Non-Coding RNAs in Castration-Resistant Prostate Cancer: Regulation of Androgen Receptor Signaling and Cancer Metabolism
Abstract
:1. Introduction
2. miRNAs in the Androgen Receptor (AR) Signaling Pathway
2.1. Androgen-Regulated miRNAs

2.1.1. miR-32
2.1.2. miR-148a
2.1.3. miR-99a
2.1.4. miR-21
2.1.5. miR-221
2.2. miRNAs Regulating AR Expression (Andro-miRs)
2.2.1. miR-34a
2.2.2. miR-205
2.2.3. miR-488*
2.2.4. miR-124
2.2.5. Let-7c
3. miRNAs in Prostate Cancer (PC) Metabolism

| miRNA | Target Gene | Up/Downregulation in PC | Metabolic Activity/Signaling Pathway | Tissues/Cell Lines | References |
|---|---|---|---|---|---|
| Glucose metabolism | |||||
| miR-1, miR-206 | G6PD, TKT, PGD, GPD2 | ↓ | NRF2 signaling; reprogramming glucose metabolism by directing carbon flux toward the PPP and the TCA cycle | DU145 prostate cancer and A549 lung carcinoma cell lines | [159] |
| Lipid metabolism | |||||
| miR-185,miR-342 | SREBP-1, SREBP-2 | ↓ | SREBP signaling; lipogenesis and cholesterogenesis | LNCaP and C4-2B prostate cancer cells | [160] |
| miR-17/92 | PPARA | ↑ | PPARA signaling; lipogenesis | LNCaP prostate cancer cells | [161] |
| miR101 | COX-2 | ↓ | COX-2/PGE2 pathway | BPH1, LNCaP, and PC3 prostate cancer cells, gastric cancer, endometrial serous adenocarcinoma, colon cancer | [162,163,164,165,166] |
| Glutamine metabolism | |||||
| miR-23a/b | GLS | ↓ | c-Myc signaling; glutamine metabolism | Burkitt lymphoma cells, P-493 and PC3 prostate cancer cells | [167] |
| miR-23b-3p | POX/PRODH | ↑ | c-Myc signaling; proline and glutamine metabolism | Burkitt lymphoma cells, P-493 and PC3 prostate cancer cells | [168] |
| Mitochondrial antioxidant metabolism | |||||
| miR-17-3p | MnSOD, GPX2, TrxR2 | ↓ | Mitochondrial antioxidant | PC3 prostate cancer cells | [169] |
3.1. miRNAs Regulating Glucose Metabolism
3.2. miRNAs Regulating Lipid Metabolism
3.3. miRNAs Regulating Glutamine Metabolism
3.4. miRNAs Regulating Mitochondrial Oxidative Metabolism
4. Long ncRNAs (lncRNAs) in Castration-Resistant Prostate Cancer (CRPC)
| lncRNA | Function | Regulation | Alteration in PC | Oncogene/Tumor Suppressor | Clinical Association | References |
|---|---|---|---|---|---|---|
| ANRIL | Functions in the DNA damage response. Represses INK4a-INK4b-ARF by binding with PRC2. | Upregulated by E2F1 | Overexpression | Oncogene | – | [188] |
| CTBP1-AS | Promotes AR transactivity by repressing the co-repressor CTBP1. Promotes both hormone-dependent and castration-resistant growth. | Androgen responsive | Overexpression | – | – | [189] |
| DRAIC (LOC145837) | Suppresses cellular transformation, migration and invasion. | Repressed by AR | Downregulated in CRPC | Tumor suppressor | Prognostic marker | [190] |
| GAS5 | Induces apoptosis. | Self regulation | Downregulated in CRPC cells | – | – | [191,192] |
| H19 | Suppresses metastasis and cell migration. Enhances stemness by regulating Oct4 and Sox2 expression. | – | Downregulated in metastatic cell lines | Tumor suppressor | – | [193,194] |
| HOTAIR | Promotes cell proliferation, migration, invasion, and survival. | – | Overexpressed in CRPC cells | Oncogene | – | [195,196] |
| Forms a complex with PRC2 and suppresses AR transcription. | ||||||
| Linc00963 | Promotes cell proliferation, migration, and invasion. | – | Overexpressed in CRPC cells | Oncogene | – | [197] |
| MALAT-1 | Promotes cell proliferation, migration, invasion, and survival. | – | Overexpression | Oncogene | Correlates with high Gleason scores, tumor stages, and CRPC | [175,198,199] |
| NEAT1 | Promotes tumor proliferation through the ERα signaling pathway | Upregulated by ERα | Overexpression | Oncogene | Associated with progression | [187] |
| PCA3 | Regulates AR signaling and cell proliferation | Androgen responsive | Overexpression | Oncogene | Diagnostic predictor of malignant patients | [200,201,202] |
| PCAT1 | Represses BRCA2 and homologous recombination, and inhibits DNA repair. Promotes proliferation through stabilization of Myc mRNA. | – | Overexpression | Oncogene | – | [173,183,203] |
| PCAT18 | Promotes cell proliferation, migration and invasion. | Upregulated by AR | Overexpressed in metastatic clinical specimens | Oncogene | – | [204] |
| PCAT29 | Suppresses cell migration and metastasis. | Downregulated by androgen and the AR | Downregulated in CRPC | Tumor suppressor | Low expression correlated with poor prognostic outcomes | [190,205] |
| PCGEM1 | Promotes cell proliferation, migration, invasion, and colony formation. Inhibits doxorubicin-induced apoptosis by attenuating p53 and p21. | Androgen dependent, upregulated by the AR; Regulated by cholesterols | Overexpressing SNP | Oncogene | Associated with high risk | [180,186,206,207,208,209,210,211,212] |
| Enhances ligand-dependent and -independent AR activation through looping. | ||||||
| Regulates tumor metabolism by enhancing Myc transactivity. | ||||||
| PlncRNA-1 | Regulates cell proliferation and apoptosis. Regulates AR mRNA. | Upregulated by the AR | Overexpression | Oncogene | – | [213] |
| PRNCR1 (PCAT8) | Enhances ligand-dependent and -independent AR activation through looping. | – | Overexpressing SNP | Oncogene | – | [184,208] |
| SChLAP1 | Promotes invasion and metastasis. Antagonizes the SWI/SNF complex. | – | Overexpression | Oncogene | Associated with progression and poor outcomes | [214,215] |
| TRPM2-AS | Regulates cellular responses to oxidative stress by controlling TRPM2 expression. | – | Overexpression | – | Associated with poor clinical outcomes and Gleason scores | [216] |
4.1. lncRNAs in AR-Dependent Oncogenicity
4.2. lncRNAs in Metabolic Regulation and Stress Responses
4.3. lncRNAs in Epigenetic Regulation
4.4. lncRNA and miRNA Interplay
5. Clinical Implications of ncRNAs in CRPC
5.1. Diagnostic Biomarkers
| Study (Year) | Sample Type and Size | No. of miRNAs Screened | Key Findings |
|---|---|---|---|
| Diagnostic biomarker | |||
| Mitchell et al. (2008) [227] | Plasma: 25 metastatic PC patients and 25 healthy controls | 6 | miR-141 can distinguish patients with PC from healthy controls. |
| Gonzales et al. (2011) [228] | Plasma: 21 PC patients | 1 | miR-141 was associated with disease progression and changes in PSA. |
| Zhang et al. (2011) [229] | Serum: 50 PC patients (20 localized PC and 30 bone metastasis) and 6 BPH | 1 | miR-141 was associated with bone-metastatic PC. |
| Bryant et al. (2012) [230] | Plasma: 78 PC patients and 28 normal controls | 742 | miR-141 and miR-375 were associated with metastatic PC, as well as recurrent disease. |
| Serum: 47 recurrent and 72 non-recurrent | |||
| Urine: 70 local cancer, 48 advanced cancer and 17 normal controls | Urine levels of miR-107 and miR-574-3p were significantly higher in PC compared to controls. | ||
| Cheng et al. (2013) [231] | Serum: 25 mCRPC patients and 25 healthy controls (cohort 1); 21 mCRPC patients and 20 age-matched healthy controls (cohort 2) | 365 | miR-141, miR-200a, miR-200c, and miR-375 were higher in mCRPC than all healthy controls. |
| Kachakova et al. (2015) [232] | Plasma: 59 PC patients and two groups of controls: 16 BPH and 11 healthy men | 4 | let-7c and miR-30c had decreased expression in PC patients compared to BPH patients. |
| Haldrup et al. (2014) [233] | Serum: 31 PC patients (11 localized, 9 metastasis, and 11 CRPC) and 13 BPH controls | 732 | Identified three miRNA panels for diagnosing and staging of prostate cancer. |
| Kelly et al. (2015) [234] | Whole blood and tissue: 75 PC cancer and 27 BPH | 12 | A panel of miRNAs (miR-141, miR-145, miR-155, and let-7a) was associated with disease progression and was superior to that of PSA. |
| Brasa et al. (2011) [235] | Serum: 7 metastatic and 14 localized PC patients | 668 | miRNA-375 and miRNA-141 can identify patients with significantly higher risk of PC. |
| Tissue: 36 PC tumors and 36 BPH | |||
| Nguyen et al. (2013) [236] | Serum: 28 patients with low-risk localized disease, 30 with high-risk localized disease, and 26 with metastatic CRPC. | 667 | miR-375, miR-378*, and miR-141 were higher in CRPC compared to low-risk localized patients |
| Prognostic biomarkers | |||
| Zhang et al. (2011) | Serum: 56 patients (20 localized PC, 20 androgen-dependent PC, 10 CRPC receiving docetaxel-based chemotherapy, and 6 BPH) | 1 | miR-21 was higher in CRPC patients who were resistant to docetaxel chemotherapy. |
| Li et al. (2014) [237] | Serum/plasma: 97 CRPC patients receiving docetaxel chemotherapy | 46 | Pre-docetaxel miR-200b levels and post-docetaxel changes in miR-20a levels were independent predictors of overall survival of CRPC patients. |
| Selth et al. (2013) [238] | Serum, tissue: 70 BCR and 31 no recurrence PC patients following a radical prostatectomy | 4 | miR-146b-3p and miR-194 were elevated in RP patients who progressed to BCR. |
| Sun et al. (2015) [239] | Serum: 128 PC patients who received an RP and 100 healthy controls | 1 | Low miR-128 expression in both PC tissues and patients’ sera had significantly shorter BCR-free survival. |
5.1.1. miRNA
5.1.2. lncRNA
5.2. Prognostic/Predictive Biomarkers
5.2.1. miRNA
5.2.2. lncRNAs
5.3. Therapeutic Targets
6. Conclusions
Acknowledgments
Author Contributions
Conflicts of Interest
References
- Howlader, N.; Noone, A.M.; Krapcho, M.; Garshell, J.; Miller, D.; Altekruse, S.F.; Kosary, C.L.; Yu, M.; Ruhl, J.; Tatalovich, Z.; et al. Seer Cancer Statistics review, 1975–2012; National Cancer Institute: Bethesda, MD, USA, 2015. [Google Scholar]
- Dickinson, L.; Ahmed, H.U.; Allen, C.; Barentsz, J.O.; Carey, B.; Futterer, J.J.; Heijmink, S.W.; Hoskin, P.J.; Kirkham, A.; Padhani, A.R.; et al. Magnetic resonance imaging for the detection, localisation, and characterisation of prostate cancer: Recommendations from a european consensus meeting. Eur. Urol. 2011, 59, 477–494. [Google Scholar] [CrossRef] [PubMed]
- Rastinehad, A.R.; Turkbey, B.; Salami, S.S.; Yaskiv, O.; George, A.K.; Fakhoury, M.; Beecher, K.; Vira, M.A.; Kavoussi, L.R.; Siegel, D.N.; et al. Improving detection of clinically significant prostate cancer: Magnetic resonance imaging/transrectal ultrasound fusion guided prostate biopsy. J. Urol. 2014, 191, 1749–1754. [Google Scholar] [CrossRef] [PubMed]
- Welch, H.G.; Albertsen, P.C. Prostate cancer diagnosis and treatment after the introduction of prostate-specific antigen screening: 1986–2005. J. Natl. Cancer Inst. 2009, 101, 1325–1329. [Google Scholar] [CrossRef] [PubMed]
- Hayes, J.H.; Barry, M.J. Screening for prostate cancer with the prostate-specific antigen test: A review of current evidence. JAMA 2014, 311, 1143–1149. [Google Scholar] [CrossRef] [PubMed]
- McLeod, D.G. Success and failure of single-modality treatment for early prostate cancer. Rev. Urol. 2004, 6 (Suppl. S2), S13–S19. [Google Scholar] [PubMed]
- Thompson, I.; Thrasher, J.B.; Aus, G.; Burnett, A.L.; Canby-Hagino, E.D.; Cookson, M.S.; D’Amico, A.V.; Dmochowski, R.R.; Eton, D.T.; Forman, J.D.; et al. Guideline for the management of clinically localized prostate cancer: 2007 update. J. Urol. 2007, 177, 2106–2131. [Google Scholar] [CrossRef] [PubMed]
- Klotz, L.; Emberton, M. Management of low risk prostate cancer-active surveillance and focal therapy. Nat. Rev. Clin. Oncol. 2014, 11, 324–334. [Google Scholar] [CrossRef] [PubMed]
- Shen, M.M.; Abate-Shen, C. Molecular genetics of prostate cancer: New prospects for old challenges. Genes Dev. 2010, 24, 1967–2000. [Google Scholar] [CrossRef] [PubMed]
- McMurtry, C.T.; McMurtry, J.M. Metastatic prostate cancer: Complications and treatment. J. Am. Geriatr. Soc. 2003, 51, 1136–1142. [Google Scholar] [CrossRef] [PubMed]
- Villavicencio, H. Quality of life of patients with advanced and metastatic prostatic carcinoma. Eur. Urol. 1993, 24 (Suppl. S2), 118–121. [Google Scholar] [PubMed]
- Petrylak, D.P.; Tangen, C.M.; Hussain, M.H.; Lara, P.N., Jr.; Jones, J.A.; Taplin, M.E.; Burch, P.A.; Berry, D.; Moinpour, C.; Kohli, M.; et al. Docetaxel and estramustine compared with mitoxantrone and prednisone for advanced refractory prostate cancer. N. Engl. J. Med. 2004, 351, 1513–1520. [Google Scholar] [CrossRef] [PubMed]
- Tannock, I.F.; de Wit, R.; Berry, W.R.; Horti, J.; Pluzanska, A.; Chi, K.N.; Oudard, S.; Theodore, C.; James, N.D.; Turesson, I.; et al. Docetaxel plus prednisone or mitoxantrone plus prednisone for advanced prostate cancer. N. Engl. J. Med. 2004, 351, 1502–1512. [Google Scholar] [CrossRef] [PubMed]
- Berthold, D.R.; Pond, G.R.; Soban, F.; de Wit, R.; Eisenberger, M.; Tannock, I.F. Docetaxel plus prednisone or mitoxantrone plus prednisone for advanced prostate cancer: Updated survival in the tax 327 study. J. Clin. Oncol. Off. J. Am. Soc. Clin. Oncol. 2008, 26, 242–245. [Google Scholar] [CrossRef] [PubMed]
- Omlin, A.; Pezaro, C.; Gillessen Sommer, S. Sequential use of novel therapeutics in advanced prostate cancer following docetaxel chemotherapy. Ther. Adv. Urol. 2014, 6, 3–14. [Google Scholar] [CrossRef] [PubMed]
- Karantanos, T.; Corn, P.G.; Thompson, T.C. Prostate cancer progression after androgen deprivation therapy: Mechanisms of castrate resistance and novel therapeutic approaches. Oncogene 2013, 32, 5501–5511. [Google Scholar] [CrossRef] [PubMed]
- Consortium, E.P.; Birney, E.; Stamatoyannopoulos, J.A.; Dutta, A.; Guigo, R.; Gingeras, T.R.; Margulies, E.H.; Weng, Z.; Snyder, M.; Dermitzakis, E.T.; et al. Identification and analysis of functional elements in 1% of the human genome by the encode pilot project. Nature 2007, 447, 799–816. [Google Scholar]
- Carninci, P.; Kasukawa, T.; Katayama, S.; Gough, J.; Frith, M.C.; Maeda, N.; Oyama, R.; Ravasi, T.; Lenhard, B.; Wells, C.; et al. The transcriptional landscape of the mammalian genome. Science 2005, 309, 1559–1563. [Google Scholar] [PubMed]
- Ponting, C.P.; Oliver, P.L.; Reik, W. Evolution and functions of long noncoding RNAs. Cell 2009, 136, 629–641. [Google Scholar] [CrossRef] [PubMed]
- Mattick, J.S.; Makunin, I.V. Non-coding RNA. Hum. Mol. Genet. 2006, 15, R17–R29. [Google Scholar] [CrossRef] [PubMed]
- Mattick, J.S. The central role of RNA in human development and cognition. FEBS Lett. 2011, 585, 1600–1616. [Google Scholar] [CrossRef] [PubMed]
- Kapranov, P.; Willingham, A.T.; Gingeras, T.R. Genome-wide transcription and the implications for genomic organization. Nat. Rev. Genet. 2007, 8, 413–423. [Google Scholar] [CrossRef] [PubMed]
- Keller, C.; Buhler, M. Chromatin-associated ncRNA activities. Chromosome Res. 2013, 21, 627–641. [Google Scholar] [CrossRef] [PubMed][Green Version]
- Luco, R.F.; Misteli, T. More than a splicing code: Integrating the role of RNA, chromatin and non-coding RNA in alternative splicing regulation. Curr. Opin. Genet. Dev. 2011, 21, 366–372. [Google Scholar] [CrossRef] [PubMed]
- Pircher, A.; Gebetsberger, J.; Polacek, N. Ribosome-associated ncRNAs: An emerging class of translation regulators. RNA Biol. 2014, 11, 1335–1339. [Google Scholar] [CrossRef] [PubMed]
- Hannon, G.J. RNA interference. Nature 2002, 418, 244–251. [Google Scholar] [CrossRef] [PubMed]
- Lewis, B.P.; Burge, C.B.; Bartel, D.P. Conserved seed pairing, often flanked by adenosines, indicates that thousands of human genes are microRNA targets. Cell 2005, 120, 15–20. [Google Scholar] [CrossRef] [PubMed]
- Brennecke, J.; Stark, A.; Russell, R.B.; Cohen, S.M. Principles of microRNA-target recognition. PLoS Biol. 2005, 3, e85. [Google Scholar] [CrossRef] [PubMed]
- Lagos-Quintana, M.; Rauhut, R.; Lendeckel, W.; Tuschl, T. Identification of novel genes coding for small expressed RNAs. Science 2001, 294, 853–858. [Google Scholar] [CrossRef] [PubMed]
- Stefani, G.; Slack, F.J. Small non-coding RNAs in animal development. Nat. Rev. Mol. Cell Biol. 2008, 9, 219–230. [Google Scholar] [CrossRef] [PubMed]
- Pasquinelli, A.E. MicroRNAs and their targets: Recognition, regulation and an emerging reciprocal relationship. Nat. Rev. Genet. 2012, 13, 271–282. [Google Scholar] [CrossRef] [PubMed]
- Derrien, T.; Johnson, R.; Bussotti, G.; Tanzer, A.; Djebali, S.; Tilgner, H.; Guernec, G.; Martin, D.; Merkel, A.; Knowles, D.G.; et al. The gencode V7 catalog of human long noncoding RNAs: Analysis of their gene structure, evolution, and expression. Genome Res. 2012, 22, 1775–1789. [Google Scholar] [CrossRef] [PubMed]
- Pang, K.C.; Frith, M.C.; Mattick, J.S. Rapid evolution of noncoding RNAs: Lack of conservation does not mean lack of function. Trends Genet. 2006, 22, 1–5. [Google Scholar] [CrossRef] [PubMed]
- Gibb, E.A.; Brown, C.J.; Lam, W.L. The functional role of long non-coding RNA in human carcinomas. Mol. Cancer 2011, 10, 38. [Google Scholar] [CrossRef] [PubMed]
- Geisler, S.; Coller, J. RNA in unexpected places: Long non-coding RNA functions in diverse cellular contexts. Nat. Rev. Mol. Cell Biol. 2013, 14, 699–712. [Google Scholar] [CrossRef] [PubMed]
- Spizzo, R.; Almeida, M.I.; Colombatti, A.; Calin, G.A. Long non-coding RNAs and cancer: A new frontier of translational research? Oncogene 2012, 31, 4577–4587. [Google Scholar] [CrossRef] [PubMed]
- Tsai, M.C.; Spitale, R.C.; Chang, H.Y. Long intergenic noncoding RNAs: New links in cancer progression. Cancer Res. 2011, 71, 3–7. [Google Scholar] [CrossRef] [PubMed]
- Heemers, H.; Maes, B.; Foufelle, F.; Heyns, W.; Verhoeven, G.; Swinnen, J.V. Androgens stimulate lipogenic gene expression in prostate cancer cells by activation of the sterol regulatory element-binding protein cleavage activating protein/sterol regulatory element-binding protein pathway. Mol. Endocrinol. 2001, 15, 1817–1828. [Google Scholar] [CrossRef] [PubMed]
- Heemers, H.; Verrijdt, G.; Organe, S.; Claessens, F.; Heyns, W.; Verhoeven, G.; Swinnen, J.V. Identification of an androgen response element in intron 8 of the sterol regulatory element-binding protein cleavage-activating protein gene allowing direct regulation by the androgen receptor. J. Biol. Chem. 2004, 279, 30880–30887. [Google Scholar] [CrossRef] [PubMed]
- Massie, C.E.; Lynch, A.; Ramos-Montoya, A.; Boren, J.; Stark, R.; Fazli, L.; Warren, A.; Scott, H.; Madhu, B.; Sharma, N.; et al. The androgen receptor fuels prostate cancer by regulating central metabolism and biosynthesis. EMBO J. 2011, 30, 2719–2733. [Google Scholar] [CrossRef] [PubMed]
- Tennakoon, J.B.; Shi, Y.; Han, J.J.; Tsouko, E.; White, M.A.; Burns, A.R.; Zhang, A.; Xia, X.; Ilkayeva, O.R.; Xin, L.; et al. Androgens regulate prostate cancer cell growth via an ampk-PGC-1α-mediated metabolic switch. Oncogene 2014, 33, 5251–5261. [Google Scholar] [CrossRef] [PubMed]
- Tsouko, E.; Khan, A.S.; White, M.A.; Han, J.J.; Shi, Y.; Merchant, F.A.; Sharpe, M.A.; Xin, L.; Frigo, D.E. Regulation of the pentose phosphate pathway by an androgen receptor-mTOR-mediated mechanism and its role in prostate cancer cell growth. Oncogenesis 2014, 3, e103. [Google Scholar] [CrossRef] [PubMed]
- Mehraein-Ghomi, F.; Basu, H.S.; Church, D.R.; Hoffmann, F.M.; Wilding, G. Androgen receptor requires jund as a coactivator to switch on an oxidative stress generation pathway in prostate cancer cells. Cancer Res. 2010, 70, 4560–4568. [Google Scholar] [CrossRef] [PubMed]
- Hanahan, D.; Weinberg, R.A. Hallmarks of cancer: The next generation. Cell 2011, 144, 646–674. [Google Scholar] [CrossRef] [PubMed]
- Oyama, N.; Akino, H.; Suzuki, Y.; Kanamaru, H.; Ishida, H.; Tanase, K.; Sadato, N.; Yonekura, Y.; Okada, K. FDG pet for evaluating the change of glucose metabolism in prostate cancer after androgen ablation. Nucl. Med. Commun. 2001, 22, 963–969. [Google Scholar] [CrossRef] [PubMed]
- Cui, Y.; Nadiminty, N.; Liu, C.; Lou, W.; Schwartz, C.T.; Gao, A.C. Upregulation of glucose metabolism by NF-κB2/p52 mediates enzalutamide resistance in castration-resistant prostate cancer cells. Endocr. Relat. Cancer 2014, 21, 435–442. [Google Scholar] [CrossRef] [PubMed]
- Wang, Q.; Tiffen, J.; Bailey, C.G.; Lehman, M.L.; Ritchie, W.; Fazli, L.; Metierre, C.; Feng, Y.J.; Li, E.; Gleave, M.; et al. Targeting amino acid transport in metastatic castration-resistant prostate cancer: Effects on cell cycle, cell growth, and tumor development. J. Natl. Cancer Inst. 2013, 105, 1463–1473. [Google Scholar] [CrossRef] [PubMed]
- Sharma, N.L.; Massie, C.E.; Ramos-Montoya, A.; Zecchini, V.; Scott, H.E.; Lamb, A.D.; MacArthur, S.; Stark, R.; Warren, A.Y.; Mills, I.G.; et al. The androgen receptor induces a distinct transcriptional program in castration-resistant prostate cancer in man. Cancer Cell 2013, 23, 35–47. [Google Scholar] [CrossRef] [PubMed]
- Koivisto, P.; Kononen, J.; Palmberg, C.; Tammela, T.; Hyytinen, E.; Isola, J.; Trapman, J.; Cleutjens, K.; Noordzij, A.; Visakorpi, T.; et al. Androgen receptor gene amplification: A possible molecular mechanism for androgen deprivation therapy failure in prostate cancer. Cancer Res. 1997, 57, 314–319. [Google Scholar] [PubMed]
- Edwards, J.; Krishna, N.S.; Witton, C.J.; Bartlett, J.M. Gene amplifications associated with the development of hormone-resistant prostate cancer. Clin. Cancer Res. 2003, 9, 5271–5281. [Google Scholar] [PubMed]
- Culig, Z.; Hobisch, A.; Cronauer, M.V.; Cato, A.C.; Hittmair, A.; Radmayr, C.; Eberle, J.; Bartsch, G.; Klocker, H. Mutant androgen receptor detected in an advanced-stage prostatic carcinoma is activated by adrenal androgens and progesterone. Mol. Endocrinol. 1993, 7, 1541–1550. [Google Scholar] [PubMed]
- Buchanan, G.; Greenberg, N.M.; Scher, H.I.; Harris, J.M.; Marshall, V.R.; Tilley, W.D. Collocation of androgen receptor gene mutations in prostate cancer. Clin. Cancer Res. 2001, 7, 1273–1281. [Google Scholar] [PubMed]
- Steketee, K.; Timmerman, L.; Ziel-van der Made, A.C.; Doesburg, P.; Brinkmann, A.O.; Trapman, J. Broadened ligand responsiveness of androgen receptor mutants obtained by random amino acid substitution of H874 and mutation hot spot T877 in prostate cancer. Int. J. Cancer 2002, 100, 309–317. [Google Scholar] [CrossRef] [PubMed]
- Chang, K.H.; Li, R.; Papari-Zareei, M.; Watumull, L.; Zhao, Y.D.; Auchus, R.J.; Sharifi, N. Dihydrotestosterone synthesis bypasses testosterone to drive castration-resistant prostate cancer. Proc. Natl. Acad. Sci. USA 2011, 108, 13728–13733. [Google Scholar] [CrossRef] [PubMed]
- Gao, J.; Arnold, J.T.; Isaacs, J.T. Conversion from a paracrine to an autocrine mechanism of androgen-stimulated growth during malignant transformation of prostatic epithelial cells. Cancer Res. 2001, 61, 5038–5044. [Google Scholar] [PubMed]
- Hofland, J.; van Weerden, W.M.; Dits, N.F.; Steenbergen, J.; van Leenders, G.J.; Jenster, G.; Schroder, F.H.; de Jong, F.H. Evidence of limited contributions for intratumoral steroidogenesis in prostate cancer. Cancer Res. 2010, 70, 1256–1264. [Google Scholar] [CrossRef] [PubMed]
- Dehm, S.M.; Schmidt, L.J.; Heemers, H.V.; Vessella, R.L.; Tindall, D.J. Splicing of a novel androgen receptor exon generates a constitutively active androgen receptor that mediates prostate cancer therapy resistance. Cancer Res. 2008, 68, 5469–5477. [Google Scholar] [CrossRef] [PubMed]
- Guo, Z.; Yang, X.; Sun, F.; Jiang, R.; Linn, D.E.; Chen, H.; Chen, H.; Kong, X.; Melamed, J.; Tepper, C.G.; et al. A novel androgen receptor splice variant is up-regulated during prostate cancer progression and promotes androgen depletion-resistant growth. Cancer Res. 2009, 69, 2305–2313. [Google Scholar] [CrossRef] [PubMed]
- Hornberg, E.; Ylitalo, E.B.; Crnalic, S.; Antti, H.; Stattin, P.; Widmark, A.; Bergh, A.; Wikstrom, P. Expression of androgen receptor splice variants in prostate cancer bone metastases is associated with castration-resistance and short survival. PLoS ONE 2011, 6, e19059. [Google Scholar] [CrossRef] [PubMed]
- Hu, R.; Dunn, T.A.; Wei, S.; Isharwal, S.; Veltri, R.W.; Humphreys, E.; Han, M.; Partin, A.W.; Vessella, R.L.; Isaacs, W.B.; et al. Ligand-independent androgen receptor variants derived from splicing of cryptic exons signify hormone-refractory prostate cancer. Cancer Res. 2009, 69, 16–22. [Google Scholar] [CrossRef] [PubMed]
- Sun, S.; Sprenger, C.C.; Vessella, R.L.; Haugk, K.; Soriano, K.; Mostaghel, E.A.; Page, S.T.; Coleman, I.M.; Nguyen, H.M.; Sun, H.; et al. Castration resistance in human prostate cancer is conferred by a frequently occurring androgen receptor splice variant. J. Clin. Investig. 2010, 120, 2715–2730. [Google Scholar] [CrossRef] [PubMed]
- Fujimoto, N.; Mizokami, A.; Harada, S.; Matsumoto, T. Different expression of androgen receptor coactivators in human prostate. Urology 2001, 58, 289–294. [Google Scholar] [CrossRef]
- Zou, J.X.; Guo, L.; Revenko, A.S.; Tepper, C.G.; Gemo, A.T.; Kung, H.J.; Chen, H.W. Androgen-induced coactivator ancca mediates specific androgen receptor signaling in prostate cancer. Cancer Res. 2009, 69, 3339–3346. [Google Scholar] [CrossRef] [PubMed]
- Dai, B.; Chen, H.; Guo, S.; Yang, X.; Linn, D.E.; Sun, F.; Li, W.; Guo, Z.; Xu, K.; Kim, O.; et al. Compensatory upregulation of tyrosine kinase ETK/BMX in response to androgen deprivation promotes castration-resistant growth of prostate cancer cells. Cancer Res. 2010, 70, 5587–5596. [Google Scholar] [CrossRef] [PubMed]
- Mahajan, K.; Challa, S.; Coppola, D.; Lawrence, H.; Luo, Y.; Gevariya, H.; Zhu, W.; Chen, Y.A.; Lawrence, N.J.; Mahajan, N.P. Effect of ACK1 tyrosine kinase inhibitor on ligand-independent androgen receptor activity. Prostate 2010, 70, 1274–1285. [Google Scholar] [CrossRef] [PubMed]
- McCall, P.; Adams, C.E.; Willder, J.M.; Bennett, L.; Qayyum, T.; Orange, C.; Underwood, M.A.; Edwards, J. Androgen receptor phosphorylation at serine 308 and serine 791 predicts enhanced survival in castrate resistant prostate cancer patients. Int. J. Mol. Sci. 2013, 14, 16656–16671. [Google Scholar] [CrossRef] [PubMed]
- McCall, P.; Gemmell, L.K.; Mukherjee, R.; Bartlett, J.M.; Edwards, J. Phosphorylation of the androgen receptor is associated with reduced survival in hormone-refractory prostate cancer patients. Br. J. Cancer 2008, 98, 1094–1101. [Google Scholar] [CrossRef] [PubMed]
- Culig, Z.; Hobisch, A.; Cronauer, M.V.; Radmayr, C.; Trapman, J.; Hittmair, A.; Bartsch, G.; Klocker, H. Androgen receptor activation in prostatic tumor cell lines by insulin-like growth factor-I, keratinocyte growth factor, and epidermal growth factor. Cancer Res. 1994, 54, 5474–5478. [Google Scholar] [PubMed]
- Mellinghoff, I.K.; Vivanco, I.; Kwon, A.; Tran, C.; Wongvipat, J.; Sawyers, C.L. HER2/NEU kinase-dependent modulation of androgen receptor function through effects on DNA binding and stability. Cancer Cell 2004, 6, 517–527. [Google Scholar] [CrossRef] [PubMed]
- Mahajan, N.P.; Liu, Y.; Majumder, S.; Warren, M.R.; Parker, C.E.; Mohler, J.L.; Earp, H.S.; Whang, Y.E. Activated CDC42-associated kinase ACK1 promotes prostate cancer progression via androgen receptor tyrosine phosphorylation. Proc. Natl. Acad. Sci. USA 2007, 104, 8438–8443. [Google Scholar] [CrossRef] [PubMed]
- Kraus, S.; Gioeli, D.; Vomastek, T.; Gordon, V.; Weber, M.J. Receptor for activated C Kinase 1 (RACK1) and SRC regulate the tyrosine phosphorylation and function of the androgen receptor. Cancer Res. 2006, 66, 11047–11054. [Google Scholar] [CrossRef] [PubMed]
- Catto, J.W.; Alcaraz, A.; Bjartell, A.S.; de Vere White, R.; Evans, C.P.; Fussel, S.; Hamdy, F.C.; Kallioniemi, O.; Mengual, L.; Schlomm, T.; et al. MicroRNA in prostate, bladder, and kidney cancer: A systematic review. Eur. Urol. 2011, 59, 671–681. [Google Scholar] [CrossRef] [PubMed]
- Coppola, V.; de Maria, R.; Bonci, D. MicroRNAs and prostate cancer. Endocr. Relat. Cancer 2010, 17, F1–F17. [Google Scholar] [CrossRef] [PubMed]
- Pang, M.; Xing, C.; Adams, N.; Rodriguez-Uribe, L.; Hughs, S.E.; Hanson, S.F.; Zhang, J. Comparative expression of miRNA genes and miRNA-based AFLP marker analysis in cultivated tetraploid cottons. J. Plant Physiol. 2011, 168, 824–830. [Google Scholar] [CrossRef] [PubMed]
- Sevli, S.; Uzumcu, A.; Solak, M.; Ittmann, M.; Ozen, M. The function of microRNAs, small but potent molecules, in human prostate cancer. Prostate Cancer Prostatic Dis. 2010, 13, 208–217. [Google Scholar] [CrossRef] [PubMed]
- Wang, Y.L.; Wu, S.; Jiang, B.; Yin, F.F.; Zheng, S.S.; Hou, S.C. Role of microRNAs in prostate cancer pathogenesis. Clin. Genitourin. Cancer 2015, 13, 261–270. [Google Scholar] [CrossRef] [PubMed]
- Jalava, S.E.; Urbanucci, A.; Latonen, L.; Waltering, K.K.; Sahu, B.; Janne, O.A.; Seppala, J.; Lahdesmaki, H.; Tammela, T.L.; Visakorpi, T. Androgen-regulated miR-32 targets BTG2 and is overexpressed in castration-resistant prostate cancer. Oncogene 2012, 31, 4460–4471. [Google Scholar] [CrossRef] [PubMed]
- Ambs, S.; Prueitt, R.L.; Yi, M.; Hudson, R.S.; Howe, T.M.; Petrocca, F.; Wallace, T.A.; Liu, C.G.; Volinia, S.; Calin, G.A.; et al. Genomic profiling of microRNA and messenger RNA reveals deregulated microRNA expression in prostate cancer. Cancer Res. 2008, 68, 6162–6170. [Google Scholar] [CrossRef] [PubMed]
- Martens-Uzunova, E.S.; Jalava, S.E.; Dits, N.F.; van Leenders, G.J.; Moller, S.; Trapman, J.; Bangma, C.H.; Litman, T.; Visakorpi, T.; Jenster, G. Diagnostic and prognostic signatures from the small non-coding RNA transcriptome in prostate cancer. Oncogene 2012, 31, 978–991. [Google Scholar] [CrossRef] [PubMed]
- Mao, B.; Zhang, Z.; Wang, G. BTG2: A rising star of tumor suppressors (review). Int. J. Oncol. 2015, 46, 459–464. [Google Scholar] [CrossRef] [PubMed]
- Ficazzola, M.A.; Fraiman, M.; Gitlin, J.; Woo, K.; Melamed, J.; Rubin, M.A.; Walden, P.D. Antiproliferative B cell translocation gene 2 protein is down-regulated post-transcriptionally as an early event in prostate carcinogenesis. Carcinogenesis 2001, 22, 1271–1279. [Google Scholar] [CrossRef] [PubMed]
- Coppola, V.; Musumeci, M.; Patrizii, M.; Cannistraci, A.; Addario, A.; Maugeri-Sacca, M.; Biffoni, M.; Francescangeli, F.; Cordenonsi, M.; Piccolo, S.; et al. BTG2 loss and miR-21 upregulation contribute to prostate cell transformation by inducing luminal markers expression and epithelial-mesenchymal transition. Oncogene 2013, 32, 1843–1853. [Google Scholar] [CrossRef] [PubMed]
- Hu, X.D.; Meng, Q.H.; Xu, J.Y.; Jiao, Y.; Ge, C.M.; Jacob, A.; Wang, P.; Rosen, E.M.; Fan, S. BTG2 is an lxxll-dependent co-repressor for androgen receptor transcriptional activity. Biochem. Biophys. Res. Commun. 2011, 404, 903–909. [Google Scholar] [CrossRef] [PubMed]
- Volinia, S.; Calin, G.A.; Liu, C.G.; Ambs, S.; Cimmino, A.; Petrocca, F.; Visone, R.; Iorio, M.; Roldo, C.; Ferracin, M.; et al. A microRNA expression signature of human solid tumors defines cancer gene targets. Proc. Natl. Acad. Sci. USA 2006, 103, 2257–2261. [Google Scholar] [CrossRef] [PubMed]
- Murata, T.; Takayama, K.; Katayama, S.; Urano, T.; Horie-Inoue, K.; Ikeda, K.; Takahashi, S.; Kawazu, C.; Hasegawa, A.; Ouchi, Y.; et al. miR-148a is an androgen-responsive microRNA that promotes LNCaP prostate cell growth by repressing its target CAND1 expression. Prostate Cancer Prostatic Dis. 2010, 13, 356–361. [Google Scholar] [CrossRef] [PubMed]
- Fujita, Y.; Kojima, K.; Ohhashi, R.; Hamada, N.; Nozawa, Y.; Kitamoto, A.; Sato, A.; Kondo, S.; Kojima, T.; Deguchi, T.; et al. miR-148a attenuates paclitaxel resistance of hormone-refractory, drug-resistant prostate cancer PC3 cells by regulating MSK1 expression. J. Biol. Chem. 2010, 285, 19076–19084. [Google Scholar] [CrossRef] [PubMed]
- Deak, M.; Clifton, A.D.; Lucocq, L.M.; Alessi, D.R. Mitogen- and stress-activated protein Kinase-1 (MSK1) is directly activated by mapk and SAPK2/p38, and may mediate activation of creb. EMBO J. 1998, 17, 4426–4441. [Google Scholar] [CrossRef] [PubMed]
- Wiggin, G.R.; Soloaga, A.; Foster, J.M.; Murray-Tait, V.; Cohen, P.; Arthur, J.S. MSK1 and MSK2 are required for the mitogen- and stress-induced phosphorylation of creb and atf1 in fibroblasts. Mol. Cell. Biol. 2002, 22, 2871–2881. [Google Scholar] [CrossRef] [PubMed]
- Sun, D.; Layer, R.; Mueller, A.C.; Cichewicz, M.A.; Negishi, M.; Paschal, B.M.; Dutta, A. Regulation of several androgen-induced genes through the repression of the miR-99a/let-7c/miR-125B-2 miRNA cluster in prostate cancer cells. Oncogene 2014, 33, 1448–1457. [Google Scholar] [CrossRef] [PubMed]
- Sun, D.; Lee, Y.S.; Malhotra, A.; Kim, H.K.; Matecic, M.; Evans, C.; Jensen, R.V.; Moskaluk, C.A.; Dutta, A. miR-99 family of microRNAs suppresses the expression of prostate-specific antigen and prostate cancer cell proliferation. Cancer Res. 2011, 71, 1313–1324. [Google Scholar] [CrossRef] [PubMed]
- Ozkan, E.E. Plasma and tissue insulin-like growth factor-I receptor (IGF-IR) as a prognostic marker for prostate cancer and anti-IGF-IR agents as novel therapeutic strategy for refractory cases: A review. Mol. Cell. Endocrinol. 2011, 344, 1–24. [Google Scholar] [CrossRef] [PubMed]
- Chen, H.X.; Sharon, E. IGF-IR as an anti-cancer target—Trials and tribulations. Chin. J. Cancer 2013, 32, 242–252. [Google Scholar] [CrossRef] [PubMed]
- Wu, J.; Yu, E. Insulin-like growth factor receptor-1 (IGF-IR) as a target for prostate cancer therapy. Cancer Metastasis Rev. 2014, 33, 607–617. [Google Scholar] [CrossRef] [PubMed]
- Edlind, M.P.; Hsieh, A.C. PI3K-AKT-mTOR signaling in prostate cancer progression and androgen deprivation therapy resistance. Asian J. Androl. 2014, 16, 378–386. [Google Scholar] [PubMed]
- Zhang, W.; Zhu, J.; Efferson, C.L.; Ware, C.; Tammam, J.; Angagaw, M.; Laskey, J.; Bettano, K.A.; Kasibhatla, S.; Reilly, J.F.; et al. Inhibition of tumor growth progression by antiandrogens and mTOR inhibitor in a pten-deficient mouse model of prostate cancer. Cancer Res. 2009, 69, 7466–7472. [Google Scholar] [CrossRef] [PubMed]
- Andersen, C.L.; Monni, O.; Wagner, U.; Kononen, J.; Barlund, M.; Bucher, C.; Haas, P.; Nocito, A.; Bissig, H.; Sauter, G.; et al. High-throughput copy number analysis of 17q23 in 3520 tissue specimens by fluorescence in situ hybridization to tissue microarrays. Am. J. Pathol. 2002, 161, 73–79. [Google Scholar] [CrossRef]
- Kasahara, K.; Taguchi, T.; Yamasaki, I.; Kamada, M.; Yuri, K.; Shuin, T. Detection of genetic alterations in advanced prostate cancer by comparative genomic hybridization. Cancer Genet. Cytogenet. 2002, 137, 59–63. [Google Scholar] [CrossRef]
- Ribas, J.; Ni, X.; Haffner, M.; Wentzel, E.A.; Salmasi, A.H.; Chowdhury, W.H.; Kudrolli, T.A.; Yegnasubramanian, S.; Luo, J.; Rodriguez, R.; et al. miR-21: An androgen receptor-regulated microRNA that promotes hormone-dependent and hormone-independent prostate cancer growth. Cancer Res. 2009, 69, 7165–7169. [Google Scholar] [CrossRef] [PubMed]
- Li, T.; Li, D.; Sha, J.; Sun, P.; Huang, Y. MicroRNA-21 directly targets marcks and promotes apoptosis resistance and invasion in prostate cancer cells. Biochem. Biophys. Res. Commun. 2009, 383, 280–285. [Google Scholar] [CrossRef] [PubMed]
- Fujita, S.; Ito, T.; Mizutani, T.; Minoguchi, S.; Yamamichi, N.; Sakurai, K.; Iba, H. miR-21 gene expression triggered by AP-1 is sustained through a double-negative feedback mechanism. J. Mol. Biol. 2008, 378, 492–504. [Google Scholar] [CrossRef] [PubMed]
- Iliopoulos, D.; Jaeger, S.A.; Hirsch, H.A.; Bulyk, M.L.; Struhl, K. STAT3 activation of miR-21 and miR-181B-1 via PTEN and CYLD are part of the epigenetic switch linking inflammation to cancer. Mol. Cell 2010, 39, 493–506. [Google Scholar] [CrossRef] [PubMed]
- Ouyang, X.; Jessen, W.J.; Al-Ahmadie, H.; Serio, A.M.; Lin, Y.; Shih, W.J.; Reuter, V.E.; Scardino, P.T.; Shen, M.M.; Aronow, B.J.; et al. Activator protein-1 transcription factors are associated with progression and recurrence of prostate cancer. Cancer Res. 2008, 68, 2132–2144. [Google Scholar] [CrossRef] [PubMed]
- Bishop, J.L.; Thaper, D.; Zoubeidi, A. The multifaceted roles of STAT3 signaling in the progression of prostate cancer. Cancers 2014, 6, 829–859. [Google Scholar] [CrossRef] [PubMed]
- Li, T.; Li, R.S.; Li, Y.H.; Zhong, S.; Chen, Y.Y.; Zhang, C.M.; Hu, M.M.; Shen, Z.J. miR-21 as an independent biochemical recurrence predictor and potential therapeutic target for prostate cancer. J. Urol. 2012, 187, 1466–1472. [Google Scholar] [CrossRef] [PubMed]
- Shen, J.; Hruby, G.W.; McKiernan, J.M.; Gurvich, I.; Lipsky, M.J.; Benson, M.C.; Santella, R.M. Dysregulation of circulating microRNAs and prediction of aggressive prostate cancer. Prostate 2012, 72, 1469–1477. [Google Scholar] [CrossRef] [PubMed]
- Zhang, H.L.; Yang, L.F.; Zhu, Y.; Yao, X.D.; Zhang, S.L.; Dai, B.; Zhu, Y.P.; Shen, Y.J.; Shi, G.H.; Ye, D.W. Serum miRNA-21: Elevated levels in patients with metastatic hormone-refractory prostate cancer and potential predictive factor for the efficacy of docetaxel-based chemotherapy. Prostate 2011, 71, 326–331. [Google Scholar] [CrossRef] [PubMed]
- Buscaglia, L.E.; Li, Y. Apoptosis and the target genes of microRNA-21. Chin. J. Cancer 2011, 30, 371–380. [Google Scholar] [CrossRef] [PubMed]
- Spahn, M.; Kneitz, S.; Scholz, C.J.; Stenger, N.; Rudiger, T.; Strobel, P.; Riedmiller, H.; Kneitz, B. Expression of microRNA-221 is progressively reduced in aggressive prostate cancer and metastasis and predicts clinical recurrence. Int. J. Cancer 2010, 127, 394–403. [Google Scholar] [PubMed]
- Gordanpour, A.; Stanimirovic, A.; Nam, R.K.; Moreno, C.S.; Sherman, C.; Sugar, L.; Seth, A. miR-221 is down-regulated in TMPRSS2: Erg fusion-positive prostate cancer. Anticancer Res. 2011, 31, 403–410. [Google Scholar] [PubMed]
- Sun, T.; Yang, M.; Chen, S.; Balk, S.; Pomerantz, M.; Hsieh, C.L.; Brown, M.; Lee, G.S.; Kantoff, P.W. The altered expression of miR-221/-222 and miR-23b/-27b is associated with the development of human castration resistant prostate cancer. Prostate 2012, 72, 1093–1103. [Google Scholar] [CrossRef] [PubMed]
- Mercatelli, N.; Coppola, V.; Bonci, D.; Miele, F.; Costantini, A.; Guadagnoli, M.; Bonanno, E.; Muto, G.; Frajese, G.V.; De Maria, R.; et al. The inhibition of the highly expressed miR-221 and miR-222 impairs the growth of prostate carcinoma xenografts in mice. PLoS ONE 2008, 3, e4029. [Google Scholar] [CrossRef] [PubMed]
- Yang, X.; Yang, Y.; Gan, R.; Zhao, L.; Li, W.; Zhou, H.; Wang, X.; Lu, J.; Meng, Q.H. Down-regulation of miR-221 and miR-222 restrain prostate cancer cell proliferation and migration that is partly mediated by activation of SIRT1. PLoS ONE 2014, 9, e98833. [Google Scholar] [CrossRef] [PubMed]
- Sun, T.; Wang, Q.; Balk, S.; Brown, M.; Lee, G.S.; Kantoff, P. The role of microRNA-221 and microRNA-222 in androgen-independent prostate cancer cell lines. Cancer Res. 2009, 69, 3356–3363. [Google Scholar] [CrossRef] [PubMed]
- Sun, T.; Wang, X.; He, H.H.; Sweeney, C.J.; Liu, S.X.; Brown, M.; Balk, S.; Lee, G.S.; Kantoff, P.W. miR-221 promotes the development of androgen independence in prostate cancer cells via downregulation of HECTD2 and RAB1A. Oncogene 2014, 33, 2790–2800. [Google Scholar] [CrossRef] [PubMed]
- Zheng, C.; Yinghao, S.; Li, J. miR-221 expression affects invasion potential of human prostate carcinoma cell lines by targeting DVL2. Med. Oncol. 2012, 29, 815–822. [Google Scholar] [CrossRef] [PubMed]
- Ostling, P.; Leivonen, S.K.; Aakula, A.; Kohonen, P.; Makela, R.; Hagman, Z.; Edsjo, A.; Kangaspeska, S.; Edgren, H.; Nicorici, D.; et al. Systematic analysis of microRNAs targeting the androgen receptor in prostate cancer cells. Cancer Res. 2011, 71, 1956–1967. [Google Scholar] [CrossRef] [PubMed]
- Kong, D.; Heath, E.; Chen, W.; Cher, M.; Powell, I.; Heilbrun, L.; Li, Y.; Ali, S.; Sethi, S.; Hassan, O.; et al. Epigenetic silencing of miR-34a in human prostate cancer cells and tumor tissue specimens can be reversed by BR-dim treatment. Am. J. Transl. Res. 2012, 4, 14–23. [Google Scholar] [PubMed]
- Hagman, Z.; Larne, O.; Edsjo, A.; Bjartell, A.; Ehrnstrom, R.A.; Ulmert, D.; Lilja, H.; Ceder, Y. miR-34c is downregulated in prostate cancer and exerts tumor suppressive functions. Int. J. Cancer 2010, 127, 2768–2776. [Google Scholar] [CrossRef] [PubMed]
- Sikand, K.; Slaibi, J.E.; Singh, R.; Slane, S.D.; Shukla, G.C. miR 488* inhibits androgen receptor expression in prostate carcinoma cells. Int. J. Cancer 2011, 129, 810–819. [Google Scholar] [CrossRef] [PubMed]
- Hagman, Z.; Haflidadottir, B.S.; Ceder, J.A.; Larne, O.; Bjartell, A.; Lilja, H.; Edsjo, A.; Ceder, Y. miR-205 negatively regulates the androgen receptor and is associated with adverse outcome of prostate cancer patients. Br. J. Cancer 2013, 108, 1668–1676. [Google Scholar] [CrossRef] [PubMed]
- Nadiminty, N.; Tummala, R.; Lou, W.; Zhu, Y.; Shi, X.B.; Zou, J.X.; Chen, H.; Zhang, J.; Chen, X.; Luo, J.; et al. MicroRNA let-7c is downregulated in prostate cancer and suppresses prostate cancer growth. PLoS ONE 2012, 7, e32832. [Google Scholar] [CrossRef] [PubMed]
- Hermeking, H. The miR-34 family in cancer and apoptosis. Cell Death Differ. 2010, 17, 193–199. [Google Scholar] [CrossRef] [PubMed]
- Chang, T.C.; Wentzel, E.A.; Kent, O.A.; Ramachandran, K.; Mullendore, M.; Lee, K.H.; Feldmann, G.; Yamakuchi, M.; Ferlito, M.; Lowenstein, C.J.; et al. Transactivation of miR-34a by p53 broadly influences gene expression and promotes apoptosis. Mol. Cell 2007, 26, 745–752. [Google Scholar] [CrossRef] [PubMed]
- He, L.; He, X.; Lim, L.P.; de Stanchina, E.; Xuan, Z.; Liang, Y.; Xue, W.; Zender, L.; Magnus, J.; Ridzon, D.; et al. A microRNA component of the p53 tumour suppressor network. Nature 2007, 447, 1130–1134. [Google Scholar] [CrossRef] [PubMed]
- Kashat, M.; Azzouz, L.; Sarkar, S.H.; Kong, D.; Li, Y.; Sarkar, F.H. Inactivation of ar and notch-1 signaling by miR-34a attenuates prostate cancer aggressiveness. Am. J. Transl. Res. 2012, 4, 432–442. [Google Scholar] [PubMed]
- Liu, C.; Kelnar, K.; Liu, B.; Chen, X.; Calhoun-Davis, T.; Li, H.; Patrawala, L.; Yan, H.; Jeter, C.; Honorio, S.; et al. The microRNA miR-34a inhibits prostate cancer stem cells and metastasis by directly repressing CD44. Nat. Med. 2011, 17, 211–215. [Google Scholar] [CrossRef] [PubMed]
- Azevedo, A.; Cunha, V.; Teixeira, A.L.; Medeiros, R. IL-6/IL-6R as a potential key signaling pathway in prostate cancer development. World J. Clin. Oncol. 2011, 2, 384–396. [Google Scholar] [CrossRef] [PubMed]
- Ueda, T.; Bruchovsky, N.; Sadar, M.D. Activation of the androgen receptor N-terminal domain by interleukin-6 via mapk and STAT3 signal transduction pathways. J. Biol. Chem. 2002, 277, 7076–7085. [Google Scholar] [CrossRef] [PubMed]
- Bakin, R.E.; Gioeli, D.; Sikes, R.A.; Bissonette, E.A.; Weber, M.J. Constitutive activation of the Ras/mitogen-activated protein kinase signaling pathway promotes androgen hypersensitivity in lncap prostate cancer cells. Cancer Res. 2003, 63, 1981–1989. [Google Scholar] [PubMed]
- Yeh, S.; Lin, H.K.; Kang, H.Y.; Thin, T.H.; Lin, M.F.; Chang, C. From HER2/NEU signal cascade to androgen receptor and its coactivators: A novel pathway by induction of androgen target genes through map kinase in prostate cancer cells. Proc. Natl. Acad. Sci. USA 1999, 96, 5458–5463. [Google Scholar] [CrossRef] [PubMed]
- Kinkade, C.W.; Castillo-Martin, M.; Puzio-Kuter, A.; Yan, J.; Foster, T.H.; Gao, H.; Sun, Y.; Ouyang, X.; Gerald, W.L.; Cordon-Cardo, C.; et al. Targeting AKT/mTOR and erk mapk signaling inhibits hormone-refractory prostate cancer in a preclinical mouse model. J. Clin. Investig. 2008, 118, 3051–3064. [Google Scholar] [CrossRef] [PubMed]
- Boll, K.; Reiche, K.; Kasack, K.; Morbt, N.; Kretzschmar, A.K.; Tomm, J.M.; Verhaegh, G.; Schalken, J.; von Bergen, M.; Horn, F.; et al. miR-130a, miR-203 and miR-205 jointly repress key oncogenic pathways and are downregulated in prostate carcinoma. Oncogene 2013, 32, 277–285. [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 cepsilon. Cancer Res. 2009, 69, 2287–2295. [Google Scholar] [CrossRef] [PubMed]
- Majid, S.; Dar, A.A.; Saini, S.; Yamamura, S.; Hirata, H.; Tanaka, Y.; Deng, G.; Dahiya, R. MicroRNA-205-directed transcriptional activation of tumor suppressor genes in prostate cancer. Cancer 2010, 116, 5637–5649. [Google Scholar] [CrossRef] [PubMed]
- Shi, X.B.; Xue, L.; Ma, A.H.; Tepper, C.G.; Gandour-Edwards, R.; Kung, H.J.; deVere White, R.W. Tumor suppressive miR-124 targets androgen receptor and inhibits proliferation of prostate cancer cells. Oncogene 2013, 32, 4130–4138. [Google Scholar] [CrossRef] [PubMed]
- Shi, Z.; Chen, Q.; Li, C.; Wang, L.; Qian, X.; Jiang, C.; Liu, X.; Wang, X.; Li, H.; Kang, C.; et al. miR-124 governs glioma growth and angiogenesis and enhances chemosensitivity by targeting R-Ras and N-Ras. Neuro Oncol. 2014, 16, 1341–1353. [Google Scholar] [CrossRef] [PubMed]
- Silber, J.; Lim, D.A.; Petritsch, C.; Persson, A.I.; Maunakea, A.K.; Yu, M.; Vandenberg, S.R.; Ginzinger, D.G.; James, C.D.; Costello, J.F.; et al. miR-124 and miR-137 inhibit proliferation of glioblastoma multiforme cells and induce differentiation of brain tumor stem cells. BMC Med. 2008, 6, 14. [Google Scholar] [CrossRef] [PubMed]
- Wang, P.; Chen, L.; Zhang, J.; Chen, H.; Fan, J.; Wang, K.; Luo, J.; Chen, Z.; Meng, Z.; Liu, L. Methylation-mediated silencing of the miR-124 genes facilitates pancreatic cancer progression and metastasis by targeting Rac1. Oncogene 2014, 33, 514–524. [Google Scholar] [CrossRef] [PubMed]
- Xia, J.; Wu, Z.; Yu, C.; He, W.; Zheng, H.; He, Y.; Jian, W.; Chen, L.; Zhang, L.; Li, W. miR-124 inhibits cell proliferation in gastric cancer through down-regulation of SPHK1. J. Pathol. 2012, 227, 470–480. [Google Scholar] [CrossRef] [PubMed]
- Zhang, H.; Wang, Q.; Zhao, Q.; Di, W. miR-124 inhibits the migration and invasion of ovarian cancer cells by targeting SPHK1. J. Ovarian Res. 2013, 6, 84. [Google Scholar] [CrossRef] [PubMed]
- Zhang, W.; Mao, Y.Q.; Wang, H.; Yin, W.J.; Zhu, S.X.; Wang, W.C. miR-124 suppresses cell motility and adhesion by targeting talin 1 in prostate cancer cells. Cancer Cell Int. 2015, 15, 49. [Google Scholar] [CrossRef] [PubMed]
- Chu, M.; Chang, Y.; Guo, Y.; Wang, N.; Cui, J.; Gao, W.Q. Regulation and methylation of tumor suppressor miR-124 by androgen receptor in prostate cancer cells. PLoS ONE 2015, 10, e0116197. [Google Scholar] [CrossRef] [PubMed]
- Shi, X.B.; Xue, L.; Yang, J.; Ma, A.H.; Zhao, J.; Xu, M.; Tepper, C.G.; Evans, C.P.; Kung, H.J.; deVere White, R.W. An androgen-regulated miRNA suppresses BAK1 expression and induces androgen-independent growth of prostate cancer cells. Proc. Natl. Acad. Sci. USA 2007, 104, 19983–19988. [Google Scholar] [CrossRef] [PubMed]
- Pierantoni, G.M.; Rinaldo, C.; Esposito, F.; Mottolese, M.; Soddu, S.; Fusco, A. High mobility group A1 (HMGA1) proteins interact with p53 and inhibit its apoptotic activity. Cell Death Differ. 2006, 13, 1554–1563. [Google Scholar] [CrossRef] [PubMed]
- Reinhart, B.J.; Slack, F.J.; Basson, M.; Pasquinelli, A.E.; Bettinger, J.C.; Rougvie, A.E.; Horvitz, H.R.; Ruvkun, G. The 21-nucleotide let-7 RNA regulates developmental timing in caenorhabditis elegans. Nature 2000, 403, 901–906. [Google Scholar] [PubMed]
- Roush, S.; Slack, F.J. The let-7 family of microRNAs. Trends Cell Biol. 2008, 18, 505–516. [Google Scholar] [CrossRef] [PubMed]
- Chu, G.C.; Zhau, H.E.; Wang, R.; Rogatko, A.; Feng, X.; Zayzafoon, M.; Liu, Y.; Farach-Carson, M.C.; You, S.; Kim, J.; et al. Rank- and C-met-mediated signal network promotes prostate cancer metastatic colonization. Endocr. Relat. Cancer 2014, 21, 311–326. [Google Scholar] [CrossRef] [PubMed]
- Gao, L.; Schwartzman, J.; Gibbs, A.; Lisac, R.; Kleinschmidt, R.; Wilmot, B.; Bottomly, D.; Coleman, I.; Nelson, P.; McWeeney, S.; et al. Androgen receptor promotes ligand-independent prostate cancer progression through c-Myc upregulation. PLoS ONE 2013, 8, e63563. [Google Scholar] [CrossRef] [PubMed]
- Nupponen, N.N.; Kakkola, L.; Koivisto, P.; Visakorpi, T. Genetic alterations in hormone-refractory recurrent prostate carcinomas. Am. J. Pathol. 1998, 153, 141–148. [Google Scholar] [CrossRef]
- Koscianska, E.; Baev, V.; Skreka, K.; Oikonomaki, K.; Rusinov, V.; Tabler, M.; Kalantidis, K. Prediction and preliminary validation of oncogene regulation by miRNAs. BMC Mol. Biol. 2007, 8, 79. [Google Scholar] [CrossRef] [PubMed]
- Boyerinas, B.; Park, S.M.; Shomron, N.; Hedegaard, M.M.; Vinther, J.; Andersen, J.S.; Feig, C.; Xu, J.; Burge, C.B.; Peter, M.E. Identification of let-7-regulated oncofetal genes. Cancer Res. 2008, 68, 2587–2591. [Google Scholar] [CrossRef] [PubMed]
- Ioannidis, P.; Mahaira, L.G.; Perez, S.A.; Gritzapis, A.D.; Sotiropoulou, P.A.; Kavalakis, G.J.; Antsaklis, A.I.; Baxevanis, C.N.; Papamichail, M. CRD-BP/IMP1 expression characterizes cord blood CD34+ stem cells and affects c-Myc and IGF-II expression in MCF-7 cancer cells. J. Biol. Chem. 2005, 280, 20086–20093. [Google Scholar] [CrossRef] [PubMed]
- Cairns, R.A.; Harris, I.S.; Mak, T.W. Regulation of cancer cell metabolism. Nat. Rev. Cancer 2011, 11, 85–95. [Google Scholar] [CrossRef] [PubMed]
- Barfeld, S.J.; Itkonen, H.M.; Urbanucci, A.; Mills, I.G. Androgen-regulated metabolism and biosynthesis in prostate cancer. Endocr. Relat. Cancer 2014, 21, T57–T66. [Google Scholar] [CrossRef] [PubMed]
- Thapar, R.; Titus, M.A. Recent advances in metabolic profiling and imaging of prostate cancer. Curr. Metabol. 2014, 2, 53–69. [Google Scholar] [CrossRef] [PubMed]
- Hatziapostolou, M.; Polytarchou, C.; Iliopoulos, D. miRNAs link metabolic reprogramming to oncogenesis. Trends Endocrinol. Metab. 2013, 24, 361–373. [Google Scholar] [CrossRef] [PubMed]
- Chan, B.; Manley, J.; Lee, J.; Singh, S.R. The emerging roles of microRNAs in cancer metabolism. Cancer Lett. 2015, 356, 301–308. [Google Scholar] [CrossRef] [PubMed]
- Chen, B.; Li, H.; Zeng, X.; Yang, P.; Liu, X.; Zhao, X.; Liang, S. Roles of microRNA on cancer cell metabolism. J. Transl. Med. 2012, 10, 228. [Google Scholar] [CrossRef] [PubMed]
- Singh, A.; Happel, C.; Manna, S.K.; Acquaah-Mensah, G.; Carrerero, J.; Kumar, S.; Nasipuri, P.; Krausz, K.W.; Wakabayashi, N.; Dewi, R.; et al. Transcription factor NRF2 regulates miR-1 and miR-206 to drive tumorigenesis. J. Clin. Investig. 2013, 123, 2921–2934. [Google Scholar] [CrossRef] [PubMed]
- Li, X.; Chen, Y.T.; Josson, S.; Mukhopadhyay, N.K.; Kim, J.; Freeman, M.R.; Huang, W.C. MicroRNA-185 and 342 inhibit tumorigenicity and induce apoptosis through blockade of the srebp metabolic pathway in prostate cancer cells. PLoS ONE 2013, 8, e70987. [Google Scholar] [CrossRef] [PubMed]
- Wang, W.L.; Welsh, J.; Tenniswood, M. 1,25-dihydroxyvitamin D3 modulates lipid metabolism in prostate cancer cells through miRNA mediated regulation of PPARA. J. Steroid Biochem. Mol. Biol. 2013, 136, 247–251. [Google Scholar] [CrossRef] [PubMed]
- He, X.P.; Shao, Y.; Li, X.L.; Xu, W.; Chen, G.S.; Sun, H.H.; Xu, H.C.; Xu, X.; Tang, D.; Zheng, X.F.; et al. Downregulation of miR-101 in gastric cancer correlates with cyclooxygenase-2 overexpression and tumor growth. FEBS J. 2012, 279, 4201–4212. [Google Scholar] [CrossRef] [PubMed]
- Hao, Y.; Gu, X.; Zhao, Y.; Greene, S.; Sha, W.; Smoot, D.T.; Califano, J.; Wu, T.C.; Pang, X. Enforced expression of miR-101 inhibits prostate cancer cell growth by modulating the COX-2 pathway in vivo. Cancer Prev. Res. 2011, 4, 1073–1083. [Google Scholar] [CrossRef] [PubMed]
- Hiroki, E.; Akahira, J.; Suzuki, F.; Nagase, S.; Ito, K.; Suzuki, T.; Sasano, H.; Yaegashi, N. Changes in microRNA expression levels correlate with clinicopathological features and prognoses in endometrial serous adenocarcinomas. Cancer Sci. 2010, 101, 241–249. [Google Scholar] [CrossRef] [PubMed]
- Wang, H.J.; Ruan, H.J.; He, X.J.; Ma, Y.Y.; Jiang, X.T.; Xia, Y.J.; Ye, Z.Y.; Tao, H.Q. MicroRNA-101 is down-regulated in gastric cancer and involved in cell migration and invasion. Eur. J. Cancer 2010, 46, 2295–2303. [Google Scholar] [CrossRef] [PubMed]
- Strillacci, A.; Griffoni, C.; Sansone, P.; Paterini, P.; Piazzi, G.; Lazzarini, G.; Spisni, E.; Pantaleo, M.A.; Biasco, G.; Tomasi, V. miR-101 downregulation is involved in cyclooxygenase-2 overexpression in human colon cancer cells. Exp. Cell Res. 2009, 315, 1439–1447. [Google Scholar] [CrossRef] [PubMed]
- Gao, P.; Tchernyshyov, I.; Chang, T.C.; Lee, Y.S.; Kita, K.; Ochi, T.; Zeller, K.I.; de Marzo, A.M.; van Eyk, J.E.; Mendell, J.T.; et al. c-Myc suppression of miR-23a/b enhances mitochondrial glutaminase expression and glutamine metabolism. Nature 2009, 458, 762–765. [Google Scholar] [CrossRef] [PubMed]
- Liu, W.; Le, A.; Hancock, C.; Lane, A.N.; Dang, C.V.; Fan, T.W.; Phang, J.M. Reprogramming of proline and glutamine metabolism contributes to the proliferative and metabolic responses regulated by oncogenic transcription factor c-Myc. Proc. Natl. Acad. Sci. USA 2012, 109, 8983–8988. [Google Scholar] [CrossRef] [PubMed]
- Xu, Y.; Fang, F.; Zhang, J.; Josson, S.; Clair, W.H., St.; Clair, D.K., St. miR-17* suppresses tumorigenicity of prostate cancer by inhibiting mitochondrial antioxidant enzymes. PLoS ONE 2010, 5, e14356. [Google Scholar] [CrossRef] [PubMed]
- Pelton, K.; Freeman, M.R.; Solomon, K.R. Cholesterol and prostate cancer. Curr. Opin. Pharmacol. 2012, 12, 751–759. [Google Scholar] [CrossRef] [PubMed]
- Singh, P.K.; Brand, R.E.; Mehla, K. MicroRNAs in pancreatic cancer metabolism. Nat. Rev. Gastroenterol. Hepatol. 2012, 9, 334–344. [Google Scholar] [CrossRef] [PubMed]
- Xu, Y.; Fang, F.; St Clair, D.K.; Josson, S.; Sompol, P.; Spasojevic, I.; Clair, W.H., St. Suppression of RelB-mediated manganese superoxide dismutase expression reveals a primary mechanism for radiosensitization effect of 1α,25-dihydroxyvitamin D3 in prostate cancer cells. Mol. Cancer Ther. 2007, 6, 2048–2056. [Google Scholar] [CrossRef] [PubMed]
- Prensner, J.R.; Iyer, M.K.; Balbin, O.A.; Dhanasekaran, S.M.; Cao, Q.; Brenner, J.C.; Laxman, B.; Asangani, I.A.; Grasso, C.S.; Kominsky, H.D.; et al. Transcriptome sequencing across a prostate cancer cohort identifies PCAT-1, an unannotated lincRNA implicated in disease progression. Nat. Biotechnol. 2011, 29, 742–749. [Google Scholar] [CrossRef] [PubMed]
- Du, Z.; Fei, T.; Verhaak, R.G.; Su, Z.; Zhang, Y.; Brown, M.; Chen, Y.; Liu, X.S. Integrative genomic analyses reveal clinically relevant long noncoding RNAs in human cancer. Nat. Struct. Mol. Biol. 2013, 20, 908–913. [Google Scholar] [CrossRef] [PubMed]
- Ren, S.; Peng, Z.; Mao, J.H.; Yu, Y.; Yin, C.; Gao, X.; Cui, Z.; Zhang, J.; Yi, K.; Xu, W.; et al. RNA-seq analysis of prostate cancer in the chinese population identifies recurrent gene fusions, cancer-associated long noncoding RNAs and aberrant alternative splicings. Cell Res. 2012, 22, 806–821. [Google Scholar] [CrossRef] [PubMed]
- Hu, C.C.; Gan, P.; Zhang, R.Y.; Xue, J.X.; Ran, L.K. Identification of prostate cancer lncRNAs by RNA-seq. Asian Pac. J. Cancer Prev. 2014, 15, 9439–9444. [Google Scholar] [CrossRef] [PubMed]
- Kim, S.T.; Cheng, Y.; Hsu, F.C.; Jin, T.; Kader, A.K.; Zheng, S.L.; Isaacs, W.B.; Xu, J.; Sun, J. Prostate cancer risk-associated variants reported from genome-wide association studies: Meta-analysis and their contribution to genetic variation. Prostate 2010, 70, 1729–1738. [Google Scholar] [CrossRef] [PubMed]
- Jin, G.; Sun, J.; Isaacs, S.D.; Wiley, K.E.; Kim, S.T.; Chu, L.W.; Zhang, Z.; Zhao, H.; Zheng, S.L.; Isaacs, W.B.; et al. Human polymorphisms at long non-coding RNAs (lncRNAs) and association with prostate cancer risk. Carcinogenesis 2011, 32, 1655–1659. [Google Scholar] [CrossRef] [PubMed]
- Al Olama, A.A.; Kote-Jarai, Z.; Giles, G.G.; Guy, M.; Morrison, J.; Severi, G.; Leongamornlert, D.A.; Tymrakiewicz, M.; Jhavar, S.; Saunders, E.; et al. Multiple loci on 8q24 associated with prostate cancer susceptibility. Nat. Genet. 2009, 41, 1058–1060. [Google Scholar] [CrossRef] [PubMed]
- He, J.H.; Zhang, J.Z.; Han, Z.P.; Wang, L.; Lv, Y.B.; Li, Y.G. Reciprocal regulation of PCGEM1 and miR-145 promote proliferation of lncap prostate cancer cells. J. Exp. Clin. Cancer Res. 2014, 33, 72. [Google Scholar] [CrossRef] [PubMed]
- Meyer, K.B.; Maia, A.T.; O’Reilly, M.; Ghoussaini, M.; Prathalingam, R.; Porter-Gill, P.; Ambs, S.; Prokunina-Olsson, L.; Carroll, J.; Ponder, B.A. A functional variant at a prostate cancer predisposition locus at 8q24 is associated with PVT1 expression. PLoS Genet. 2011, 7, e1002165. [Google Scholar] [CrossRef] [PubMed]
- Tseng, Y.Y.; Moriarity, B.S.; Gong, W.; Akiyama, R.; Tiwari, A.; Kawakami, H.; Ronning, P.; Reuland, B.; Guenther, K.; Beadnell, T.C.; et al. PVT1 dependence in cancer with myc copy-number increase. Nature 2014, 512, 82–86. [Google Scholar] [CrossRef] [PubMed]
- Prensner, J.R.; Chen, W.; Han, S.; Iyer, M.K.; Cao, Q.; Kothari, V.; Evans, J.R.; Knudsen, K.E.; Paulsen, M.T.; Ljungman, M.; et al. The long non-coding RNA PCAT-1 promotes prostate cancer cell proliferation through cmyc. Neoplasia 2014, 16, 900–908. [Google Scholar] [CrossRef] [PubMed]
- Chung, S.; Nakagawa, H.; Uemura, M.; Piao, L.; Ashikawa, K.; Hosono, N.; Takata, R.; Akamatsu, S.; Kawaguchi, T.; Morizono, T.; et al. Association of a novel long non-coding RNA in 8q24 with prostate cancer susceptibility. Cancer Sci. 2011, 102, 245–252. [Google Scholar] [CrossRef] [PubMed]
- Cao, D.L.; Gu, C.Y.; Zhu, Y.; Dai, B.; Zhang, H.L.; Shi, G.H.; Shen, Y.J.; Zhu, Y.P.; Ma, C.G.; Xiao, W.J.; et al. Polymorphisms at long non-coding RNAs and prostate cancer risk in an eastern chinese population. Prostate Cancer Prostatic Dis. 2014, 17, 315–319. [Google Scholar] [CrossRef] [PubMed]
- Xue, Y.; Wang, M.; Kang, M.; Wang, Q.; Wu, B.; Chu, H.; Zhong, D.; Qin, C.; Yin, C.; Zhang, Z.; et al. Association between lncRNA PCGEM1 polymorphisms and prostate cancer risk. Prostate Cancer Prostatic Dis. 2013, 16, 139–144. [Google Scholar] [CrossRef] [PubMed]
- Chakravarty, D.; Sboner, A.; Nair, S.S.; Giannopoulou, E.; Li, R.; Hennig, S.; Mosquera, J.M.; Pauwels, J.; Park, K.; Kossai, M.; et al. The oestrogen receptor α-regulated lncRNA NEAT1 is a critical modulator of prostate cancer. Nat. Commun. 2014, 5, 5383. [Google Scholar] [CrossRef] [PubMed]
- Yap, K.L.; Li, S.; Munoz-Cabello, A.M.; Raguz, S.; Zeng, L.; Mujtaba, S.; Gil, J.; Walsh, M.J.; Zhou, M.M. Molecular interplay of the noncoding RNA anril and methylated histone H3 lysine 27 by polycomb CBX7 in transcriptional silencing of INK4a. Mol. Cell 2010, 38, 662–674. [Google Scholar] [CrossRef] [PubMed]
- Takayama, K.; Horie-Inoue, K.; Katayama, S.; Suzuki, T.; Tsutsumi, S.; Ikeda, K.; Urano, T.; Fujimura, T.; Takagi, K.; Takahashi, S.; et al. Androgen-responsive long noncoding RNA CTBP1-as promotes prostate cancer. EMBO J. 2013, 32, 1665–1680. [Google Scholar] [CrossRef] [PubMed]
- Sakurai, K.; Reon, B.J.; Anaya, J.; Dutta, A. The lncRNA DRAIC/PCAT29 locus constitutes a tumor-suppressive nexus. Mol. Cancer Res. 2015, 13, 828–838. [Google Scholar] [CrossRef] [PubMed]
- Pickard, M.R.; Mourtada-Maarabouni, M.; Williams, G.T. Long non-coding RNA GAS5 regulates apoptosis in prostate cancer cell lines. Biochim. Biophys. Acta 2013, 1832, 1613–1623. [Google Scholar] [CrossRef] [PubMed]
- Yacqub-Usman, K.; Pickard, M.R.; Williams, G.T. Reciprocal regulation of GAS5 lncRNA levels and mTOR inhibitor action in prostate cancer cells. Prostate 2015, 75, 693–705. [Google Scholar] [CrossRef] [PubMed]
- Zhu, M.; Chen, Q.; Liu, X.; Sun, Q.; Zhao, X.; Deng, R.; Wang, Y.; Huang, J.; Xu, M.; Yan, J.; et al. LncRNA h19/miR-675 axis represses prostate cancer metastasis by targeting TGFBI. FEBS J. 2014, 281, 3766–3775. [Google Scholar] [CrossRef] [PubMed]
- Bauderlique-Le Roy, H.; Vennin, C.; Brocqueville, G.; Spruyt, N.; Adriaenssens, E.; Bourette, R.P. Enrichment of human stem-like prostate cells with s-ship promoter activity uncovers a role in stemness for the long noncoding RNA H19. Stem Cells Dev. 2015, 24, 1252–1262. [Google Scholar] [CrossRef] [PubMed]
- Scaravilli, M.; Porkka, K.P.; Brofeldt, A.; Annala, M.; Tammela, T.L.; Jenster, G.W.; Nykter, M.; Visakorpi, T. miR-1247-5p is overexpressed in castration resistant prostate cancer and targets MYCBP2. Prostate 2015, 75, 798–805. [Google Scholar] [CrossRef] [PubMed]
- Chiyomaru, T.; Yamamura, S.; Fukuhara, S.; Yoshino, H.; Kinoshita, T.; Majid, S.; Saini, S.; Chang, I.; Tanaka, Y.; Enokida, H.; et al. Genistein inhibits prostate cancer cell growth by targeting miR-34a and oncogenic hotair. PLoS ONE 2013, 8, e70372. [Google Scholar] [CrossRef] [PubMed]
- Wang, L.; Han, S.; Jin, G.; Zhou, X.; Li, M.; Ying, X.; Wang, L.; Wu, H.; Zhu, Q. Linc00963: A novel, long non-coding RNA involved in the transition of prostate cancer from androgen-dependence to androgen-independence. Int. J. Oncol. 2014, 44, 2041–2049. [Google Scholar] [CrossRef] [PubMed]
- Ren, S.; Liu, Y.; Xu, W.; Sun, Y.; Lu, J.; Wang, F.; Wei, M.; Shen, J.; Hou, J.; Gao, X.; et al. Long noncoding RNA MALAT-1 is a new potential therapeutic target for castration resistant prostate cancer. J. Urol. 2013, 190, 2278–2287. [Google Scholar] [CrossRef] [PubMed]
- Wang, F.; Ren, S.; Chen, R.; Lu, J.; Shi, X.; Zhu, Y.; Zhang, W.; Jing, T.; Zhang, C.; Shen, J.; et al. Development and prospective multicenter evaluation of the long noncoding RNA MALAT-1 as a diagnostic urinary biomarker for prostate cancer. Oncotarget 2014, 5, 11091–11102. [Google Scholar] [CrossRef] [PubMed]
- Salameh, A.; Lee, A.K.; Cardo-Vila, M.; Nunes, D.N.; Efstathiou, E.; Staquicini, F.I.; Dobroff, A.S.; Marchio, S.; Navone, N.M.; Hosoya, H.; et al. PRUNE2 is a human prostate cancer suppressor regulated by the intronic long noncoding RNA PCA3. Proc. Natl. Acad. Sci. USA 2015, 112, 8403–8408. [Google Scholar] [CrossRef] [PubMed]
- Ferreira, L.B.; Palumbo, A.; de Mello, K.D.; Sternberg, C.; Caetano, M.S.; de Oliveira, F.L.; Neves, A.F.; Nasciutti, L.E.; Goulart, L.R.; Gimba, E.R. PCA3 noncoding RNA is involved in the control of prostate-cancer cell survival and modulates androgen receptor signaling. BMC Cancer 2012, 12, 507. [Google Scholar] [CrossRef] [PubMed]
- Bussemakers, M.J.; van Bokhoven, A.; Verhaegh, G.W.; Smit, F.P.; Karthaus, H.F.; Schalken, J.A.; Debruyne, F.M.; Ru, N.; Isaacs, W.B. DD3: A new prostate-specific gene, highly overexpressed in prostate cancer. Cancer Res. 1999, 59, 5975–5979. [Google Scholar] [PubMed]
- Prensner, J.R.; Chen, W.; Iyer, M.K.; Cao, Q.; Ma, T.; Han, S.; Sahu, A.; Malik, R.; Wilder-Romans, K.; Navone, N.; et al. PCAT-1, a long noncoding RNA, regulates BRCA2 and controls homologous recombination in cancer. Cancer Res. 2014, 74, 1651–1660. [Google Scholar] [CrossRef] [PubMed]
- Crea, F.; Watahiki, A.; Quagliata, L.; Xue, H.; Pikor, L.; Parolia, A.; Wang, Y.; Lin, D.; Lam, W.L.; Farrar, W.L.; et al. Identification of a long non-coding RNA as a novel biomarker and potential therapeutic target for metastatic prostate cancer. Oncotarget 2014, 5, 764–774. [Google Scholar] [CrossRef] [PubMed]
- Malik, R.; Patel, L.; Prensner, J.R.; Shi, Y.; Iyer, M.K.; Subramaniyan, S.; Carley, A.; Niknafs, Y.S.; Sahu, A.; Han, S.; et al. The lncRNA PCAT29 inhibits oncogenic phenotypes in prostate cancer. Mol. Cancer Res. 2014, 12, 1081–1087. [Google Scholar] [CrossRef] [PubMed]
- Fu, X.; Ravindranath, L.; Tran, N.; Petrovics, G.; Srivastava, S. Regulation of apoptosis by a prostate-specific and prostate cancer-associated noncoding gene, PCGEM1. DNA Cell Biol. 2006, 25, 135–141. [Google Scholar] [CrossRef] [PubMed]
- Parolia, A.; Crea, F.; Xue, H.; Wang, Y.; Mo, F.; Ramnarine, V.R.; Liu, H.H.; Lin, D.; Saidy, N.R.; Clermont, P.L.; et al. The long non-coding RNA PCGEM1 is regulated by androgen receptor activity in vivo. Mol. Cancer 2015, 14, 46. [Google Scholar] [CrossRef] [PubMed]
- Yang, L.; Lin, C.; Jin, C.; Yang, J.C.; Tanasa, B.; Li, W.; Merkurjev, D.; Ohgi, K.A.; Meng, D.; Zhang, J.; et al. LncRNA-dependent mechanisms of androgen-receptor-regulated gene activation programs. Nature 2013, 500, 598–602. [Google Scholar] [CrossRef] [PubMed]
- Petrovics, G.; Zhang, W.; Makarem, M.; Street, J.P.; Connelly, R.; Sun, L.; Sesterhenn, I.A.; Srikantan, V.; Moul, J.W.; Srivastava, S. Elevated expression of PCGEM1, a prostate-specific gene with cell growth-promoting function, is associated with high-risk prostate cancer patients. Oncogene 2004, 23, 605–611. [Google Scholar] [CrossRef] [PubMed]
- Prensner, J.R.; Sahu, A.; Iyer, M.K.; Malik, R.; Chandler, B.; Asangani, I.A.; Poliakov, A.; Vergara, I.A.; Alshalalfa, M.; Jenkins, R.B.; et al. The incRNAs PCGEM1 and PRNCR1 are not implicated in castration resistant prostate cancer. Oncotarget 2014, 5, 1434–1438. [Google Scholar] [CrossRef] [PubMed]
- Hung, C.L.; Wang, L.Y.; Yu, Y.L.; Chen, H.W.; Srivastava, S.; Petrovics, G.; Kung, H.J. A long noncoding RNA connects c-Myc to tumor metabolism. Proc. Natl. Acad. Sci. USA 2014, 111, 18697–18702. [Google Scholar] [CrossRef] [PubMed]
- Srikantan, V.; Zou, Z.; Petrovics, G.; Xu, L.; Augustus, M.; Davis, L.; Livezey, J.R.; Connell, T.; Sesterhenn, I.A.; Yoshino, K.; et al. PCGEM1, a prostate-specific gene, is overexpressed in prostate cancer. Proc. Natl. Acad. Sci. USA 2000, 97, 12216–12221. [Google Scholar] [CrossRef] [PubMed]
- Cui, Z.; Ren, S.; Lu, J.; Wang, F.; Xu, W.; Sun, Y.; Wei, M.; Chen, J.; Gao, X.; Xu, C.; et al. The prostate cancer-up-regulated long noncoding RNA plncRNA-1 modulates apoptosis and proliferation through reciprocal regulation of androgen receptor. Urol. Oncol. 2013, 31, 1117–1123. [Google Scholar] [CrossRef] [PubMed]
- Lee, R.S.; Roberts, C.W. Linking the SWI/SNF complex to prostate cancer. Nat. Genet. 2013, 45, 1268–1269. [Google Scholar] [CrossRef] [PubMed]
- Mehra, R.; Shi, Y.; Udager, A.M.; Prensner, J.R.; Sahu, A.; Iyer, M.K.; Siddiqui, J.; Cao, X.; Wei, J.; Jiang, H.; et al. A novel RNA in situ hybridization assay for the long noncoding RNA SChLAP1 predicts poor clinical outcome after radical prostatectomy in clinically localized prostate cancer. Neoplasia 2014, 16, 1121–1127. [Google Scholar] [CrossRef] [PubMed]
- Orfanelli, U.; Jachetti, E.; Chiacchiera, F.; Grioni, M.; Brambilla, P.; Briganti, A.; Freschi, M.; Martinelli-Boneschi, F.; Doglioni, C.; Montorsi, F.; et al. Antisense transcription at the TRPM2 locus as a novel prognostic marker and therapeutic target in prostate cancer. Oncogene 2015, 34, 2094–2102. [Google Scholar] [CrossRef] [PubMed]
- Yang, F.; Zhang, H.; Mei, Y.; Wu, M. Reciprocal regulation of HIF-1α and lincRNA-p21 modulates the warburg effect. Mol. Cell 2014, 53, 88–100. [Google Scholar] [CrossRef] [PubMed]
- Isin, M.; Uysaler, E.; Ozgur, E.; Koseoglu, H.; Sanli, O.; Yucel, O.B.; Gezer, U.; Dalay, N. Exosomal lncRNA-p21 levels may help to distinguish prostate cancer from benign disease. Front. Genet. 2015, 6, 168. [Google Scholar] [PubMed]
- Wan, G.; Mathur, R.; Hu, X.; Liu, Y.; Zhang, X.; Peng, G.; Lu, X. Long non-coding RNA anril (CDKN2b-as) is induced by the ATM-E2F1 signaling pathway. Cell Signal. 2013, 25, 1086–1095. [Google Scholar] [CrossRef] [PubMed]
- Kotake, Y.; Nakagawa, T.; Kitagawa, K.; Suzuki, S.; Liu, N.; Kitagawa, M.; Xiong, Y. Long non-coding RNA anril is required for the PRC2 recruitment to and silencing of p15(INK4b) tumor suppressor gene. Oncogene 2011, 30, 1956–1962. [Google Scholar] [CrossRef] [PubMed]
- Roberts, C.W.; Orkin, S.H. The SWI/SNF complex—chromatin and cancer. Nat. Rev. Cancer 2004, 4, 133–142. [Google Scholar] [CrossRef] [PubMed]
- Salmena, L.; Poliseno, L.; Tay, Y.; Kats, L.; Pandolfi, P.P. A ceRNA hypothesis: The rosetta stone of a hidden RNA language? Cell 2011, 146, 353–358. [Google Scholar] [CrossRef] [PubMed]
- Hansen, T.B.; Jensen, T.I.; Clausen, B.H.; Bramsen, J.B.; Finsen, B.; Damgaard, C.K.; Kjems, J. Natural RNA circles function as efficient microRNA sponges. Nature 2013, 495, 384–388. [Google Scholar] [CrossRef] [PubMed]
- Poliseno, L.; Salmena, L.; Zhang, J.; Carver, B.; Haveman, W.J.; Pandolfi, P.P. A coding-independent function of gene and pseudogene mRNAs regulates tumour biology. Nature 2010, 465, 1033–1038. [Google Scholar] [CrossRef] [PubMed]
- Ferraldeschi, R.; Nava Rodrigues, D.; Riisnaes, R.; Miranda, S.; Figueiredo, I.; Rescigno, P.; Ravi, P.; Pezaro, C.; Omlin, A.; Lorente, D.; et al. Pten protein loss and clinical outcome from castration-resistant prostate cancer treated with abiraterone acetate. Eur. Urol. 2015, 67, 795–802. [Google Scholar] [CrossRef] [PubMed]
- Armstrong, A.J.; Eisenberger, M.A.; Halabi, S.; Oudard, S.; Nanus, D.M.; Petrylak, D.P.; Sartor, A.O.; Scher, H.I. Biomarkers in the management and treatment of men with metastatic castration-resistant prostate cancer. Eur. Urol. 2012, 61, 549–559. [Google Scholar] [CrossRef] [PubMed]
- Mitchell, P.S.; Parkin, R.K.; Kroh, E.M.; Fritz, B.R.; Wyman, S.K.; Pogosova-Agadjanyan, E.L.; Peterson, A.; Noteboom, J.; O’Briant, K.C.; Allen, A.; et al. Circulating microRNAs as stable blood-based markers for cancer detection. Proc. Natl. Acad. Sci. USA 2008, 105, 10513–10518. [Google Scholar] [CrossRef] [PubMed]
- Gonzales, J.C.; Fink, L.M.; Goodman, O.B., Jr.; Symanowski, J.T.; Vogelzang, N.J.; Ward, D.C. Comparison of circulating microRNA 141 to circulating tumor cells, lactate dehydrogenase, and prostate-specific antigen for determining treatment response in patients with metastatic prostate cancer. Clin. Genitourin. Cancer 2011, 9, 39–45. [Google Scholar] [CrossRef] [PubMed]
- Zhang, H.L.; Qin, X.J.; Cao, D.L.; Zhu, Y.; Yao, X.D.; Zhang, S.L.; Dai, B.; Ye, D.W. An elevated serum miR-141 level in patients with bone-metastatic prostate cancer is correlated with more bone lesions. Asian J. Androl. 2013, 15, 231–235. [Google Scholar] [CrossRef] [PubMed]
- Bryant, R.J.; Pawlowski, T.; Catto, J.W.; Marsden, G.; Vessella, R.L.; Rhees, B.; Kuslich, C.; Visakorpi, T.; Hamdy, F.C. Changes in circulating microRNA levels associated with prostate cancer. Br. J. Cancer 2012, 106, 768–774. [Google Scholar] [CrossRef] [PubMed]
- Cheng, H.H.; Mitchell, P.S.; Kroh, E.M.; Dowell, A.E.; Chery, L.; Siddiqui, J.; Nelson, P.S.; Vessella, R.L.; Knudsen, B.S.; Chinnaiyan, A.M.; et al. Circulating microRNA profiling identifies a subset of metastatic prostate cancer patients with evidence of cancer-associated hypoxia. PLoS ONE 2013, 8, e69239. [Google Scholar] [CrossRef] [PubMed]
- Kachakova, D.; Mitkova, A.; Popov, E.; Popov, I.; Vlahova, A.; Dikov, T.; Christova, S.; Mitev, V.; Slavov, C.; Kaneva, R. Combinations of serum prostate-specific antigen and plasma expression levels of let-7c, miR-30c, miR-141, and miR-375 as potential better diagnostic biomarkers for prostate cancer. DNA Cell Biol. 2015, 34, 189–200. [Google Scholar] [CrossRef] [PubMed]
- Haldrup, C.; Kosaka, N.; Ochiya, T.; Borre, M.; Hoyer, S.; Orntoft, T.F.; Sorensen, K.D. Profiling of circulating microRNAs for prostate cancer biomarker discovery. Drug Deliv. Transl. Res. 2014, 4, 19–30. [Google Scholar] [CrossRef] [PubMed]
- Kelly, B.D.; Miller, N.; Sweeney, K.J.; Durkan, G.C.; Rogers, E.; Walsh, K.; Kerin, M.J. A circulating microRNA signature as a biomarker for prostate cancer in a high risk group. J. Clin. Med. 2015, 4, 1369–1379. [Google Scholar] [CrossRef] [PubMed]
- Brase, J.C.; Johannes, M.; Schlomm, T.; Falth, M.; Haese, A.; Steuber, T.; Beissbarth, T.; Kuner, R.; Sultmann, H. Circulating miRNAs are correlated with tumor progression in prostate cancer. Int. J. Cancer 2011, 128, 608–616. [Google Scholar] [CrossRef] [PubMed]
- Nguyen, H.C.; Xie, W.; Yang, M.; Hsieh, C.L.; Drouin, S.; Lee, G.S.; Kantoff, P.W. Expression differences of circulating microRNAs in metastatic castration resistant prostate cancer and low-risk, localized prostate cancer. Prostate 2013, 73, 346–354. [Google Scholar] [CrossRef] [PubMed]
- Lin, H.M.; Castillo, L.; Mahon, K.L.; Chiam, K.; Lee, B.Y.; Nguyen, Q.; Boyer, M.J.; Stockler, M.R.; Pavlakis, N.; Marx, G.; et al. Circulating microRNAs are associated with docetaxel chemotherapy outcome in castration-resistant prostate cancer. Br. J. Cancer 2014, 110, 2462–2471. [Google Scholar] [CrossRef] [PubMed]
- Selth, L.A.; Townley, S.L.; Bert, A.G.; Stricker, P.D.; Sutherland, P.D.; Horvath, L.G.; Goodall, G.J.; Butler, L.M.; Tilley, W.D. Circulating microRNAs predict biochemical recurrence in prostate cancer patients. Br. J. Cancer 2013, 109, 641–650. [Google Scholar] [CrossRef] [PubMed]
- Sun, X.; Yang, Z.; Zhang, Y.; He, J.; Wang, F.; Su, P.; Han, J.; Song, Z.; Fei, Y. Prognostic implications of tissue and serum levels of microRNA-128 in human prostate cancer. Int. J. Clin. Exp. Pathol. 2015, 8, 8394–8401. [Google Scholar] [PubMed]
- Hessels, D.; Klein Gunnewiek, J.M.; van Oort, I.; Karthaus, H.F.; van Leenders, G.J.; van Balken, B.; Kiemeney, L.A.; Witjes, J.A.; Schalken, J.A. DD3(PCA3)-based molecular urine analysis for the diagnosis of prostate cancer. Eur. Urol. 2003, 44, 8–15. [Google Scholar] [CrossRef]
- Hessels, D.; Schalken, J.A. The use of PCA3 in the diagnosis of prostate cancer. Nat. Rev. Urol. 2009, 6, 255–261. [Google Scholar] [CrossRef] [PubMed]
- Ploussard, G.; Durand, X.; Xylinas, E.; Moutereau, S.; Radulescu, C.; Forgue, A.; Nicolaiew, N.; Terry, S.; Allory, Y.; Loric, S.; et al. Prostate cancer antigen 3 score accurately predicts tumour volume and might help in selecting prostate cancer patients for active surveillance. Eur. Urol. 2011, 59, 422–429. [Google Scholar] [CrossRef] [PubMed]
- Martinez-Pineiro, L.; Schalken, J.A.; Cabri, P.; Maisonobe, P.; de la Taille, A.; Triptocare Study, G. Evaluation of urinary prostate cancer antigen-3 (PCA3) and TMPRSS2-ERG score changes when starting androgen-deprivation therapy with triptorelin 6-month formulation in patients with locally advanced and metastatic prostate cancer. Br. J. Urol. Int. 2014, 114, 608–616. [Google Scholar] [CrossRef] [PubMed]
- Bourdoumis, A.; Chrisofos, M.; Stasinou, T.; Christopoulos, P.; Mourmouris, P.; Kostakopoulos, A.; Deliveliotis, C. The role of PCA3 as a prognostic factor in patients with castration-resistant prostate cancer (CRPC) treated with docetaxel. Anticancer Res. 2015, 35, 3075–3079. [Google Scholar] [PubMed]
- Ren, S.; Wang, F.; Shen, J.; Sun, Y.; Xu, W.; Lu, J.; Wei, M.; Xu, C.; Wu, C.; Zhang, Z.; et al. Long non-coding RNA metastasis associated in lung adenocarcinoma transcript 1 derived miniRNA as a novel plasma-based biomarker for diagnosing prostate cancer. Eur. J. Cancer 2013, 49, 2949–2959. [Google Scholar] [CrossRef] [PubMed]
- Zhang, W.; Ren, S.C.; Shi, X.L.; Liu, Y.W.; Zhu, Y.S.; Jing, T.L.; Wang, F.B.; Chen, R.; Xu, C.L.; Wang, H.Q.; et al. A novel urinary long non-coding RNA transcript improves diagnostic accuracy in patients undergoing prostate biopsy. Prostate 2015, 75, 653–661. [Google Scholar] [CrossRef] [PubMed]
- Lee, B.; Mazar, J.; Aftab, M.N.; Qi, F.; Shelley, J.; Li, J.L.; Govindarajan, S.; Valerio, F.; Rivera, I.; Thurn, T.; et al. Long noncoding RNAs as putative biomarkers for prostate cancer detection. J. Mol. Diagn 2014, 16, 615–626. [Google Scholar] [CrossRef] [PubMed]
- Prensner, J.R.; Zhao, S.; Erho, N.; Schipper, M.; Iyer, M.K.; Dhanasekaran, S.M.; Magi-Galluzzi, C.; Mehra, R.; Sahu, A.; Siddiqui, J.; et al. RNA biomarkers associated with metastatic progression in prostate cancer: A multi-institutional high-throughput analysis of schlap1. Lancet Oncol. 2014, 15, 1469–1480. [Google Scholar] [CrossRef]
- Li, Z.; Rana, T.M. Therapeutic targeting of microRNAs: Current status and future challenges. Nat. Rev. Drug Discov. 2014, 13, 622–638. [Google Scholar] [CrossRef] [PubMed]
- Ochiya, T.; Nagahara, S.; Sano, A.; Itoh, H.; Terada, M. Biomaterials for gene delivery: Atelocollagen-mediated controlled release of molecular medicines. Curr. Gene Ther. 2001, 1, 31–52. [Google Scholar] [CrossRef] [PubMed]
- Takeshita, F.; Patrawala, L.; Osaki, M.; Takahashi, R.U.; Yamamoto, Y.; Kosaka, N.; Kawamata, M.; Kelnar, K.; Bader, A.G.; Brown, D.; et al. Systemic delivery of synthetic microRNA-16 inhibits the growth of metastatic prostate tumors via downregulation of multiple cell-cycle genes. Mol. Ther. 2010, 18, 181–187. [Google Scholar] [CrossRef] [PubMed]
- Bouchie, A. First microRNA mimic enters clinic. Nat. Biotechnol. 2013, 31, 577. [Google Scholar] [CrossRef] [PubMed]
© 2015 by the authors; licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons by Attribution (CC-BY) license (http://creativecommons.org/licenses/by/4.0/).
Share and Cite
Shih, J.-W.; Wang, L.-Y.; Hung, C.-L.; Kung, H.-J.; Hsieh, C.-L. Non-Coding RNAs in Castration-Resistant Prostate Cancer: Regulation of Androgen Receptor Signaling and Cancer Metabolism. Int. J. Mol. Sci. 2015, 16, 28943-28978. https://doi.org/10.3390/ijms161226138
Shih J-W, Wang L-Y, Hung C-L, Kung H-J, Hsieh C-L. Non-Coding RNAs in Castration-Resistant Prostate Cancer: Regulation of Androgen Receptor Signaling and Cancer Metabolism. International Journal of Molecular Sciences. 2015; 16(12):28943-28978. https://doi.org/10.3390/ijms161226138
Chicago/Turabian StyleShih, Jing-Wen, Ling-Yu Wang, Chiu-Lien Hung, Hsing-Jien Kung, and Chia-Ling Hsieh. 2015. "Non-Coding RNAs in Castration-Resistant Prostate Cancer: Regulation of Androgen Receptor Signaling and Cancer Metabolism" International Journal of Molecular Sciences 16, no. 12: 28943-28978. https://doi.org/10.3390/ijms161226138
APA StyleShih, J.-W., Wang, L.-Y., Hung, C.-L., Kung, H.-J., & Hsieh, C.-L. (2015). Non-Coding RNAs in Castration-Resistant Prostate Cancer: Regulation of Androgen Receptor Signaling and Cancer Metabolism. International Journal of Molecular Sciences, 16(12), 28943-28978. https://doi.org/10.3390/ijms161226138
