidence and mortality worldwide: Sources, methods and major patterns in GLOBOCAN 2012. Int. J. Cancer 2015, 136, E359–E386. [Google Scholar] [CrossRef] [PubMed]
- Matuszak, E.A.; Kyprianou, N. Androgen regulation of epithelial–mesenchymal transition in prostate tumorigenesis. Expert Rev. Endocrinol. Metab. 2011, 6, 469–482. [Google Scholar] [CrossRef] [Green Version]
- Lamouille, S.; Xu, J.; Derynck, R. Molecular mechanisms of epithelial–mesenchymal transition. Nat. Rev. Mol. Cell Biol. 2014, 15, 178–196. [Google Scholar] [CrossRef] [Green Version]
- Kalluri, R.; Weinberg, R.A. The basics of epithelial-mesenchymal transition. J. Clin. Investig. 2009, 119, 1420–1428. [Google Scholar] [CrossRef] [Green Version]
- Thiery, J.P.; Sleeman, J.P. Complex networks orchestrate epithelial–mesenchymal transitions. Nat. Rev. Mol. Cell Biol. 2006, 7, 131–142. [Google Scholar] [CrossRef]
- Thiery, J.P.; Acloque, H.; Huang, R.Y.-J.; Nieto, M.A. Epithelial-Mesenchymal Transitions in Development and Disease. Cell 2009, 139, 871–890. [Google Scholar] [CrossRef]
- Thiery, J.P. Epithelial–mesenchymal transitions in tumour progression. Nat. Rev. Cancer 2002, 2, 442–454. [Google Scholar] [CrossRef] [PubMed]
- Loh, C.-Y.; Chai, J.; Tang, T.; Wong, W.; Sethi, G.; Shanmugam, M.; Chong, P.; Looi, C. The E-Cadherin and N-Cadherin Switch in Epithelial-to-Mesenchymal Transition: Signaling, Therapeutic Implications, and Challenges. Cells 2019, 8, 1118. [Google Scholar] [CrossRef] [Green Version]
- Pu, H.; Begemann, D.E.; Kyprianou, N. Aberrant TGF-β Signaling Drives Castration-Resistant Prostate Cancer in a Male Mouse Model of Prostate Tumorigenesis. Endocrinology 2017, 158, 1612–1622. [Google Scholar] [CrossRef] [Green Version]
- Røsland, G.V.; Dyrstad, S.E.; Tusubira, D.; Helwa, R.; Tan, T.Z.; Lotsberg, M.L.; Pettersen, I.K.N.; Berg, A.; Kindt, C.; Hoel, F.; et al. Epithelial to mesenchymal transition (EMT) is associated with attenuation of succinate dehydrogenase (SDH) in breast cancer through reduced expression of SDHC. Cancer Metab. 2019, 7, 6. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Jia, D.; Li, X.; Bocci, F.; Tripathi, S.; Deng, Y.; Jolly, M.K.; Onuchic, J.N.; Levine, H. Quantifying Cancer Epithelial-Mesenchymal Plasticity and its Association with Stemness and Immune Response. J. Clin. Med. 2019, 8, 725. [Google Scholar] [CrossRef] [Green Version]
- De Craene, B.; Berx, G. Regulatory networks defining EMT during cancer initiation and progression. Nat. Rev. Cancer 2013, 13, 97–110. [Google Scholar] [CrossRef]
- Moreno-Bueno, G.; Portillo, F.; Cano, A. Transcriptional regulation of cell polarity in EMT and cancer. Oncogene 2008, 27, 6958–6969. [Google Scholar] [CrossRef] [Green Version]
- Watanabe, K.; Villarreal-Ponce, A.; Sun, P.; Salmans, M.L.; Fallahi, M.; Andersen, B.; Dai, X. Mammary Morphogenesis and Regeneration Require the Inhibition of EMT at Terminal End Buds by Ovol2 Transcriptional Repressor. Dev. Cell 2014, 29, 59–74. [Google Scholar] [CrossRef] [Green Version]
- Li, C.; Hong, T.; Nie, Q. Quantifying the landscape and kinetic paths for epithelial-mesenchymal transition from a core circuit. Phys. Chem. 2016, 18, 17949–17956. [Google Scholar] [CrossRef] [Green Version]
- Li, C.; Wang, J. Quantifying the Landscape for Development and Cancer from a Core Cancer Stem Cell Circuit. Cancer Res. 2015, 75, 2607–2618. [Google Scholar] [CrossRef] [Green Version]
- Boareto, M.; Jolly, M.K.; Goldman, A.; Pietilä, M.; Mani, S.; Sengupta, S.; Ben-Jacob, E.; Levine, H.; Onuchic, J.N. Notch-Jagged signalling can give rise to clusters of cells exhibiting a hybrid epithelial/mesenchymal phenotype. J. R. Soc. Interface 2016, 13, 20151106. [Google Scholar] [CrossRef]
- Bocci, F.; Jolly, M.K.; Tripathi, S.C.; Aguilar, M.; Hanash, S.M.; Levine, H.; Onuchic, J.N. Numb prevents a complete epithelial–mesenchymal transition by modulating Notch signalling. J. R. Soc. Interface 2017, 14, 20170512. [Google Scholar] [CrossRef] [Green Version]
- Hasegawa, S.; Nagano, H.; Konno, M.; Eguchi, H.; Tomokuni, A.; Tomimaru, Y.; Asaoka, T.; Wada, H.; Hama, N.; Kawamoto, K.; et al. A crucial epithelial to mesenchymal transition regulator, Sox4/Ezh2 axis is closely related to the clinical outcome in pancreatic cancer patients. Int. J. Oncol. 2016, 48, 145–152. [Google Scholar] [CrossRef] [Green Version]
- Thompson, E.W.; Newgreen, D.F.; Tarin, D. Carcinoma Invasion and Metastasis: A Role for Epithelial-Mesenchymal Transition? Cancer Res. 2005, 65, 5991–5995. [Google Scholar] [CrossRef] [Green Version]
- Tsai, J.H.; Yang, J. Epithelial-mesenchymal plasticity in carcinoma metastasis. Genes Dev. 2013, 27, 2192–2206. [Google Scholar] [CrossRef] [Green Version]
- Liao, T.-T.; Yang, M.-H. Hybrid Epithelial/Mesenchymal State in Cancer Metastasis: Clinical Significance and Regulatory Mechanisms. Cells 2020, 9, 623. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kolijn, K.; Verhoef, E.; van Leenders, G.J. Morphological and immunohistochemical identification of epithelial-to-mesenchymal transition in clinical prostate cancer. Oncotarget 2015, 6, 24488–24498. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Morris, S.M.; Carter, K.T.; Baek, J.Y.; Koszarek, A.; Yeh, M.M.; Knoblaugh, S.E.; Grady, W.M. TGF-β signaling alters the pattern of liver tumorigenesis induced by Pten inactivation. Oncogene 2015, 34, 3273–3282. [Google Scholar] [CrossRef] [Green Version]
- Horie, Y.; Suzuki, A.; Kataoka, E.; Sasaki, T.; Hamada, K.; Sasaki, J.; Mizuno, K.; Hasegawa, G.; Kishimoto, H.; Iizuka, M.; et al. Hepatocyte-specific Pten deficiency results in steatohepatitis and hepatocellular carcinomas. J. Clin. Investig. 2004, 113, 1774–1783. [Google Scholar] [CrossRef] [Green Version]
- Attisano, L.; Wrana, J.L. Signal integration in TGF-β, WNT, and Hippo pathways. F1000Prime Rep. 2013, 5, 17. [Google Scholar] [CrossRef]
- Ikushima, H.; Miyazono, K. TGFβ signalling: A complex web in cancer progression. Nat. Rev. Cancer 2010, 10, 415–424. [Google Scholar] [CrossRef]
- Derynck, R.; Akhurst, R.J.; Balmain, A. TGF-β signaling in tumor suppression and cancer progression. Nat. Genet. 2001, 29, 117–129. [Google Scholar] [CrossRef]
- Kiyono, K.; Suzuki, H.I.; Morishita, Y.; Komuro, A.; Iwata, C.; Yashiro, M.; Hirakawa, K.; Kano, M.R.; Miyazono, K. c-Ski overexpression promotes tumor growth and angiogenesis through inhibition of transforming growth factor-β signaling in diffuse-type gastric carcinoma. Cancer Sci. 2009, 100, 1809–1816. [Google Scholar] [CrossRef] [PubMed]
- Komuro, A.; Yashiro, M.; Iwata, C.; Morishita, Y.; Johansson, E.; Matsumoto, Y.; Watanabe, A.; Aburatani, H.; Miyoshi, H.; Kiyono, K.; et al. Diffuse-Type Gastric Carcinoma: Progression, Angiogenesis, and Transforming Growth Factor β Signaling. J. Natl. Cancer Inst. 2009, 101, 592–604. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Roberts, A.B.; Wakefield, L. The two faces of transforming growth factor in carcinogenesis. Proc. Natl. Acad. Sci. USA 2003, 100, 8621–8623. [Google Scholar] [CrossRef] [Green Version]
- Massagué, J. TGF-β Signal Transduction. Annu. Rev. Biochem. 1998, 67, 753–791. [Google Scholar] [CrossRef]
- Liu, S.; Chen, S.; Zeng, J. TGF-β signaling: A complex role in tumorigenesis (Review). Mol. Med. Rep. 2018, 17, 699–704. [Google Scholar] [CrossRef] [Green Version]
- Yu, Q.; Stamenkovic, I. Cell surface localized matrix metalloproteinase 9 proteolytically activates TGF beta and promotes tu-mor invasion and angiogenesis. Genes Dev. 2000, 14, 163–176. [Google Scholar]
- Yu, J.S.L.; Ramasamy, T.S.; Murphy, N.; Holt, M.; Czapiewski, R.; Wei, S.-K.; Cui, W. PI3K/mTORC2 regulates TGF-β/Activin signalling by modulating Smad2/3 activity via linker phosphorylation. Nat. Commun. 2015, 6, 7212. [Google Scholar] [CrossRef] [Green Version]
- Vo, B.T.; Morton, D.; Komaragiri, S.; Millena, A.C.; Leath, C.; Khan, S.A. TGF-β Effects on Prostate Cancer Cell Migration and Invasion Are Mediated by PGE2 through Activation of PI3K/AKT/mTOR Pathway. Endocrinology 2013, 154, 1768–1779. [Google Scholar] [CrossRef]
- Singha, P.K.; Pandeswara, S.; Geng, H.; Lan, R.; Venkatachalam, M.A.; Saikumar, P. TGF-β induced TMEPAI/PMEPA1 inhibits canonical Smad signaling through R-Smad sequestration and promotes non-canonical PI3K/Akt signaling by reducing PTEN in triple negative breast cancer. Genes Cancer 2014, 5, 320–336. [Google Scholar] [CrossRef] [Green Version]
- Xie, F.; Liu, J.; Li, C.; Zhao, Y. Simvastatin blocks TGF-β1-induced epithelial-mesenchymal transition in human prostate cancer cells. Oncol. Lett. 2016, 11, 3377–3383. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Jin, H.; He, Y.; Zhao, P.; Hu, Y.; Tao, J.; Chen, J.; Huang, Y. Targeting lipid metabolism to overcome EMT-associated drug resistance via integrin β3/FAK pathway and tumor-associated macrophage repolarization using legumain-activatable delivery. Theranostics 2019, 9, 265–278. [Google Scholar] [CrossRef]
- Odero-Marah, V.; Hawsawi, O.; Henderson, V.; Sweeney, J. Epithelial-Mesenchymal Transition (EMT) and Prostate Cancer. In Cell & Molecular Biology of Prostate Cancer; Schatten, H., Ed.; Springer: Berlin/Heidelberg, Germany, 2018; pp. 101–110. [Google Scholar]
- Shiao, S.L.; Chu, G.C.-Y.; Chung, L.W. Regulation of prostate cancer progression by the tumor microenvironment. Cancer Lett. 2016, 380, 340–348. [Google Scholar] [CrossRef] [Green Version]
- Nguyen, D.P.; Li, J.; Yadav, S.S.; Tewari, A.K. Recent insights into NF-κB signalling pathways and the link between inflammation and prostate cancer. BJU Int. 2014, 114, 168–176. [Google Scholar] [CrossRef]
- Zhang, Q.; Helfand, B.T.; Jang, T.L.; Zhu, L.J.; Chen, L.; Yang, X.J.; Kozlowski, J.; Smith, N.; Kundu, S.D.; Yang, G.; et al. Nuclear Factor-κB-Mediated Transforming Growth Factor-β-Induced Expression of Vimentin Is an Independent Predictor of Biochemical Recurrence after Radical Prostatectomy. Clin. Cancer Res. 2009, 15, 3557–3567. [Google Scholar] [CrossRef] [Green Version]
- Locke, J.A.; Guns, E.S.; Lubik, A.A.; Adomat, H.H.; Hendy, S.C.; Wood, C.A.; Ettinger, S.L.; Gleave, M.E.; Nelson, C.C. Androgen Levels Increase by Intratumoral De novo Steroidogenesis during Progression of Castration-Resistant Prostate Cancer. Cancer Res. 2008, 68, 6407–6415. [Google Scholar] [CrossRef] [Green Version]
- Ye, H.; Cheng, J.; Tang, Y.; Liu, Z.; Xu, C.; Liu, Y.; Sun, Y. Human Bone Marrow-Derived Mesenchymal Stem Cells produced TGFbeta Contributes to Progression and Metastasis of Prostate Cancer. Cancer Investig. 2012, 30, 513–518. [Google Scholar] [CrossRef]
- Song, B.; Park, S.-H.; Zhao, J.C.; Fong, K.-W.; Li, S.; Lee, Y.; Yang, Y.A.; Sridhar, S.; Lu, X.; Abdulkadir, S.A.; et al. Targeting FOXA1-mediated repression of TGF-β signaling suppresses castration-resistant prostate cancer progression. J. Clin. Investig. 2019, 129, 569–582. [Google Scholar] [CrossRef] [Green Version]
- Haque, S.; Morris, J.C. Transforming growth factor-β: A therapeutic target for cancer. Hum. Vaccines Immunother. 2017, 13, 1741–1750. [Google Scholar] [CrossRef] [PubMed]
- Yoon, G.; Kim, J.Y.; Choi, Y.K.; Won, Y.S.; Lim, I.K. Direct activation of TGF-β1 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. [Google Scholar] [CrossRef]
- Wang, H.; Song, K.; Sponseller, T.L.; Danielpour, D. Novel Function of Androgen Receptor-associated Protein 55/Hic-5 as a Negative Regulator of Smad3 Signaling. J. Biol. Chem. 2005, 280, 5154–5162. [Google Scholar] [CrossRef] [Green Version]
- Song, K.; Wang, H.; Krebs, T.L.; Wang, B.; Kelley, T.J.; Danielpour, D. DHT Selectively Reverses Smad3-Mediated/TGF-β-Induced Responses through Transcriptional Down-Regulation of Smad3 in Prostate Epithelial Cells. Mol. Endocrinol. 2010, 24, 2019–2029. [Google Scholar] [CrossRef] [Green Version]
- Qi, W.; Gao, S.; Chu, J.; Zhou, L.; Wang, Z. Negative Androgen-Response Elements Mediate Androgen-Dependent Transcriptional Inhibition of TGF- 1 and CDK2 Promoters in the Prostate Gland. J. Androl. 2012, 33, 27–36. [Google Scholar] [CrossRef]
- Schaeffer, E.M.; Marchionni, L.; Huang, Z.; Simons, B.; Blackman, A.; Yu, W.; Parmigiani, G.; Berman, D.M. Androgen-induced programs for prostate epithelial growth and invasion arise in embryogenesis and are reactivated in cancer. Oncogene 2008, 27, 7180–7191. [Google Scholar] [CrossRef] [Green Version]
- Harris, W.P.; Mostaghel, E.A.; Nelson, P.S.; Montgomery, B. Androgen deprivation therapy: Progress in understanding mechanisms of resistance and optimizing androgen depletion. Nat. Clin. Pract. Urol. 2009, 6, 76–85. [Google Scholar] [CrossRef]
- Nieto, M.; Finn, S.; Loda, M.; Hahn, W.C. Prostate cancer: Re-focusing on androgen receptor signaling. Int. J. Biochem. Cell Biol. 2007, 39, 1562–1568. [Google Scholar] [CrossRef] [Green Version]
- Zhu, M.; Kyprianou, N. Role of androgens and the androgen receptor in epithelial-mesenchymal transition and invasion of prostate cancer cells. FASEB J. 2010, 24, 769–777. [Google Scholar] [CrossRef] [Green Version]
- Liu, Y.-N.; Liu, Y.; Lee, H.-J.; Hsu, Y.-H.; Chen, J.-H. Activated Androgen Receptor Downregulates E-Cadherin Gene Expression and Promotes Tumor Metastasis. Mol. Cell. Biol. 2008, 28, 7096–7108. [Google Scholar] [CrossRef] [Green Version]
- Zhu, S.; Zhao, D.; Li, C.; Li, Q.; Jiang, W.; Liu, Q.; Wang, R.; Fazli, L.; Li, Y.; Zhang, L.; et al. BMI1 is directly regulated by androgen receptor to promote castration-resistance in prostate cancer. Oncogene 2020, 39, 17–29. [Google Scholar] [CrossRef]
- Sun, Y.; Wang, B.-E.; Leong, K.G.; Yue, P.; Li, L.; Jhunjhunwala, S.; Chen, D.; Seo, K.; Modrusan, Z.; Gao, W.-Q.; et al. Androgen Deprivation Causes Epithelial–Mesenchymal Transition in the Prostate: Implications for Androgen-Deprivation Therapy. Cancer Res. 2012, 72, 527–536. [Google Scholar] [CrossRef] [Green Version]
- Jennbacken, K.; Tešan, T.; Wang, W.; Gustavsson, H.; Damber, J.-E.; Welén, K. N-cadherin increases after androgen deprivation and is associated with metastasis in prostate cancer. Endocr. Relat. Cancer 2010, 17, 469–479. [Google Scholar] [CrossRef] [Green Version]
- Tanaka, H.; Kono, E.; Tran, C.P.; Miyazaki, H.; Yamashiro, J.; Shimomura, T.; Fazli, L.; Wada, R.; Huang, J.; Vessella, R.L.; et al. Monoclonal antibody targeting of N-cadherin inhibits prostate cancer growth, metastasis and castration resistance. Nat. Med. 2010, 16, 1414–1420. [Google Scholar] [CrossRef] [PubMed]
- Li, H.; Price, D.K.; Figg, W.D. ADH1, an N-cadherin inhibitor, evaluated in preclinical models of angiogenesis and androgen-independent prostate cancer. Anti-Cancer Drugs 2007, 18, 563–568. [Google Scholar] [CrossRef]
- Jaggi, M.; Nazemi, T.; Abrahams, N.A.; Baker, J.J.; Galich, A.; Smith, L.M.; Balaji, K. N-cadherin switching occurs in high Gleason grade prostate cancer. Prostate 2006, 66, 193–199. [Google Scholar] [CrossRef] [PubMed]
- Blaschuk, O.W.; Devemy, E. Cadherins as novel targets for anti-cancer therapy. Eur. J. Pharmacol. 2009, 625, 195–198. [Google Scholar] [CrossRef]
- Tsai, Y.-C.; Chen, W.-Y.; Abou-Kheir, W.; Zeng, T.; Yin, J.J.; Bahmad, H.; Lee, Y.-C.; Liu, Y.-N. Androgen deprivation therapy-induced epithelial-mesenchymal transition of prostate cancer through downregulating SPDEF and activating CCL2. Biochim. Biophys. Acta Mol. Basis Dis. 2018, 1864, 1717–1727. [Google Scholar] [CrossRef] [PubMed]
- Parimi, V.; Goyal, R.; Poropatich, K.; Yang, X.J. Neuroendocrine differentiation of prostate cancer: A review. Am. J. Clin. Exp. Urol. 2014, 2, 273–285. [Google Scholar]
- Dicken, H.; Hensley, P.; Kyprianou, N. Prostate tumor neuroendocrine differentiation via EMT: The road less traveled. Asian J. Urol. 2019, 6, 82–90. [Google Scholar] [CrossRef] [PubMed]
- Tiwari, R.; Manzar, N.; Bhatia, V.; Yadav, A.; Nengroo, M.A.; Datta, D.; Carskadon, S.; Gupta, N.; Sigouros, M.; Khani, F.; et al. Androgen deprivation upregulates SPINK1 expression and potentiates cellular plasticity in prostate cancer. Nat. Commun. 2020, 11, 384. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Yeh, Y.; Guo, Q.; Connelly, Z.; Cheng, S.; Yang, S.; Prieto-Domínguez, N.; Yu, X. Wnt/Beta-Catenin Signaling and Prostate Cancer Therapy Resistance. Adv. Exp. Med. Biol. 2019, 1210, 351–378. [Google Scholar] [CrossRef]
- Montanari, M.; Rossetti, S.; Cavaliere, C.; D’Aniello, C.; Malzone, M.G.; Vanacore, D.; di Franco, R.; La Mantia, E.; Iovane, G.; Piscitelli, R.; et al. Epithelial-mesenchymal transition in prostate cancer: An overview. Oncotarget 2017, 8, 35376–35389. [Google Scholar] [CrossRef] [Green Version]
- Di Zazzo, E.; Galasso, G.; Giovannelli, P.; di Donato, M.; Bilancio, A.; Perillo, B.; Sinisi, A.A.; Migliaccio, A.; Castoria, G. Estrogen Receptors in Epithelial-Mesenchymal Transition of Prostate Cancer. Cancers 2019, 11, 1418. [Google Scholar] [CrossRef] [Green Version]
- Gyftopoulos, K.; Sotiropoulou, G.; Varakis, I.; Barbalias, G. Cellular Distribution of Retinoic Acid Receptor–α in Benign Hyperplastic and Malignant Human Prostates: Comparison with Androgen, Estrogen and Progesterone Receptor Status. Eur. Urol. 2000, 38, 323–330. [Google Scholar] [CrossRef] [PubMed]
- Bonkhoff, H.; Fixemer, T.; Hunsicker, I.; Remberger, K. Estrogen Receptor Expression in Prostate Cancer and Premalignant Prostatic Lesions. Am. J. Pathol. 1999, 155, 641–647. [Google Scholar] [CrossRef] [Green Version]
- Bonkhoff, H. Estrogen receptor signaling in prostate cancer: Implications for carcinogenesis and tumor progression. Prostate 2018, 78, 2–10. [Google Scholar] [CrossRef] [PubMed]
- Chakravarty, D.; Sboner, A.; Nair, S.S.; Giannopoulou, E.G.; Li, R.; Hennig, S.; Mosquera, J.M.; Pauwels, J.; Park, K.; Kossai, M.; et al. The oestrogen receptor alpha-regulated lncRNA NEAT1 is a critical modulator of prostate cancer. Nat. Commun. 2014, 5, 5383. [Google Scholar] [CrossRef] [PubMed]
- Shen, Y.; Cao, J.; Liang, Z.; Lin, Q.; Wang, J.; Yang, X.; Zhang, R.; Zong, J.; Du, X.; Peng, Y.; et al. Estrogen receptor α-NOTCH1 axis enhances basal stem-like cells and epithelial-mesenchymal transition phenotypes in prostate cancer. Cell Commun. Signal. 2019, 17, 50. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wang, L.; Yang, X.; Chang, Y.W.Y.; Qi, M.; Zhou, Z.; Zhang, J.; Han, B. SOX4 is associated with poor prognosis in prostate cancer and promotes epithelial–mesenchymal transition in vitro. Prostate Cancer Prostatic Dis. 2013, 16, 301–307. [Google Scholar] [CrossRef] [PubMed]
- Dey, P.; Velazquez-Villegas, L.A.; Faria, M.; Turner, A.; Jonsson, P.; Webb, P.; Williams, C.; Gustafsson, J.-Å.; Ström, A.M. Estrogen Receptor β2 Induces Hypoxia Signature of Gene Expression by Stabilizing HIF-1α in Prostate Cancer. PLoS ONE 2015, 10, e0128239. [Google Scholar] [CrossRef]
- Gadkar, S.; Nair, S.; Patil, S.; Kalamani, S.; Bandivdekar, A.; Patel, V.; Chaudhari, U.; Sachdeva, G. Membrane-initiated estrogen signaling in prostate cancer: A route to epithelial-to-mesenchymal transition. Mol. Carcinog. 2019, 58, 2077–2090. [Google Scholar] [CrossRef]
- Lafront, C.; Germain, L.; Weidmann, C.; Audet-Walsh, É. A Systematic Study of the Impact of Estrogens and Selective Estrogen Receptor Modulators on Prostate Cancer Cell Proliferation. Sci. Rep. 2020, 10, 1–12. [Google Scholar] [CrossRef] [Green Version]
- Varkaris, A.; Katsiampoura, A.D.; Araujo, J.C.; Gallick, G.E.; Corn, P.G. Src signaling pathways in prostate cancer. Cancer Metastasis Rev. 2014, 33, 595–606. [Google Scholar] [CrossRef] [Green Version]
- Thomas, S.M.; Brugge, J.S. Cellular Functions Regulated by Src Family Kinases. Annu. Rev. Cell Dev. Biol. 1997, 13, 513–609. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Marzia, M.; Sims, N.A.; Voit, S.; Migliaccio, S.; Taranta, A.; Bernardini, S.; Faraggiana, T.; Yoneda, T.; Mundy, G.R.; Boyce, B.F.; et al. Decreased C-Src Expression Enhances Osteoblast Differentiation and Bone Formation. J. Cell Biol. 2000, 151, 311–320. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Roskoski, R. Src protein–tyrosine kinase structure and regulation. Biochem. Biophys. Res. Commun. 2004, 324, 1155–1164. [Google Scholar] [CrossRef]
- Manetti, F.; Botta, M. Src Inhibitors and Angiogenesis. Curr. Pharm. Des. 2007, 13, 2118–2128. [Google Scholar] [CrossRef]
- Guarino, M. Src signaling in cancer invasion. J. Cell. Physiol. 2009, 223, 14–26. [Google Scholar] [CrossRef]
- Fizazi, K. The role of Src in prostate cancer. Ann. Oncol. 2007, 18, 1765–1773. [Google Scholar] [CrossRef] [PubMed]
- Gelman, I.H. Androgen Receptor Activation in Castration-Recurrent Prostate Cancer: The Role of Src-Family and Ack1 Tyrosine Kinases. Int. J. Biol. Sci. 2014, 10, 620–626. [Google Scholar] [CrossRef]
- Dai, Y.; Siemann, D. c-Src is required for hypoxia-induced metastasis-associated functions in prostate cancer cells. OncoTargets Ther. 2019, 12, 3519–3529. [Google Scholar] [CrossRef] [Green Version]
- Lu, Y.; Dong, B.; Xu, F.; Xu, Y.; Pan, J.; Song, J.; Zhang, J.; Huang, Y.; Xue, W. CXCL1-LCN2 paracrine axis promotes progression of prostate cancer via the Src activation and epithelial-mesenchymal transition. Cell Commun. Signal. 2019, 17, 1–15. [Google Scholar] [CrossRef] [Green Version]
- Alwanian, W.M.; Tyner, A.L. Protein tyrosine kinase 6 signaling in prostate cancer. Am. J. Clin. Exp. Urol. 2020, 8, 1–8. [Google Scholar]
- Baron, S.; Manin, M.; Beaudoin, C.; Leotoing, L.; Communal, Y.; Veyssiere, G.; Morel, L. Androgen Receptor Mediates Non-genomic Activation of Phosphatidylinositol 3-OH Kinase in Androgen-sensitive Epithelial Cells. J. Biol. Chem. 2004, 279, 14579–14586. [Google Scholar] [CrossRef] [Green Version]
- Zamagni, A.; Cortesi, M.; Zanoni, M.; Tesei, A. Non-nuclear AR Signaling in Prostate Cancer. Front. Chem. 2019, 7, 651. [Google Scholar] [CrossRef]
- Liu, P.; Wakamiya, M.; Shea, M.J.; Albrecht, U.; Behringer, R.R.; Bradley, A. Requirement for Wnt3 in vertebrate axis formation. Nat. Genet. 1999, 22, 361–365. [Google Scholar] [CrossRef]
- Popperl, H.; Schmidt, C.; Wilson, V.; Hume, C.R.; Dodd, J.; Krumlauf, R.; Beddington, R.S. Misexpression of Wnt8C in the mouse induces an ectopic embry-onic axis and causes a truncation of the anterior neuroectoderm. Development 1997, 124, 2997–3005. [Google Scholar] [CrossRef]
- Garcia-Castro, M.I.; Marcelle, C.; Bronner-Fraser, M. Ectodermal Wnt function as a neural crest inducer. Science 2002, 297, 848–851. [Google Scholar]
- Carmona-Fontaine, C.; Matthews, H.K.; Kuriyama, S.; Moreno, M.; Dunn, G.A.; Parsons, M.; Stern, C.D.; Mayor, R. Contact inhibition of locomotion in vivo controls neural crest directional migration. Nature 2008, 456, 957–961. [Google Scholar] [CrossRef] [Green Version]
- Brabletz, T.; Jung, A.; Reu, S.; Porzner, M.; Hlubek, F.; Kunz-Schughart, L.A.; Knuechel, R.; Kirchner, T. Variable -catenin expression in colorectal cancers indicates tumor progression driven by the tumor environment. Proc. Natl. Acad. Sci. USA 2001, 98, 10356–10361. [Google Scholar] [CrossRef] [Green Version]
- Yilmaz, M.; Christofori, G. EMT, the cytoskeleton, and cancer cell invasion. Cancer Metastasis Rev. 2009, 28, 15–33. [Google Scholar] [CrossRef] [Green Version]
- Robinson, D.R.; Zylstra, C.R.; Williams, B. Wnt Signaling and Prostate Cancer. Curr. Drug Targets 2008, 9, 271–580. [Google Scholar] [CrossRef]
- Li, Y.; Wang, L.; Zhang, M.; Melamed, J.; Liu, X.; Reiter, R.; Wei, J.; Peng, Y.; Zou, X.; Pellicer, A.; et al. LEF1 in Androgen-Independent Prostate Cancer: Regulation of Androgen Receptor Expression, Prostate Cancer Growth, and Invasion. Cancer Res. 2009, 69, 3332–3338. [Google Scholar] [CrossRef] [Green Version]
- Wang, G.; Wang, J.; Sadar, M.D. Crosstalk between the Androgen Receptor and β-Catenin in Castrate-Resistant Prostate Cancer. Cancer Res. 2008, 68, 9918–9927. [Google Scholar] [CrossRef] [Green Version]
- Schmalhofer, O.; Brabletz, S.; Brabletz, T. E-cadherin, β-catenin, and ZEB1 in malignant progression of cancer. Cancer Metastasis Rev. 2009, 28, 151–166. [Google Scholar] [CrossRef]
- Polette, M.; Mestdagt, M.; Bindels, S.; Nawrocki-Raby, B.; Hunziker, W.; Foidart, J.-M.; Birembaut, P.; Gilles, C. β-Catenin and ZO-1: Shuttle Molecules Involved in Tumor Invasion-Associated Epithelial-Mesenchymal Transition Processes. Cells Tissues Organs 2007, 185, 61–65. [Google Scholar] [CrossRef] [PubMed]
- Whitaker, H.C.; Girling, J.; Warren, A.Y.; Leung, H.; Mills, I.G.; Neal, D.E. Alterations in β-catenin expression and localization in prostate cancer. Prostate 2008, 68, 1196–1205. [Google Scholar] [CrossRef]
- Saha, B.; Arase, A.; Imam, S.S.; Tsao-Wei, D.; Naritoku, W.Y.; Groshen, S.; Jones, L.W. Overexpression of E-cadherin and β-Catenin proteins in metastatic prostate cancer cells in bone. Prostate 2008, 68, 78–84. [Google Scholar] [CrossRef]
- Li, Q.; Xia, D.; Wang, Z.; Liu, B.; Zhang, J.; Peng, P.; Tang, Q.; Dong, J.; Guo, J.; Kuang, D.; et al. Circadian Rhythm Gene PER3 Negatively Regulates Stemness of Prostate Cancer Stem Cells via WNT/β-Catenin Signaling in Tumor Microenvironment. Front. Cell Dev. Biol. 2021, 9. [Google Scholar] [CrossRef]
- Khan, A.Q.; Ahmed, E.I.; Elareer, N.R.; Junejo, K.; Steinhoff, M.; Uddin, S. Role of miRNA-Regulated Cancer Stem Cells in the Pathogenesis of Human Malignancies. Cells 2019, 8, 840. [Google Scholar] [CrossRef] [Green Version]
- Briscoe, J.; Therond, P. The mechanisms of Hedgehog signalling and its roles in development and disease. Nat. Rev. Mol. Cell Biol. 2013, 14, 416–429. [Google Scholar] [CrossRef]
- Chiang, C.; Litingtung, Y.; Lee, E.; Young, K.E.; Corden, J.L.; Westphal, H.; Beachy, P.A. Cyclopia and defective axial patterning in mice lacking Sonic hedgehog gene function. Nature 1996, 383, 407–413. [Google Scholar] [CrossRef] [PubMed]
- Monsoro-Burq, A.-H. Sclerotome development and morphogenesis: When experimental embryology meets genetics. Int. J. Dev. Biol. 2005, 49, 301–308. [Google Scholar] [CrossRef]
- Li, X.; Deng, W.; Nail, C.D.; Bailey, S.K.; Kraus, M.H.; Ruppert, J.M.; Lobo-Ruppert, S.M. Snail induction is an early response to Gli1 that determines the efficiency of epithelial transformation. Oncogene 2006, 25, 609–621. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Gonnissen, A.; Isebaert, S.; Haustermans, K. Hedgehog Signaling in Prostate Cancer and Its Therapeutic Implication. Int. J. Mol. Sci. 2013, 14, 13979. [Google Scholar] [CrossRef]
- Fendrich, V.; Waldmann, J.; Esni, F.; Ramaswamy, A.; Mullendore, M.; Buchholz, M.; Maitra, A.; Feldmann, G. Snail and Sonic Hedgehog activation in neuroendocrine tumors of the ileum. Endocr. Relat. Cancer 2007, 14, 865–874. [Google Scholar] [CrossRef] [Green Version]
- Yauch, R.L.; Gould, S.E.; Scales, S.; Tang, T.; Tian, H.; Ahn, C.P.; Marshall, D.; Fu, L.; Januario, T.; Kallop, D.; et al. A paracrine requirement for hedgehog signalling in cancer. Nature 2008, 455, 406–410. [Google Scholar] [CrossRef]
- Tian, H.; Callahan, C.A.; DuPree, K.J.; Darbonne, W.C.; Ahn, C.P.; Scales, S.J.; de Sauvage, F.J. Hedgehog signaling is restricted to the stromal compartment during pancreatic carcinogenesis. Proc. Natl. Acad. Sci. USA 2009, 106, 4254–4259. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Fan, L.; Pepicelli, C.V.; Dibble, C.C.; Catbagan, W.; Zarycki, J.L.; Laciak, R.; Gipp, J.; Shaw, A.; Lamm, M.L.G.; Munoz, A.; et al. Hedgehog Signaling Promotes Prostate Xenograft Tumor Growth. Endocrinology 2004, 145, 3961–3970. [Google Scholar] [CrossRef] [Green Version]
- Scales, S.; de Sauvage, F.J. Mechanisms of Hedgehog pathway activation in cancer and implications for therapy. Trends Pharmacol. Sci. 2009, 30, 303–312. [Google Scholar] [CrossRef] [PubMed]
- Sheng, T.; Li, C.; Zhang, X.; Chi, S.; He, N.; Chen, K.; McCormick, F.; Gatalica, Z.; Xie, J. Activation of the hedgehog pathway in advanced prostate cancer. Mol. Cancer 2004, 3, 29. [Google Scholar] [CrossRef] [Green Version]
- Datta, S.; Datta, M.W. Sonic Hedgehog signaling in advanced prostate cancer. Cell. Mol. Life Sci. 2006, 63, 435–448. [Google Scholar] [CrossRef]
- Sanchez, P.; Hernández, A.M.; Stecca, B.; Kahler, A.J.; deGueme, A.M.; Barrett, A.; Beyna, M.; Datta, M.W.; Datta, S.; i Altaba, A.R. Inhibition of prostate cancer proliferation by interference with Sonic Hedgehog-GLI1 signaling. Proc. Natl. Acad. Sci. USA 2004, 101, 12561–12566. [Google Scholar] [CrossRef] [Green Version]
- Tzelepi, V.; Karlou, M.; Wen, S.; Hoang, A.; Logothetis, C.; Troncoso, P.; Efstathiou, E. Expression of hedgehog pathway components in prostate carcinoma microenvironment: Shifting the balance towards autocrine signalling. Histopathology 2011, 58, 1037–1047. [Google Scholar] [CrossRef] [Green Version]
- Azoulay, S.; Terry, S.; Chimingqi, M.; Sirab, N.; Faucon, H.; Medina, S.G.D.D.; Moutereau, S.; Maillé, P.; Soyeux, P.; Abbou, C.; et al. Comparative expression of Hedgehog ligands at different stages of prostate carcinoma progression. J. Pathol. 2008, 216, 460–470. [Google Scholar] [CrossRef] [PubMed]
- Kim, T.-J.; Lee, J.Y.; Hwang, T.-K.; Kang, C.S.; Choi, Y.-J. Hedgehog signaling protein expression and its association with prognostic parameters in prostate cancer: A retrospective study from the view point of new 2010 anatomic stage/prognostic groups. J. Surg. Oncol. 2011, 104, 472–479. [Google Scholar] [CrossRef] [PubMed]
- Lubik, A.A.; Nouri, M.; Truong, S.; Ghaffari, M.; Adomat, H.H.; Corey, E.; Cox, M.E.; Li, N.; Guns, E.S.; Yenki, P.; et al. Paracrine sonic hedgehog signaling contributes significantly to acquired steroidogenesis in the prostate tumor microenvironment. Int. J. Cancer 2017, 140, 358–369. [Google Scholar] [CrossRef] [PubMed]
- Ishii, A.; Shigemura, K.; Kitagawa, K.; Sung, S.-Y.; Chen, K.-C.; Yi-Te, C.; Liu, M.-C.; Fujisawa, M. Anti-tumor Effect of Hedgehog Signaling Inhibitor, Vismodegib, on Castration-resistant Prostate Cancer. Anticancer. Res. 2020, 40, 5107–5114. [Google Scholar] [CrossRef] [PubMed]
- Su, Q.; Xin, L. Notch signaling in prostate cancer: Refining a therapeutic opportunity. Histol. Histopathol. 2016, 31, 149–157. [Google Scholar] [CrossRef] [PubMed]
- Deng, G.; Ma, L.; Meng, Q.; Ju, X.; Jiang, K.; Jiang, P.; Yu, Z. Notch signaling in the prostate: Critical roles during development and in the hallmarks of prostate cancer biology. J. Cancer Res. Clin. Oncol. 2015, 142, 531–547. [Google Scholar] [CrossRef]
- Leong, K.G.; Gao, W.-Q. The Notch pathway in prostate development and cancer. Differentiation 2008, 76, 699–716. [Google Scholar] [CrossRef] [PubMed]
- Zhang, L.; Sha, J.; Yang, G.; Huang, X.; Bo, J.; Huang, Y. Activation of Notch pathway is linked with epithelial-mesenchymal transition in prostate cancer cells. Cell Cycle 2017, 16, 999–1007. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zhu, H.; Zhou, X.; Redfield, S.; Lewin, J.; Miele, L. Elevated Jagged-1 and Notch-1 expression in high grade and metastatic prostate cancers. Am. J. Transl. Res. 2013, 5, 368–378. [Google Scholar] [PubMed]
- Zhang, J.; Kuang, Y.; Wang, Y.; Xu, Q.; Ren, Q. Notch-4 silencing inhibits prostate cancer growth and EMT via the NF-κB pathway. Apoptosis 2017, 22, 877–884. [Google Scholar] [CrossRef]
- Day, K.C.; Hiles, G.L.; Kozminsky, M.; Dawsey, S.J.; Paul, A.; Broses, L.J.; Shah, R.; Kunja, L.P.; Hall, C.; Palanisamy, N.; et al. HER2 and EGFR Overexpression Support Metastatic Progression of Prostate Cancer to Bone. Cancer Res. 2017, 77, 74–85. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Hardbower, D.M.; Coburn, L.A.; Asim, M.; Singh, K.; Sierra, J.C.; Barry, D.P.; Gobert, A.P.; Piazuelo, M.B.; Washington, M.K.; Wilson, K.T. EGFR-mediated macrophage activation promotes colitis-associated tumorigenesis. Oncogene 2017, 36, 3807–3819. [Google Scholar] [CrossRef] [Green Version]
- Jia, Y.; Yun, C.-H.; Park, E.; Ercan, D.; Manuia, M.; Juarez, J.; Xu, C.; Rhee, K.; Chen, T.; Zhang, H.; et al. Overcoming EGFR(T790M) and EGFR(C797S) resistance with mutant-selective allosteric inhibitors. Nature 2016, 534, 129–132. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Cargnello, M.; Roux, P.P. Activation and Function of the MAPKs and Their Substrates, the MAPK-Activated Protein Kinases. Microbiol. Mol. Biol. Rev. 2011, 75, 50–83. [Google Scholar] [CrossRef] [Green Version]
- Lo, H.-W.; Xia, W.; Wei, Y.; Ali-Seyed, M.; Huang, S.-F.; Hung, M.-C. Novel prognostic value of nuclear epidermal growth factor receptor in breast cancer. Cancer Res. 2005, 65, 338–348. [Google Scholar]
- Marti, U.; Ruchti, C.; Kämpf, J.; Thomas, G.A.; Williams, E.D.; Peter, H.J.; Gerber, H.; Bürgi, U. Nuclear Localization of Epidermal Growth Factor and Epidermal Growth Factor Receptors in Human Thyroid Tissues. Thyroid 2001, 11, 137–145. [Google Scholar] [CrossRef]
- Psyrri, A.; Yu, Z.; Weinberger, P.M.; Sasaki, C.; Haffty, B.; Camp, R.; Rimm, D.; Burtness, B.A. Quantitative Determination of Nuclear and Cytoplasmic Epidermal Growth Factor Receptor Expression in Oropharyngeal Squamous Cell Cancer by Using Automated Quantitative Analysis. Clin. Cancer Res. 2005, 11, 5856–5862. [Google Scholar] [CrossRef] [Green Version]
- Dittmann, K.; Mayer, C.; Fehrenbacher, B.; Schaller, M.; Kehlbach, R.; Rodemann, H.P. Nuclear EGFR shuttling induced by ionizing radiation is regulated by phosphorylation at residue Thr654. FEBS Lett. 2010, 584, 3878–3884. [Google Scholar] [CrossRef] [Green Version]
- Liccardi, G.; Hartley, J.A.; Hochhauser, D. EGFR Nuclear Translocation Modulates DNA Repair following Cisplatin and Ionizing Radiation Treatment. Cancer Res. 2011, 71, 1103–1114. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Hsu, S.-C.; Miller, S.A.; Wang, Y.; Hung, M.-C. Nuclear EGFR is required for cisplatin resistance and DNA repair. Am. J. Transl. Res. 2009, 1, 249–258. [Google Scholar]
- Salomon, D.S.; Brandt, R.; Ciardiello, F.; Normanno, N. Epidermal growth factor-related peptides and their receptors in human malignancies. Crit. Rev. Oncol. Hematol. 1995, 19, 183–232. [Google Scholar] [CrossRef]
- Shah, R.B.; Ghosh, D.; Elder, J.T. Epidermal growth factor receptor (ErbB1) expression in prostate cancer progression: Correlation with androgen independence. Prostate 2006, 66, 1437–1444. [Google Scholar] [CrossRef] [Green Version]
- Di Lorenzo, G.; Tortora, G.; d’Armiento, F.P.; de Rosa, G.; Staibano, S.; Autorino, R.; d’Armiento, M.; de Laurentiis, M.; de Placido, S.; Catalano, G.; et al. Expression of epidermal growth factor receptor correlates with disease relapse and progression to androgen-independence in human prostate cancer. Clin. Cancer Res. 2002, 8, 3438–3444. [Google Scholar] [PubMed]
- Traish, A.M.; Morgentaler, A. Epidermal growth factor receptor expression escapes androgen regulation in prostate cancer: A potential molecular switch for tumour growth. Br. J. Cancer 2009, 101, 1949–1956. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Frawley, T.; Piskareva, O. Extracellular Vesicle Dissemination of Epidermal Growth Factor Receptor and Ligands and Its Role in Cancer Progression. Cancers 2020, 12, 3200. [Google Scholar] [CrossRef]
- Liao, Y.; Guo, Z.; Xia, X.; Liu, Y.; Huang, C.; Jiang, L.; Wang, X.; Liu, J.; Huang, H. Inhibition of EGFR signaling with Spautin-1 represents a novel therapeutics for prostate cancer. J. Exp. Clin. Cancer Res. 2019, 38, 1–16. [Google Scholar] [CrossRef]
- Correa, R.J.; Valdes, Y.R.; Peart, T.M.; Fazio, E.N.; Bertrand, M.; McGee, J.; Préfontaine, M.; Sugimoto, A.; DiMattia, G.E.; Shepherd, T.G. Combination of AKT inhibition with autophagy blockade effectively reduces ascites-derived ovarian cancer cell viability. Carcinogenesis 2014, 35, 1951–1961. [Google Scholar] [CrossRef] [Green Version]
- Horie, R.; Nakamura, O.; Yamagami, Y.; Mori, M.; Nishimura, H.; Fukuoka, N.; Yamamoto, T. Apoptosis and antitumor effects induced by the combination of an mTOR inhibitor and an autophagy inhibitor in human osteosarcoma MG63 cells. Int. J. Oncol. 2016, 48, 37–44. [Google Scholar] [CrossRef] [Green Version]
- Shao, S.; Li, S.; Qin, Y.; Wang, X.; Yang, Y.; Bai, H.; Zhou, L.; Zhao, C.; Wang, C. Spautin-1, a novel autophagy inhibitor, enhances imatinib-induced apoptosis in chronic myeloid leukemia. Int. J. Oncol. 2014, 44, 1661–1668. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Bach, D.-H.; Long, N.P.; Luu, T.-T.-T.; Anh, N.H.; Kwon, S.W.; Lee, S.K. The Dominant Role of Forkhead Box Proteins in Cancer. Int. J. Mol. Sci. 2018, 19, 3279. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wang, T.; Zheng, L.; Wang, Q.; Hu, Y.-W. Emerging roles and mechanisms of FOXC2 in cancer. Clin. Chim. Acta 2018, 479, 84–93. [Google Scholar] [CrossRef]
- Hollier, B.G.; Tinnirello, A.A.; Werden, S.J.; Evans, K.W.; Taube, J.; Sarkar, T.R.; Sphyris, N.; Shariati, M.; Kumar, S.V.; Battula, V.L.; et al. FOXC2 Expression Links Epithelial–Mesenchymal Transition and Stem Cell Properties in Breast Cancer. Cancer Res. 2013, 73, 1981–1992. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Li, C.; Ding, H.; Tian, J.; Wu, L.; Wang, Y.; Xing, Y.; Chen, M. Forkhead Box Protein C2 Promotes Epithelial-Mesenchymal Transition, Migration and Invasion in Cisplatin-Resistant Human Ovarian Cancer Cell Line (SKOV3/CDDP). Cell. Physiol. Biochem. 2016, 39, 1098–1110. [Google Scholar] [CrossRef] [Green Version]
- Børretzen, A.; Gravdal, K.; Haukaas, S.A.; Beisland, C.; Akslen, L.A.; Halvorsen, O.J. FOXC2 expression and epithelial–mesenchymal phenotypes are associated with castration resistance, metastasis and survival in prostate cancer. J. Pathol. Clin. Res. 2019, 5, 272–286. [Google Scholar] [CrossRef]
- Paranjape, A.N.; Soundararajan, R.; Werden, S.J.; Joseph, R.; Taube, J.; Liu, H.; Rodriguez-Canales, J.; Sphyris, N.; Wistuba, I.; Miura, N.; et al. Inhibition of FOXC2 restores epithelial phenotype and drug sensitivity in prostate cancer cells with stem-cell properties. Oncogene 2016, 35, 5963–5976. [Google Scholar] [CrossRef] [Green Version]
- Teng, M.; Zhou, S.; Cai, C.; Lupien, M.; He, H.H. Pioneer of prostate cancer: Past, present and the future of FOXA1. Protein Cell 2021, 12, 29–38. [Google Scholar] [CrossRef]
- Gao, S.; Chen, S.; Han, D.; Wang, Z.; Li, M.; Han, W.; Besschetnova, A.; Liu, M.; Zhou, F.; Barrett, D.; et al. Chromatin binding of FOXA1 is promoted by LSD1-mediated demethylation in prostate cancer. Nat. Genet. 2020, 52, 1011–1017. [Google Scholar] [CrossRef]
- Ma, Z.; Xin, Z.; Hu, W.; Jiang, S.; Yang, Z.; Yan, X.; Li, X.; Yang, Y.; Chen, F. Forkhead box O proteins: Crucial regulators of cancer EMT. Semin. Cancer Biol. 2018, 50, 21–31. [Google Scholar] [CrossRef]
- Alwhaibi, A.; Verma, A.; Artham, S.; Adil, M.S.; Somanath, P.R. Nodal pathway activation due to Akt1 suppression is a molecular switch for prostate cancer cell epithelial-to-mesenchymal transition and metastasis. Biochem. Pharmacol. 2019, 168, 1–13. [Google Scholar] [CrossRef] [PubMed]
- Saad, F.; Shore, N.; Zhang, T.; Sharma, S.; Cho, H.K.; Jacobs, I.A. Emerging therapeutic targets for patients with advanced prostate cancer. Cancer Treat. Rev. 2019, 76, 1–9. [Google Scholar] [CrossRef] [PubMed]
- Gan, L.; Xu, M.; Hua, R.; Tan, C.; Zhang, J.; Gong, Y.; Wu, Z.; Weng, W.; Sheng, W.; Guo, W. The polycomb group protein EZH2 induces epithelial–mesenchymal transition and pluripotent phenotype of gastric cancer cells by binding to PTEN promoter. J. Hematol. Oncol. 2018, 11, 9. [Google Scholar] [CrossRef]
- Perotti, V.; Baldassari, P.; Molla, A.; Nicolini, G.; Bersani, I.; Grazia, G.; Benigni, F.; Maurichi, A.; Santinami, M.; Anichini, A.; et al. An actionable axis linking NFATc2 to EZH2 controls the EMT-like program of melanoma cells. Oncogene 2019, 38, 4384–4396. [Google Scholar] [CrossRef]
- Lobo, J.; Rodrigues, Â.; Antunes, L.; Graça, I.; Ramalho-Carvalho, J.; Vieira, F.Q.; Martins, A.T.; Oliveira, J.; Jeronimo, C.; Henrique, R. High immunoexpression of Ki67, EZH2, and SMYD3 in diagnostic prostate biopsies independently predicts outcome in patients with prostate cancer. Urol. Oncol. Semin. Orig. Investig. 2018, 36, 161.e7–161.e17. [Google Scholar] [CrossRef] [PubMed]
- Xu, K.; Wu, Z.J.; Groner, A.C.; He, H.H.; Cai, C.; Lis, R.T.; Wu, X.; Stack, E.C.; Loda, M.; Liu, T.; et al. EZH2 Oncogenic Activity in Castration-Resistant Prostate Cancer Cells Is Polycomb-Independent. Science 2012, 338, 1465–1469. [Google Scholar] [CrossRef] [Green Version]
- Yang, Y.A.; Yu, J. EZH2, an epigenetic driver of prostate cancer. Protein Cell 2013, 4, 331–341. [Google Scholar] [CrossRef]
- Kim, J.; Lee, Y.; Lu, X.; Song, B.; Fong, K.-W.; Cao, Q.; Licht, J.D.; Zhao, J.C.; Yu, J. Polycomb- and Methylation-Independent Roles of EZH2 as a Transcription Activator. Cell Rep. 2018, 25, 2808–2820.e4. [Google Scholar] [CrossRef] [Green Version]
- Wang, J.; He, C.; Gao, P.; Wang, S.; Lv, R.; Zhou, H.; Zhou, Q.; Zhang, K.; Sun, J.; Fan, C.; et al. HNF1B-mediated repression of SLUG is suppressed by EZH2 in aggressive prostate cancer. Oncogene 2020, 39, 1335–1346. [Google Scholar] [CrossRef] [Green Version]
- Tao, J.; Shi, L.; Huang, L.; Shi, H.; Chen, H.; Wang, Y.; Wang, T. EZH2 is involved in silencing of WNT5A during epithelial–mesenchymal transition of colon cancer cell line. J. Cancer Res. Clin. Oncol. 2017, 143, 2211–2219. [Google Scholar] [CrossRef]
- Bai, Y.; Zhang, Z.; Cheng, L.; Wang, R.; Chen, X.; Kong, Y.; Feng, F.; Ahmad, N.; Li, L.; Liu, X. Inhibition of enhancer of zeste homolog 2 (EZH2) overcomes enzalutamide resistance in castration-resistant prostate cancer. J. Biol. Chem. 2019, 294, 9911–9923. [Google Scholar] [CrossRef] [PubMed]
- Taplin, M.-E.; Hussain, A.; Shah, S.; Shore, N.D.; Agrawal, M.; Clark, W.; Edenfield, W.J.; Nordquist, L.T.; Sartor, O.A.; Butrynski, J.E.; et al. ProSTAR: A phase Ib/II study of CPI-1205, a small molecule inhibitor of EZH2, combined with enzalutamide (E) or abiraterone/prednisone (A/P) in patients with metastatic castration-resistant prostate cancer (mCRPC). J. Clin. Oncol. 2019, 37 (Suppl. 7), TPS335. [Google Scholar] [CrossRef]
- Qiu, X.; Wang, W.; Li, B.; Cheng, B.; Lin, K.; Bai, J.; Li, H.; Yang, G. Targeting Ezh2 could overcome docetaxel resistance in prostate cancer cells. BMC Cancer 2019, 19, 27. [Google Scholar] [CrossRef] [PubMed]
- Elsir, T.; Smits, A.; Lindström, M.S.; Nistér, M. Transcription factor PROX1: Its role in development and cancer. Cancer Metastasis Rev. 2012, 31, 793–805. [Google Scholar] [CrossRef]
- Lu, M.-H.; Huang, C.-C.; Pan, M.-R.; Chen, H.-H.; Hung, W.-C. Prospero Homeobox 1 Promotes Epithelial–Mesenchymal Transition in Colon Cancer Cells by Inhibiting E-cadherin via miR-9. Clin. Cancer Res. 2012, 18, 6416–6425. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Liu, Y.; Zhang, J.-B.; Qin, Y.; Wang, W.; Wei, L.; Teng, Y.; Guo, L.; Zhang, B.; Lin, Z.; Liu, J.; et al. PROX1 promotes hepatocellular carcinoma metastasis by way of up-regulating hypoxia-inducible factor 1α expression and protein stability. Hepatology 2013, 58, 692–705. [Google Scholar] [CrossRef] [Green Version]
- Wang, B.; Huang, J.; Zhou, J.; Hui, K.; Xu, S.; Fan, J.; Li, L.; Wang, X.; Hsieh, J.-T.; He, D.; et al. DAB2IP regulates EMT and metastasis of prostate cancer through targeting PROX1 transcription and destabilizing HIF1α protein. Cell. Signal. 2016, 28, 1623–1630. [Google Scholar] [CrossRef]
- Nelson, J.; Bagnato, A.; Battistini, B.; Nisen, P. The endothelin axis: Emerging role in cancer. Nat. Rev. Cancer 2003, 3, 110–116. [Google Scholar] [CrossRef] [PubMed]
- McKenzie, G.A.G.; Hinsley, E.E.; Hunter, K.; Lambert, D.W. The endothelin axis in head and neck cancer: A promising therapeutic opportunity? J. Oral Pathol. Med. 2014, 43, 395–404. [Google Scholar] [CrossRef]
- Gu, X.; Han, S.; Cui, M.; Xue, J.; Ai, L.; Sun, L.; Zhu, X.; Wang, Y.; Liu, C. Knockdown of endothelin receptor B inhibits the progression of triple-negative breast cancer. Ann. N. Y. Acad. Sci. 2019, 1448, 5–18. [Google Scholar] [CrossRef]
- Cianfrocca, R.; Rosanò, L.; Tocci, P.; Sestito, R.; Caprara, V.; di Castro, V.; de Maria, R.; Bagnato, A. Blocking endothelin-1-receptor/β-catenin circuit sensitizes to chemotherapy in colorectal cancer. Cell Death Differ. 2017, 24, 1811–1820. [Google Scholar] [CrossRef] [Green Version]
- Bagnato, A.; Rosanò, L. Epithelial-Mesenchymal Transition in Ovarian Cancer Progression: A Crucial Role for the Endothelin Axis. Cells Tissues Organs 2007, 185, 85–94. [Google Scholar] [CrossRef]
- Gyftopoulos, K.; Papanikolaou, S.; Bravou, V.; Papadaki, H. The role of the endothelin axis in promoting epithelial to mesenchymal transition and lymph node metastasis in prostate adenocarcinoma. Urol. Ann. 2017, 9, 372–379. [Google Scholar] [CrossRef]
- Qi, P.; Chen, M.; Zhang, L.-X.; Song, R.-X.; He, Z.-H.; Wang, Z.-P. A Meta-Analysis and Indirect Comparison of Endothelin A Receptor Antagonist for Castration-Resistant Prostate Cancer. PLoS ONE 2015, 10, e0133803. [Google Scholar] [CrossRef] [Green Version]
- Moon, H.H.; Clines, K.L.; Cooks, M.A.; Cialek, C.A.; Esvelt, M.A.; Clines, G.A. Castration Determines the Efficacy of ETAR Blockade in a Mouse Model of Prostate Cancer Bone Metastasis. Endocrinology 2019, 160, 1786–1796. [Google Scholar] [CrossRef] [PubMed]
- Xu, K.; Liao, Y. Epigenetic regulation of prostate cancer: The theories and the clinical implications. Asian J. Androl. 2019, 21, 279–290. [Google Scholar] [CrossRef]
- Jones, P.A. Functions of DNA methylation: Islands, start sites, gene bodies and beyond. Nat. Rev. Genet. 2012, 13, 484–492. [Google Scholar] [CrossRef] [PubMed]
- Curradi, M.; Izzo, A.; Badaracco, G.; Landsberger, N. Molecular Mechanisms of Gene Silencing Mediated by DNA Methylation. Mol. Cell. Biol. 2002, 22, 3157–3173. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kumar, S.; Cheng, X.; Klimasauskas, S.; Sha, M.; Posfai, J.; Roberts, R.; Wilson, G.G. The DNA (cytosine-5) methyltransferases. Nucleic Acids Res. 1994, 22, 1–10. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Cheung, H.H.; Lee, T.-L.; Rennert, O.M.; Chan, W.-Y. DNA methylation of cancer genome. Birth Defects Res. Part C Embryo Today Rev. 2009, 87, 335–350. [Google Scholar] [CrossRef] [Green Version]
- Ikeda, Y.; Kinoshita, T. DNA demethylation: A lesson from the garden. Chromosomes 2008, 118, 37–41. [Google Scholar] [CrossRef]
- Pistore, C.; Giannoni, E.; Colangelo, T.; Rizzo, F.; Magnani, E.; Muccillo, L.; Giurato, G.; Mancini, M.; Rizzo, S.; Riccardi, M.; et al. DNA methylation variations are required for epithelial-to-mesenchymal transition induced by cancer-associated fibroblasts in prostate cancer cells. Oncogene 2017, 36, 5551–5566. [Google Scholar] [CrossRef]
- Lin, P.-C.; Giannopoulou, E.G.; Park, K.; Mosquera, J.M.; Sboner, A.; Tewari, A.K.; Garraway, L.A.; Beltran, H.; Rubin, M.A.; Elemento, O. Epigenomic Alterations in Localized and Advanced Prostate Cancer. Neoplasia 2013, 15, 373–383, IN2–IN5. [Google Scholar] [CrossRef] [Green Version]
- Kang, G.H.; Lee, S.; Lee, H.J.; Hwang, K.S. Aberrant CpG island hypermethylation of multiple genes in prostate cancer and prostatic intraepithelial neoplasia. J. Pathol. 2004, 202, 233–240. [Google Scholar] [CrossRef]
- Mahon, K.L.; PRIMe Consortium; Qu, W.; Devaney, J.M.; Paul, C.; Castillo, L.; Wykes, R.J.; Chatfield, M.; Boyer, M.; Stockler, M.R.; et al. Methylated Glutathione S-transferase 1 (mGSTP1) is a potential plasma free DNA epigenetic marker of prognosis and response to chemotherapy in castrate-resistant prostate cancer. Br. J. Cancer 2014, 111, 1802–1809. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Suzuki, H.; Freije, D.; Nusskern, D.R.; Okami, K.; Cairns, P.; Sidransky, D.; Isaacs, W.B.; Bova, G.S. Interfocal heterogeneity of PTEN/MMAC1 gene alterations in multiple metastatic prostate cancer tissues. Cancer Res. 1998, 58, 9. [Google Scholar]
- Jarrard, D.F.; Bova, G.S.; Ewing, C.M.; Pin, S.S.; Nguyen, S.H.; Baylin, S.B.; Cairns, P.; Sidransky, D.; Herman, J.G.; Isaacs, W.B. Deletional, mutational, and methylation analyses of CDKN2 (p16/MTS1) in primary and metastatic prostate cancer. Genes Chromosom. Cancer 1997, 19, 90–96. [Google Scholar] [CrossRef]
- Pakneshan, P.; Xing, R.H.; Rabbani, S.A. Methylation status of uPA promoter as a molecular mechanism regulating prostate cancer invasion and growth in vitro and in vivo. FASEB J. 2003, 17, 1081–1088. [Google Scholar] [CrossRef] [PubMed]
- Lee, E.; Wang, J.; Yumoto, K.; Jung, Y.; Cackowski, F.C.; Decker, A.M.; Li, Y.; Franceschi, R.T.; Pienta, K.J.; Taichman, R.S. DNMT1 Regulates Epithelial-Mesenchymal Transition and Cancer Stem Cells, Which Promotes Prostate Cancer Metastasis. Neoplasia 2016, 18, 553–566. [Google Scholar] [CrossRef] [Green Version]
- Alhamwe, B.A.; Khalaila, R.; Wolf, J.; von Bülow, V.; Harb, H.; Alhamdan, F.; Hii, C.S.; Prescott, S.L.; Ferrante, A.; Renz, H.; et al. Histone modifications and their role in epigenetics of atopy and allergic diseases. Allergy Asthma Clin. Immunol. 2018, 14, 1–16. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Harb, H.; Alhamwe, B.A.; Garn, H.; Renz, H.; Potaczek, D.P. Recent developments in epigenetics of pediatric asthma. Curr. Opin. Pediatr. 2016, 28, 754–763. [Google Scholar] [CrossRef] [PubMed]
- Yu, T.; Wang, C.; Yang, J.; Guo, Y.; Wu, Y.; Li, X. Metformin inhibits SUV39H1-mediated migration of prostate cancer cells. Oncogene 2017, 6, e324. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Li, Q.; Li, Y.; Wang, Y.; Cui, Z.; Gong, L.; Qu, Z.; Zhong, Y.; Zhou, J.; Zhou, Y.; Gao, Y.; et al. Quantitative proteomic study of human prostate cancer cells with different metastatic potentials. Int. J. Oncol. 2016, 48, 1437–1446. [Google Scholar] [CrossRef] [Green Version]
- Askew, E.B.; Bai, S.; Parris, A.B.; Minges, J.T.; Wilson, E.M. Androgen receptor regulation by histone methyltransferase Suppressor of variegation 3-9 homolog 2 and Melanoma antigen-A11. Mol. Cell. Endocrinol. 2017, 443, 42–51. [Google Scholar] [CrossRef] [Green Version]
- Stopa, N.; Krebs, J.E.; Shechter, D. The PRMT5 arginine methyltransferase: Many roles in development, cancer and beyond. Cell. Mol. Life Sci. 2015, 72, 2041–2059. [Google Scholar] [CrossRef]
- Li, Y.; Trojer, P.; Xu, C.-F.; Cheung, P.; Kuo, A.; Drury, W.J.; Qiao, Q.; Neubert, T.A.; Xu, R.-M.; Gozani, O.; et al. The Target of the NSD Family of Histone Lysine Methyltransferases Depends on the Nature of the Substrate. J. Biol. Chem. 2009, 284, 34283–34295. [Google Scholar] [CrossRef] [Green Version]
- Ezponda, T.; Popovic, R.; Shah, M.Y.; Martinez-Garcia, E.; Zheng, Y.; Min, D.-J.; Will, C.; Neri, A.; Kelleher, N.L.; Yu, J.; et al. The histone methyltransferase MMSET/WHSC1 activates TWIST1 to promote an epithelial–mesenchymal transition and invasive properties of prostate cancer. Oncogene 2013, 32, 2882–2890. [Google Scholar] [CrossRef] [Green Version]
- Crea, F.; Clermont, P.-L.; Mai, A.; Helgason, C. Histone Modifications, Stem Cells and Prostate Cancer. Curr. Pharm. Des. 2014, 20, 1687–1697. [Google Scholar] [CrossRef]
- Cohen, I.; Poręba, E.; Kamieniarz, K.; Schneider, R. Histone Modifiers in Cancer: Friends or Foes? Genes Cancer 2011, 2, 631–647. [Google Scholar] [CrossRef] [Green Version]
- Lee, E.; Wang, J.; Jung, Y.; Cackowski, F.C.; Taichman, R.S. Reduction of two histone marks, H3k9me3 and H3k27me3 by epidrug induces neuroendocrine differentiation in prostate cancer. J. Cell. Biochem. 2018, 119, 3697–3705. [Google Scholar] [CrossRef]
- Sun, L.; Fang, J. Epigenetic regulation of epithelial–mesenchymal transition. Cell. Mol. Life Sci. 2016, 73, 4493–4515. [Google Scholar] [CrossRef]
- Sekhon, K.; Bucay, N.; Majid, S.; Dahiya, R.; Saini, S. MicroRNAs and epithelial-mesenchymal transition in prostate cancer. Oncotarget 2016, 7, 67597–67611. [Google Scholar] [CrossRef] [Green Version]
- Elmageed, Z.A.; Yang, Y.; Thomas, R.; Ranjan, M.; Mondal, D.; Moroz, K.; Fang, Z.; Rezk, B.M.; Moparty, K.; Sikka, S.C.; et al. Neoplastic Reprogramming of Patient-Derived Adipose Stem Cells by Prostate Cancer Cell-Associated Exosomes. STEM Cells 2014, 32, 983–997. [Google Scholar] [CrossRef] [Green Version]
- Seashols-Williams, S.J.; Budd, W.; Clark, G.C.; Wu, Q.; Daniel, R.; Dragoescu, E.; Zehner, Z.E. miR-9 Acts as an OncomiR in Prostate Cancer through Multiple Pathways That Drive Tumour Progression and Metastasis. PLoS ONE 2016, 11, e0159601. [Google Scholar] [CrossRef] [Green Version]
- Fletcher, C.E.; Dart, D.A.; Sita-Lumsden, A.; Cheng, H.; Rennie, P.S.; Bevan, C.L. Androgen-regulated processing of the oncomir MiR-27a, which targets Prohibitin in prostate cancer. Hum. Mol. Genet. 2012, 21, 3112–3127. [Google Scholar] [CrossRef] [Green Version]
- Hsu, T.-I.; Hsu, C.-H.; Lee, K.-H.; Lin, J.-T.; Chen, C.-S.; Chang, K.-C.; Su, C.-Y.; Hsiao, M.; Lu, P.-J. MicroRNA-18a is elevated in prostate cancer and promotes tumorigenesis through suppressing STK4 in vitro and in vivo. Oncogenesis 2014, 3, e99. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Liu, Y.-N.; Yin, J.J.; Abou-Kheir, W.; Hynes, P.G.; Casey, O.M.; Fang, L.; Yi, M.; Stephens, R.M.; Seng, V.; Sheppard-Tillman, H.; et al. MiR-1 and miR-200 inhibit EMT via Slug-dependent and tumorigenesis via Slug-independent mechanisms. Oncogene 2013, 32, 296–306. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Steele, R.; Mott, J.L.; Ray, R.B. MBP-1 Upregulates miR-29b, Which Represses Mcl-1, Collagens, and Matrix Metalloproteinase-2 in Prostate Cancer Cells. Genes Cancer 2010, 1, 381–387. [Google Scholar] [CrossRef] [PubMed]
- Gandellini, P.; Folini, M.; Longoni, N.; Pennati, M.; Binda, M.; Colecchia, M.; Salvioni, R.; Supino, R.; Moretti, R.; Limonta, P.; et al. miR-205 Exerts Tumor-Suppressive Functions in Human Prostate through Down-regulation of Protein Kinase Cε. Cancer Res. 2009, 69, 2287–2295. [Google Scholar] [CrossRef] [Green Version]
- Gandellini, P.; Profumo, V.; Casamichele, A.; Fenderico, N.; Borrelli, S.; Petrovich, G.; Santilli, G.; Callari, M.; Colecchia, M.; Pozzi, S.; et al. miR-205 regulates basement membrane deposition in human prostate: Implications for cancer development. Cell Death Differ. 2012, 19, 1750–1760. [Google Scholar] [CrossRef] [Green Version]
- Xu, B.; Wang, N.; Wang, X.; Tong, N.; Shao, N.; Tao, J.; Li, P.; Niu, X.; Feng, N.; Zhang, L.; et al. MiR-146a suppresses tumor growth and progression by targeting EGFR pathway and in a p-ERK-dependent manner in castration-resistant prostate cancer. Prostate 2012, 72, 1171–1178. [Google Scholar] [CrossRef]
- Dai, Y.; Wu, Z.; Lang, C.; Zhang, X.; He, S.; Yang, Q.; Guo, W.; Lai, Y.; Du, H.; Peng, X.; et al. Copy number gain of ZEB1 mediates a double-negative feedback loop with miR-33a-5p that regulates EMT and bone metastasis of prostate cancer dependent on TGF-β signaling. Theranostics 2019, 9, 6063–6079. [Google Scholar] [CrossRef] [PubMed]
- Ren, D.; Wang, M.; Guo, W.; Huang, S.; Wang, Z.; Zhao, X.; Du, H.; Song, L.; Peng, X. Double-negative feedback loop between ZEB2 and miR-145 regulates epithelial-mesenchymal transition and stem cell properties in prostate cancer cells. Cell Tissue Res. 2014, 358, 763–778. [Google Scholar] [CrossRef] [PubMed]
- Lo, U.-G.; Lee, C.-F.; Lee, M.-S.; Hsieh, J.-T. The Role and Mechanism of Epithelial-to-Mesenchymal Transition in Prostate Cancer Progression. Int. J. Mol. Sci. 2017, 18, 2079. [Google Scholar] [CrossRef] [Green Version]
- He, J.-H.; Li, B.-X.; Han, Z.-P.; Zou, M.-X.; Wang, L.; Lv, Y.-B.; Zhou, J.-B.; Cao, M.-R.; Li, Y.-G.; Zhang, J.-Z. Snail-activated long non-coding RNA PCA3 up-regulates PRKD3 expression by miR-1261 sponging, thereby promotes invasion and migration of prostate cancer cells. Tumor Biol. 2016, 37, 16163–16176. [Google Scholar] [CrossRef]
- Zhang, Y.; Zhang, J.; Liang, S.; Lang, G.; Liu, G.; Liu, P.; Deng, X. Long non-coding RNA VIM-AS1 promotes prostate cancer growth and invasion by regulating epithelial-mesenchymal transition. Off. J. Balk. Union Oncol. 2019, 24, 2090–2098. [Google Scholar]
- Wang, D.; Ding, L.; Wang, L.; Zhao, Y.; Sun, Z.; Karnes, R.J.; Zhang, J.; Huang, H. LncRNA MALAT1 enhances oncogenic activities of EZH2 in castration-resistant prostate cancer. Oncotarget 2015, 6, 41045–41055. [Google Scholar] [CrossRef]
- Chang, Z.; Cui, J.; Song, Y. Long noncoding RNA PVT1 promotes EMT via mediating microRNA-186 targeting of Twist1 in prostate cancer. Gene 2018, 654, 36–42. [Google Scholar] [CrossRef]
- Yang, B.; Gao, G.; Wang, Z.; Sun, D.; Wei, X.; Ma, Y.; Ding, Y. Long non-coding RNA HOTTIP promotes prostate cancer cells proliferation and migration by sponging miR-216a-5p. Biosci. Rep. 2018, 38. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Jiborn, T.; Bjartell, A.; Abrahamsson, P.-A. Neuroendocrine Differentiation in Prostatic Carcinoma During Hormonal Treatment. Urology 1998, 51, 585–589. [Google Scholar] [CrossRef]
- Arumugam, T.; Ramachandran, V.; Fournier, K.F.; Wang, H.; Marquis, L.; Abbruzzese, J.L.; Gallick, G.E.; Logsdon, C.D.; McConkey, D.J.; Choi, W. Epithelial to Mesenchymal Transition Contributes to Drug Resistance in Pancreatic Cancer. Cancer Res. 2009, 69, 5820–5828. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Smith, B.N.; Bhowmick, N.A. Role of EMT in Metastasis and Therapy Resistance. J. Clin. Med. 2016, 5, 17. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Culig, Z. Epithelial mesenchymal transition and resistance in endocrine-related cancers. Biochim. Biophys. Acta Mol. Cell Res. 2019, 1866, 1368–1375. [Google Scholar] [CrossRef] [PubMed]
- Shibue, T.; Weinberg, T.S.R.A. EMT, CSCs, and drug resistance: The mechanistic link and clinical implications. Nat. Rev. Clin. Oncol. 2017, 14, 611–629. [Google Scholar] [CrossRef] [Green Version]
- Shiota, M.; Itsumi, M.; Takeuchi, A.; Imada, K.; Yokomizo, A.; Kuruma, H.; Inokuchi, J.; Tatsugami, K.; Uchiumi, T.; Oda, Y.; et al. Crosstalk between epithelial-mesenchymal transition and castration resistance mediated by Twist1/AR signaling in prostate cancer. Endocr. Relat. Cancer 2015, 22, 889–900. [Google Scholar] [CrossRef] [Green Version]
- Paller, C.; Pu, H.; Begemann, D.E.; Wade, C.A.; Hensley, P.; Kyprianou, N. TGF-β receptor I inhibitor enhances response to enzalutamide in a pre-clinical model of advanced prostate cancer. Prostate 2019, 79, 31–43. [Google Scholar] [CrossRef] [Green Version]
- Shao, C.; Yu, B.; Liu, Y. Androgen receptor splicing variant 7: Beyond being a constitutively active variant. Life Sci. 2019, 234, 116768. [Google Scholar] [CrossRef]
- Ramamurthy, V.P.; Ramalingam, S.; Gediya, L.K.; Njar, V.C.O. The retinamide VNLG -152 inhibits f-AR/AR -V7 and MNK—eIF 4E signaling pathways to suppress EMT and castration-resistant prostate cancer xenograft growth. FEBS J. 2018, 285, 1051–1063. [Google Scholar] [CrossRef]
- Borgmann, H.; Lallous, N.; Ozistanbullu, D.; Beraldi, E.; Paul, N.; Dalal, K.; Fazli, L.; Haferkamp, A.; Lejeune, P.; Cherkasov, A.; et al. Moving Towards Precision Urologic Oncology: Targeting Enzalutamide-resistant Prostate Cancer and Mutated Forms of the Androgen Receptor Using the Novel Inhibitor Darolutamide (ODM-201). Eur. Urol. 2018, 73, 4–8. [Google Scholar] [CrossRef]
- Nam, R.K.; Benatar, T.; Amemiya, Y.; Wallis, C.J.; Romero, J.M.; Tsagaris, M.; Sherman, C.; Sugar, L.; Seth, A. MicroRNA-652 induces NED in LNCaP and EMT in PC3 prostate cancer cells. Oncotarget 2018, 9, 19159–19176. [Google Scholar] [CrossRef] [Green Version]
- Doldi, V.; Pennati, M.; Forte, B.; Gandellini, P.; Zaffaroni, N. Dissecting the role of microRNAs in prostate cancer metastasis: Implications for the design of novel therapeutic approaches. Cell. Mol. Life Sci. 2016, 73, 2531–2542. [Google Scholar] [CrossRef]
- Kolijn, K.; Verhoef, E.I.; Smid, M.; Böttcher, R.; Jenster, G.W.; Debets, R.; van Leenders, G.J. Epithelial–Mesenchymal Transition in Human Prostate Cancer Demonstrates Enhanced Immune Evasion Marked by IDO1 Expression. Cancer Res. 2018, 78, 4671–4679. [Google Scholar] [CrossRef] [Green Version]
- Tsai, Y.-S.; Jou, Y.-C.; Tsai, H.-T.; Cheong, I.-S.; Tzai, T.-S. Indoleamine-2,3-dioxygenase-1 expression predicts poorer survival and up-regulates ZEB2 expression in human early stage bladder cancer. Urol. Oncol. Semin. Orig. Investig. 2019, 37, 810.e17–810.e27. [Google Scholar] [CrossRef] [PubMed]
- Li, F.; Mahato, R.I. MicroRNAs and Drug Resistance in Prostate Cancers. Mol. Pharm. 2014, 11, 2539–2552. [Google Scholar] [CrossRef]
- Puhr, M.; Hoefer, J.; Schäfer, G.; Erb, H.H.; Oh, S.J.; Klocker, H.; Heidegger, I.; Neuwirt, H.; Culig, Z. Epithelial-to-Mesenchymal Transition Leads to Docetaxel Resistance in Prostate Cancer and Is Mediated by Reduced Expression of miR-200c and miR-205. Am. J. Pathol. 2012, 181, 2188–2201. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wang, P.; Henning, S.M.; Heber, D.; Vadgama, J.V. Sensitization to docetaxel in prostate cancer cells by green tea and quercetin. J. Nutr. Biochem. 2015, 26, 408–415. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lu, X.; Yang, F.; Chen, D.; Zhao, Q.; Chen, D.; Ping, H.; Xing, N. Quercetin reverses docetaxel resistance in prostate cancer via androgen receptor and PI3K/Akt signaling pathways. Int. J. Biol. Sci. 2020, 16, 1121–1134. [Google Scholar] [CrossRef]
- Lu, X.; Chen, D.; Yang, F.; Xing, N. Quercetin Inhibits Epithelial-to-Mesenchymal Transition (EMT) Process and Promotes Apoptosis in Prostate Cancer via Downregulating lncRNA MALAT1. Cancer Manag. Res. 2020, 12, 1741–1750. [Google Scholar] [CrossRef] [Green Version]
- Xue, Y.; Ms, B.Y.; Liu, Y.; Guo, R.; Li, J.; Ms, L.Z.; Su, J.; Sun, L.; Li, Y. Zinc promotes prostate cancer cell chemosensitivity to paclitaxel by inhibiting epithelial-mesenchymal transition and inducing apoptosis. Prostate 2019, 79, 647–656. [Google Scholar] [CrossRef]
- Murata, K.; Saga, R.; Monzen, S.; Tsuruga, E.; Hasegawa, K.; Hosokawa, Y. Understanding the mechanism underlying the acquisition of radioresistance in human prostate cancer cells. Oncol. Lett. 2019, 17, 5830–5838. [Google Scholar] [CrossRef] [Green Version]
- Tsao, T.; Beretov, J.; Ni, J.; Bai, X.; Bucci, J.; Graham, P.; Li, Y. Cancer stem cells in prostate cancer radioresistance. Cancer Lett. 2019, 465, 94–104. [Google Scholar] [CrossRef] [PubMed]
- Deng, X.; Elzey, B.D.; Poulson, J.M.; Morrison, W.B.; Ko, S.-C.; Hahn, N.M.; Ratliff, T.L.; Hu, C.-D. Ionizing radiation induces neuroendocrine differentiation of prostate cancer cells in vitro, in vivo and in prostate cancer patients. Am. J. Cancer Res. 2011, 1, 834–844. [Google Scholar]
- Chaiswing, L.; Weiss, H.L.; Jayswal, R.D.; Clair, D.K.S.; Kyprianou, N. Profiles of Radioresistance Mechanisms in Prostate Cancer. Crit. Rev. Oncog. 2018, 23, 39–67. [Google Scholar] [CrossRef]
- Wu, X.; Scott, H.; Carlsson, S.V.; Sjoberg, D.D.; Cerundolo, L.; Lilja, H.; Prevo, R.; Rieunier, G.; Macaulay, V.; Higgins, G.S.; et al. Increased EZH2 expression in prostate cancer is associated with metastatic recurrence following external beam radiotherapy. Prostate 2019, 79, 1079–1089. [Google Scholar] [CrossRef] [Green Version]
- Yadav, S.; Kowolik, C.M.; Lin, M.; Zuro, D.; Hui, S.K.; Riggs, A.; Horne, D.A. SMC1A is associated with radioresistance in prostate cancer and acts by regulating epithelial-mesenchymal transition and cancer stem-like properties. Mol. Carcinog. 2019, 58, 113–125. [Google Scholar] [CrossRef] [Green Version]
- Xie, P.; Yu, H.; Wang, F.; Yan, F.; He, X. Inhibition of LOXL2 Enhances the Radiosensitivity of Castration-Resistant Prostate Cancer Cells Associated with the Reversal of the EMT Process. BioMed Res. Int. 2019, 2019, 1–10. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wu, T.; Dai, Y. Tumor microenvironment and therapeutic response. Cancer Lett. 2017, 387, 61–68. [Google Scholar] [CrossRef] [PubMed]
- Taddei, M.L.; Cavallini, L.; Ramazzotti, M.; Comito, G.; Pietrovito, L.; Morandi, A.; Giannoni, E.; Raugei, G.; Chiarugi, P. Stromal-induced downregulation of miR-1247 promotes prostate cancer malignancy. J. Cell. Physiol. 2019, 234, 8274–8285. [Google Scholar] [CrossRef]
- Su, F.; Ahn, S.; Saha, A.; DiGiovanni, J.; Kolonin, M.G. Adipose stromal cell targeting suppresses prostate cancer epithelial-mesenchymal transition and chemoresistance. Oncogene 2019, 38, 1979–1988. [Google Scholar] [CrossRef]
- Su, F.; Daquinag, A.C.; Ahn, S.; Saha, A.; Dai, Y.; Zhao, Z.; DiGiovanni, J.; Kolonin, M.G. Progression of prostate carcinoma is promoted by adipose stromal cell-secreted CXCL12 signaling in prostate epithelium. NPJ Precis. Oncol. 2021, 5, 1–10. [Google Scholar] [CrossRef]
- Shi, J.; Wang, L.; Zou, C.; Xia, Y.; Qin, S.; Keller, E.; Mizokami, A.; Zhang, J.; Lu, Y. Tumor microenvironment promotes prostate cancer cell dissemination via the Akt/mTOR pathway. Oncotarget 2018, 9, 9206–9218. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lilis, I.; Giopanou, I.; Papadaki, H.; Gyftopoulos, K. The expression of p-mTOR and COUP-TFII correlates with increased lymphangiogenesis and lymph node metastasis in prostate adenocarcinoma. Urol. Oncol. Semin. Orig. Investig. 2018, 36, 311.e27–311.e35. [Google Scholar] [CrossRef]
- Chen, X.; Cheng, H.; Pan, T.; Liu, Y.; Su, Y.; Ren, C.; Huang, D.; Zha, X.; Liang, C. mTOR regulate EMT through RhoA and Rac1 pathway in prostate cancer. Mol. Carcinog. 2015, 54, 1086–1095. [Google Scholar] [CrossRef] [PubMed]
- Li, S.; Sheng, J.; Liu, Z.; Fan, Y.; Zhang, C.; Lv, T.; Hu, S.; Jin, J.; Yu, W.; Song, Y. Potent antitumour of the mTORC1/2 dual inhibitor AZD2014 in docetaxel-sensitive and docetaxel-resistant castration-resistant prostate cancer cells. J. Cell. Mol. Med. 2021, 25, 2436–2449. [Google Scholar] [CrossRef] [PubMed]
- Desbats, M.A.; Giacomini, I.; Prayer-Galetti, T.; Montopoli, M. Metabolic Plasticity in Chemotherapy Resistance. Front. Oncol. 2020, 10, 281. [Google Scholar] [CrossRef]
- Ehsani, R.; Drabløs, F. Enhanced identification of significant regulators of gene expression. BMC Bioinform. 2020, 21, 1–14. [Google Scholar] [CrossRef] [Green Version]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations. |
© 2021 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).
Share and Cite
Papanikolaou, S.; Vourda, A.; Syggelos, S.; Gyftopoulos, K. Cell Plasticity and Prostate Cancer: The Role of Epithelial–Mesenchymal Transition in Tumor Progression, Invasion, Metastasis and Cancer Therapy Resistance. Cancers 2021, 13, 2795. https://doi.org/10.3390/cancers13112795
Papanikolaou S, Vourda A, Syggelos S, Gyftopoulos K. Cell Plasticity and Prostate Cancer: The Role of Epithelial–Mesenchymal Transition in Tumor Progression, Invasion, Metastasis and Cancer Therapy Resistance. Cancers. 2021; 13(11):2795. https://doi.org/10.3390/cancers13112795
Chicago/Turabian StylePapanikolaou, Sofia, Aikaterini Vourda, Spyros Syggelos, and Kostis Gyftopoulos. 2021. "Cell Plasticity and Prostate Cancer: The Role of Epithelial–Mesenchymal Transition in Tumor Progression, Invasion, Metastasis and Cancer Therapy Resistance" Cancers 13, no. 11: 2795. https://doi.org/10.3390/cancers13112795
APA StylePapanikolaou, S., Vourda, A., Syggelos, S., & Gyftopoulos, K. (2021). Cell Plasticity and Prostate Cancer: The Role of Epithelial–Mesenchymal Transition in Tumor Progression, Invasion, Metastasis and Cancer Therapy Resistance. Cancers, 13(11), 2795. https://doi.org/10.3390/cancers13112795