Regulation of Myogenesis by a Na/K-ATPase α1 Caveolin-Binding Motif

Author:

Huang Minqi1,Wang Xiaoliang12,Banerjee Moumita1,Mukherji Shreya T1,Kutz Laura C1,Zhao Aijie1,Sepanski Michael3,Fan Chen-Ming3,Zhu Guo-Zhang4,Tian Jiang12,Wang Da-Zhi5,Zhu Hua6,Xie Zi-Jian1,Pierre Sandrine V1,Cai Liquan1

Affiliation:

1. Marshall Institute for Interdisciplinary Research (MIIR) at Marshall University, Huntington, WV 25703, USA

2. Joan C. Edwards School of Medicine at Marshall University, Huntington, WV 25703, USA

3. Department of Embryology, Carnegie Institution for Science, 3520 San Martin Drive, Baltimore, MD 21218, USA

4. Department of Biological Sciences, Marshall University, Huntington, WV 25703, USA

5. University of South Florida Health Heart Institute, Morsani College of Medicine, University of South Florida, 560 Channelside Drive, Tampa, FL 33602, USA

6. Department of Surgery, The Ohio State University, 396 Biomedical Research Tower, 460 W. 12th Avenue, Columbus, OH 43210, USA

Abstract

Abstract The N-terminal caveolin-binding motif (CBM) in Na/K-ATPase (NKA) α1 subunit is essential for cell signaling and somitogenesis in animals. To further investigate the molecular mechanism, we have generated CBM mutant human-induced pluripotent stem cells (iPSCs) through CRISPR/Cas9 genome editing and examined their ability to differentiate into skeletal muscle (Skm) cells. Compared with the parental wild-type human iPSCs, the CBM mutant cells lost their ability of Skm differentiation, which was evidenced by the absence of spontaneous cell contraction, marker gene expression, and subcellular myofiber banding structures in the final differentiated induced Skm cells. Another NKA functional mutant, A420P, which lacks NKA/Src signaling function, did not produce a similar defect. Indeed, A420P mutant iPSCs retained intact pluripotency and ability of Skm differentiation. Mechanistically, the myogenic transcription factor MYOD was greatly suppressed by the CBM mutation. Overexpression of a mouse Myod cDNA through lentiviral delivery restored the CBM mutant cells’ ability to differentiate into Skm. Upstream of MYOD, Wnt signaling was demonstrated from the TOPFlash assay to have a similar inhibition. This effect on Wnt activity was further confirmed functionally by defective induction of the presomitic mesoderm marker genes BRACHYURY (T) and MESOGENIN1 (MSGN1) by Wnt3a ligand or the GSK3 inhibitor/Wnt pathway activator CHIR. Further investigation through immunofluorescence imaging and cell fractionation revealed a shifted membrane localization of β-catenin in CBM mutant iPSCs, revealing a novel molecular component of NKA-Wnt regulation. This study sheds light on a genetic regulation of myogenesis through the CBM of NKA and control of Wnt/β-catenin signaling.

Funder

Marshall Institute for Interdisciplinary Research

NIH

Publisher

Oxford University Press (OUP)

Subject

Cell Biology,Developmental Biology,Molecular Medicine

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