Prostate cancer (PCa) is the most commonly diagnosed non-skin cancer in American men. While organ-confined PCa can be effectively eradicated through surgical and radiation therapies, metastatic disease is essentially incurable. Androgen signaling mediated through the androgen receptor (AR) is one of the most important pathways in PCa initiation and progression. Hormone-deprivation has thus been the standard, first-line treatment for metastatic PCa. Interestingly, in the late-stage castration-resistant PCa (CRPC), AR has become activated, rather than insensitive, in a low androgen environment, thus being responsible for castration resistance. Therefore, understanding and targeting AR pathway remains a central challenge in PCa research. While AR signaling has been studied for decades, it is incompletely understood with past focus largely on genes induced by the AR. Interestingly, recent studies, including ours, have started to show AR also as a globally acting transcriptional repressor. Further, our data suggest that this repression is mediated by the polycomb group protein EZH2 and repressive chromatin remodeling around the target genes. However, a model gene that is representative of this innovative role of AR is needed to further delineate the mechanistic details of AR-mediated repression and, most importantly, to appreciate the functional relevance and therapeutic importance of this repression. Through meta-analysis of androgen-regulated expression microarray data, we nominated NOV (CCN3) as a top candidate. Our preliminary data suggest that NOV is a novel gene of great importance in PCa. It is inhibited by AR and EZH2 and is markedly down-regulated in advanced PCa. Functional analysis suggests that NOV inhibits PCa progression and its loss-of-function drives castration resistance. We thus hypothesize that NOV is directly suppressed by AR through repressive chromatin remodeling and plays essential roles in PCa progression. To address this hypothesis, three specific aims are proposed.
In Aim 1, we will determine androgen regulation and NOV expression in prostate cancer, including patient samples.
In Aim 2, we will examine how AR inhibits NOV expression in prostate cancer cells. We will determine if AR directly binds to the NOV gene promoter and/or enhancer, map chromatin changes around the gene, and examine whether/how EZH2 is response for these changes and thus gene repression.
Aim 3 will investigate the functional role of NOV in regulating PCa proliferation, migration, invasion/metastasis, and castration resistance using cell line models and nude mice.

Public Health Relevance

Prostate cancer is a leading cause of cancer-related deaths in American men. This proposal will investigate the expression, regulation, and function of an important androgen-repressed gene NOV in prostate cancer. Not only will this study enhance our understanding of androgen receptor signaling but it also may provide novel therapeutic strategies.

Agency
National Institute of Health (NIH)
Institute
National Cancer Institute (NCI)
Type
Research Project (R01)
Project #
4R01CA172384-04
Application #
9038328
Study Section
Tumor Cell Biology Study Section (TCB)
Program Officer
Ault, Grace S
Project Start
2013-04-01
Project End
2018-03-31
Budget Start
2016-04-01
Budget End
2017-03-31
Support Year
4
Fiscal Year
2016
Total Cost
Indirect Cost
Name
Northwestern University at Chicago
Department
Internal Medicine/Medicine
Type
Schools of Medicine
DUNS #
005436803
City
Chicago
State
IL
Country
United States
Zip Code
60611
Fong, Ka-Wing; Zhao, Jonathan C; Song, Bing et al. (2018) TRIM28 protects TRIM24 from SPOP-mediated degradation and promotes prostate cancer progression. Nat Commun 9:5007
Fong, Ka-Wing; Zhao, Jonathan C; Kim, Jung et al. (2017) Polycomb-Mediated Disruption of an Androgen Receptor Feedback Loop Drives Castration-Resistant Prostate Cancer. Cancer Res 77:412-422
Kim, J; Jin, H; Zhao, J C et al. (2017) FOXA1 inhibits prostate cancer neuroendocrine differentiation. Oncogene 36:4072-4080
Yang, Yeqing A; Zhao, Jonathan C; Fong, Ka-Wing et al. (2016) FOXA1 potentiates lineage-specific enhancer activation through modulating TET1 expression and function. Nucleic Acids Res 44:8153-64
Yang, Yeqing Angela; Yu, Jindan (2015) Current perspectives on FOXA1 regulation of androgen receptor signaling and prostate cancer. Genes Dis 2:144-151
Zhang, Ali; Zhao, Jonathan C; Kim, Jung et al. (2015) LncRNA HOTAIR Enhances the Androgen-Receptor-Mediated Transcriptional Program and Drives Castration-Resistant Prostate Cancer. Cell Rep 13:209-221
Jin, Hong-Jian; Zhao, Jonathan C; Wu, Longtao et al. (2014) Cooperativity and equilibrium with FOXA1 define the androgen receptor transcriptional program. Nat Commun 5:3972
Kim, J; Wu, L; Zhao, J C et al. (2014) TMPRSS2-ERG gene fusions induce prostate tumorigenesis by modulating microRNA miR-200c. Oncogene 33:5183-92
Wu, L; Runkle, C; Jin, H-J et al. (2014) CCN3/NOV gene expression in human prostate cancer is directly suppressed by the androgen receptor. Oncogene 33:504-13
Wu, Longtao; Zhao, Jonathan C; Kim, Jung et al. (2013) ERG is a critical regulator of Wnt/LEF1 signaling in prostate cancer. Cancer Res 73:6068-79

Showing the most recent 10 out of 11 publications