4.6 Article

Protein Kinase Cα Promotes Cell Migration through a PDZ-Dependent Interaction with its Novel Substrate Discs Large Homolog 1 (DLG1)

期刊

JOURNAL OF BIOLOGICAL CHEMISTRY
卷 286, 期 50, 页码 43559-43568

出版社

AMER SOC BIOCHEMISTRY MOLECULAR BIOLOGY INC
DOI: 10.1074/jbc.M111.294603

关键词

-

资金

  1. National Institutes of Health [GM43154, P01 DK54441, CA081436, 1F31GM083628]
  2. University of California at San Diego through NIGMS, National Institutes of Health [T32 GM007752]

向作者/读者索取更多资源

Protein scaffolds maintain precision in kinase signaling by coordinating kinases with components of specific signaling pathways. Such spatial segregation is particularly important in allowing specificity of signaling mediated by the 10-member family of protein kinase C (PKC) isozymes. Here we identified a novel interaction between PKC alpha and the Discs large homolog (DLG) family of scaffolds that is mediated by a class I C-terminal PDZ (PSD-95, disheveled, and ZO1) ligand unique to this PKC isozyme. Specifically, use of a proteomic array containing 96 purified PDZ domains identified the third PDZ domains of DLG1/SAP97 and DLG4/PSD95 as interaction partners for the PDZ binding motif of PKC alpha. Co-immunoprecipitation experiments verified that PKC alpha and DLG1 interact in cells by a mechanism dependent on an intact PDZ ligand. Functional assays revealed that the interaction of PKC alpha with DLG1 promotes wound healing; scratch assays using cells depleted of PKC alpha and/or DLG1 have impaired cellular migration that is no longer sensitive to PKC inhibition, and the ability of exogenous PKC alpha to rescue cellular migration is dependent on the presence of its PDZ ligand. Furthermore, we identified Thr-656 as a novel phosphorylation site in the SH3-Hook region of DLG1 that acts as a marker for PKC alpha activity at this scaffold. Increased phosphorylation of Thr-656 is correlated with increased invasiveness in non-small cell lung cancer lines from the NCI-60, consistent with this phosphorylation site serving as a marker of PKC alpha mediated invasion. Taken together, these data establish the requirement of scaffolding to DLG1 for PKC alpha to promote cellular migration.

作者

我是这篇论文的作者
点击您的名字以认领此论文并将其添加到您的个人资料中。

评论

主要评分

4.6
评分不足

次要评分

新颖性
-
重要性
-
科学严谨性
-
评价这篇论文

推荐

暂无数据
暂无数据