GONUTS has been updated to MW1.31 Most things seem to be working but be sure to report problems.

Have any questions? Please email us at ecoliwiki@gmail.com

TableEdit

Jump to: navigation, search

PMID:22174871

You don't have sufficient rights on this wiki to edit tables. Perhaps you need to log in. Changes you make in the Table editor will not be saved back to the wiki

See Help for Help on this wiki. See the documentation for how to use the table editor

Citation

Ravenscroft, G, Jackaman, C, Sewry, CA, McNamara, E, Squire, SE, Potter, AC, Papadimitriou, J, Griffiths, LM, Bakker, AJ, Davies, KE, Laing, NG and Nowak, KJ (2011) Actin nemaline myopathy mouse reproduces disease, suggests other actin disease phenotypes and provides cautionary note on muscle transgene expression. PLoS ONE 6:e28699

Abstract

Mutations in the skeletal muscle α-actin gene (ACTA1) cause congenital myopathies including nemaline myopathy, actin aggregate myopathy and rod-core disease. The majority of patients with ACTA1 mutations have severe hypotonia and do not survive beyond the age of one. A transgenic mouse model was generated expressing an autosomal dominant mutant (D286G) of ACTA1 (identified in a severe nemaline myopathy patient) fused with EGFP. Nemaline bodies were observed in multiple skeletal muscles, with serial sections showing these correlated to aggregates of the mutant skeletal muscle α-actin-EGFP. Isolated extensor digitorum longus and soleus muscles were significantly weaker than wild-type (WT) muscle at 4 weeks of age, coinciding with the peak in structural lesions. These 4 week-old mice were ~30% less active on voluntary running wheels than WT mice. The α-actin-EGFP protein clearly demonstrated that the transgene was expressed equally in all myosin heavy chain (MHC) fibre types during the early postnatal period, but subsequently became largely confined to MHCIIB fibres. Ringbinden fibres, internal nuclei and myofibrillar myopathy pathologies, not typical features in nemaline myopathy or patients with ACTA1 mutations, were frequently observed. Ringbinden were found in fast fibre predominant muscles of adult mice and were exclusively MHCIIB-positive fibres. Thus, this mouse model presents a reliable model for the investigation of the pathobiology of nemaline body formation and muscle weakness and for evaluation of potential therapeutic interventions. The occurrence of core-like regions, internal nuclei and ringbinden will allow analysis of the mechanisms underlying these lesions. The occurrence of ringbinden and features of myofibrillar myopathy in this mouse model of ACTA1 disease suggests that patients with these pathologies and no genetic explanation should be screened for ACTA1 mutations.

Links

PubMed PMC3235150 Online version:10.1371/journal.pone.0028699

Keywords

Actins/metabolism; Animals; Behavior, Animal; Disease Models, Animal; Gene Expression; Green Fluorescent Proteins/metabolism; Mice; Mice, Inbred C57BL; Mice, Transgenic; Muscle Contraction/physiology; Muscle Fibers, Skeletal/pathology; Muscle Fibers, Skeletal/ultrastructure; Muscle, Skeletal/metabolism; Muscle, Skeletal/pathology; Muscle, Skeletal/physiopathology; Muscle, Skeletal/ultrastructure; Myopathies, Nemaline/pathology; Myopathies, Nemaline/physiopathology; Myosin Heavy Chains/metabolism; Phenotype; Recombinant Fusion Proteins/metabolism; Transgenes/genetics

public



Cancel