PRMT5 methylating Smad4 activates TGF-β signaling and promotes colorectal cancer metastasis

Author:

Wang Guihua1ORCID,Liu Anyi1,Yu Chengxin1,Qiu Cheng1,Wu Qi1,Huang Changsheng1,Li Xun1,She Xiaowei1,Wan Kairui,Liu Lang1,Li Mao1,Wang Zhihong1,Chen Yaqi1,Hu Fuqing1,Song Da1,Li Kangdi1,zhao chong,Deng Haiteng2ORCID,Sun Xuling3,Xu Feng,Lai Senyan,Luo Xuelai4,Hu Junbo5ORCID

Affiliation:

1. Tongji Hospital, Huazhong University of Science and Technology

2. Tsinghua University

3. Huazhong University of Science and Technology

4. Tongji Cancer Research Institute

5. Cancer Biology Research Center (Key laboratory of the ministry of education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan

Abstract

Abstract Perturbations in transforming growth factor- β (TGF-β) signaling can lead to a plethora of diseases, including cancer. Mutations and posttranslational modifications (PTMs) of the partner of Smad complexes contribute to the dysregulation of TGF-β signaling. Here, we reported a PTM of Smad4, R361 methylation, that was critical for Smad complexes formation and TGF-β signaling activation. Through mass spectrometric, co-immunoprecipitation (Co-IP) and immunofluorescent (IF) assays, we found that oncogene protein arginine methyltransferase 5 (PRMT5) interacted with Smad4 under TGF-β1 treatment. Mechanically, PRMT5 triggered Smad4 methylation at R361 and induced Smad complexes formation and nuclear import. Furthermore, we emphasized that PRMT5 interacting and methylating Smad4 was required for TGF-β1-induced epithelial-mesenchymal transition (EMT) and colorectal cancer (CRC) metastasis, and Smad4 R361 mutation diminished PRMT5 and TGF-β1-induced metastasis. In addition, highly expressed PRMT5 or high level of Smad4 R361 methylation indicated worse outcomes in clinical specimens analysis. Collectively, our study highlights the critical interaction of PRMT5 and Smad4 and the roles of Smad4 R361 methylation for controlling TGF-β signaling during metastasis. We provided a new insight for Smad4 activation. And this study indicated that blocking PRMT5-Smad4 signaling might be an effective targeting strategy in Smad4 wide type CRC.

Publisher

Research Square Platform LLC

Reference47 articles.

1. Transforming Growth Factor-β Signaling in Immunity and Cancer;Batlle E;Immunity,2019

2. Massagué J. TGFβ in Cancer. Cell. 2008;134(2):215–30.

3. The role of TGF-β/SMAD4 signaling in cancer;Zhao M;Int J Biol Sci,2018

4. Targeting TGF-β Signaling in Cancer;Colak S;Trends in Cancer,2017

5. TGF-Β-induced epithelial to mesenchymal transition;Xu J;Cell Res,2009

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