Show simple item record

dc.contributor.authorRavenscroft, G.
dc.contributor.authorJackaman, Connie
dc.contributor.authorSewry, C.
dc.contributor.authorMcnamara, E.
dc.contributor.authorSquire, S.
dc.contributor.authorPotter, A.
dc.contributor.authorPapadimitriou, J.
dc.contributor.authorGriffiths, L.
dc.contributor.authorBakker, A.
dc.contributor.authorDavies, K.
dc.contributor.authorLaing, N.
dc.contributor.authorNowak, K.
dc.date.accessioned2017-03-15T22:03:55Z
dc.date.available2017-03-15T22:03:55Z
dc.date.created2017-02-24T00:09:08Z
dc.date.issued2011
dc.identifier.citationRavenscroft, G. and Jackaman, C. and Sewry, C. and Mcnamara, E. and Squire, S. and Potter, A. and Papadimitriou, J. et al. 2011. Actin Nemaline Myopathy Mouse Reproduces disease, suggests other actin disease phenotypes and provides cautionary note on muscle transgene expression. PLoS ONE. 6 (12): e28699.
dc.identifier.urihttp://hdl.handle.net/20.500.11937/49290
dc.identifier.doi10.1371/journal.pone.0028699
dc.description.abstract

Mutations in the skeletal muscle a-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 a-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 a-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.

dc.publisherPublic Library of Science
dc.titleActin Nemaline Myopathy Mouse Reproduces disease, suggests other actin disease phenotypes and provides cautionary note on muscle transgene expression
dc.typeJournal Article
dcterms.source.volume6
dcterms.source.number12
dcterms.source.startPagee28699
dcterms.source.endPage1
dcterms.source.issn19326203
dcterms.source.titlePLoS ONE
curtin.departmentSchool of Biomedical Sciences
curtin.accessStatusOpen access via publisher


Files in this item

Thumbnail

This item appears in the following Collection(s)

Show simple item record