Kindlin-2 promotes rear focal adhesion disassembly and directional persistence in cell migration

Author:

Liu Jie1,Liu Zhongzhen1,Chen Keng1,Chen Wei1,Fang Xiyuan1,Li Meng1,Zhou Xuening1,Ding Ning1,Lei Huan1,Guo Chen1,Qian Tao1,Wang Yilin2,Liu Lin3,Chen Yonglong1,Zhao Hui4,Sun Ying1,Deng Yi1ORCID,Wu Chuanyue5

Affiliation:

1. Department of Biology, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, and Shenzhen Key Laboratory of Cell Microenvironment, Southern University of Science and Technology, Shenzhen 518055, China

2. Core Research Facilities, Southern University of Science and Technology, Shenzhen 518055, China

3. College of Life Sciences, Nan Kai University, Tianjin, 300071, China

4. School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong

5. Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA

Abstract

Cell migration involves front-rear asymmetric FA dynamics, which facilitates trailing edge detachment and directional persistence. Here we show that kinldin-2 is critical for FA sliding and disassembly in migrating cells. Loss of kindlin-2 markedly reduced FA number and selectively impaired rear FA sliding and disassembly, resulting in defective rear retraction and reduced directional persistence during cell migration. Kindlin-2 deficient cells failed to develop serum-induced actomyosin-dependent tension at FAs. At the molecular level, kindlin-2 directly interacted with myosin light chain kinase (MLCK), which was enhanced in response to serum stimulation. Serum deprivation inhibited rear FA disassembly, which was released in response to serum stimulation. Overexpression of the MLCK-binding kindlin-2 F0F1 fragment (aa 1-167), which inhibits the interaction of endogenous kindlin-2 with MLCK, phenocopied kindlin-2 deficiency-induced migration defects. Inhibition of MLCK, like loss of kindlin-2, also impaired trailing edge detachment, rear FA disassembly and directional persistence. These results suggest a role of kindlin-2 in promoting actomyosin contractility at FAs, leading to increased rear FA sliding and disassembly and directional persistence in cell migration.

Funder

Natural Science Foundation of Guangdong Province

Ministry of Science and Technology of the People's Republic of China

National Natural Science Foundation of China

Shenzhen Municipal Science and Technology Innovation Council

Research Grants Council, University Grants Committee

National Key R&D Program of China

Publisher

The Company of Biologists

Subject

Cell Biology

同舟云学术

1.学者识别学者识别

2.学术分析学术分析

3.人才评估人才评估

"同舟云学术"是以全球学者为主线,采集、加工和组织学术论文而形成的新型学术文献查询和分析系统,可以对全球学者进行文献检索和人才价值评估。用户可以通过关注某些学科领域的顶尖人物而持续追踪该领域的学科进展和研究前沿。经过近期的数据扩容,当前同舟云学术共收录了国内外主流学术期刊6万余种,收集的期刊论文及会议论文总量共计约1.5亿篇,并以每天添加12000余篇中外论文的速度递增。我们也可以为用户提供个性化、定制化的学者数据。欢迎来电咨询!咨询电话:010-8811{复制后删除}0370

www.globalauthorid.com

TOP

Copyright © 2019-2024 北京同舟云网络信息技术有限公司
京公网安备11010802033243号  京ICP备18003416号-3