Prostate cancer (PCa) is the most commonly diagnosed malignancy and the second leading cause of cancer death in American men. However, the etiology of this disease is not fully understood, which hampers the improvement of treatment for a large segment of PCa patients. The TMPRSS2-ERG gene fusion results in aberrant overexpression of fusion/truncation ERG in approximately 50% of all human PCa, suggesting a causal role of ERG in PCa. Notably, overexpression of fusion ERG alone is insufficient to induce PCa in mice; but can cooperate with other lesions such as deletion of the Pten tumor suppressor gene in prostate tumorigenesis, although the underlying mechanisms are largely unknown. Forkhead protein FOXO1 functions as a key downstream effector of PTEN by transcriptional upregulation of genes that induce apoptosis and cell cycle arrest, implying that FOXO1 is a tumor suppressor. This concept is supported by findings that FOXO1 is frequently inactivated in human cancers, such as PCa, by AKT and CDK1 and CDK2-mediated phosphorylation/nuclear exclusion due to PTEN loss, genomic deletion and transcriptional downregulation. To date, however, the precise role of FOXO1 inactivation in prostate tumorigenesis remains elusive. Our preliminary data demonstrated that FOXO1 binds directly to and inhibits the transcriptional and oncogenic activities of fusion ERG in PCa cells. We showed that silencing of FOXO1 upregulates the expression of endogenous fusion ERG target genes and increases the recruitment of fusion ERG onto its target gene loci. We further showed that depletion of FOXO1 in combination with fusion ERG overexpression promotes prostatic epithelial cell transformation in vitro and tumor formation in immune-deficient mice. Additionally, our ChIP-on-chip analyses in mouse embryo stem (ES) cells identified Foxo1 as a putative target of the Polycomb protein Ezh2, activation of which promotes gene silencing by catalyzing trimethylation on histone H3 lysine 27 (H3K27me3). We further demonstrated that EZH2 knockdown or pharmacological inhibition of EZH2 increases FOXO1 expression in human PCa cells. These preliminary data lead to our central hypothesis that inactivation of FOXO1 results in aberrant activation of fusion ERG and that aberrantly activated fusion ERG cooperates with FOXO1 deficiency to promote prostate tumorigenesis. We further hypothesize that EZH2 plays an essential role in mediating transcriptional downregulation of FOXO1 in PCa cells. To test these hypotheses, first we will determine the molecular mechanism, role in tumorigenesis and the clinical significance of FOXO1 inhibition of fusion ERG in PCa (Aim 1); second we will determine the mechanism, disease relevance and preclinical therapeutic application of EZH2 repression of FOXO1 in PCa (Aim 2). Upon completion, findings from this innovative application will not only yield basic insights into genetic and biochemical mechanisms that drive PCa development and progression; but also have significant clinical implications in identification of new targets and development of novel therapeutics for PCa therapy, thus strongly influencing the PCa field both scientifically and clinically.

Public Health Relevance

Despite the high mortality of prostate cancer, the etiology of this disease is not fully understood. This application is designed to determine how mechanistically the FOXO1 tumor suppressor is transcriptionally downregulated and how FOXO1 inactivation leads to aberrant activation of the oncogenic fusion ERG protein and to define the mechanisms by which FOXO1 inactivation and aberrantly activated fusion ERG work in concert to promote prostate cancer initiation and progression. This knowledge will lay the foundation for development of new therapeutic strategies to improve the clinical outcome of prostate cancer patients.

Agency
National Institute of Health (NIH)
Institute
National Cancer Institute (NCI)
Type
Research Project (R01)
Project #
5R01CA134514-09
Application #
9288120
Study Section
Molecular Oncogenesis Study Section (MONC)
Program Officer
Mietz, Judy
Project Start
2009-06-16
Project End
2020-06-30
Budget Start
2017-07-01
Budget End
2018-06-30
Support Year
9
Fiscal Year
2017
Total Cost
Indirect Cost
Name
Mayo Clinic, Rochester
Department
Type
DUNS #
006471700
City
Rochester
State
MN
Country
United States
Zip Code
55905
Yan, Yuqian; An, Jian; Yang, Yinhui et al. (2018) Dual inhibition of AKT-mTOR and AR signaling by targeting HDAC3 in PTEN- or SPOP-mutated prostate cancer. EMBO Mol Med 10:
Blee, Alexandra M; He, Yundong; Yang, Yinhui et al. (2018) TMPRSS2-ERG Controls Luminal Epithelial Lineage and Antiandrogen Sensitivity in PTEN and TP53-Mutated Prostate Cancer. Clin Cancer Res 24:4551-4565
Yang, Yinhui; Bai, Yang; He, Yundong et al. (2018) PTEN Loss Promotes Intratumoral Androgen Synthesis and Tumor Microenvironment Remodeling via Aberrant Activation of RUNX2 in Castration-Resistant Prostate Cancer. Clin Cancer Res 24:834-846
Ye, Zhenqing; Dong, Haidong; Li, Ying et al. (2018) Prevalent Homozygous Deletions of Type I Interferon and Defensin Genes in Human Cancers Associate with Immunotherapy Resistance. Clin Cancer Res 24:3299-3308
Jin, Xin; Yan, Yuqian; Wang, Dejie et al. (2018) DUB3 Promotes BET Inhibitor Resistance and Cancer Progression by Deubiquitinating BRD4. Mol Cell 71:592-605.e4
Yang, Yinhui; Blee, Alexandra M; Wang, Dejie et al. (2017) Loss of FOXO1 Cooperates with TMPRSS2-ERG Overexpression to Promote Prostate Tumorigenesis and Cell Invasion. Cancer Res 77:6524-6537
Kohli, Manish; Ho, Yeung; Hillman, David W et al. (2017) Androgen Receptor Variant AR-V9 Is Coexpressed with AR-V7 in Prostate Cancer Metastases and Predicts Abiraterone Resistance. Clin Cancer Res 23:4704-4715
Fan, Long; Zhu, Qingyi; Liu, Li et al. (2017) CXCL13 is androgen-responsive and involved in androgen induced prostate cancer cell migration and invasion. Oncotarget 8:53244-53261
Yang, Jing; Jin, Xin; Yan, Yuqian et al. (2017) Inhibiting histone deacetylases suppresses glucose metabolism and hepatocellular carcinoma growth by restoring FBP1 expression. Sci Rep 7:43864
Zhang, Pingzhao; Wang, Dejie; Zhao, Yu et al. (2017) Intrinsic BET inhibitor resistance in SPOP-mutated prostate cancer is mediated by BET protein stabilization and AKT-mTORC1 activation. Nat Med 23:1055-1062

Showing the most recent 10 out of 38 publications