[HTML][HTML] Activation of Rac1 by Src-dependent phosphorylation of Dock180Y1811 mediates PDGFRα-stimulated glioma tumorigenesis in mice and humans

H Feng, B Hu, KW Liu, Y Li, X Lu… - The Journal of …, 2011 - Am Soc Clin Investig
H Feng, B Hu, KW Liu, Y Li, X Lu, T Cheng, JJ Yiin, S Lu, S Keezer, T Fenton, FB Furnari
The Journal of clinical investigation, 2011Am Soc Clin Investig
Two hallmarks of glioblastoma multiforme, the most common malignant brain cancer in
humans, are aggressive growth and the ability of single glioma cells to disperse throughout
the brain. These characteristics render tumors resistant to current therapies and account for
the poor prognosis of patients. Although it is known that oncogenic signaling caused by
overexpression of genes such as PDGFRA is responsible for robust glioma growth and cell
infiltration, the mechanisms underlying glioblastoma malignancy remain largely elusive …
Two hallmarks of glioblastoma multiforme, the most common malignant brain cancer in humans, are aggressive growth and the ability of single glioma cells to disperse throughout the brain. These characteristics render tumors resistant to current therapies and account for the poor prognosis of patients. Although it is known that oncogenic signaling caused by overexpression of genes such as PDGFRA is responsible for robust glioma growth and cell infiltration, the mechanisms underlying glioblastoma malignancy remain largely elusive. Here, we report that PDGFRα signaling in glioblastomas leads to Src-dependent phosphorylation of the guanine nucleotide exchange factor Dock180 at tyrosine 1811 (Dock180Y1811) that results in activation of the GTPase Rac1 and subsequent cell growth and invasion. In human glioma cells, knockdown of Dock180 and reversion with an RNAi-resistant Dock180Y1811F abrogated, whereas an RNAi-resistant Dock180WT rescued, PDGFRα-promoted glioma growth, survival, and invasion. Phosphorylation of Dock180Y1811 enhanced its association with CrkII and p130Cas, causing activation of Rac1 and consequent cell motility. Dock180 also associated with PDGFRα to promote cell migration. Finally, phosphorylated Dock180Y1811 was detected in clinical samples of gliomas and various types of human cancers, and coexpression of phosphorylated Dock180Y1811, phosphorylated SrcY418, and PDGFRα was predictive of extremely poor prognosis of patients with gliomas. Taken together, our findings provide insight into PDGFRα-stimulated gliomagenesis and suggest that phosphorylated Dock180Y1811 contributes to activation of Rac1 in human cancers with PDGFRA amplification.
The Journal of Clinical Investigation