4.8 Article

TRAF4-mediated ubiquitination of NGF receptor TrkA regulates prostate cancer metastasis

Journal

JOURNAL OF CLINICAL INVESTIGATION
Volume 128, Issue 7, Pages 3129-3143

Publisher

AMER SOC CLINICAL INVESTIGATION INC
DOI: 10.1172/JCI96060

Keywords

-

Funding

  1. Department of Defense [W81XWH-15-1-0536, W81XWH-16-1-0297]
  2. NCI Cancer Center Support grant [P30CA125123]
  3. Dan L. Duncan Comprehensive Cancer Center [P30 CA125123]
  4. Cancer Prevention & Research Institute of Texas Proteomics and Metabolomics Core Facility Support Award [RP170005]

Ask authors/readers for more resources

Receptor tyrosine kinases (RTKs) are important drivers of cancers. In addition to genomic alterations, aberrant activation of WT RTKs plays an important role in driving cancer progression. However, the mechanisms underlying how RTKs drive prostate cancer remain incompletely characterized. Here we show that non-proteolytic ubiquitination of RTK regulates its kinase activity and contributes to RTK-mediated prostate cancer metastasis. TRAF4, an E3 ubiquitin ligase, is highly expressed in metastatic prostate cancer. We demonstrated here that it is a key player in regulating RTK-mediated prostate cancer metastasis. We further identified TrkA, a neurotrophin RTK, as a TRAF4-targeted ubiquitination substrate that promotes cancer cell invasion and found that inhibition of TrkA activity abolished TRAF4-dependent cell invasion. TRAF4 promoted K27-and K29-linked ubiquitination at the TrkA kinase domain and increased its kinase activity. Mutation of TRAF4-targeted ubiquitination sites abolished TrkA tyrosine autophosphorylation and its interaction with downstream proteins. TRAF4 knockdown also suppressed nerve growth factor (NGF) stimulated TrkA downstream p38 MAPK activation and invasion-associated gene expression. Furthermore, elevated TRAF4 levels significantly correlated with increased NGF-stimulated invasion-associated gene expression in prostate cancer patients, indicating that this signaling axis is significantly activated during oncogenesis. Our results revealed a posttranslational modification mechanism contributing to aberrant non-mutated RTK activation in cancer cells.

Authors

I am an author on this paper
Click your name to claim this paper and add it to your profile.

Reviews

Primary Rating

4.8
Not enough ratings

Secondary Ratings

Novelty
-
Significance
-
Scientific rigor
-
Rate this paper

Recommended

No Data Available
No Data Available