Prostate cancer-associated mutations in speckle-type POZ protein (SPOP) regulate steroid receptor coactivator 3 protein turnover.

TitleProstate cancer-associated mutations in speckle-type POZ protein (SPOP) regulate steroid receptor coactivator 3 protein turnover.
Publication TypeJournal Article
Year of Publication2013
AuthorsGeng C, He B, Xu L, Barbieri CE, Eedunuri VKumar, Chew SAnne, Zimmermann M, Bond R, Shou J, Li C, Blattner M, Lonard DM, Demichelis F, Coarfa C, Rubin MA, Zhou P, O'Malley BW, Mitsiades N
JournalProc Natl Acad Sci U S A
Volume110
Issue17
Pagination6997-7002
Date Published2013 Apr 23
ISSN1091-6490
KeywordsAnalysis of Variance, Cell Line, Tumor, Electrophoresis, Polyacrylamide Gel, Gene Expression Regulation, Genetic Vectors, HEK293 Cells, Humans, Immunoblotting, Immunoprecipitation, Lentivirus, Male, Mutation, Missense, Nuclear Proteins, Nuclear Receptor Coactivator 3, Prostatic Neoplasms, Real-Time Polymerase Chain Reaction, Receptors, Androgen, Repressor Proteins, Reverse Transcriptase Polymerase Chain Reaction, Tetrazolium Salts, Thiazoles
Abstract

The p160 steroid receptor coactivators (SRCs) SRC-1, SRC-2 [nuclear receptor coactivator (NCOA)2], and SRC-3 [amplified in breast cancer 1 (AIB1)/NCOA3] are key pleiotropic "master regulators" of transcription factor activity necessary for cancer cell proliferation, survival, metabolism, and metastasis. SRC overexpression and overactivation occur in numerous human cancers and are associated with poor clinical outcomes and resistance to therapy. In prostate cancer (PC), the p160 SRCs play critical roles in androgen receptor transcriptional activity, cell proliferation, and resistance to androgen deprivation therapy. We recently demonstrated that the E3 ubiquitin ligase adaptor speckle-type poxvirus and zinc finger (POZ) domain protein (SPOP) interacts directly with SRC-3 and promotes its cullin 3-dependent ubiquitination and proteolysis in breast cancer, thus functioning as a potential tumor suppressor. Interestingly, somatic heterozygous missense mutations in the SPOP substrate-binding cleft recently were identified in up to 15% of human PCs (making SPOP the gene most commonly affected by nonsynonymous point mutations in PC), but their contribution to PC pathophysiology remains unknown. We now report that PC-associated SPOP mutants cannot interact with SRC-3 protein or promote its ubiquitination and degradation. Our data suggest that wild-type SPOP plays a critical tumor suppressor role in PC cells, promoting the turnover of SRC-3 protein and suppressing androgen receptor transcriptional activity. This tumor suppressor effect is abrogated by the PC-associated SPOP mutations. These studies provide a possible explanation for the role of SPOP mutations in PC, and highlight the potential of SRC-3 as a therapeutic target in PC.

DOI10.1073/pnas.1304502110
Alternate JournalProc Natl Acad Sci U S A
PubMed ID23559371
PubMed Central IDPMC3637757
Grant ListP30CA125123 / CA / NCI NIH HHS / United States
NICHD 8818 / / PHS HHS / United States
P30DK079638 / DK / NIDDK NIH HHS / United States
Related Lab: 
Related Faculty: 
Pengbo Zhou, Ph.D.

Pathology & Laboratory Medicine 1300 York Avenue New York, NY 10065 Phone: (212) 746-6464
Surgical Pathology: (212) 746-2700