Oncotarget

Research Papers:

DGC-specific RHOA mutations maintained cancer cell survival and promoted cell migration via ROCK inactivation

Takashi Nishizawa _, Kiyotaka Nakano, Aya Harada, Miwako Kakiuchi, Shin-Ichi Funahashi, Masami Suzuki, Shumpei Ishikawa and Hiroyuki Aburatani

PDF  |  HTML  |  Supplementary Files  |  How to cite

Oncotarget. 2018; 9:23198-23207. https://doi.org/10.18632/oncotarget.25269

Metrics: PDF 2108 views  |   HTML 3669 views  |   ?  


Abstract

Takashi Nishizawa1, Kiyotaka Nakano1, Aya Harada1, Miwako Kakiuchi2, Shin-Ichi Funahashi1, Masami Suzuki1, Shumpei Ishikawa2,3 and Hiroyuki Aburatani2

1Department for Research, Forerunner Pharma Research Co., Ltd., Tokyo, Japan

2Genome Science Division, Research Center for Advanced Science and Technology, The University of Tokyo, Tokyo, Japan

3Department of Genomic Pathology, Medical Research Institute, Tokyo Medical and Dental University, Tokyo, Japan

Correspondence to:

Takashi Nishizawa, email: [email protected]

Keywords: RHOA; ROCK; diffuse gastric cancer; mutation; ARHGAP

Received: November 26, 2017     Accepted: April 06, 2018     Published: May 01, 2018

ABSTRACT

RHOA missense mutations exist specifically in diffuse type gastric cancers (DGC) and are considered one of the DGC driver genes, but it is not fully understood how RHOA mutations contribute to DGC development. Here we examined how RHOA mutations affect cancer cell survival and cell motility. We revealed that cell survival was maintained by specific mutation sites, namely G17, Y42, and L57. Because these functional mutations suppressed MLC2 phosphorylation and actin stress fiber formation, we realized they act in a dominant-negative fashion against the ROCK pathway. Through the same inactivating mechanism that maintained cell survival, RHOA mutations also increased cell migration activity. Cell survival and migration studies on CLDN18-ARHGAP (CLG) fusions, which are known to be mutually exclusive to RHOA mutations, showed that CLG fusions complemented cell survival under RHOA knockdown condition and also induced cell migration. Site-directed mutagenesis analysis revealed the importance of the GAP domain and indicated that CLG fusions maintained RHOA in the inactive form. Taken together, these findings show that the inactivation of ROCK would be a key step in DGC development, so ROCK activation might provide novel therapeutic opportunities.


Creative Commons License All site content, except where otherwise noted, is licensed under a Creative Commons Attribution 4.0 License.
PII: 25269