Your browser doesn't support javascript.
loading
ß-catenin regulates muscle glucose transport via actin remodelling and M-cadherin binding.
Masson, Stewart W C; Sorrenson, Brie; Shepherd, Peter R; Merry, Troy L.
Afiliação
  • Masson SWC; Discipline of Nutrition, Faculty of Medical and Health Sciences, The University of Auckland, Auckland, New Zealand; Maurice Wilkins Centre for Molecular Biodiscovery, The University of Auckland, Auckland, New Zealand.
  • Sorrenson B; Department of Molecular Medicine and Pathology, Faculty of Medical and Health Sciences, The University of Auckland, Auckland, New Zealand.
  • Shepherd PR; Maurice Wilkins Centre for Molecular Biodiscovery, The University of Auckland, Auckland, New Zealand; Department of Molecular Medicine and Pathology, Faculty of Medical and Health Sciences, The University of Auckland, Auckland, New Zealand.
  • Merry TL; Discipline of Nutrition, Faculty of Medical and Health Sciences, The University of Auckland, Auckland, New Zealand; Maurice Wilkins Centre for Molecular Biodiscovery, The University of Auckland, Auckland, New Zealand. Electronic address: t.merry@auckland.ac.nz.
Mol Metab ; 42: 101091, 2020 12.
Article em En | MEDLINE | ID: mdl-33011305
ABSTRACT

OBJECTIVE:

Skeletal muscle glucose disposal following a meal is mediated through insulin-stimulated movement of the GLUT4-containing vesicles to the cell surface. The highly conserved scaffold-protein ß-catenin is an emerging regulator of vesicle trafficking in other tissues. Here, we investigated the involvement of ß-catenin in skeletal muscle insulin-stimulated glucose transport.

METHODS:

Glucose homeostasis and transport was investigated in inducible muscle specific ß-catenin knockout (BCAT-mKO) mice. The effect of ß-catenin deletion and mutation of ß-catenin serine 552 on signal transduction, glucose uptake and protein-protein interactions were determined in L6-G4-myc cells, and ß-catenin insulin-responsive binding partners were identified via immunoprecipitation coupled to label-free proteomics.

RESULTS:

Skeletal muscle specific deletion of ß-catenin impaired whole-body insulin sensitivity and insulin-stimulated glucose uptake into muscle independent of canonical Wnt signalling. In response to insulin, ß-catenin was phosphorylated at serine 552 in an Akt-dependent manner, and in L6-G4-myc cells, mutation of ß-cateninS552 impaired insulin-induced actin-polymerisation, resulting in attenuated insulin-induced glucose transport and GLUT4 translocation. ß-catenin was found to interact with M-cadherin in an insulin-dependent ß-cateninS552-phosphorylation dependent manner, and loss of M-cadherin in L6-G4-myc cells attenuated insulin-induced actin-polymerisation and glucose transport.

CONCLUSIONS:

Our data suggest that ß-catenin is a novel mediator of glucose transport in skeletal muscle and may contribute to insulin-induced actin-cytoskeleton remodelling to support GLUT4 translocation.
Assuntos
Palavras-chave

Texto completo: 1 Base de dados: MEDLINE Assunto principal: Actinas / Beta Catenina / Proteínas Facilitadoras de Transporte de Glucose Limite: Animals Idioma: En Revista: Mol Metab Ano de publicação: 2020 Tipo de documento: Article País de afiliação: Nova Zelândia

Texto completo: 1 Base de dados: MEDLINE Assunto principal: Actinas / Beta Catenina / Proteínas Facilitadoras de Transporte de Glucose Limite: Animals Idioma: En Revista: Mol Metab Ano de publicação: 2020 Tipo de documento: Article País de afiliação: Nova Zelândia