Articles

< Previous         Next >  
Acetylation of ezrin regulates membrane–cytoskeleton interaction underlying CCL18-elicited cell migration
Xiaoyu Song1,2,3 , Wanjuan Wang1,2 , Haowei Wang2,4 , Xiao Yuan2 , Fengrui Yang2,3 , Lingli Zhao2,3 , McKay Mullen2,3 , Shihao Du1,2 , Najdat Zohbi2,3 , Saravanakumar Muthusamy2,3 , Yalei Cao1,2 , Jiying Jiang2 , Peng Xia2 , Ping He2 , Mingrui Ding2,3 , Nerimah Emmett3 , Mingming Ma2 , Quan Wu2 , Hadiyah-Nicole Green1,3 , Xia Ding1,2,3,* , Dongmei Wang2,* , Fengsong Wang2,5,* , Xing Liu1,2,3,*
1School of Traditional Medicine, Beijing University of Chinese Medicine, Beijing, China
2MOE Key Laboratory for Membraneless Organelles & Cellular Dynamics, Hefei National Center for Physical Sciences at the Microscale, Hefei, China
3Morehouse School of Medicine, Keck Center for Organoids Plasticity, Atlanta, GA, USA
4Optics and Optical Engineering, University of Science and Technology of China, Hefei, China
5School of Life Science, Anhui Medical University, Hefei, China
These authors contributed equally to this work.
*Correspondence to:Xia Ding , Email:dingx@bucm.edu.cn; Dongmei Wang , Email:wangdm@ustc.edu.cn; Fengsong Wang , Email:fengsongw@ahmu.edu.cn Xing Liu , Email:xing1017@ustc.edu.cn
J Mol Cell Biol, Volume 12, Issue 6, June 2020, 424-437,  https://doi.org/10.1093/jmcb/mjz099
Keyword: ezrin, acetylation, phosphorylation, actin, cell migration

Ezrin, a membrane–cytoskeleton linker protein, plays an essential role in cell polarity establishment, cell migration, and division. Recent studies show that ezrin phosphorylation regulates breast cancer metastasis by promoting cancer cell survivor and promotes intrahepatic metastasis via cell migration. However, it was less characterized whether there are additional post-translational modifications and/or post-translational crosstalks on ezrin underlying context-dependent breast cancer cell migration and invasion. Here we show that ezrin is acetylated by p300/CBP-associated factor (PCAF) in breast cancer cells in response to CCL18 stimulation. Ezrin physically interacts with PCAF and is a cognate substrate of PCAF. The acetylation site of ezrin was mapped by mass spectrometric analyses, and dynamic acetylation of ezrin is essential for CCL18-induced breast cancer cell migration and invasion. Mechanistically, the acetylation reduced the lipid-binding activity of ezrin to ensure a robust and dynamic cycling between the plasma membrane and cytosol in response to CCL18 stimulation. Biochemical analyses show that ezrin acetylation prevents the phosphorylation of Thr567. Using atomic force microscopic measurements, our study revealed that acetylation of ezrin induced its unfolding into a dominant structure, which prevents ezrin phosphorylation at Thr567. Thus, these results present a previously undefined mechanism by which CCL18-elicited crosstalks between the acetylation and phosphorylation on ezrin control breast cancer cell migration and invasion. This suggests that targeting PCAF signaling could be a potential therapeutic strategy for combating hyperactive ezrin-driven cancer progression.