151
|
Meeker AK. Cancer telomeres and white crows. AMERICAN JOURNAL OF CLINICAL AND EXPERIMENTAL UROLOGY 2018; 6:93-100. [PMID: 29666837 PMCID: PMC5902727] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 03/12/2018] [Accepted: 03/19/2018] [Indexed: 06/08/2023]
Abstract
This mini-review article discusses past and present prostate-focused research on telomere and telomerase biology conducted at Johns Hopkins, through the eyes of a Donald S Coffey trainee. Included are past discoveries of abnormalities in telomere biology in the context of prostate cancer and its pre-malignant precursor prostatic intraepithelial neoplasia (PIN); the finding that telomerase activity is androgen-regulated in the prostate, and the potential role of telomerase in prostate epithelial stem cells. Also reviewed are more recent results showing that in situ telomere length measurements in patient tissue specimens may have utility in risk assessment and as a prognostic biomarker. Highlighted throughout the article are some of the training and mentorship approaches employed by the late Dr. Coffey, former Director of Urologic Research at the Brady Urological Research Institute, which inspired new research ideas, team science, and discovery.
Collapse
Affiliation(s)
- Alan K Meeker
- The Department of Pathology, Johns Hopkins School of MedicineBaltimore, MD, USA
- The James Buchanan Brady Urologic Institute and Department of Urology, Johns Hopkins School of MedicineBaltimore, MD, USA
- The Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, School of Medicine, Johns Hopkins UniversityBaltimore, MD, USA
- The Department of Biochemistry and Molecular Biology, The Johns Hopkins Bloomberg School of Public HealthBaltimore, MD, USA
| |
Collapse
|
152
|
Singh KB, Hahm ER, Rigatti LH, Normolle DP, Yuan JM, Singh SV. Inhibition of Glycolysis in Prostate Cancer Chemoprevention by Phenethyl Isothiocyanate. Cancer Prev Res (Phila) 2018; 11:337-346. [PMID: 29545400 DOI: 10.1158/1940-6207.capr-17-0389] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2017] [Revised: 01/30/2018] [Accepted: 03/01/2018] [Indexed: 12/17/2022]
Abstract
We have shown previously that dietary administration of phenethyl isothiocyanate (PEITC), a small molecule from edible cruciferous vegetables, significantly decreases the incidence of poorly differentiated prostate cancer in Transgenic Adenocarcinoma of Mouse Prostate (TRAMP) mice without any side effects. In this study, we investigated the role of c-Myc-regulated glycolysis in prostate cancer chemoprevention by PEITC. Exposure of LNCaP (androgen-responsive) and 22Rv1 (castration-resistant) human prostate cancer cells to PEITC resulted in suppression of expression as well as transcriptional activity of c-Myc. Prostate cancer cell growth inhibition by PEITC was significantly attenuated by stable overexpression of c-Myc. Analysis of the RNA-Seq data from The Cancer Genome Atlas indicated a significant positive association between Myc expression and gene expression of many glycolysis-related genes, including hexokinase II and lactate dehydrogenase A Expression of these enzyme proteins and lactate levels were decreased upon PEITC treatment in prostate cancer cells, and these effects were significantly attenuated by ectopic expression of c-Myc. A normal prostate stromal cell line (PrSC) was resistant to lactic acid suppression by PEITC treatment. Prostate cancer chemoprevention by PEITC in TRAMP mice was associated with a significant decrease in plasma lactate and pyruvate levels. However, a 1-week intervention with 10 mg PEITC (orally, 4 times/day) was not sufficient to decrease lactate levels in the serum of human subjects. These results indicated that although prostate cancer prevention by PEITC in TRAMP mice was associated with suppression of glycolysis, longer than 1-week intervention might be necessary to observe such an effect in human subjects. Cancer Prev Res; 11(6); 337-46. ©2018 AACR.
Collapse
Affiliation(s)
- Krishna B Singh
- Department of Pharmacology & Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Eun-Ryeong Hahm
- Department of Pharmacology & Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Lora H Rigatti
- UPMC Hillman Cancer Center, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Daniel P Normolle
- UPMC Hillman Cancer Center, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania.,Department of Biostatistics, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Jian-Min Yuan
- UPMC Hillman Cancer Center, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania.,Department of Epidemiology, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Shivendra V Singh
- Department of Pharmacology & Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania. .,UPMC Hillman Cancer Center, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| |
Collapse
|
153
|
The histone demethylase KDM3A regulates the transcriptional program of the androgen receptor in prostate cancer cells. Oncotarget 2018; 8:30328-30343. [PMID: 28416760 PMCID: PMC5444746 DOI: 10.18632/oncotarget.15681] [Citation(s) in RCA: 72] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2016] [Accepted: 09/09/2016] [Indexed: 01/07/2023] Open
Abstract
The lysine demethylase 3A (KDM3A, JMJD1A or JHDM2A) controls transcriptional networks in a variety of biological processes such as spermatogenesis, metabolism, stem cell activity, and tumor progression. We matched transcriptomic and ChIP-Seq profiles to decipher a genome-wide regulatory network of epigenetic control by KDM3A in prostate cancer cells. ChIP-Seq experiments monitoring histone 3 lysine 9 (H3K9) methylation marks show global histone demethylation effects of KDM3A. Combined assessment of histone demethylation events and gene expression changes presented major transcriptional activation suggesting that distinct oncogenic regulators may synergize with the epigenetic patterns by KDM3A. Pathway enrichment analysis of cells with KDM3A knockdown prioritized androgen signaling indicating that KDM3A plays a key role in regulating androgen receptor activity. Matched ChIP-Seq and knockdown experiments of KDM3A in combination with ChIP-Seq of the androgen receptor resulted in a gain of H3K9 methylation marks around androgen receptor binding sites of selected transcriptional targets in androgen signaling including positive regulation of KRT19, NKX3-1, KLK3, NDRG1, MAF, CREB3L4, MYC, INPP4B, PTK2B, MAPK1, MAP2K1, IGF1, E2F1, HSP90AA1, HIF1A, and ACSL3. The cancer systems biology analysis of KDM3A-dependent genes identifies an epigenetic and transcriptional network in androgen response, hypoxia, glycolysis, and lipid metabolism. Genome-wide ChIP-Seq data highlights specific gene targets and the ability of epigenetic master regulators to control oncogenic pathways and cancer progression.
Collapse
|
154
|
Pettersson A, Gerke T, Penney KL, Lis RT, Stack EC, Pértega-Gomes N, Zadra G, Tyekucheva S, Giovannucci EL, Mucci LA, Loda M. MYC Overexpression at the Protein and mRNA Level and Cancer Outcomes among Men Treated with Radical Prostatectomy for Prostate Cancer. Cancer Epidemiol Biomarkers Prev 2018; 27:201-207. [PMID: 29141848 PMCID: PMC5831163 DOI: 10.1158/1055-9965.epi-17-0637] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2017] [Revised: 09/22/2017] [Accepted: 11/09/2017] [Indexed: 12/16/2022] Open
Abstract
Background: The proto-oncogene MYC is implicated in prostate cancer progression. Whether MYC tumor expression at the protein or mRNA level is associated with poorer prognosis has not been well studied.Methods: We conducted a cohort study including 634 men from the Physicians' Health Study and Health Professionals Follow-up Study treated with radical prostatectomy for prostate cancer in 1983-2004 and followed up for a median of 13.7 years. MYC protein expression was evaluated using IHC, and we used Cox regression to calculate HRs and 95% confidence intervals (CIs) of its association with lethal prostate cancer (distant metastases/prostate cancer-related death). We assessed the association between MYC mRNA expression and lethal prostate cancer in a case-control study, including 113 lethal cases and 291 indolent controls.Results: MYC nuclear protein expression was present in 97% of tumors. MYC protein expression was positively correlated with tumor proliferation rate (r = 0.37; P < 0.001) and negatively correlated with apoptotic count (r = -0.17; P < 0.001). There were no significant associations between MYC protein expression and stage, grade, or PSA level at diagnosis. The multivariable HR for lethal prostate cancer among men in the top versus bottom quartile of MYC protein expression was 1.09 (95% CI, 0.50-2.35). There was no significant association between MYC mRNA expression and lethal prostate cancer.Conclusions: Neither MYC protein overexpression nor MYC mRNA overexpression are strong prognostic markers in men treated with radical prostatectomy for prostate cancer.Impact: This is the largest study to examine the prognostic role of MYC protein and mRNA expression in prostate cancer. Cancer Epidemiol Biomarkers Prev; 27(2); 201-7. ©2017 AACR.
Collapse
Affiliation(s)
- Andreas Pettersson
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
- Clinical Epidemiology Unit, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
| | - Travis Gerke
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
- Department of Cancer Epidemiology, Moffitt Cancer Center, Tampa, Florida
| | - Kathryn L Penney
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Rosina T Lis
- Department of Oncologic Pathology, Dana-Farber Cancer Institute, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Edward C Stack
- Department of Oncologic Pathology, Dana-Farber Cancer Institute, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Nelma Pértega-Gomes
- Department of Oncologic Pathology, Dana-Farber Cancer Institute, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Giorgia Zadra
- Department of Oncologic Pathology, Dana-Farber Cancer Institute, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Svitlana Tyekucheva
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
- Departments of Biostatistics and Computational Biology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Edward L Giovannucci
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
| | - Lorelei A Mucci
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
| | - Massimo Loda
- Department of Oncologic Pathology, Dana-Farber Cancer Institute, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts.
| |
Collapse
|
155
|
Rickman DS, Schulte JH, Eilers M. The Expanding World of N-MYC–Driven Tumors. Cancer Discov 2018; 8:150-163. [DOI: 10.1158/2159-8290.cd-17-0273] [Citation(s) in RCA: 169] [Impact Index Per Article: 24.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2017] [Revised: 08/04/2017] [Accepted: 10/18/2017] [Indexed: 11/16/2022]
|
156
|
Chua CW, Epsi NJ, Leung EY, Xuan S, Lei M, Li BI, Bergren SK, Hibshoosh H, Mitrofanova A, Shen MM. Differential requirements of androgen receptor in luminal progenitors during prostate regeneration and tumor initiation. eLife 2018; 7:28768. [PMID: 29334357 PMCID: PMC5807048 DOI: 10.7554/elife.28768] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2017] [Accepted: 01/12/2018] [Indexed: 12/16/2022] Open
Abstract
Master regulatory genes of tissue specification play key roles in stem/progenitor cells and are often important in cancer. In the prostate, androgen receptor (AR) is a master regulator essential for development and tumorigenesis, but its specific functions in prostate stem/progenitor cells have not been elucidated. We have investigated AR function in CARNs (CAstration-Resistant Nkx3.1-expressing cells), a luminal stem/progenitor cell that functions in prostate regeneration. Using genetically--engineered mouse models and novel prostate epithelial cell lines, we find that progenitor properties of CARNs are largely unaffected by AR deletion, apart from decreased proliferation in vivo. Furthermore, AR loss suppresses tumor formation after deletion of the Pten tumor suppressor in CARNs; however, combined Pten deletion and activation of oncogenic Kras in AR-deleted CARNs result in tumors with focal neuroendocrine differentiation. Our findings show that AR modulates specific progenitor properties of CARNs, including their ability to serve as a cell of origin for prostate cancer.
Collapse
Affiliation(s)
- Chee Wai Chua
- Department of Medicine, Columbia University Medical Center, New York, United States.,Department of Genetics and Development, Columbia University Medical Center, New York, United States.,Department of Urology, Columbia University Medical Center, New York, United States.,Department of Systems Biology, Columbia University Medical Center, New York, United States.,Herbert Irving Comprehensive Cancer Center, Columbia University Medical Center, New York, United States
| | - Nusrat J Epsi
- Department of Health Informatics, Rutgers School of Health Professions, Rutgers, The State University of New Jersey, Newark, United States.,Rutgers Biomedical and Health Sciences, Rutgers, The State University of New Jersey, Newark, United States
| | - Eva Y Leung
- Department of Medicine, Columbia University Medical Center, New York, United States.,Department of Genetics and Development, Columbia University Medical Center, New York, United States.,Department of Urology, Columbia University Medical Center, New York, United States.,Department of Systems Biology, Columbia University Medical Center, New York, United States.,Herbert Irving Comprehensive Cancer Center, Columbia University Medical Center, New York, United States
| | - Shouhong Xuan
- Department of Medicine, Columbia University Medical Center, New York, United States.,Department of Genetics and Development, Columbia University Medical Center, New York, United States.,Department of Urology, Columbia University Medical Center, New York, United States.,Department of Systems Biology, Columbia University Medical Center, New York, United States.,Herbert Irving Comprehensive Cancer Center, Columbia University Medical Center, New York, United States
| | - Ming Lei
- Department of Medicine, Columbia University Medical Center, New York, United States.,Department of Genetics and Development, Columbia University Medical Center, New York, United States.,Department of Urology, Columbia University Medical Center, New York, United States.,Department of Systems Biology, Columbia University Medical Center, New York, United States.,Herbert Irving Comprehensive Cancer Center, Columbia University Medical Center, New York, United States
| | - Bo I Li
- Department of Medicine, Columbia University Medical Center, New York, United States.,Department of Genetics and Development, Columbia University Medical Center, New York, United States.,Department of Urology, Columbia University Medical Center, New York, United States.,Department of Systems Biology, Columbia University Medical Center, New York, United States.,Herbert Irving Comprehensive Cancer Center, Columbia University Medical Center, New York, United States
| | - Sarah K Bergren
- Department of Medicine, Columbia University Medical Center, New York, United States.,Department of Genetics and Development, Columbia University Medical Center, New York, United States.,Department of Urology, Columbia University Medical Center, New York, United States.,Department of Systems Biology, Columbia University Medical Center, New York, United States.,Herbert Irving Comprehensive Cancer Center, Columbia University Medical Center, New York, United States
| | - Hanina Hibshoosh
- Herbert Irving Comprehensive Cancer Center, Columbia University Medical Center, New York, United States.,Department of Pathology and Cell Biology, Columbia University Medical Center, New York, United States
| | - Antonina Mitrofanova
- Department of Health Informatics, Rutgers School of Health Professions, Rutgers, The State University of New Jersey, Newark, United States.,Rutgers Biomedical and Health Sciences, Rutgers, The State University of New Jersey, Newark, United States
| | - Michael M Shen
- Department of Medicine, Columbia University Medical Center, New York, United States.,Department of Genetics and Development, Columbia University Medical Center, New York, United States.,Department of Urology, Columbia University Medical Center, New York, United States.,Department of Systems Biology, Columbia University Medical Center, New York, United States.,Herbert Irving Comprehensive Cancer Center, Columbia University Medical Center, New York, United States
| |
Collapse
|
157
|
Netto GJ, Eich ML, Varambally S. Prostate Cancer: An Update on Molecular Pathology with Clinical Implications. EUR UROL SUPPL 2017. [DOI: 10.1016/j.eursup.2017.10.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
|
158
|
Lu Y, Hu Z, Mangala LS, Stine ZE, Hu X, Jiang D, Xiang Y, Zhang Y, Pradeep S, Rodriguez-Aguayo C, Lopez-Berestein G, DeMarzo AM, Sood AK, Zhang L, Dang CV. MYC Targeted Long Noncoding RNA DANCR Promotes Cancer in Part by Reducing p21 Levels. Cancer Res 2017; 78:64-74. [PMID: 29180471 DOI: 10.1158/0008-5472.can-17-0815] [Citation(s) in RCA: 75] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2017] [Revised: 09/20/2017] [Accepted: 11/03/2017] [Indexed: 01/10/2023]
Abstract
The MYC oncogene broadly promotes transcription mediated by all nuclear RNA polymerases, thereby acting as a positive modifier of global gene expression. Here, we report that MYC stimulates the transcription of DANCR, a long noncoding RNA (lncRNA) that is widely overexpressed in human cancer. We identified DANCR through its overexpression in a transgenic model of MYC-induced lymphoma, but found that it was broadly upregulated in many human cancer cell lines and cancers, including most notably in prostate and ovarian cancers. Mechanistic investigations indicated that DANCR limited the expression of cell-cycle inhibitor p21 (CDKN1A) and that the inhibitory effects of DANCR loss on cell proliferation could be partially rescued by p21 silencing. In a xenograft model of human ovarian cancer, a nanoparticle-mediated siRNA strategy to target DANCR in vivo was sufficient to strongly inhibit tumor growth. Our observations expand knowledge of how MYC drives cancer cell proliferation by identifying DANCR as a critical lncRNA widely overexpressed in human cancers.Significance: These findings expand knowledge of how MYC drives cancer cell proliferation by identifying an oncogenic long noncoding RNA that is widely overexpressed in human cancers. Cancer Res; 78(1); 64-74. ©2017 AACR.
Collapse
Affiliation(s)
- Yunqi Lu
- Abramson Family Cancer Research Institute, Abramson Cancer Center and Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Zhongyi Hu
- Center for Research on Reproduction and Women's Health, and Department of Obstetrics and Gynecology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Lingegowda S Mangala
- Department of Gynecologic Oncology, and Center for RNA Interference and Non-Coding RNA, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Zachary E Stine
- Abramson Family Cancer Research Institute, Abramson Cancer Center and Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Xiaowen Hu
- Center for Research on Reproduction and Women's Health, and Department of Obstetrics and Gynecology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Dahai Jiang
- Department of Gynecologic Oncology, and Center for RNA Interference and Non-Coding RNA, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Yan Xiang
- Abramson Family Cancer Research Institute, Abramson Cancer Center and Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Youyou Zhang
- Center for Research on Reproduction and Women's Health, and Department of Obstetrics and Gynecology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Sunila Pradeep
- Department of Gynecologic Oncology, and Center for RNA Interference and Non-Coding RNA, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Cristian Rodriguez-Aguayo
- Department of Experimental Therapeutics, and Center for RNA Interference and Non-Coding RNA, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Gabriel Lopez-Berestein
- Department of Experimental Therapeutics, and Center for RNA Interference and Non-Coding RNA, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Angelo M DeMarzo
- Departments of Pathology, Urology and Oncology, The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Anil K Sood
- Department of Gynecologic Oncology, and Center for RNA Interference and Non-Coding RNA, University of Texas MD Anderson Cancer Center, Houston, Texas.
| | - Lin Zhang
- Center for Research on Reproduction and Women's Health, and Department of Obstetrics and Gynecology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania.
| | - Chi V Dang
- Abramson Family Cancer Research Institute, Abramson Cancer Center and Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania. .,Ludwig Institute for Cancer Research, New York, New York.,The Wistar Institute, Philadelphia, Pennsylvania
| |
Collapse
|
159
|
Li X, Wu JB, Li Q, Shigemura K, Chung LWK, Huang WC. SREBP-2 promotes stem cell-like properties and metastasis by transcriptional activation of c-Myc in prostate cancer. Oncotarget 2017; 7:12869-84. [PMID: 26883200 PMCID: PMC4914327 DOI: 10.18632/oncotarget.7331] [Citation(s) in RCA: 79] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2015] [Accepted: 01/27/2016] [Indexed: 12/14/2022] Open
Abstract
Sterol regulatory element-binding protein-2 (SREBP-2) transcription factor mainly controls cholesterol biosynthesis and homeostasis in normal cells. The role of SREBP-2 in lethal prostate cancer (PCa) progression remains to be elucidated. Here, we showed that expression of SREBP-2 was elevated in advanced pathologic grade and metastatic PCa and significantly associated with poor clinical outcomes. Biofunctional analyses demonstrated that SREBP-2 induced PCa cell proliferation, invasion and migration. Furthermore, overexpression of SREBP-2 increased the PCa stem cell population, prostasphere-forming ability and tumor-initiating capability, whereas genetic silencing of SREBP-2 inhibited PCa cell growth, stemness, and xenograft tumor growth and metastasis. Clinical and mechanistic data showed that SREBP-2 was positively correlated with c-Myc and induced c-Myc activation by directly interacting with an SREBP-2-binding element in the 5′-flanking c-Myc promoter region to drive stemness and metastasis. Collectively, these clinical and experimental results reveal a novel role of SREBP-2 in the induction of a stem cell-like phenotype and PCa metastasis, which sheds light on translational potential by targeting SREBP-2 as a promising therapeutic approach in PCa.
Collapse
Affiliation(s)
- Xiangyan Li
- Uro-Oncology Research Program, Department of Medicine, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Jason Boyang Wu
- Uro-Oncology Research Program, Department of Medicine, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Qinlong Li
- Uro-Oncology Research Program, Department of Medicine, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA.,Department of Pathology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Katsumi Shigemura
- Department of Urology, Kobe University Graduate School of Medicine, Chuo-Ku, Kobe, Japan
| | - Leland W K Chung
- Uro-Oncology Research Program, Department of Medicine, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Wen-Chin Huang
- Uro-Oncology Research Program, Department of Medicine, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
| |
Collapse
|
160
|
Baena-Del Valle JA, Zheng Q, Esopi DM, Rubenstein M, Hubbard GK, Moncaliano MC, Hruszkewycz A, Vaghasia A, Yegnasubramanian S, Wheelan SJ, Meeker AK, Heaphy CM, Graham MK, De Marzo AM. MYC drives overexpression of telomerase RNA (hTR/TERC) in prostate cancer. J Pathol 2017; 244:11-24. [PMID: 28888037 DOI: 10.1002/path.4980] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2016] [Revised: 08/07/2017] [Accepted: 08/24/2017] [Indexed: 01/21/2023]
Abstract
Telomerase consists of at least two essential elements, an RNA component hTR or TERC that contains the template for telomere DNA addition and a catalytic reverse transcriptase (TERT). While expression of TERT has been considered the key rate-limiting component for telomerase activity, increasing evidence suggests an important role for the regulation of TERC in telomere maintenance and perhaps other functions in human cancer. By using three orthogonal methods including RNAseq, RT-qPCR, and an analytically validated chromogenic RNA in situ hybridization assay, we report consistent overexpression of TERC in prostate cancer. This overexpression occurs at the precursor stage (e.g. high-grade prostatic intraepithelial neoplasia or PIN) and persists throughout all stages of disease progression. Levels of TERC correlate with levels of MYC (a known driver of prostate cancer) in clinical samples and we also show the following: forced reductions of MYC result in decreased TERC levels in eight cancer cell lines (prostate, lung, breast, and colorectal); forced overexpression of MYC in PCa cell lines, and in the mouse prostate, results in increased TERC levels; human TERC promoter activity is decreased after MYC silencing; and MYC occupies the TERC locus as assessed by chromatin immunoprecipitation (ChIP). Finally, we show that knockdown of TERC by siRNA results in reduced proliferation of prostate cancer cell lines. These studies indicate that TERC is consistently overexpressed in all stages of prostatic adenocarcinoma and that its expression is regulated by MYC. These findings nominate TERC as a novel prostate cancer biomarker and therapeutic target. Copyright © 2017 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Javier A Baena-Del Valle
- Department of Pathology, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.,Department of Pathology and Laboratory Medicine, Fundacion Santa Fe De Bogota University Hospital, Bogota, DC, Colombia
| | - Qizhi Zheng
- Department of Pathology, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - David M Esopi
- Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Michael Rubenstein
- Department of Biological Sciences, University of Maryland, Baltimore County, Baltimore, Maryland, USA
| | - Gretchen K Hubbard
- Department of Pathology, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Maria C Moncaliano
- Department of Pathology, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Andrew Hruszkewycz
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, Maryland, USA
| | - Ajay Vaghasia
- Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Srinivasan Yegnasubramanian
- Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.,Departments of Urology and Oncology, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.,The Brady Urological Research Institute, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Sarah J Wheelan
- Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.,Departments of Urology and Oncology, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.,The Brady Urological Research Institute, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Alan K Meeker
- Department of Pathology, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.,Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.,The Brady Urological Research Institute, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Christopher M Heaphy
- Department of Pathology, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.,Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.,The Brady Urological Research Institute, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Mindy K Graham
- Department of Pathology, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.,Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.,The Brady Urological Research Institute, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Angelo M De Marzo
- Department of Pathology, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.,Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.,Departments of Urology and Oncology, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.,The Brady Urological Research Institute, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
161
|
Baena-Del Valle JA, Zheng Q, Hicks JL, Fedor H, Trock BJ, Morrissey C, Corey E, Cornish TC, Sfanos KS, De Marzo AM. Rapid Loss of RNA Detection by In Situ Hybridization in Stored Tissue Blocks and Preservation by Cold Storage of Unstained Slides. Am J Clin Pathol 2017; 148:398-415. [PMID: 29106457 DOI: 10.1093/ajcp/aqx094] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
OBJECTIVES Recent commercialization of methods for in situ hybridization using Z-pair probe/branched DNA amplification has led to increasing adoption of this technology for interrogating RNA expression in formalin-fixed, paraffin-embedded (FFPE) tissues. Current practice for FFPE block storage is to maintain them at room temperature, often for many years. METHODS To examine the effects of block storage time on FFPE tissues using a number of RNA in situ probes with the Advanced Cellular Diagnostic's RNAscope assay. RESULTS We report marked reductions in signals after 5 years and significant reductions often after 1 year. Furthermore, storing unstained slides cut from recent cases (<1 year old) at -20°C can preserve hybridization signals significantly better than storing the blocks at room temperature and cutting the slides fresh when needed. CONCLUSIONS We submit that the standard practice of storing FFPE tissue blocks at room temperature should be reevaluated to better preserve RNA for in situ hybridization.
Collapse
Affiliation(s)
- Javier A Baena-Del Valle
- From the Department of Pathology
- Department of Pathology and Laboratory Medicine, Fundacion Santa Fe de Bogota University Hospital, Bogota DC, Colombia
| | | | | | | | - Bruce J Trock
- Departments of Urology and Oncology
- Sidney Kimmel Comprehensive Cancer Center
- The Brady Urological Research Institute, The Johns Hopkins University School of Medicine, Baltimore, MD
| | - Colm Morrissey
- Department of Urology, University of Washington, Seattle
| | - Eva Corey
- Department of Urology, University of Washington, Seattle
| | - Toby C Cornish
- From the Department of Pathology
- Department of Pathology, University of Colorado School of Medicine, Aurora
| | - Karen S Sfanos
- From the Department of Pathology
- Departments of Urology and Oncology
- Sidney Kimmel Comprehensive Cancer Center
- The Brady Urological Research Institute, The Johns Hopkins University School of Medicine, Baltimore, MD
| | - Angelo M De Marzo
- From the Department of Pathology
- Departments of Urology and Oncology
- Sidney Kimmel Comprehensive Cancer Center
- The Brady Urological Research Institute, The Johns Hopkins University School of Medicine, Baltimore, MD
| |
Collapse
|
162
|
Sfanos KS, Yegnasubramanian S, Nelson WG, De Marzo AM. The inflammatory microenvironment and microbiome in prostate cancer development. Nat Rev Urol 2017; 15:11-24. [PMID: 29089606 DOI: 10.1038/nrurol.2017.167] [Citation(s) in RCA: 314] [Impact Index Per Article: 39.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Chronic inflammation promotes the development of several types of solid cancers and might contribute to prostate carcinogenesis. This hypothesis partly originates in the frequent observation of inflammatory cells in the prostate microenvironment of adult men. Inflammation is associated with putative prostate cancer precursor lesions, termed proliferative inflammatory atrophy. Inflammation might drive prostate carcinogenesis via oxidative stress and generation of reactive oxygen species that induce mutagenesis. Additionally, inflammatory stress might cause epigenetic alterations that promote neoplastic transformation. Proliferative inflammatory atrophy is enriched for proliferative luminal epithelial cells of intermediate phenotype that might be prone to genomic alterations leading to prostatic intraepithelial neoplasia and prostate cancer. Studies in animals suggest that inflammatory changes in the prostate microenvironment contribute to reprogramming of prostate epithelial cells, a possible step in tumour initiation. Prostatic infection, concurrent with epithelial barrier disruption, might be a key driver of an inflammatory microenvironment; the discovery of a urinary microbiome indicates a potential source of frequent exposure of the prostate to a diverse number of microorganisms. Hence, current evidence suggests that inflammation and atrophy are involved in prostate carcinogenesis and suggests a role for the microbiome in establishing an inflammatory prostate microenvironment that might promote prostate cancer development and progression.
Collapse
Affiliation(s)
- Karen S Sfanos
- Department of Pathology, Johns Hopkins University School of Medicine.,Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Johns Hopkins University School of Medicine, 1550 Orleans Street, Baltimore, Maryland 21231, USA.,Department of Urology, James Buchanan Brady Urological Institute, Johns Hopkins University School of Medicine, 600 North Wolfe Street, Baltimore, Maryland 21287, USA
| | - Srinivasan Yegnasubramanian
- Department of Pathology, Johns Hopkins University School of Medicine.,Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Johns Hopkins University School of Medicine, 1550 Orleans Street, Baltimore, Maryland 21231, USA
| | - William G Nelson
- Department of Pathology, Johns Hopkins University School of Medicine.,Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Johns Hopkins University School of Medicine, 1550 Orleans Street, Baltimore, Maryland 21231, USA.,Department of Urology, James Buchanan Brady Urological Institute, Johns Hopkins University School of Medicine, 600 North Wolfe Street, Baltimore, Maryland 21287, USA
| | - Angelo M De Marzo
- Department of Pathology, Johns Hopkins University School of Medicine.,Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Johns Hopkins University School of Medicine, 1550 Orleans Street, Baltimore, Maryland 21231, USA.,Department of Urology, James Buchanan Brady Urological Institute, Johns Hopkins University School of Medicine, 600 North Wolfe Street, Baltimore, Maryland 21287, USA
| |
Collapse
|
163
|
Chang C, Liu J, He W, Qu M, Huang X, Deng Y, Shen L, Zhao X, Guo H, Jiang J, Fu XY, Huang R, Zhang D, Yan J. A regulatory circuit HP1γ/miR-451a/c-Myc promotes prostate cancer progression. Oncogene 2017; 37:415-426. [PMID: 28967902 DOI: 10.1038/onc.2017.332] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2016] [Revised: 06/08/2017] [Accepted: 07/13/2017] [Indexed: 02/06/2023]
Abstract
Heterochromatin protein 1γ (HP1γ) has been implicated in carcinogenesis of various cancer types. However, the role of HP1γ in prostate cancer (PCa) progression and the underlying molecular mechanisms remain largely unknown. We found that HP1γ is upregulated in PCa and elevated levels of HP1γ in PCa predict poor outcome. In addition, depletion of HP1γ in PCa cells not only repressed proliferation and induced apoptosis but also impaired tumorigenicity. We also found that c-Myc was capable of upregulating HP1γ by directly binding to the E-box element in the first intron of HP1γ gene, and the upregulated HP1γ, in turn, repressed the expression of miR-451a by enhancing H3K9 methylation at the promoter region of miR-451a. Furthermore, reduction of miR-451a significantly reversed HP1γ loss-induced PCa cell apoptosis, whereas miR-451a overexpression repressed cell survival by targeting and downregulating c-Myc. The association among c-Myc, HP1γ and miR-451a was further confirmed in human clinical samples. Therefore, we propose that an HP1γ/miR-451a/c-Myc regulatory circuitry exists in PCa cells and this circuit has a crucial role in PCa progression.
Collapse
Affiliation(s)
- C Chang
- State Key Laboratory of Pharmaceutical Biotechnology and MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Nanjing University, Nanjing, China.,Collaborative Innovation Center of Genetics and Development, Shanghai, China
| | - J Liu
- State Key Laboratory of Pharmaceutical Biotechnology and MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Nanjing University, Nanjing, China.,Collaborative Innovation Center of Genetics and Development, Shanghai, China
| | - W He
- State Key Laboratory of Pharmaceutical Biotechnology and MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Nanjing University, Nanjing, China
| | - M Qu
- Department of Bioscience and Bioengineering, School of Chemistry and Life Science, Jinling College of Nanjing University, Nanjing, China
| | - X Huang
- State Key Laboratory of Pharmaceutical Biotechnology and MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Nanjing University, Nanjing, China.,Collaborative Innovation Center of Genetics and Development, Shanghai, China
| | - Y Deng
- Department of Urology, Nanjing Drum Tower Hospital, Nanjing University Medical School, Institute of Urology, Nanjing University, Nanjing, China
| | - L Shen
- State Key Laboratory of Pharmaceutical Biotechnology and MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Nanjing University, Nanjing, China.,Collaborative Innovation Center of Genetics and Development, Shanghai, China
| | - X Zhao
- Department of Urology, Nanjing Drum Tower Hospital, Nanjing University Medical School, Institute of Urology, Nanjing University, Nanjing, China
| | - H Guo
- Department of Urology, Nanjing Drum Tower Hospital, Nanjing University Medical School, Institute of Urology, Nanjing University, Nanjing, China
| | - J Jiang
- Department of Urology, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing, China
| | - X Y Fu
- State Key Laboratory of Pharmaceutical Biotechnology and MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Nanjing University, Nanjing, China
| | - R Huang
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - D Zhang
- Department of Bio-Medical Sciences, Philadelphia College of Osteopathic Medicine, Philadelphia, PA, USA
| | - J Yan
- State Key Laboratory of Pharmaceutical Biotechnology and MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Nanjing University, Nanjing, China.,Collaborative Innovation Center of Genetics and Development, Shanghai, China.,State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| |
Collapse
|
164
|
Melis MHM, Nevedomskaya E, van Burgsteden J, Cioni B, van Zeeburg HJT, Song JY, Zevenhoven J, Hawinkels LJAC, de Visser KE, Bergman AM. The adaptive immune system promotes initiation of prostate carcinogenesis in a human c-Myc transgenic mouse model. Oncotarget 2017; 8:93867-93877. [PMID: 29212195 PMCID: PMC5706841 DOI: 10.18632/oncotarget.21305] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2017] [Accepted: 08/26/2017] [Indexed: 12/14/2022] Open
Abstract
Increasing evidence from epidemiological and pathological studies suggests a role of the immune system in the initiation and progression of multiple cancers, including prostate cancer. Reports on the contribution of the adaptive immune system are contradictive, since both suppression and acceleration of disease development have been reported. This study addresses the functional role of lymphocytes in prostate cancer development using a genetically engineered mouse model (GEMM) of human c-Myc driven prostate cancer (Hi-Myc mice) combined with B and T cell deficiency (RAG1-/- mice). From a pre-cancerous stage on, Hi-Myc mice showed higher accumulation of immune cells in their prostates then wild-type mice, of which macrophages were the most abundant. The onset of invasive adenocarcinoma was delayed in Hi-MycRAG1-/- compared to Hi-Myc mice and associated with decreased infiltration of leukocytes into the prostate. In addition, lower levels of the cytokines CXCL2, CCL5 and TGF-β1 were detected in Hi-MycRAG1-/- compared to Hi-Myc mouse prostates. These results from a GEMM of prostate cancer provide new insights into the promoting role of the adaptive immune system in prostate cancer development. Our findings indicate that the endogenous adaptive immune system does not protect against de novo prostate carcinogenesis in Hi-Myc transgenic mice, but rather accelerates the formation of invasive adenocarcinomas. This may have implications for the development of novel treatment strategies.
Collapse
Affiliation(s)
- Monique H M Melis
- Division of Molecular Genetics, Netherlands Cancer Institute, The Netherlands
| | | | | | - Bianca Cioni
- Division of Molecular Genetics, Netherlands Cancer Institute, The Netherlands
| | | | - Ji-Ying Song
- Division of Experimental Animal Pathology, Netherlands Cancer Institute, The Netherlands
| | - John Zevenhoven
- Division of Molecular Genetics, Netherlands Cancer Institute, The Netherlands
| | - Lukas J A C Hawinkels
- Division of Gastroenterology-Hepatology and Molecular Cell biology, Leiden university medical center, (LUMC), Netherlands
| | - Karin E de Visser
- Division of Immunology, Netherlands Cancer Institute, The Netherlands
| | - Andries M Bergman
- Division of Molecular Genetics, Netherlands Cancer Institute, The Netherlands.,Division of Medical Oncology, Netherlands Cancer Institute, The Netherlands
| |
Collapse
|
165
|
Nowacka-Zawisza M, Wiśnik E. DNA methylation and histone modifications as epigenetic regulation in prostate cancer (Review). Oncol Rep 2017; 38:2587-2596. [PMID: 29048620 DOI: 10.3892/or.2017.5972] [Citation(s) in RCA: 82] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2017] [Accepted: 07/24/2017] [Indexed: 11/06/2022] Open
Abstract
Prostate cancer is the second most commonly diagnosed cancer in men in Poland after lung cancer and the third leading cause of cancer-related mortality after lung and colon cancer. The etiology of most cases of prostate cancer are not fully known, and therefore it is essential to search for the molecular basis of prostate cancer and markers for the early diagnosis of this type of cancer. Epigenetics deals with changes in gene expression that are not determined by changes in the DNA sequence. Epigenetic changes refer to changes in the structure of DNA, which are the result of DNA modification after replication and/or post-translational modification of proteins associated with DNA. In contrast to mutations, epigenetic changes are reversible and occur very rapidly. The major epigenetic mechanisms include DNA methylation, modification of histone proteins, chemical modification and chromatin remodeling changes in gene expression caused by microRNAs (miRNAs). Epigenetic changes play an important role in malignant transformation and can be specific to types of cancers including prostate cancer.
Collapse
Affiliation(s)
- Maria Nowacka-Zawisza
- Department of Cytobiochemistry, Faculty of Biology and Environmental Protection, University of Lodz, 90-236 Lodz, Poland
| | - Ewelina Wiśnik
- Department of Cytobiochemistry, Faculty of Biology and Environmental Protection, University of Lodz, 90-236 Lodz, Poland
| |
Collapse
|
166
|
Fu J, Liu Y, Wang X, Yuan B, Zhang Y. Role of DHX33 in c-Myc-induced cancers. Carcinogenesis 2017; 38:649-660. [PMID: 28498893 DOI: 10.1093/carcin/bgx041] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2016] [Accepted: 04/12/2017] [Indexed: 12/25/2022] Open
Abstract
Oncogene c-Myc is frequently amplified and activated in human cancers. Deregulation of c-Myc protein has been shown to occur in 30% of all human cancers, especially in hematopoietic malignancies. As a transcription factor, c-Myc has been shown to regulate up to 15% of all human genome genes, controlling diverse cellular activities including cell cycle, ribosome biogenesis, protein synthesis, metabolism, apoptosis and angiogenesis. In this report, we provide evidence that the RNA helicase DHX33 is a critical downstream target of c-Myc. Myc binds to DHX33 upstream promoter region and stimulates its transcription. Elevated DHX33 protein is pivotal for c-Myc to drive tumor formation. Knockdown of DHX33 to basal levels in c-Myc overexpressing cells significantly reduced cell proliferation, cell migration and anchorage-independent cell growth in vitro and in vivo. Additionally, we found that DHX33 promotes MMP9, MMP14 and urokinase-type plasminogen activator (PLAU) transcription by directly binding to their promoters, thus promoting cancer cell migration. DHX33 protein was overexpressed in a certain subset of human non-Hodgkin's lymphoma tissues. Finally, knockdown of DHX33 significantly inhibits the development of Myc-induced acute myeloid leukemia. Overall, our results implicate the important role for DHX33 in Myc-induced cancer and point toward its potential therapeutic value in Myc driven cancers.
Collapse
Affiliation(s)
- Jijun Fu
- Department of Biology, Southern University of Science and Technology, Shenzhen, China
| | - Yuchu Liu
- Department of Biology, Southern University of Science and Technology, Shenzhen, China
| | - Xingshun Wang
- Department of Biology, Southern University of Science and Technology, Shenzhen, China
| | - Baolei Yuan
- Department of Biology, Southern University of Science and Technology, Shenzhen, China
| | - Yandong Zhang
- Department of Biology, Southern University of Science and Technology, Shenzhen, China
| |
Collapse
|
167
|
The immunosuppressive effect of the tick protein, Salp15, is long-lasting and persists in a murine model of hematopoietic transplant. Sci Rep 2017; 7:10740. [PMID: 28878331 PMCID: PMC5587732 DOI: 10.1038/s41598-017-11354-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2017] [Accepted: 08/23/2017] [Indexed: 12/22/2022] Open
Abstract
Salp15, a salivary protein of Ixodes ticks, inhibits the activation of naïve CD4 T cells. Treatment with Salp15 results in the inhibition of early signaling events and the production of the autocrine growth factor, interleukin-2. The fate of the CD4 T cells activated in the presence of Salp15 or its long-term effects are, however, unknown. We now show that Salp15 binding to CD4 is persistent and induces a long-lasting immunomodulatory effect. The activity of Salp15 results in sustained diminished cross-antigenic antibody production even after interruption of the treatment with the protein. Transcriptionally, the salivary protein provokes an acute effect that includes known activation markers, such as Il2 or Cd44, and that fades over time. The long-term effects exerted by Salp15 do not involve the induction of either anergy traits nor increased populations of regulatory T cells. Similarly, the treatment with Salp15 does not result in B cell anergy or the generation of myeloid suppressor cells. However, Salp15 induces the increased expression of the ectoenzyme, CD73, in regulatory T cells and increased production of adenosine. Our study provides a profound characterization of the immunomodulatory activity of Salp15 and suggests that its long-term effects are due to the specific regulation of CD73.
Collapse
|
168
|
Geng C, Kaochar S, Li M, Rajapakshe K, Fiskus W, Dong J, Foley C, Dong B, Zhang L, Kwon OJ, Shah SS, Bolaki M, Xin L, Ittmann M, O’Malley BW, Coarfa C, Mitsiades N. SPOP regulates prostate epithelial cell proliferation and promotes ubiquitination and turnover of c-MYC oncoprotein. Oncogene 2017; 36:4767-4777. [PMID: 28414305 PMCID: PMC5887163 DOI: 10.1038/onc.2017.80] [Citation(s) in RCA: 91] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2016] [Revised: 01/16/2017] [Accepted: 02/26/2017] [Indexed: 12/13/2022]
Abstract
The E3 ubiquitin ligase adaptor speckle-type POZ protein (SPOP) is frequently dysregulated in prostate adenocarcinoma (PC), via either somatic mutations or mRNA downregulation, suggesting an important tumour suppressor function. To examine its physiologic role in the prostate epithelium in vivo, we generated mice with prostate-specific biallelic ablation of Spop. These mice exhibited increased prostate mass, prostate epithelial cell proliferation, and expression of c-MYC protein compared to littermate controls, and eventually developed prostatic intraepithelial neoplasia (PIN). We found that SPOPWT can physically interact with c-MYC protein and, upon exogenous expression in vitro, can promote c-MYC ubiquitination and degradation. This effect was attenuated in PC cells by introducing PC-associated SPOP mutants or upon knockdown of SPOP via short-hairpin-RNA, suggesting that SPOP inactivation directly increases c-MYC protein levels. Gene Set Enrichment Analysis revealed enrichment of Myc-induced genes in transcriptomic signatures associated with SPOPMT. Likewise, we observed strong inverse correlation between c-MYC activity and SPOP mRNA levels in two independent PC patient cohorts. The core SPOPMT;MYCHigh transcriptomic response, defined by the overlap between the SPOPMT and c-MYC transcriptomic programmes, was also associated with inferior clinical outcome in human PCs. Finally, the organoid-forming capacity of Spop-null murine prostate cells was more sensitive to c-MYC inhibition than that of Spop-WT cells, suggesting that c-MYC upregulation functionally contributes to the proliferative phenotype of Spop knock-out prostates. Taken together, our data highlight SPOP as an important regulator of luminal epithelial cell proliferation and c-MYC expression in prostate physiology, identify c-MYC as a novel bona fide SPOP substrate, and help explain the frequent inactivation of SPOP in human PC. We propose SPOPMT-induced stabilization of c-MYC protein as a novel mechanism that can increase total c-MYC levels in PC cells, in addition to amplification of c-MYC locus.
Collapse
Affiliation(s)
- Chuandong Geng
- Dept. of Medicine, Houston, TX 77030
- Dept. of Molecular and Cellular Biology, Houston, TX 77030
| | - Salma Kaochar
- Dept. of Medicine, Houston, TX 77030
- Dept. of Molecular and Cellular Biology, Houston, TX 77030
| | - Min Li
- Dept. of Medicine, Houston, TX 77030
- Dept. of Molecular and Cellular Biology, Houston, TX 77030
| | | | - Warren Fiskus
- Dept. of Medicine, Houston, TX 77030
- Dept. of Molecular and Cellular Biology, Houston, TX 77030
| | - Jianrong Dong
- Dept. of Molecular and Cellular Biology, Houston, TX 77030
| | - Christopher Foley
- Dept. of Medicine, Houston, TX 77030
- Dept. of Molecular and Cellular Biology, Houston, TX 77030
| | - Boming Dong
- Dept. of Medicine, Houston, TX 77030
- Dept. of Molecular and Cellular Biology, Houston, TX 77030
| | - Li Zhang
- Dept. of Molecular and Cellular Biology, Houston, TX 77030
| | - Oh-Joon Kwon
- Dept. of Molecular and Cellular Biology, Houston, TX 77030
| | - Shrijal S. Shah
- Dept. of Medicine, Houston, TX 77030
- Dept. of Molecular and Cellular Biology, Houston, TX 77030
| | - Menaka Bolaki
- Dept. of Medicine, Houston, TX 77030
- Dept. of Molecular and Cellular Biology, Houston, TX 77030
| | - Li Xin
- Dept. of Molecular and Cellular Biology, Houston, TX 77030
- Dan L. Duncan Cancer Center, Houston, TX 77030
| | - Michael Ittmann
- Dan L. Duncan Cancer Center, Houston, TX 77030
- Dept. of Pathology and Immunology and Michael E. DeBakey Veterans Affairs Medical Center, Houston, TX 77030
| | | | - Cristian Coarfa
- Dept. of Molecular and Cellular Biology, Houston, TX 77030
- Dan L. Duncan Cancer Center, Houston, TX 77030
| | - Nicholas Mitsiades
- Dept. of Medicine, Houston, TX 77030
- Dept. of Molecular and Cellular Biology, Houston, TX 77030
- Dan L. Duncan Cancer Center, Houston, TX 77030
- Center for Drug Discovery, Baylor College of Medicine, Houston, TX 77030
| |
Collapse
|
169
|
Abstract
CONTEXT - Precursor lesions of urologic malignancies are established histopathologic entities, which are important not only to recognize for clinical purposes, but also to further investigate at the molecular level in order to gain a better understanding of the pathogenesis of these malignancies. OBJECTIVE - To provide a brief overview of precursor lesions to the most common malignancies that develop within the genitourinary tract with a focus on their clinical implications, histologic features, and molecular characteristics. DATA SOURCES - Literature review from PubMed, urologic pathology textbooks, and the 4th edition of the World Health Organization Classification of Tumours of the Urinary System and Male Genital Organs. All photomicrographs were taken from cases seen at Weill Cornell Medicine or from the authors' personal slide collections. CONCLUSIONS - The clinical importance and histologic criteria are well established for the known precursor lesions of the most common malignancies throughout the genitourinary tract, but further investigation is warranted at the molecular level to better understand the pathogenesis of these lesions. Such investigation may lead to better risk stratification of patients and potentially novel treatments.
Collapse
|
170
|
Saha A, Ahn S, Blando J, Su F, Kolonin MG, DiGiovanni J. Proinflammatory CXCL12-CXCR4/CXCR7 Signaling Axis Drives Myc-Induced Prostate Cancer in Obese Mice. Cancer Res 2017; 77:5158-5168. [PMID: 28687617 DOI: 10.1158/0008-5472.can-17-0284] [Citation(s) in RCA: 72] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2017] [Revised: 06/07/2017] [Accepted: 07/03/2017] [Indexed: 01/11/2023]
Abstract
Obesity is a prognostic risk factor in the progression of prostate cancer; however, the molecular mechanisms involved are unclear. In this study, we provide preclinical proof of concept for the role of a proinflammatory CXCL12-CXCR4/CXCR7 signaling axis in an obesity-driven mouse model of myc-induced prostate cancer. Analysis of the stromal vascular fraction from periprostatic white adipose tissue from obese HiMyc mice at 6 months of age revealed a dramatic increase in mRNAs encoding various chemokines, cytokines, growth factors, and angiogenesis mediators, with CXCL12 among the most significantly upregulated genes. Immunofluorescence staining of ventral prostate tissue from obese HiMyc mice revealed high levels of CXCL12 in the stromal compartment as well as high staining for CXCR4 and CXCR7 in the epithelial compartment of tumors. Prostate cancer cell lines derived from HiMyc tumors (HMVP2 and derivative cell lines) displayed increased protein expression of both CXCR4 and CXCR7 compared with protein lysates from a nontumorigenic prostate epithelial cell line (NMVP cells). CXCL12 treatment stimulated migration and invasion of HMVP2 cells but not NMVP cells. These effects of CXCL12 on HMVP2 cells were inhibited by the CXCR4 antagonist AMD3100 as well as knockdown of either CXCR4 or CXCR7. CXCL12 treatment also produced rapid activation of STAT3, NFκB, and MAPK signaling in HMVP2 cells, which was again attenuated by either AMD3100 or knockdown of CXCR4 or CXCR7. Collectively, these data suggest that CXCL12 secreted by stromal cells activates invasiveness of prostate cancer cells and may play a role in driving tumor progression in obesity. Targeting the CXCL12-CXCR4/CXCR7 axis could lead to novel approaches for offsetting the effects of obesity on prostate cancer progression. Cancer Res; 77(18); 5158-68. ©2017 AACR.
Collapse
Affiliation(s)
- Achinto Saha
- Division of Pharmacology and Toxicology, Dell Pediatric Research Institute, The University of Texas at Austin, Austin, Texas
| | - Songyeon Ahn
- Division of Pharmacology and Toxicology, Dell Pediatric Research Institute, The University of Texas at Austin, Austin, Texas
| | - Jorge Blando
- Division of Pharmacology and Toxicology, Dell Pediatric Research Institute, The University of Texas at Austin, Austin, Texas
| | - Fei Su
- The Brown Foundation Institute of Molecular Medicine for the Prevention of Disease, The University of Texas Health Sciences Center at Houston, Houston, Texas
| | - Mikhail G Kolonin
- The Brown Foundation Institute of Molecular Medicine for the Prevention of Disease, The University of Texas Health Sciences Center at Houston, Houston, Texas
| | - John DiGiovanni
- Division of Pharmacology and Toxicology, Dell Pediatric Research Institute, The University of Texas at Austin, Austin, Texas.
| |
Collapse
|
171
|
Abstract
Aberrations in telomere biology are among the earliest events in prostate cancer tumorigenesis and continue during tumour progression. Substantial telomere shortening occurs in prostate cancer cells and high-grade prostatic intraepithelial neoplasia. Not all mechanisms of telomere shortening are understood, but oxidative stress from local inflammation might accelerate prostatic telomere loss. Critically short telomeres can drive the accumulation of tumour-promoting genomic alterations; however, continued telomere erosion is unsustainable and must be mitigated to ensure cancer cell survival and unlimited replication potential. Prostate cancers predominantly maintain telomeres by activating telomerase, but alternative mechanisms of telomere extension can occur in metastatic disease. Telomerase activity and telomere length assessment might be useful in prostate cancer diagnosis and prognosis. Telomere shortening in normal stromal cells has been associated with prostate cancer, whereas variable telomere lengths in prostate cancer cells and telomere shortening in cancer-associated stromal cells correlated with lethal disease. Single-agent telomerase-targeted treatments for solid cancers were ineffective in clinical trials but have not been investigated in prostate cancer and might be useful in combination with established regimens. Telomere-directed strategies have not been explored as extensively. Telomere deprotection strategies have the advantage of being effective in both telomerase-dependent and telomerase-independent cancers. Disruption of androgen receptor function in prostate cancer cells results in telomere dysfunction, indicating telomeres and telomerase as potential therapeutic targets in prostate cancer.
Collapse
|
172
|
White MA, Lin C, Rajapakshe K, Dong J, Shi Y, Tsouko E, Mukhopadhyay R, Jasso D, Dawood W, Coarfa C, Frigo DE. Glutamine Transporters Are Targets of Multiple Oncogenic Signaling Pathways in Prostate Cancer. Mol Cancer Res 2017; 15:1017-1028. [PMID: 28507054 DOI: 10.1158/1541-7786.mcr-16-0480] [Citation(s) in RCA: 70] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2016] [Revised: 04/11/2017] [Accepted: 05/09/2017] [Indexed: 01/07/2023]
Abstract
Despite the known importance of androgen receptor (AR) signaling in prostate cancer, the processes downstream of AR that drive disease development and progression remain poorly understood. This knowledge gap has thus limited the ability to treat cancer. Here, it is demonstrated that androgens increase the metabolism of glutamine in prostate cancer cells. This metabolism was required for maximal cell growth under conditions of serum starvation. Mechanistically, AR signaling promoted glutamine metabolism by increasing the expression of the glutamine transporters SLC1A4 and SLC1A5, genes commonly overexpressed in prostate cancer. Correspondingly, gene expression signatures of AR activity correlated with SLC1A4 and SLC1A5 mRNA levels in clinical cohorts. Interestingly, MYC, a canonical oncogene in prostate cancer and previously described master regulator of glutamine metabolism, was only a context-dependent regulator of SLC1A4 and SLC1A5 levels, being unable to regulate either transporter in PTEN wild-type cells. In contrast, rapamycin was able to decrease the androgen-mediated expression of SLC1A4 and SLC1A5 independent of PTEN status, indicating that mTOR complex 1 (mTORC1) was needed for maximal AR-mediated glutamine uptake and prostate cancer cell growth. Taken together, these data indicate that three well-established oncogenic drivers (AR, MYC, and mTOR) function by converging to collectively increase the expression of glutamine transporters, thereby promoting glutamine uptake and subsequent prostate cancer cell growth.Implications: AR, MYC, and mTOR converge to increase glutamine uptake and metabolism in prostate cancer through increasing the levels of glutamine transporters. Mol Cancer Res; 15(8); 1017-28. ©2017 AACR.
Collapse
Affiliation(s)
- Mark A White
- Center for Nuclear Receptors and Cell Signaling, Department of Biology and Biochemistry, University of Houston, Houston, Texas
| | - Chenchu Lin
- Center for Nuclear Receptors and Cell Signaling, Department of Biology and Biochemistry, University of Houston, Houston, Texas
| | - Kimal Rajapakshe
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas
| | - Jianrong Dong
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas
| | - Yan Shi
- Center for Nuclear Receptors and Cell Signaling, Department of Biology and Biochemistry, University of Houston, Houston, Texas
| | - Efrosini Tsouko
- Center for Nuclear Receptors and Cell Signaling, Department of Biology and Biochemistry, University of Houston, Houston, Texas
| | - Ratna Mukhopadhyay
- Center for Nuclear Receptors and Cell Signaling, Department of Biology and Biochemistry, University of Houston, Houston, Texas
| | - Diana Jasso
- Center for Nuclear Receptors and Cell Signaling, Department of Biology and Biochemistry, University of Houston, Houston, Texas
| | - Wajahat Dawood
- Center for Nuclear Receptors and Cell Signaling, Department of Biology and Biochemistry, University of Houston, Houston, Texas
| | - Cristian Coarfa
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas
| | - Daniel E Frigo
- Center for Nuclear Receptors and Cell Signaling, Department of Biology and Biochemistry, University of Houston, Houston, Texas. .,Molecular Medicine Program, The Houston Methodist Research Institute, Houston, Texas
| |
Collapse
|
173
|
Schuster DM, Nanni C, Fanti S. Evaluation of Prostate Cancer with Radiolabeled Amino Acid Analogs. J Nucl Med 2017; 57:61S-66S. [PMID: 27694174 DOI: 10.2967/jnumed.115.170209] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2016] [Accepted: 05/20/2016] [Indexed: 12/14/2022] Open
Abstract
Conventional imaging of prostate cancer has limitations related to the frequently indolent biology of the disease. PET is a functional imaging method that can exploit various aspects of tumor biology to enable greater detection of prostate cancer than can be provided by morphologic imaging alone. Radiotracers that are in use or under investigation for targeting salient features of prostate cancer include those directed to glucose, choline, acetate, prostate-specific membrane antigen, bombesin, and amino acids. The tumor imaging features of this last class of radiotracers mirror the upregulation of transmembrane amino acid transport that is necessary in carcinomas because of increased amino acid use for energy requirements and protein synthesis. Natural and synthetic amino acids radiolabeled for PET imaging have been investigated in prostate cancer patients. Early work with naturally occurring amino acid-derived radiotracers, such as l-11C-methionine and l-1-11C-5-hydroxytryptophan, demonstrated promising results, including greater sensitivity than 18F-FDG for intraprostatic and extraprostatic cancer detection. However, limitations with naturally occurring amino acid-derived compounds, including metabolism of the radiotracer itself, led to the development of synthetic amino acid radiotracers, which are not metabolized and therefore more accurately reflect transmembrane amino acid transport. Of the synthetic amino acid-derived PET radiotracers, anti-1-amino-3-18F-fluorocyclobutane-1-carboxylic acid (18F-FACBC or 18F-fluciclovine) has undergone the most promising translation to human use, including the availability of simplified radiosynthesis. Several studies have indicated advantageous biodistribution in the abdomen and pelvis with little renal excretion and bladder activity-characteristics beneficial for prostate cancer imaging. Studies have demonstrated improved lesion detection and diagnostic performance of 18F-fluciclovine in comparison with conventional imaging, especially for recurrent prostate cancer, although issues with nonspecific uptake limit the potential role of 18F-fluciclovine in the diagnosis of primary prostate cancer. Although work is ongoing, recently published intrapatient comparisons of 18F-fluciclovine with 11C-choline reported higher overall diagnostic performance of the former, especially for the detection of disease relapse. This review is aimed at providing a detailed overview of amino acid-derived PET compounds that have been studied for use in prostate cancer imaging.
Collapse
Affiliation(s)
- David M Schuster
- Department of Radiology and Imaging Sciences, Emory University, Atlanta, Georgia; and
| | - Cristina Nanni
- Department of Nuclear Medicine, Policlinico S. Orsola, University of Bologna, Bologna, Italy
| | - Stefano Fanti
- Department of Nuclear Medicine, Policlinico S. Orsola, University of Bologna, Bologna, Italy
| |
Collapse
|
174
|
Barfeld SJ, Urbanucci A, Itkonen HM, Fazli L, Hicks JL, Thiede B, Rennie PS, Yegnasubramanian S, DeMarzo AM, Mills IG. c-Myc Antagonises the Transcriptional Activity of the Androgen Receptor in Prostate Cancer Affecting Key Gene Networks. EBioMedicine 2017; 18:83-93. [PMID: 28412251 PMCID: PMC5405195 DOI: 10.1016/j.ebiom.2017.04.006] [Citation(s) in RCA: 80] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2017] [Accepted: 04/04/2017] [Indexed: 12/25/2022] Open
Abstract
Prostate cancer (PCa) is the most common non-cutaneous cancer in men. The androgen receptor (AR), a ligand-activated transcription factor, constitutes the main drug target for advanced cases of the disease. However, a variety of other transcription factors and signaling networks have been shown to be altered in patients and to influence AR activity. Amongst these, the oncogenic transcription factor c-Myc has been studied extensively in multiple malignancies and elevated protein levels of c-Myc are commonly observed in PCa. Its impact on AR activity, however, remains elusive. In this study, we assessed the impact of c-Myc overexpression on AR activity and transcriptional output in a PCa cell line model and validated the antagonistic effect of c-MYC on AR-targets in patient samples. We found that c-Myc overexpression partially reprogrammed AR chromatin occupancy and was associated with altered histone marks distribution, most notably H3K4me1 and H3K27me3. We found c-Myc and the AR co-occupy a substantial number of binding sites and these exhibited enhancer-like characteristics. Interestingly, c-Myc overexpression antagonised clinically relevant AR target genes. Therefore, as an example, we validated the antagonistic relationship between c-Myc and two AR target genes, KLK3 (alias PSA, prostate specific antigen), and Glycine N-Methyltransferase (GNMT), in patient samples. Our findings provide unbiased evidence that MYC overexpression deregulates the AR transcriptional program, which is thought to be a driving force in PCa. c-MYC and AR share one third of chromatin binding with enhancer-like features. c-MYC can repress the expression of a subset prostate cancer biomarkers, including PSA. c-MYC and AR antagonize the expression of, Glycine N-Methyltransferase (GNMT), responsible for sarcosine biosynthesis.
Prostate cancer is a heterogeneous disease. The most frequently used biomarker in clinical setting, a well described androgen receptor target gene, PSA, still performs poorly in stratifying patients at real risk of death due to the disease. Despite this, therapeutic approaches focus on suppressing androgen receptor signaling. However, this is only one of the recurrent alterations found in patients. This study focuses on c-MYC and the effects of its deregulation in advanced prostate cancer. We find that there is an inverse relationship between established biomarkers expression, including PSA. This inverse relationship could be used in clinics to select beneficial therapeutic approaches for a subset of prostate cancer cases.
Collapse
Affiliation(s)
- Stefan J Barfeld
- Centre for Molecular Medicine Norway (NCMM), Nordic EMBL Partnership, University of Oslo, Oslo, Norway.
| | - Alfonso Urbanucci
- Centre for Molecular Medicine Norway (NCMM), Nordic EMBL Partnership, University of Oslo, Oslo, Norway; Department of Molecular Oncology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway.
| | - Harri M Itkonen
- Centre for Molecular Medicine Norway (NCMM), Nordic EMBL Partnership, University of Oslo, Oslo, Norway
| | - Ladan Fazli
- The Vancouver Prostate Centre, University of British Columbia, Canada
| | | | - Bernd Thiede
- Department of Biosciences, University of Oslo, Oslo, Norway
| | - Paul S Rennie
- The Vancouver Prostate Centre, University of British Columbia, Canada
| | | | - Angelo M DeMarzo
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Ian G Mills
- Centre for Molecular Medicine Norway (NCMM), Nordic EMBL Partnership, University of Oslo, Oslo, Norway; Department of Molecular Oncology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway; PCUK/Movember Centre of Excellence, CCRCB, Queen's University, Belfast, UK.
| |
Collapse
|
175
|
McNair C, Urbanucci A, Comstock CES, Augello MA, Goodwin JF, Launchbury R, Zhao SG, Schiewer MJ, Ertel A, Karnes J, Davicioni E, Wang L, Wang Q, Mills IG, Feng FY, Li W, Carroll JS, Knudsen KE. Cell cycle-coupled expansion of AR activity promotes cancer progression. Oncogene 2017; 36:1655-1668. [PMID: 27669432 PMCID: PMC5364060 DOI: 10.1038/onc.2016.334] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2016] [Accepted: 08/03/2016] [Indexed: 12/13/2022]
Abstract
The androgen receptor (AR) is required for prostate cancer (PCa) survival and progression, and ablation of AR activity is the first line of therapeutic intervention for disseminated disease. While initially effective, recurrent tumors ultimately arise for which there is no durable cure. Despite the dependence of PCa on AR activity throughout the course of disease, delineation of the AR-dependent transcriptional network that governs disease progression remains elusive, and the function of AR in mitotically active cells is not well understood. Analyzing AR activity as a function of cell cycle revealed an unexpected and highly expanded repertoire of AR-regulated gene networks in actively cycling cells. New AR functions segregated into two major clusters: those that are specific to cycling cells and retained throughout the mitotic cell cycle ('Cell Cycle Common'), versus those that were specifically enriched in a subset of cell cycle phases ('Phase Restricted'). Further analyses identified previously unrecognized AR functions in major pathways associated with clinical PCa progression. Illustrating the impact of these unmasked AR-driven pathways, dihydroceramide desaturase 1 was identified as an AR-regulated gene in mitotically active cells that promoted pro-metastatic phenotypes, and in advanced PCa proved to be highly associated with development of metastases, recurrence after therapeutic intervention and reduced overall survival. Taken together, these findings delineate AR function in mitotically active tumor cells, thus providing critical insight into the molecular basis by which AR promotes development of lethal PCa and nominate new avenues for therapeutic intervention.
Collapse
Affiliation(s)
- C McNair
- Department of Cancer Biology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, USA
| | - A Urbanucci
- Prostate Cancer Research Group, Centre for Molecular Medicine Norway (NCMM), Nordic EMBL Partnership, University of Oslo and Oslo University Hospitals, Oslo, Norway
- Department of Molecular Oncology, Institute for Cancer Research, Oslo University Hospitals, Oslo, Norway
| | - C E S Comstock
- Department of Cancer Biology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, USA
| | - M A Augello
- Department of Cancer Biology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, USA
| | - J F Goodwin
- Department of Cancer Biology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, USA
| | - R Launchbury
- Cambridge Research Institute, Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK
| | - S G Zhao
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI, USA
| | - M J Schiewer
- Department of Cancer Biology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, USA
| | - A Ertel
- Department of Cancer Biology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, USA
| | - J Karnes
- Division of Biomedical Statistics and Informatics, Mayo Clinic College of Medicine, Rochester, MN, USA
| | | | - L Wang
- Division of Biomedical Statistics and Informatics, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - Q Wang
- Ohio State University College of Medicine, Columbus, OH, USA
| | - I G Mills
- Prostate Cancer Research Group, Centre for Molecular Medicine Norway (NCMM), Nordic EMBL Partnership, University of Oslo and Oslo University Hospitals, Oslo, Norway
- Department of Molecular Oncology, Institute for Cancer Research, Oslo University Hospitals, Oslo, Norway
- Prostate Cancer UK/Movember Centre of Excellence for Prostate Cancer Research, Centre for Cancer Research and Cell Biology, Queen's University Belfast, Belfast, UK
| | - F Y Feng
- Department of Radiation Oncology, Urology, and Medicine and Helen Diller Family Comprehensive Cancer Center, University of California at San Francisco, San Francisco, CA, USA
| | - W Li
- Dan L. Duncan Cancer Center and Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| | - J S Carroll
- Cambridge Research Institute, Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK
| | - K E Knudsen
- Department of Cancer Biology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, USA
| |
Collapse
|
176
|
Kumar B, Lupold SE. MicroRNA expression and function in prostate cancer: a review of current knowledge and opportunities for discovery. Asian J Androl 2017; 18:559-67. [PMID: 27056344 PMCID: PMC4955179 DOI: 10.4103/1008-682x.177839] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
MicroRNAs (miRNAs) are well-conserved noncoding RNAs that broadly regulate gene expression through posttranscriptional silencing of coding genes. Dysregulated miRNA expression in prostate and other cancers implicates their role in cancer biology. Moreover, functional studies provide support for the contribution of miRNAs to several key pathways in cancer initiation and progression. Comparative analyses of miRNA gene expression between malignant and nonmalignant prostate tissues, healthy controls and prostate cancer (PCa) patients, as well as less aggressive versus more aggressive disease indicate that miRNAs may be future diagnostic or prognostic biomarkers in tumor tissue, blood, or urine. Further, miRNAs may be future therapeutics or therapeutic targets. In this review, we examine the miRNAs most commonly observed to be de-regulated in PCa gene expression analyses and review the potential contribution of these miRNAs to important pathways in PCa initiation and progression.
Collapse
Affiliation(s)
- Binod Kumar
- The James Buchanan Brady Urological Institute, Department of Urology, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Shawn E Lupold
- The James Buchanan Brady Urological Institute, Department of Urology, Johns Hopkins School of Medicine, Baltimore, MD, USA
| |
Collapse
|
177
|
Rebello RJ, Pearson RB, Hannan RD, Furic L. Therapeutic Approaches Targeting MYC-Driven Prostate Cancer. Genes (Basel) 2017; 8:genes8020071. [PMID: 28212321 PMCID: PMC5333060 DOI: 10.3390/genes8020071] [Citation(s) in RCA: 77] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2016] [Revised: 02/06/2017] [Accepted: 02/09/2017] [Indexed: 02/02/2023] Open
Abstract
The transcript encoding the proto-oncogene MYC is commonly overexpressed in prostate cancer (PC). MYC protein abundance is also increased in the majority of cases of advanced and metastatic castrate-resistant PC (mCRPC). Accordingly, the MYC-directed transcriptional program directly contributes to PC by upregulating the expression of a number of pro-tumorigenic factors involved in cell growth and proliferation. A key cellular process downstream of MYC activity is the regulation of ribosome biogenesis which sustains tumor growth. MYC activity also cooperates with the dysregulation of the phosphoinositol-3-kinase (PI3K)/AKT/mTOR pathway to promote PC cell survival. Recent advances in the understanding of these interactions through the use of animal models have provided significant insight into the therapeutic efficacy of targeting MYC activity by interfering with its transcriptional program, and indirectly by targeting downstream cellular events linked to MYC transformation potential.
Collapse
Affiliation(s)
- Richard J Rebello
- Prostate Cancer Translational Research Laboratory, Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia.
- Cancer Program, Biomedicine Discovery Institute and Department of Anatomy & Developmental Biology, Monash University, Melbourne, VIC 3800, Australia.
| | - Richard B Pearson
- Oncogenic Signalling and Growth Control Program, Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia.
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, VIC 3010, Australia.
- Department of Biochemistry and Molecular Biology, University of Melbourne, Parkville, VIC 3010, Australia.
- Department of Biochemistry and Molecular Biology, Monash University, Melbourne, VIC 3800, Australia.
| | - Ross D Hannan
- Oncogenic Signalling and Growth Control Program, Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia.
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, VIC 3010, Australia.
- Department of Biochemistry and Molecular Biology, University of Melbourne, Parkville, VIC 3010, Australia.
- Department of Biochemistry and Molecular Biology, Monash University, Melbourne, VIC 3800, Australia.
- The ACRF Department of Cancer Biology and Therapeutics, The John Curtin School of Medical Research, The Australian National University, Acton, ACT 2601, Australia.
- School of Biomedical Sciences, University of Queensland, Brisbane, QLD 4072, Australia.
| | - Luc Furic
- Prostate Cancer Translational Research Laboratory, Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia.
- Cancer Program, Biomedicine Discovery Institute and Department of Anatomy & Developmental Biology, Monash University, Melbourne, VIC 3800, Australia.
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, VIC 3010, Australia.
| |
Collapse
|
178
|
Guner G, Sirajuddin P, Zheng Q, Bai B, Brodie A, Liu H, Af Hällström T, Kulac I, Laiho M, De Marzo AM. Novel Assay to Detect RNA Polymerase I Activity In Vivo. Mol Cancer Res 2017; 15:577-584. [PMID: 28119429 DOI: 10.1158/1541-7786.mcr-16-0246] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2016] [Revised: 12/08/2016] [Accepted: 01/02/2017] [Indexed: 01/22/2023]
Abstract
This report develops an analytically validated chromogenic in situ hybridization (CISH) assay using branched DNA signal amplification (RNAscope) for detecting the expression of the 5' external transcribed spacer (ETS) of the 45S ribosomal (r) RNA precursor in formalin-fixed and paraffin-embedded (FFPE) human tissues. 5'ETS/45S CISH was performed on standard clinical specimens and tissue microarrays (TMA) from untreated prostate carcinomas, high-grade prostatic intraepithelial neoplasia (PIN), and matched benign prostatic tissues. Signals were quantified using image analysis software. The 5'ETS rRNA signal was restricted to the nucleolus. The signal was markedly attenuated in cell lines and in prostate tissue slices after pharmacologic inhibition of RNA polymerase I (Pol I) using BMH-21 or actinomycin D, and by RNAi depletion of Pol I, demonstrating validity as a measure of Pol I activity. Clinical human prostate FFPE tissue sections and TMAs showed a marked increase in the signal in the presumptive precursor lesion (high-grade PIN) and invasive adenocarcinoma lesions (P = 0.0001 and P = 0.0001, respectively) compared with non-neoplastic luminal epithelium. The increase in 5'ETS rRNA signal was present throughout all Gleason scores and pathologic stages at radical prostatectomy, with no marked difference among these. This precursor rRNA assay has potential utility for detection of increased rRNA production in various tumor types and as a novel companion diagnostic for clinical trials involving Pol I inhibition.Implications: Increased rRNA production, a possible therapeutic target for multiple cancers, can be detected with a new, validated assay that also serves as a pharmacodynamic marker for Pol I inhibitors. Mol Cancer Res; 15(5); 577-84. ©2017 AACR.
Collapse
Affiliation(s)
- Gunes Guner
- Department of Pathology, Urology and Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Paul Sirajuddin
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Qizhi Zheng
- Department of Pathology, Urology and Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Baoyan Bai
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Alexandra Brodie
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Hester Liu
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Taija Af Hällström
- Institute for Molecular Medicine Finland, University of Helsinki, Helsinki, Finland
| | - Ibrahim Kulac
- Department of Pathology, Urology and Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Marikki Laiho
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland. .,Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Angelo M De Marzo
- Department of Pathology, Urology and Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland. .,Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
| |
Collapse
|
179
|
Chakravarthi BVSK, Goswami MT, Pathi SS, Dodson M, Chandrashekar DS, Agarwal S, Nepal S, Hodigere Balasubramanya SA, Siddiqui J, Lonigro RJ, Chinnaiyan AM, Kunju LP, Palanisamy N, Varambally S. Expression and Role of PAICS, a De Novo Purine Biosynthetic Gene in Prostate Cancer. Prostate 2017; 77:10-21. [PMID: 27550065 DOI: 10.1002/pros.23243] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2016] [Accepted: 07/25/2016] [Indexed: 01/10/2023]
Abstract
BACKGROUND Our goal was to investigate de novo purine biosynthetic gene PAICS expression and evaluate its role in prostate cancer progression. METHODS Next-generation sequencing, qRTPCR and immunoblot analysis revealed an elevated expression of a de novo purine biosynthetic gene, Phosphoribosylaminoimidazole Carboxylase, Phosphoribosylaminoimidazole Succinocarboxamide Synthetase (PAICS) in a progressive manner in prostate cancer. Functional analyses were performed using prostate cancer cell lines- DU145, PC3, LnCaP, and VCaP. The oncogenic properties of PAICS were studied both by transient and stable knockdown strategies, in vivo chicken chorioallantoic membrane (CAM) and murine xenograft models. Effect of BET bromodomain inhibitor JQ1 on the expression level of PAICS was also studied. RESULTS Molecular staging of prostate cancer is important factor in effective diagnosis, prognosis and therapy. In this study, we identified a de novo purine biosynthetic gene; PAICS is overexpressed in PCa and its expression correlated with disease aggressiveness. Through several in vitro and in vivo functional studies, we show that PAICS is necessary for proliferation and invasion in prostate cancer cells. We identified JQ1, a BET bromodomain inhibitor previously implicated in regulating MYC expression and demonstrated role in prostate cancer, abrogates PAICS expression in several prostate cancer cells. Furthermore, we observe loss of MYC occupancy on PAICS promoter in presence of JQ1. CONCLUSIONS Here, we report that evaluation of PAICS in prostate cancer progression and its role in prostate cancer cell proliferation and invasion and suggest it as a valid therapeutic target. We suggest JQ1, a BET-domain inhibitor, as possible therapeutic option in targeting PAICS in prostate cancer. Prostate 77:10-21, 2017. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Balabhadrapatruni V S K Chakravarthi
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, Michigan
- Department of Pathology, University of Michigan, Ann Arbor, Michigan
- Department of Pathology, University of Alabama at Birmingham, Birmingham, Alabama
- Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, Alabama
| | - Moloy T Goswami
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, Michigan
- Department of Pathology, University of Michigan, Ann Arbor, Michigan
| | - Satya S Pathi
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, Michigan
- Department of Pathology, University of Michigan, Ann Arbor, Michigan
| | - Matthew Dodson
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, Michigan
| | | | - Sumit Agarwal
- Department of Pathology, University of Alabama at Birmingham, Birmingham, Alabama
| | - Saroj Nepal
- Department of Pathology, University of Alabama at Birmingham, Birmingham, Alabama
| | | | - Javed Siddiqui
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, Michigan
| | - Robert J Lonigro
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, Michigan
- Comprehensive Cancer Center, University of Michigan Medical School, Ann Arbor, Michigan
| | - Arul M Chinnaiyan
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, Michigan
- Department of Pathology, University of Michigan, Ann Arbor, Michigan
- Comprehensive Cancer Center, University of Michigan Medical School, Ann Arbor, Michigan
- Department of Urology, University of Michigan, Ann Arbor, Michigan
- Howard Hughes Medical Institute, University of Michigan Medical School, Ann Arbor, Michigan
| | - Lakshmi P Kunju
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, Michigan
- Department of Pathology, University of Michigan, Ann Arbor, Michigan
| | - Nallasivam Palanisamy
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, Michigan
| | - Sooryanarayana Varambally
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, Michigan
- Department of Pathology, University of Alabama at Birmingham, Birmingham, Alabama
- Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, Alabama
| |
Collapse
|
180
|
Sadeghi M, Ranjbar B, Ganjalikhany MR, M. Khan F, Schmitz U, Wolkenhauer O, Gupta SK. MicroRNA and Transcription Factor Gene Regulatory Network Analysis Reveals Key Regulatory Elements Associated with Prostate Cancer Progression. PLoS One 2016; 11:e0168760. [PMID: 28005952 PMCID: PMC5179129 DOI: 10.1371/journal.pone.0168760] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2016] [Accepted: 11/21/2016] [Indexed: 11/18/2022] Open
Abstract
Technological and methodological advances in multi-omics data generation and integration approaches help elucidate genetic features of complex biological traits and diseases such as prostate cancer. Due to its heterogeneity, the identification of key functional components involved in the regulation and progression of prostate cancer is a methodological challenge. In this study, we identified key regulatory interactions responsible for primary to metastasis transitions in prostate cancer using network inference approaches by integrating patient derived transcriptomic and miRomics data into gene/miRNA/transcription factor regulatory networks. One such network was derived for each of the clinical states of prostate cancer based on differentially expressed and significantly correlated gene, miRNA and TF pairs from the patient data. We identified key elements of each network using a network analysis approach and validated our results using patient survival analysis. We observed that HOXD10, BCL2 and PGR are the most important factors affected in primary prostate samples, whereas, in the metastatic state, STAT3, JUN and JUNB are playing a central role. Benefiting integrative networks our analysis suggests that some of these molecules were targeted by several overexpressed miRNAs which may have a major effect on the dysregulation of these molecules. For example, in the metastatic tumors five miRNAs (miR-671-5p, miR-665, miR-663, miR-512-3p and miR-371-5p) are mainly responsible for the dysregulation of STAT3 and hence can provide an opportunity for early detection of metastasis and development of alternative therapeutic approaches. Our findings deliver new details on key functional components in prostate cancer progression and provide opportunities for the development of alternative therapeutic approaches.
Collapse
Affiliation(s)
- Mehdi Sadeghi
- Department of Biophysics, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Bijan Ranjbar
- Department of Biophysics, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | | | - Faiz M. Khan
- Department of Systems Biology and Bioinformatics, University of Rostock, Rostock, Germany
| | - Ulf Schmitz
- Gene and Stem Cell Therapy Program, Centenary Institute, University of Sydney, Camperdown, Australia
- Sydney Medical School, University of Sydney, Camperdown, Australia
| | - Olaf Wolkenhauer
- Department of Systems Biology and Bioinformatics, University of Rostock, Rostock, Germany
- Stellenbosch Institute for Advanced Study (STIAS), Wallenberg Research Centre at Stellenbosch University, Stellenbosch, South Africa
| | - Shailendra K. Gupta
- Department of Systems Biology and Bioinformatics, University of Rostock, Rostock, Germany
- Department of Bioinformatics, CSIR-Indian Institute of Toxicology Research, Lucknow, India
| |
Collapse
|
181
|
Guo Y, Alexander K, Clark AG, Grimson A, Yu H. Integrated network analysis reveals distinct regulatory roles of transcription factors and microRNAs. RNA (NEW YORK, N.Y.) 2016; 22:1663-1672. [PMID: 27604961 PMCID: PMC5066619 DOI: 10.1261/rna.048025.114] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/08/2014] [Accepted: 07/25/2016] [Indexed: 06/06/2023]
Abstract
Analysis of transcription regulatory networks has revealed many principal features that govern gene expression regulation. MicroRNAs (miRNAs) have emerged as another major class of gene regulators that influence gene expression post-transcriptionally, but there remains a need to assess quantitatively their global roles in gene regulation. Here, we have constructed an integrated gene regulatory network comprised of transcription factors (TFs), miRNAs, and their target genes and analyzed the effect of regulation on target mRNA expression, target protein expression, protein-protein interaction, and disease association. We found that while target genes regulated by the same TFs tend to be co-expressed, co-regulation by miRNAs does not lead to co-expression assessed at either mRNA or protein levels. Analysis of interacting protein pairs in the regulatory network revealed that compared to genes co-regulated by miRNAs, a higher fraction of genes co-regulated by TFs encode proteins in the same complex. Although these results suggest that genes co-regulated by TFs are more functionally related than those co-regulated by miRNAs, genes that share either TF or miRNA regulators are more likely to cause the same disease. Further analysis on the interplay between TFs and miRNAs suggests that TFs tend to regulate intramodule/pathway clusters, while miRNAs tend to regulate intermodule/pathway clusters. These results demonstrate that although TFs and miRNAs both regulate gene expression, they occupy distinct niches in the overall regulatory network within the cell.
Collapse
Affiliation(s)
- Yu Guo
- Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, New York 14853, USA
- Department of Biological Statistics and Computational Biology, Cornell University, Ithaca, New York 14853, USA
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, New York 14853, USA
| | - Katherine Alexander
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, New York 14853, USA
| | - Andrew G Clark
- Department of Biological Statistics and Computational Biology, Cornell University, Ithaca, New York 14853, USA
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, New York 14853, USA
| | - Andrew Grimson
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, New York 14853, USA
| | - Haiyuan Yu
- Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, New York 14853, USA
- Department of Biological Statistics and Computational Biology, Cornell University, Ithaca, New York 14853, USA
| |
Collapse
|
182
|
Abstract
Conventional anatomical imaging with CT and MRI has limitations in the evaluation of prostate cancer. PET is a powerful imaging technique, which can be directed toward molecular targets as diverse as glucose metabolism, density of prostate-specific membrane antigen receptors, and skeletal osteoblastic activity. Although 2-deoxy-2-18F-FDG-PET is the mainstay of molecular imaging, FDG has limitations in typically indolent prostate cancer. Yet, there are many useful and emerging PET tracers beyond FDG, which provide added value. These include radiotracers interrogating prostate cancer via molecular mechanisms related to the biology of choline, acetate, amino acids, bombesin, and dihydrotestosterone, among others. Choline is used for cell membrane synthesis and its metabolism is upregulated in prostate cancer. 11C-choline and 18F-choline are in wide clinical use outside the United States, and they have proven most beneficial for detection of recurrent prostate cancer. 11C-acetate is an indirect biomarker of fatty acid synthesis, which is also upregulated in prostate cancer. Imaging of prostate cancer with 11C-acetate is overall similar to the choline radiotracers yet is not as widely used. Upregulation of amino acid transport in prostate cancer provides the biologic basis for amino acid-based radiotracers. Most recent progress has been made with the nonnatural alicyclic amino acid analogue radiotracer anti-1-amino-3-18F-fluorocyclobutane-1-carboxylic acid (FACBC or fluciclovine) also proven most useful for the detection of recurrent prostate cancer. Other emerging PET radiotracers for prostate cancer include the bombesin group directed to the gastrin-releasing peptide receptor, 16β-18F-fluoro-5α-dihydrotestosterone (FDHT) that binds to the androgen receptor, and those targeting the vasoactive intestinal polypeptide receptor 1 (VPAC-1) and urokinase plasminogen activator receptor (uPAR), which are also overexpressed in prostate cancer.
Collapse
Affiliation(s)
- David M Schuster
- Department of Radiology and Imaging Sciences, Emory University, Atlanta, GA.
| | - Cristina Nanni
- Department of Nuclear Medicine, Policlinico S. Orsola, University of Bologna, Bologna, Italy
| | - Stefano Fanti
- Department of Nuclear Medicine, Policlinico S. Orsola, University of Bologna, Bologna, Italy
| |
Collapse
|
183
|
The combination of 13N-ammonia and 18F-FDG whole-body PET/CT on the same day for diagnosis of advanced prostate cancer. Nucl Med Commun 2016; 37:239-46. [PMID: 26588068 PMCID: PMC4727500 DOI: 10.1097/mnm.0000000000000444] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Purpose The aim of the study was to evaluate the efficacy of 13N-ammonia and 18F-fluorodeoxyglucose (18F-FDG) PET performed on the same day in the detection of advanced prostate cancer (PC) and its metastases. Patients and methods Twenty-six patients with high-risk PC [Gleason score 8–10 or prostate-specific antigen (PSA)>20 ng/ml or clinical tumor extension≥T2c] were recruited into the study. 13N-Ammonia and 18F-FDG PET/CT were performed on the same day (18F-FDG followed ammonia, with an interval of a minimum of 2 h). Lesions were interpreted as positive, negative, or equivocal. Patient-based and field-based performance characteristics for both imaging techniques were reported. Results There was significant correlation between 13N-ammonia and 18F-FDG PET/CT in the detection of primary PC (κ=0.425, P=0.001) and no significant difference in sensitivity (60.2 vs. 54.5%) and specificity (100 vs. 83.3%). The maximum standard uptake values and corresponding target-to-background ratio values of the concordantly positive lesions in prostate glands in the two studies did not differ significantly (P=0.124 and 0.075, respectively). The sensitivity and specificity of PET imaging using 13N-ammonia for lymph node metastases were 77.5 and 96.3%, respectively, whereas the values were 75 and 44.4% using 18F-FDG. The two modalities were highly correlated with respect to the detection of lymph nodes and bone metastases. Conclusion The concordance between the two imaging modalities suggests a clinical impact of 13N-ammonia PET/CT in advanced PC patients as well as of 18F-FDG. 13N-Ammonia is a useful PET tracer and a complement to 18F-FDG for detecting primary focus and distant metastases in PC. The combination of these two tracers on the same day can accurately detect advanced PC.
Collapse
|
184
|
Puhr M, De Marzo A, Isaacs W, Lucia MS, Sfanos K, Yegnasubramanian S, Culig Z. Inflammation, Microbiota, and Prostate Cancer. Eur Urol Focus 2016; 2:374-382. [DOI: 10.1016/j.euf.2016.08.010] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2016] [Accepted: 08/18/2016] [Indexed: 01/31/2023]
|
185
|
Activation of Notch1 synergizes with multiple pathways in promoting castration-resistant prostate cancer. Proc Natl Acad Sci U S A 2016; 113:E6457-E6466. [PMID: 27694579 DOI: 10.1073/pnas.1614529113] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Metastatic castration-resistant prostate cancer (CRPC) is the primary cause of prostate cancer-specific mortality. Defining new mechanisms that can predict recurrence and drive lethal CRPC is critical. Here, we demonstrate that localized high-risk prostate cancer and metastatic CRPC, but not benign prostate tissues or low/intermediate-risk prostate cancer, express high levels of nuclear Notch homolog 1, translocation-associated (Notch1) receptor intracellular domain. Chronic activation of Notch1 synergizes with multiple oncogenic pathways altered in early disease to promote the development of prostate adenocarcinoma. These tumors display features of epithelial-to-mesenchymal transition, a cellular state associated with increased tumor aggressiveness. Consistent with its activation in clinical CRPC, tumors driven by Notch1 intracellular domain in combination with multiple pathways altered in prostate cancer are metastatic and resistant to androgen deprivation. Our study provides functional evidence that the Notch1 signaling axis synergizes with alternative pathways in promoting metastatic CRPC and may represent a new therapeutic target for advanced prostate cancer.
Collapse
|
186
|
Ellis L, Ku S, Li Q, Azabdaftari G, Seliski J, Olson B, Netherby CS, Tang DG, Abrams SI, Goodrich DW, Pili R. Generation of a C57BL/6 MYC-Driven Mouse Model and Cell Line of Prostate Cancer. Prostate 2016; 76:1192-202. [PMID: 27225803 PMCID: PMC6123824 DOI: 10.1002/pros.23206] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2016] [Accepted: 05/09/2016] [Indexed: 01/06/2023]
Abstract
INTRODUCTION Transgenic mouse modeling is a favorable tool to reflect human prostate tumorigenesis and interactions between prostate cancer and the microenvironment. The use of GEMMs and derived cell lines represent powerful tools to study prostate cancer initiation and progression with an associated tumor microenvironment. Notably, such models provide the capacity for rapid preclinical therapy studies including immune therapies for prostate cancer treatment. METHODS Backcrossing FVB Hi-MYC mice with C57BL/6N mice, we established a Hi-MYC transgenic mouse model on a C57BL/6 background (B6MYC). In addition, using a conditional reprogramming method, a novel C57BL/6 MYC driven prostate adenocarcinoma cell line was generated. RESULTS Our results demonstrate that disease progression is significantly delayed in B6MYC when compared to their FVB counterparts. Current data also indicates infiltrating immune cells are present in pre-cancer lesions, prostate intraepithelial neoplasia (PIN). Further, immunophenotyping of this immune infiltrate demonstrates the predominant population as myeloid-derived suppressor cells (MDSC). Also, we successfully generated a B6MYC-CaP cell line, and determined that this new PCa cell line express markers of luminal epithelial lineage. DISCUSSION This novel model of PCa provides a new platform to understand the cross talk between MYC driven prostate cancer and the microenvironment. Importantly, these models will be an ideal tool to support the clinical development of immunotherapy as well as other novel therapeutic strategies for prostate cancer treatment. Prostate 76:1192-1202, 2016. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Leigh Ellis
- Genitourinary Program, Roswell Park Cancer Institute, Buffalo, New York
- Department of Pharmacology and Therapeutics, Roswell Park Cancer Institute, Buffalo, New York
| | - ShengYu Ku
- Genitourinary Program, Roswell Park Cancer Institute, Buffalo, New York
- Department of Pharmacology and Therapeutics, Roswell Park Cancer Institute, Buffalo, New York
| | - Qiuhui Li
- Department of Pharmacology and Therapeutics, Roswell Park Cancer Institute, Buffalo, New York
| | - Gissou Azabdaftari
- Genitourinary Program, Roswell Park Cancer Institute, Buffalo, New York
- Department of Pathology, Roswell Park Cancer Institute, Buffalo, New York
| | - Joseph Seliski
- University of Wisconsin Carbone Cancer Center, Madison, Wisconsin
| | - Brian Olson
- University of Wisconsin Carbone Cancer Center, Madison, Wisconsin
| | | | - Dean G. Tang
- Department of Pharmacology and Therapeutics, Roswell Park Cancer Institute, Buffalo, New York
| | - Scott I. Abrams
- Department of Immunology, Roswell Park Cancer Institute, Buffalo, New York
| | - David W. Goodrich
- Department of Pharmacology and Therapeutics, Roswell Park Cancer Institute, Buffalo, New York
| | - Roberto Pili
- Department of Medicine, Indiana University-Simon Cancer Center, Indianapolis, Indiana
| |
Collapse
|
187
|
Gerrin SJ, Sowalsky AG, Balk SP, Ye H. Mutation Profiling Indicates High Grade Prostatic Intraepithelial Neoplasia as Distant Precursors of Adjacent Invasive Prostatic Adenocarcinoma. Prostate 2016; 76:1227-36. [PMID: 27272561 PMCID: PMC5507580 DOI: 10.1002/pros.23212] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2016] [Accepted: 05/16/2016] [Indexed: 12/23/2022]
Abstract
INTRODUCTION High Grade Prostatic Intraepithelial Neoplasia (HGPIN) is the putative precursor lesion to prostatic adenocarcinoma (PCa), but the precise relationship between HGPIN and PCa remains unclear. METHODS We performed a molecular case study in which we studied mutation profiles of six tumor-associated HGPIN lesions in a single case of TMPRSS2:ERG fusion positive Gleason score 7 PCa that we had previously mapped for somatic mutations in adjacent Gleason patterns 3 and 4 foci, using microdissection and targeted deep-sequencing. RESULTS A total of 32 tumor-specific mutated sites were successfully amplified and sequenced, including 25 truncal mutations and 7 mutations that were specific to either the Gleason pattern 3 or pattern 4 foci. All six HGPIN foci shared the same tumor-specific TMPRSS2:ERG fusion breakpoint, establishing that they were all clonally related to the adjacent invasive tumor. Among the 32 gene targets mutated in the invasive tumor, only mutation of the OR2AP1 gene, a truncal mutation, was found in a single focus of HGPIN. The remaining gene targets that were successfully sequenced were wild-type in all other HGPIN foci. DISCUSSION This study demonstrates the feasibility of targeted mutation profiling of HGPIN lesions, which will be important to understand PCa tumorigenesis. The results in this case, showing a remarkable absence of truncal mutations in HGPIN lesions bearing the tumor-specific ERG fusion, indicate HGPIN lesions may be relatively stable genetically and argue against a stepwise clonal evolution model of HGPIN to PCa. Prostate 76:1227-1236, 2016. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Sean J. Gerrin
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215
| | - Adam G. Sowalsky
- Hematology-Oncology Division, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215
- Laboratory of Genitourinary Cancer Pathogenesis, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Steven P. Balk
- Hematology-Oncology Division, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215
| | - Huihui Ye
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215
| |
Collapse
|
188
|
Rebello RJ, Kusnadi E, Cameron DP, Pearson HB, Lesmana A, Devlin JR, Drygin D, Clark AK, Porter L, Pedersen J, Sandhu S, Risbridger GP, Pearson RB, Hannan RD, Furic L. The Dual Inhibition of RNA Pol I Transcription and PIM Kinase as a New Therapeutic Approach to Treat Advanced Prostate Cancer. Clin Cancer Res 2016; 22:5539-5552. [PMID: 27486174 DOI: 10.1158/1078-0432.ccr-16-0124] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2016] [Revised: 07/15/2016] [Accepted: 07/21/2016] [Indexed: 11/16/2022]
Abstract
PURPOSE The MYC oncogene is frequently overexpressed in prostate cancer. Upregulation of ribosome biogenesis and function is characteristic of MYC-driven tumors. In addition, PIM kinases activate MYC signaling and mRNA translation in prostate cancer and cooperate with MYC to accelerate tumorigenesis. Here, we investigate the efficacy of a single and dual approach targeting ribosome biogenesis and function to treat prostate cancer. EXPERIMENTAL DESIGN The inhibition of ribosomal RNA (rRNA) synthesis with CX-5461, a potent, selective, and orally bioavailable inhibitor of RNA polymerase I (Pol I) transcription, has been successfully exploited therapeutically but only in models of hematologic malignancy. CX-5461 and CX-6258, a pan-PIM kinase inhibitor, were tested alone and in combination in prostate cancer cell lines, in Hi-MYC- and PTEN-deficient mouse models and in patient-derived xenografts (PDX) of metastatic tissue obtained from a patient with castration-resistant prostate cancer. RESULTS CX-5461 inhibited anchorage-independent growth and induced cell-cycle arrest in prostate cancer cell lines at nanomolar concentrations. Oral administration of 50 mg/kg CX-5461 induced TP53 expression and activity and reduced proliferation (MKI67) and invasion (loss of ductal actin) in Hi-MYC tumors, but not in PTEN-null (low MYC) tumors. While 100 mg/kg CX-6258 showed limited effect alone, its combination with CX-5461 further suppressed proliferation and dramatically reduced large invasive lesions in both models. This rational combination strategy significantly inhibited proliferation and induced cell death in PDX of prostate cancer. CONCLUSIONS Our results demonstrate preclinical efficacy of targeting the ribosome at multiple levels and provide a new approach for the treatment of prostate cancer. Clin Cancer Res; 22(22); 5539-52. ©2016 AACR.
Collapse
Affiliation(s)
- Richard J Rebello
- Cancer Program, Biomedicine Discovery Institute and Department of Anatomy & Developmental Biology, Monash University, Victoria, Australia
| | - Eric Kusnadi
- Oncogenic Signaling and Growth Control Program, Peter MacCallum Cancer Centre, St Andrews Place, East Melbourne, Victoria, Australia
| | - Donald P Cameron
- Oncogenic Signaling and Growth Control Program, Peter MacCallum Cancer Centre, St Andrews Place, East Melbourne, Victoria, Australia.,Department of Cancer Biology and Therapeutics, The John Curtin School of Medical Research, The Australian National University, Canberra, Australia
| | - Helen B Pearson
- Oncogenic Signaling and Growth Control Program, Peter MacCallum Cancer Centre, St Andrews Place, East Melbourne, Victoria, Australia
| | - Analia Lesmana
- Oncogenic Signaling and Growth Control Program, Peter MacCallum Cancer Centre, St Andrews Place, East Melbourne, Victoria, Australia
| | - Jennifer R Devlin
- Oncogenic Signaling and Growth Control Program, Peter MacCallum Cancer Centre, St Andrews Place, East Melbourne, Victoria, Australia
| | | | - Ashlee K Clark
- Cancer Program, Biomedicine Discovery Institute and Department of Anatomy & Developmental Biology, Monash University, Victoria, Australia
| | - Laura Porter
- Cancer Program, Biomedicine Discovery Institute and Department of Anatomy & Developmental Biology, Monash University, Victoria, Australia
| | | | - Shahneen Sandhu
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Victoria, Australia
| | - Gail P Risbridger
- Cancer Program, Biomedicine Discovery Institute and Department of Anatomy & Developmental Biology, Monash University, Victoria, Australia
| | - Richard B Pearson
- Oncogenic Signaling and Growth Control Program, Peter MacCallum Cancer Centre, St Andrews Place, East Melbourne, Victoria, Australia. .,Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Victoria, Australia.,Department of Biochemistry and Molecular Biology, University of Melbourne, Parkville, Victoria, Australia.,Department of Biochemistry and Molecular Biology, Monash University, Victoria, Australia
| | - Ross D Hannan
- Oncogenic Signaling and Growth Control Program, Peter MacCallum Cancer Centre, St Andrews Place, East Melbourne, Victoria, Australia. .,Department of Cancer Biology and Therapeutics, The John Curtin School of Medical Research, The Australian National University, Canberra, Australia.,Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Victoria, Australia.,Department of Biochemistry and Molecular Biology, University of Melbourne, Parkville, Victoria, Australia.,Department of Biochemistry and Molecular Biology, Monash University, Victoria, Australia.,School of Biomedical Sciences, University of Queensland, Brisbane, Queensland, Australia
| | - Luc Furic
- Cancer Program, Biomedicine Discovery Institute and Department of Anatomy & Developmental Biology, Monash University, Victoria, Australia.
| |
Collapse
|
189
|
Baker AM, Van Noorden S, Rodriguez-Justo M, Cohen P, Wright NA, Lampert IA. Distribution of the c-MYC gene product in colorectal neoplasia. Histopathology 2016; 69:222-9. [PMID: 26826706 PMCID: PMC4949543 DOI: 10.1111/his.12939] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2015] [Accepted: 01/27/2016] [Indexed: 12/14/2022]
Abstract
AIMS Recent attempts to study MYC distribution in human samples have been confounded by a lack of agreement in immunohistochemical staining between antibodies targeting the N-terminus and those targeting the C-terminus of the MYC protein. The aim of this study was to use a novel in-situ hybridization (ISH) approach to detect MYC mRNA in clinically relevant samples, and thereby determine the reliability of MYC-targeting antibodies. METHODS AND RESULTS We performed immunohistochemistry on human formalin-fixed paraffin embedded normal colon (n = 15), hyperplastic polyp (n = 4) and neoplastic colon samples (n = 55), using the N-terminally directed antibody Y69, and the C-terminally directed antibody 9E10. The MYC protein distributions were then compared with the location of MYC mRNA, determined by ISH. We found that the localization of MYC mRNA correlated well with the protein distribution determined with the N-terminally directed antibody Y69, and was also associated with expression of the proliferation marker Ki67. The protein distribution determined with the C-terminally directed antibody 9E10 was not always associated with MYC mRNA, Y69, or Ki67, and indeed often showed a reciprocal pattern of expression, with staining being strongest in non-proliferating cells. CONCLUSIONS The observed discrepancy between the staining patterns suggests that the significance of 9E10 in immunohistochemical staining is currently uncertain, and therefore should be interpreted with caution.
Collapse
Affiliation(s)
- Ann-Marie Baker
- Centre for Tumour Biology, Barts Cancer Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Susan Van Noorden
- Department of Histopathology, Imperial College London, Hammersmith Hospital, London, UK
| | | | - Patrizia Cohen
- Department of Cellular Pathology, Clarence Memorial Wing, St Mary's Hospital, London, UK
| | - Nicholas A Wright
- Centre for Tumour Biology, Barts Cancer Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Irvin A Lampert
- Department of Histopathology, West Middlesex University Hospital, Isleworth, UK
| |
Collapse
|
190
|
Hahm ER, Singh KB, Singh SV. c-Myc is a novel target of cell cycle arrest by honokiol in prostate cancer cells. Cell Cycle 2016; 15:2309-20. [PMID: 27341160 DOI: 10.1080/15384101.2016.1201253] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022] Open
Abstract
Honokiol (HNK), a highly promising phytochemical derived from Magnolia officinalis plant, exhibits in vitro and in vivo anticancer activity against prostate cancer but the underlying mechanism is not fully clear. This study was undertaken to delineate the role of c-Myc in anticancer effects of HNK. Exposure of prostate cancer cells to plasma achievable doses of HNK resulted in a marked decrease in levels of total and/or phosphorylated c-Myc protein as well as its mRNA expression. We also observed suppression of c-Myc protein in PC-3 xenografts upon oral HNK administration. Stable overexpression of c-Myc in PC-3 and 22Rv1 cells conferred significant protection against HNK-mediated growth inhibition and G0-G1 phase cell cycle arrest. HNK treatment decreased expression of c-Myc downstream targets including Cyclin D1 and Enhancer of Zeste Homolog 2 (EZH2), and these effects were partially restored upon c-Myc overexpression. In addition, PC-3 and DU145 cells with stable knockdown of EZH2 were relatively more sensitive to growth inhibition by HNK compared with control cells. Finally, androgen receptor overexpression abrogated HNK-mediated downregulation of c-Myc and its targets particularly EZH2. The present study indicates that c-Myc, which is often overexpressed in early and late stages of human prostate cancer, is a novel target of prostate cancer growth inhibition by HNK.
Collapse
Affiliation(s)
- Eun-Ryeong Hahm
- a Department of Pharmacology & Chemical Biology , University of Pittsburgh School of Medicine , Pittsburgh , Pennsylvania , USA.,b University of Pittsburgh Cancer Institute, University of Pittsburgh School of Medicine , Pittsburgh , Pennsylvania , USA
| | - Krishna Beer Singh
- a Department of Pharmacology & Chemical Biology , University of Pittsburgh School of Medicine , Pittsburgh , Pennsylvania , USA.,b University of Pittsburgh Cancer Institute, University of Pittsburgh School of Medicine , Pittsburgh , Pennsylvania , USA
| | - Shivendra V Singh
- a Department of Pharmacology & Chemical Biology , University of Pittsburgh School of Medicine , Pittsburgh , Pennsylvania , USA.,b University of Pittsburgh Cancer Institute, University of Pittsburgh School of Medicine , Pittsburgh , Pennsylvania , USA
| |
Collapse
|
191
|
Udager AM, De Marzo AM, Shi Y, Hicks JL, Cao X, Siddiqui J, Jiang H, Chinnaiyan AM, Mehra R. Concurrent nuclear ERG and MYC protein overexpression defines a subset of locally advanced prostate cancer: Potential opportunities for synergistic targeted therapeutics. Prostate 2016; 76:845-53. [PMID: 27159573 PMCID: PMC4975940 DOI: 10.1002/pros.23175] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2015] [Accepted: 02/16/2016] [Indexed: 12/28/2022]
Abstract
BACKGROUND Recurrent ERG gene fusions, the most common genetic alterations in prostate cancer, drive overexpression of the nuclear transcription factor ERG, and are early clonal events in prostate cancer progression. The nuclear transcription factor MYC is also frequently overexpressed in prostate cancer and may play a role in tumor initiation and/or progression. The relationship between nuclear ERG and MYC protein overexpression in prostate cancer, as well as the clinicopathologic characteristics and prognosis of ERG-positive/MYC high tumors, is not well understood. METHODS Immunohistochemistry (IHC) for ERG and MYC was performed on formalin-fixed, paraffin-embedded tissue from prostate cancer tissue microarrays (TMAs), and nuclear staining was scored semi-quantitatively (IHC product score range = 0-300). Correlation between nuclear ERG and MYC protein expression and association with clinicopathologic parameters and biochemical recurrence after radical prostatectomy was assessed. RESULTS 29.1% of all tumor nodules showed concurrent nuclear ERG and MYC protein overexpression (i.e., ERG-positive/MYC high), including 35.0% of secondary nodules. Overall, there was weak positive correlation between ERG and MYC expression across all tumor nodules (rpb = 0.149, P = 0.045), although this correlation was strongest in secondary nodules (rpb = 0.520, P = 0.019). In radical prostatectomy specimens, ERG-positive/MYC high tumors were positively associated with the presence of extraprostatic extension (EPE), relative to all other ERG/MYC expression subgroups, however, there was no significant association between concurrent nuclear ERG and MYC protein overexpression and time to biochemical recurrence. CONCLUSIONS Concurrent nuclear ERG and MYC protein overexpression is common in prostate cancer and defines a subset of locally advanced tumors. Recent data indicates that BET bromodomain proteins regulate ERG gene fusion and MYC gene expression in prostate cancer, suggesting possible synergistic targeted therapeutics in ERG-positive/MYC high tumors. Prostate 76:845-853, 2016. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Aaron M. Udager
- Department of Pathology, University of Michigan Health System, Ann Arbor, MI
| | - Angelo M. De Marzo
- Department of Pathology, The Sidney Kimmel Comprehensive Cancer Center and The James Buchanan Brady Urological Institute, Johns Hopkins School of Medicine, Baltimore, MD
| | - Yang Shi
- Department of Biostatistics, University of Michigan School of Public Health, Ann Arbor, MI
- Michigan Center for Translational Pathology, Ann Arbor, MI
| | - Jessica L. Hicks
- Department of Pathology, The Sidney Kimmel Comprehensive Cancer Center and The James Buchanan Brady Urological Institute, Johns Hopkins School of Medicine, Baltimore, MD
| | - Xuhong Cao
- Michigan Center for Translational Pathology, Ann Arbor, MI
| | - Javed Siddiqui
- Michigan Center for Translational Pathology, Ann Arbor, MI
| | - Hui Jiang
- Department of Biostatistics, University of Michigan School of Public Health, Ann Arbor, MI
| | - Arul M. Chinnaiyan
- Department of Pathology, University of Michigan Health System, Ann Arbor, MI
- Michigan Center for Translational Pathology, Ann Arbor, MI
- Department of Urology, University of Michigan Health System, Ann Arbor, MI
- Comprehensive Cancer Center, University of Michigan Health System, Ann Arbor, MI
- Howard Hughes Medical Institute, Ann Arbor, MI
| | - Rohit Mehra
- Department of Pathology, University of Michigan Health System, Ann Arbor, MI
- Michigan Center for Translational Pathology, Ann Arbor, MI
- Comprehensive Cancer Center, University of Michigan Health System, Ann Arbor, MI
| |
Collapse
|
192
|
Vyas AR, Moura MB, Hahm ER, Singh KB, Singh SV. Sulforaphane Inhibits c-Myc-Mediated Prostate Cancer Stem-Like Traits. J Cell Biochem 2016; 117:2482-95. [PMID: 26990292 DOI: 10.1002/jcb.25541] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2015] [Accepted: 03/11/2016] [Indexed: 12/21/2022]
Abstract
Preventive and therapeutic efficiencies of dietary sulforaphane (SFN) against human prostate cancer have been demonstrated in vivo, but the underlying mechanism(s) by which this occurs is poorly understood. Here, we show that the prostate cancer stem cell (pCSC)-like traits, such as accelerated activity of aldehyde dehydrogenase 1 (ALDH1), enrichment of CD49f+ fraction, and sphere forming efficiency, are attenuated by SFN treatment. Interestingly, the expression of c-Myc, an oncogenic transcription factor that is frequently deregulated in prostate cancer cells, was markedly suppressed by SFN both in vitro and in vivo. This is biologically relevant, because the lessening of pCSC-like phenotypes mediated by SFN was attenuated when c-Myc was overexpressed. Naturally occurring thio, sulfinyl, and sulfonyl analogs of SFN were also effective in causing suppression of c-Myc protein level. However, basal glycolysis, a basic metabolic pathway that can also be promoted by c-Myc overexpression, was not largely suppressed by SFN, implying that, in addition to c-Myc, there might be another SFN-sensitive cellular factor, which is not directly involved in basal glycolysis, but cooperates with c-Myc to sustain pCSC-like phenotypes. Our study suggests that oncogenic c-Myc is a target of SFN to prevent and eliminate the onset of human prostate cancer. J. Cell. Biochem. 117: 2482-2495, 2016. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Avani R Vyas
- Pharmacology and Chemical Biology, University of Pittsburgh Cancer Institute, University of Pittsburgh School of Medicine, Pittsburgh 15213, Pennsylvania
| | - Michelle B Moura
- Pharmacology and Chemical Biology, University of Pittsburgh Cancer Institute, University of Pittsburgh School of Medicine, Pittsburgh 15213, Pennsylvania
| | - Eun-Ryeong Hahm
- Pharmacology and Chemical Biology, University of Pittsburgh Cancer Institute, University of Pittsburgh School of Medicine, Pittsburgh 15213, Pennsylvania
| | - Krishna Beer Singh
- Pharmacology and Chemical Biology, University of Pittsburgh Cancer Institute, University of Pittsburgh School of Medicine, Pittsburgh 15213, Pennsylvania
| | - Shivendra V Singh
- Pharmacology and Chemical Biology, University of Pittsburgh Cancer Institute, University of Pittsburgh School of Medicine, Pittsburgh 15213, Pennsylvania.
| |
Collapse
|
193
|
Fulford L, Milewski D, Ustiyan V, Ravishankar N, Cai Y, Le T, Masineni S, Kasper S, Aronow B, Kalinichenko VV, Kalin TV. The transcription factor FOXF1 promotes prostate cancer by stimulating the mitogen-activated protein kinase ERK5. Sci Signal 2016; 9:ra48. [DOI: 10.1126/scisignal.aad5582] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
194
|
Miyamoto KI, Kobayashi Y, Maeshima AM, Taniguchi H, Nomoto J, Kitahara H, Fukuhara S, Munakata W, Maruyama D, Tobinai K. Clinicopathological prognostic factors of 24 patients with B-cell lymphoma, unclassifiable, with features intermediate between diffuse large B-cell lymphoma and Burkitt lymphoma. Int J Hematol 2016; 103:693-702. [PMID: 27095041 DOI: 10.1007/s12185-016-1989-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2015] [Revised: 03/09/2016] [Accepted: 03/15/2016] [Indexed: 01/28/2023]
Abstract
B-cell lymphoma, unclassifiable, with features intermediate between diffuse large B-cell lymphoma and Burkitt lymphoma (iBL/DLBCL), is a rare, but an aggressive subtype. In iBL/DLBCL, clinicopathological prognostic factors, including MYC and BCL2 translocations (double hit translocation, DHT) and the expression of both MYC and BCL2 (double hit score 2, DHS2), have not been studied thoroughly. We retrospectively analyzed the prognostic impact of clinicopathological factors, including MYC split, IGH/BCL2 fusion, MYC and BCL2 expressions, in 24 iBL/DLBCL patients (median age: 47 years). Fifteen patients (62 %) underwent intensive chemotherapy, and nine patients (38 %) underwent rituximab-cyclophosphamide, doxorubicin, vincristine, and prednisolone (R-CHOP). The 5-year progression-free (PFS) and overall survival (OS) rates of intensive chemotherapy and R-CHOP were 57 and 72 %, respectively. PFS was significantly shorter in patients with high IPI score (P < .0001), stage IV (P = .001), aged ≥60 years (P = .042), IGH/BCL2 fusion (P = .029), DHS2 (P = .015), and DHT (P = .03). OS was significantly shorter in patients with high IPI score (P < .0001) and aged ≥60 years (P = .008). In iBL/DLBCL, IGH/BCL2 fusion, DHS2, and DHT were pathological prognostic factors for poor PFS, while IPI remained as more predictive for PFS and OS.
Collapse
Affiliation(s)
- Ken-Ichi Miyamoto
- Department of Hematology, National Cancer Center Hospital, Tsukiji 5-1-1, Chuo-ku, Tokyo, 104-0045, Japan
| | - Yukio Kobayashi
- Department of Hematology, National Cancer Center Hospital, Tsukiji 5-1-1, Chuo-ku, Tokyo, 104-0045, Japan.
| | | | | | - Junko Nomoto
- Department of Hematology, National Cancer Center Hospital, Tsukiji 5-1-1, Chuo-ku, Tokyo, 104-0045, Japan
| | - Hideaki Kitahara
- Department of Hematology, National Cancer Center Hospital, Tsukiji 5-1-1, Chuo-ku, Tokyo, 104-0045, Japan
| | - Suguru Fukuhara
- Department of Hematology, National Cancer Center Hospital, Tsukiji 5-1-1, Chuo-ku, Tokyo, 104-0045, Japan
| | - Wataru Munakata
- Department of Hematology, National Cancer Center Hospital, Tsukiji 5-1-1, Chuo-ku, Tokyo, 104-0045, Japan
| | - Dai Maruyama
- Department of Hematology, National Cancer Center Hospital, Tsukiji 5-1-1, Chuo-ku, Tokyo, 104-0045, Japan
| | - Kensei Tobinai
- Department of Hematology, National Cancer Center Hospital, Tsukiji 5-1-1, Chuo-ku, Tokyo, 104-0045, Japan
| |
Collapse
|
195
|
Bradshaw A, Wickremsekera A, Tan ST, Peng L, Davis PF, Itinteang T. Cancer Stem Cell Hierarchy in Glioblastoma Multiforme. Front Surg 2016; 3:21. [PMID: 27148537 PMCID: PMC4831983 DOI: 10.3389/fsurg.2016.00021] [Citation(s) in RCA: 153] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2016] [Accepted: 03/29/2016] [Indexed: 12/17/2022] Open
Abstract
Glioblastoma multiforme (GBM), an aggressive tumor that typically exhibits treatment failure with high mortality rates, is associated with the presence of cancer stem cells (CSCs) within the tumor. CSCs possess the ability for perpetual self-renewal and proliferation, producing downstream progenitor cells that drive tumor growth. Studies of many cancer types have identified CSCs using specific markers, but it is still unclear as to where in the stem cell hierarchy these markers fall. This is compounded further by the presence of multiple GBM and glioblastoma cancer stem cell subtypes, making investigation and establishment of a universal treatment difficult. This review examines the current knowledge on the CSC markers SALL4, OCT-4, SOX2, STAT3, NANOG, c-Myc, KLF4, CD133, CD44, nestin, and glial fibrillary acidic protein, specifically focusing on their use and validity in GBM research and how they may be utilized for investigations into GBM's cancer biology.
Collapse
Affiliation(s)
- Amy Bradshaw
- Gillies McIndoe Research Institute , Wellington , New Zealand
| | - Agadha Wickremsekera
- Gillies McIndoe Research Institute, Wellington, New Zealand; Department of Neurosurgery, Wellington Regional Hospital, Wellington, New Zealand
| | - Swee T Tan
- Gillies McIndoe Research Institute , Wellington , New Zealand
| | - Lifeng Peng
- Centre for Biodiscovery, School of Biological Sciences, Victoria University of Wellington , Wellington , New Zealand
| | - Paul F Davis
- Gillies McIndoe Research Institute , Wellington , New Zealand
| | - Tinte Itinteang
- Gillies McIndoe Research Institute , Wellington , New Zealand
| |
Collapse
|
196
|
Prostate epithelial cell of origin determines cancer differentiation state in an organoid transformation assay. Proc Natl Acad Sci U S A 2016; 113:4482-7. [PMID: 27044116 DOI: 10.1073/pnas.1603645113] [Citation(s) in RCA: 93] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The cell of origin for prostate cancer remains a subject of debate. Genetically engineered mouse models have demonstrated that both basal and luminal cells can serve as cells of origin for prostate cancer. Using a human prostate regeneration and transformation assay, our group previously demonstrated that basal cells can serve as efficient targets for transformation. Recently, a subpopulation of multipotent human luminal cells defined by CD26 expression that retains progenitor activity in a defined organoid culture was identified. We transduced primary human prostate basal and luminal cells with lentiviruses expressing c-Myc and activated AKT1 (myristoylated AKT1 or myrAKT1) to mimic theMYCamplification andPTENloss commonly detected in human prostate cancer. These cells were propagated in organoid culture before being transplanted into immunodeficient mice. We found that c-Myc/myrAKT1-transduced luminal xenografts exhibited histological features of well-differentiated acinar adenocarcinoma, with strong androgen receptor (AR) and prostate-specific antigen (PSA) expression. In contrast, c-Myc/myrAKT1-transduced basal xenografts were histologically more aggressive, with a loss of acinar structures and low/absent AR and PSA expression. Our findings imply that distinct subtypes of prostate cancer may arise from luminal and basal epithelial cell types subjected to the same oncogenic insults. This study provides a platform for the functional evaluation of oncogenes in basal and luminal epithelial populations of the human prostate. Tumors derived in this fashion with defined genetics can be used in the preclinical development of targeted therapeutics.
Collapse
|
197
|
Myc-dependent purine biosynthesis affects nucleolar stress and therapy response in prostate cancer. Oncotarget 2016; 6:12587-602. [PMID: 25869206 PMCID: PMC4494960 DOI: 10.18632/oncotarget.3494] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2014] [Accepted: 03/07/2015] [Indexed: 11/25/2022] Open
Abstract
The androgen receptor is a key transcription factor contributing to the development of all stages of prostate cancer (PCa). In addition, other transcription factors have been associated with poor prognosis in PCa, amongst which c-Myc (MYC) is a well-established oncogene in many other cancers. We have previously reported that the AR promotes glycolysis and anabolic metabolism; many of these metabolic pathways are also MYC-regulated in other cancers. In this study, we report that in PCa cells de novo purine biosynthesis and the subsequent conversion to XMP is tightly regulated by MYC and independent of AR activity. We characterized two enzymes, PAICS and IMPDH2, within the pathway as PCa biomarkers in tissue samples and report increased efficacy of established anti-androgens in combination with a clinically approved IMPDH inhibitor, mycophenolic acid (MPA). Treatment with MPA led to a significant reduction in cellular guanosine triphosphate (GTP) levels accompanied by nucleolar stress and p53 stabilization. In conclusion, targeting purine biosynthesis provides an opportunity to perturb PCa metabolism and enhance tumour suppressive stress responses.
Collapse
|
198
|
Cai M, Kim S, Wang K, Farnham PJ, Coetzee GA, Lu W. 4C-seq revealed long-range interactions of a functional enhancer at the 8q24 prostate cancer risk locus. Sci Rep 2016; 6:22462. [PMID: 26934861 PMCID: PMC4776156 DOI: 10.1038/srep22462] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2015] [Accepted: 02/15/2016] [Indexed: 02/07/2023] Open
Abstract
Genome-wide association studies (GWAS) have identified >100 independent susceptibility loci for prostate cancer, including the hot spot at 8q24. However, how genetic variants at this locus confer disease risk hasn’t been fully characterized. Using circularized chromosome conformation capture (4C) coupled with next-generation sequencing and an enhancer at 8q24 as “bait”, we identified genome-wide partners interacting with this enhancer in cell lines LNCaP and C4-2B. These 4C-identified regions are distributed in open nuclear compartments, featuring active histone marks (H3K4me1, H3K4me2 and H3K27Ac). Transcription factors NKX3-1, FOXA1 and AR (androgen receptor) tend to occupy these 4C regions. We identified genes located at the interacting regions, and found them linked to positive regulation of mesenchymal cell proliferation in LNCaP and C4-2B, and several pathways (TGF beta signaling pathway in LNCaP and p53 pathway in C4-2B). Common genes (e.g. MYC and POU5F1B) were identified in both prostate cancer cell lines. However, each cell line also had exclusive genes (e.g. ELAC2 and PTEN in LNCaP and BRCA2 and ZFHX3 in C4-2B). In addition, BCL-2 identified in C4-2B might contribute to the progression of androgen-refractory prostate cancer. Overall, our work reveals key genes and pathways involved in prostate cancer onset and progression.
Collapse
Affiliation(s)
- Mingyang Cai
- Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, Department of Stem Cell Biology and Regenerative Medicine, University of Southern California, Los Angeles, CA 90033, USA.,Zilkha Neurogenetic Institute, University of Southern California, Los Angeles, CA 90033, USA.,Division of Biostatistics, Department of Preventive Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Sewoon Kim
- Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, Department of Stem Cell Biology and Regenerative Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Kai Wang
- Zilkha Neurogenetic Institute, University of Southern California, Los Angeles, CA 90033, USA.,Division of Biostatistics, Department of Preventive Medicine, University of Southern California, Los Angeles, CA 90033, USA.,Department of Psychiatry, University of Southern California, Los Angeles, CA 90033, USA
| | - Peggy J Farnham
- Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA 90033, USA.,Department of Biochemistry and Molecular Biology, University of Southern California, Los Angeles, CA 90033, USA
| | - Gerhard A Coetzee
- Department of Preventive Medicine, University of Southern California, Los Angeles, CA 90033, USA.,Department of Urology, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Wange Lu
- Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, Department of Stem Cell Biology and Regenerative Medicine, University of Southern California, Los Angeles, CA 90033, USA
| |
Collapse
|
199
|
Li HX, Gao JM, Liang JQ, Xi JM, Fu M, Wu YJ. Vitamin D3 potentiates the growth inhibitory effects of metformin in DU145 human prostate cancer cells mediated by AMPK/mTOR signalling pathway. Clin Exp Pharmacol Physiol 2016; 42:711-7. [PMID: 25903858 DOI: 10.1111/1440-1681.12409] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2014] [Revised: 04/10/2015] [Accepted: 04/13/2015] [Indexed: 12/30/2022]
Abstract
Metformin and vitamin D₃ both exhibit a strong antiproliferative action in numerous cancer cell lines, including in human prostate cancer cells. Here we showed that the combination of the two drugs had a much stronger effect on DU145 human prostate cancer cell growth than either drug alone. In this research, cell proliferation was measured by methylthiazol tetrazolium (MTT) assay. Cell apoptosis was determined with Hoechst 33342 staining. Western blotting and cell cycle analyses were used to elucidate potential mechanisms of interaction between the drugs. It is shown that in cultured DU145 cells, vitamin D₃ combined with metformin exhibits synergistic effects on cell proliferation and apoptosis. The underlying antitumor mechanisms may involve altered cycle distribution with a G1/S cell cycle arrest, activation of phospho-AMPK with subsequent inhibition of downstream mTOR signalling pathway, down-regulate c-Myc expression, and reducing the level of anti-apoptotic protein p-Bcl-2. In conclusion, metformin and vitamin D₃ synergistically inhibit DU145 cell growth, indicating a promising clinical therapeutic strategy for the treatment of androgen-independent prostate cancer.
Collapse
Affiliation(s)
- Hong-Xia Li
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, Department of Pharmacology, School of Basic Medicine, Lanzhou University, Lanzhou, China
| | - Jing-Miao Gao
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, Department of Pharmacology, School of Basic Medicine, Lanzhou University, Lanzhou, China
| | - Jia-Qi Liang
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, Department of Pharmacology, School of Basic Medicine, Lanzhou University, Lanzhou, China
| | - Jun-Min Xi
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, Department of Pharmacology, School of Basic Medicine, Lanzhou University, Lanzhou, China
| | - Meng Fu
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, Department of Pharmacology, School of Basic Medicine, Lanzhou University, Lanzhou, China
| | - Yong-Jie Wu
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, Department of Pharmacology, School of Basic Medicine, Lanzhou University, Lanzhou, China
| |
Collapse
|
200
|
The cancer-promoting gene fatty acid-binding protein 5 (FABP5) is epigenetically regulated during human prostate carcinogenesis. Biochem J 2016; 473:449-61. [DOI: 10.1042/bj20150926] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2015] [Accepted: 11/27/2015] [Indexed: 12/12/2022]
Abstract
The DNA methylation status of CpG islands in the FABP5 promoter is critical for its expression. Epigenetic regulation of FABP5 gene expression plays an important role during human prostate carcinogenesis, along with up-regulation of c-Myc and Sp1.
Collapse
|