RhoA Regulates G1-S Progression of Gastric Cancer Cells by Modulation of Multiple INK4 Family Tumor Suppressors

S Zhang, Q Tang, F Xu, Y Xue, Z Zhen, Y Deng… - Molecular Cancer …, 2009 - AACR
S Zhang, Q Tang, F Xu, Y Xue, Z Zhen, Y Deng, M Liu, J Chen, S Liu, M Qiu, Z Liao, Z Li…
Molecular Cancer Research, 2009AACR
RhoA, a member of the Rho GTPase family, has been extensively studied in the regulation
of cytoskeletal dynamics, gene transcription, cell cycle progression, and cell transformation.
Overexpression of RhoA is found in many malignancies and elevated RhoA activity is
associated with proliferation phenotypes of cancer cells. We reported previously that RhoA
was hyperactivated in gastric cancer tissues and suppression of RhoA activity could partially
reverse the proliferation phenotype of gastric cancer cells, but the underlying mechanism …
Abstract
RhoA, a member of the Rho GTPase family, has been extensively studied in the regulation of cytoskeletal dynamics, gene transcription, cell cycle progression, and cell transformation. Overexpression of RhoA is found in many malignancies and elevated RhoA activity is associated with proliferation phenotypes of cancer cells. We reported previously that RhoA was hyperactivated in gastric cancer tissues and suppression of RhoA activity could partially reverse the proliferation phenotype of gastric cancer cells, but the underlying mechanism has yet to be elucidated. It has been reported that RhoA activation is crucial for the cell cycle G1-S procession through the regulation of Cip/Kip family tumor suppressors in benign cell lines. In this study, we found that selective suppression of RhoA or its effectors mammalian Diaphanous 1 and Rho kinase (ROCK) by small interfering RNA and a pharmacologic inhibitor effectively inhibited proliferation and cell cycle G1-S transition in gastric cancer lines. Down-regulation of RhoA-mammalian Diaphanous 1 pathway, but not RhoA-ROCK pathway, caused an increase in the expression of p21Waf1/Cip1 and p27Kip1, which are coupled with reduced expression and activity of CDK2 and a cytoplasmic mislocalization of p27Kip1. Suppression of RhoA-ROCK pathway, on the other hand, resulted in an accumulation of p15INK4b, p16INK4a, p18INK4c, and p19INK4d, leading to reduced expression and activities of CDK4 and CDK6. Thus, RhoA may use two distinct effector pathways in regulating the G1-S progression of gastric cancer cells.(Mol Cancer Res 2009;7(4):570–80)
AACR