We recently identified mouse and human RTVP-1/GLIPR1 (Glipr1 and GLIPR1, respectively) as direct p53 target genes and showed that GLIPR1 encodes a secreted protein and that GLIPR1 expression is down- regulated during prostate cancer progression through epigenetic mechanisms. Based on sequence homology we identified and characterized a novel p53 target gene cluster located on human chromosome 12q21 that includes GLIPR1 together with two GLIPR1-like (GLIPR1L) genes, and on mouse chromosome 10D1 that includes Glipr1 and three Glipr1-like (Glipr1l) genes. Functional analysis demonstrated that GLIPR1 or GLIPR1L2 gene overexpression or treatment with GLIPR1 protein induces growth arrest in G0/G1 and/or apoptosis in various mouse and human cancer cell lines in vitro and in vivo. To test the tumor suppressor activities of GLIPR1 we generated mice that harbored an inactivating mutation of the Glipr1 gene. Long-term cohort analysis demonstrated that loss of Glipr1 function led to reduced tumor free survival resulting from a unique spectrum of malignancies. Mechanistic studies demonstrated that GLIPR1 expression leads to down-regulation of c-myc mRNA, suggesting a critical control point in GLIPR1 mediated cell cycle control. Interestingly, GLIPR1 overexpression leads to reduced levels of cyclin A and cyclin D and increased levels of p27Kip1 in prostate and bladder cancer cells. We identified and used chemical and molecular inhibitors to confirm a pro-apoptotic pathway associated with GLIPR1 expression that involves generation of ROS, activation of ASK1-MEK4/7-JNK pathway, suppression of Bcl-2 and broad based caspase activation. Analysis of MEF revealed that treatment with DNA damaging agents resulted in Glipr1 induction, elevated ROS levels and JNK activities, and increased apoptosis in Glipr1+/+ compared to Glipr1-/- MEF. In addition, Glipr1-/- MEF had increased susceptibility to ras + myc induced transformation in vitro. Our results establish GLIPR1 as a novel, wide-spectrum tumor suppressor that mediates pro-apoptotic activities through generation of ROS JNK signaling. We now propose to: (1) Identify the mechanisms of GLIPR1 or GLIPR1L21 mediated growth arrest in prostate and bladder cancer cells;(2) Analyze the molecular pathways that lead to increased ROS-ASK1-MEK4/7-JNK-apoptosis following GLIPR1 or GLIPR1L21 expression;(3) Identify GLIPR1 binding proteins/membrane receptor and characterize GLIPR1 protein uptake and (4) Generate ARR2PB-c-myc transgenic mice, intercross these mice with Glipr1-/- mice and use the bigenic mice to analyze the genetic activities that underlie the capacity of endogenous Glipr1 to suppress the development of pre-malignant and malignant prostatic lesions in vivo. Project Narrative: This project seeks to understand the mechanism(s) through which GLIPR1, a newly identified tumor suppressor gene, inhibits the growth of prostate and bladder cancer. The results of our studies may identify new methods for diagnosing and/or lead to new therapies for these malignancies.

Agency
National Institute of Health (NIH)
Institute
National Cancer Institute (NCI)
Type
Research Project (R01)
Project #
5R01CA050588-22
Application #
7808835
Study Section
Tumor Progression and Metastasis Study Section (TPM)
Program Officer
Snyderwine, Elizabeth G
Project Start
1989-09-08
Project End
2012-05-31
Budget Start
2010-06-01
Budget End
2011-05-31
Support Year
22
Fiscal Year
2010
Total Cost
$325,941
Indirect Cost
Name
University of Texas MD Anderson Cancer Center
Department
Internal Medicine/Medicine
Type
Other Domestic Higher Education
DUNS #
800772139
City
Houston
State
TX
Country
United States
Zip Code
77030
Fujita, Tetsuo; Satoh, Takefumi; Timme, Terry L et al. (2014) Combined therapeutic effects of adenoviral vector-mediated GLIPR1 gene therapy and radiotherapy in prostate and bladder cancer models. Urol Oncol 32:92-100
Li, Likun; Yang, Guang; Ren, Chengzhen et al. (2013) Glioma pathogenesis-related protein 1 induces prostate cancer cell death through Hsc70-mediated suppression of AURKA and TPX2. Mol Oncol 7:484-96
Yang, Guang; Goltsov, Alexei A; Ren, Chengzhen et al. (2012) Caveolin-1 upregulation contributes to c-Myc-induced high-grade prostatic intraepithelial neoplasia and prostate cancer. Mol Cancer Res 10:218-29
Floryk, Daniel; Kurosaka, Shinji; Tanimoto, Ryuta et al. (2011) Castration-induced changes in mouse epididymal white adipose tissue. Mol Cell Endocrinol 345:58-67
Tabata, K; Kurosaka, S; Watanabe, M et al. (2011) Tumor growth and metastasis suppression by Glipr1 gene-modified macrophages in a metastatic prostate cancer model. Gene Ther 18:969-78
Li, Likun; Ren, Chengzhen; Yang, Guang et al. (2011) GLIPR1 suppresses prostate cancer development through targeted oncoprotein destruction. Cancer Res 71:7694-704
Thompson, Timothy C (2010) Glioma pathogenesis-related protein 1: tumor-suppressor activities and therapeutic potential. Yonsei Med J 51:479-83
Langeberg, Wendy J; Tahir, Salahaldin A; Feng, Ziding et al. (2010) Association of caveolin-1 and -2 genetic variants and post-treatment serum caveolin-1 with prostate cancer risk and outcomes. Prostate 70:1020-35
Floryk, Daniel; Thompson, Timothy C (2008) Perifosine induces differentiation and cell death in prostate cancer cells. Cancer Lett 266:216-26
Tahir, Salahaldin A; Ren, Chengzhen; Timme, Terry L et al. (2003) Development of an immunoassay for serum caveolin-1: a novel biomarker for prostate cancer. Clin Cancer Res 9:3653-9

Showing the most recent 10 out of 65 publications