Your browser doesn't support javascript.
loading
FHL1 mutants that cause clinically distinct human myopathies form protein aggregates and impair myoblast differentiation.
Wilding, Brendan R; McGrath, Meagan J; Bonne, Gisèle; Mitchell, Christina A.
Afiliação
  • Wilding BR; Department of Biochemistry & Molecular Biology, Faculty of Medicine, Nursing & Health Sciences, Monash University, Clayton, VIC 3800, Australia.
  • McGrath MJ; Department of Biochemistry & Molecular Biology, Faculty of Medicine, Nursing & Health Sciences, Monash University, Clayton, VIC 3800, Australia.
  • Bonne G; Inserm, U974, Paris, F-75013, France Université Pierre et Marie Curie-Paris 6, UM 76, CNRS, UMR7215, Institut de Myologie, IFR14, Paris, F-75013, France AP-HP, Groupe Hospitalier Pitié-Salpêtrière, U.F. Cardiogénétique et Myogénétique Moléculaire, Service de Biochimie Métabolique, Paris, F-75013, Fr
  • Mitchell CA; Department of Biochemistry & Molecular Biology, Faculty of Medicine, Nursing & Health Sciences, Monash University, Clayton, VIC 3800, Australia christina.mitchell@monash.edu.
J Cell Sci ; 127(Pt 10): 2269-81, 2014 May 15.
Article em En | MEDLINE | ID: mdl-24634512
ABSTRACT
FHL1 mutations cause several clinically heterogeneous myopathies, including reducing body myopathy (RBM), scapuloperoneal myopathy (SPM) and X-linked myopathy with postural muscle atrophy (XMPMA). The molecular mechanisms underlying the pathogenesis of FHL1 myopathies are unknown. Protein aggregates, designated 'reducing bodies', that contain mutant FHL1 are detected in RBM muscle but not in several other FHL1 myopathies. Here, RBM, SPM and XMPMA FHL1 mutants were expressed in C2C12 cells and showed equivalent protein expression to wild-type FHL1. These mutants formed aggregates that were positive for the reducing body stain Menadione-NBT, analogous to RBM muscle aggregates. However, hypertrophic cardiomyopathy (HCM) and Emery-Dreifuss muscular dystrophy (EDMD) FHL1 mutants generally exhibited reduced expression. Wild-type FHL1 promotes myoblast differentiation; however, RBM, SPM and XMPMA mutations impaired differentiation, consistent with a loss of normal FHL1 function. Furthermore, SPM and XMPMA FHL1 mutants retarded myotube formation relative to vector control, consistent with a dominant-negative or toxic function. Mutant FHL1 myotube formation was partially rescued by expression of a constitutively active FHL1-binding partner, NFATc1. This is the first study to show that FHL1 mutations identified in several clinically distinct myopathies lead to similar protein aggregation and impair myotube formation, suggesting a common pathogenic mechanism despite heterogeneous clinical features.
Assuntos
Palavras-chave

Texto completo: 1 Coleções: 01-internacional Base de dados: MEDLINE Assunto principal: Mioblastos / Peptídeos e Proteínas de Sinalização Intracelular / Proteínas com Domínio LIM / Proteínas Musculares Limite: Humans Idioma: En Revista: J Cell Sci Ano de publicação: 2014 Tipo de documento: Article País de afiliação: Austrália

Texto completo: 1 Coleções: 01-internacional Base de dados: MEDLINE Assunto principal: Mioblastos / Peptídeos e Proteínas de Sinalização Intracelular / Proteínas com Domínio LIM / Proteínas Musculares Limite: Humans Idioma: En Revista: J Cell Sci Ano de publicação: 2014 Tipo de documento: Article País de afiliação: Austrália