Actin nemaline myopathy mouse reproduces disease, suggests other actin disease phenotypes and provides cautionary note on muscle transgene expression.

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 gen...

Full description

Bibliographic Details
Main Authors: Gianina Ravenscroft, Connie Jackaman, Caroline A Sewry, Elyshia McNamara, Sarah E Squire, Allyson C Potter, John Papadimitriou, Lisa M Griffiths, Anthony J Bakker, Kay E Davies, Nigel G Laing, Kristen J Nowak
Format: Article
Language:English
Published: Public Library of Science (PLoS) 2011-01-01
Series:PLoS ONE
Online Access:http://europepmc.org/articles/PMC3235150?pdf=render
id doaj-8753ec1ece004cf593775b53dd3a6757
record_format Article
spelling doaj-8753ec1ece004cf593775b53dd3a67572020-11-25T00:08:49ZengPublic Library of Science (PLoS)PLoS ONE1932-62032011-01-01612e2869910.1371/journal.pone.0028699Actin nemaline myopathy mouse reproduces disease, suggests other actin disease phenotypes and provides cautionary note on muscle transgene expression.Gianina RavenscroftConnie JackamanCaroline A SewryElyshia McNamaraSarah E SquireAllyson C PotterJohn PapadimitriouLisa M GriffithsAnthony J BakkerKay E DaviesNigel G LaingKristen J NowakMutations 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.http://europepmc.org/articles/PMC3235150?pdf=render
collection DOAJ
language English
format Article
sources DOAJ
author Gianina Ravenscroft
Connie Jackaman
Caroline A Sewry
Elyshia McNamara
Sarah E Squire
Allyson C Potter
John Papadimitriou
Lisa M Griffiths
Anthony J Bakker
Kay E Davies
Nigel G Laing
Kristen J Nowak
spellingShingle Gianina Ravenscroft
Connie Jackaman
Caroline A Sewry
Elyshia McNamara
Sarah E Squire
Allyson C Potter
John Papadimitriou
Lisa M Griffiths
Anthony J Bakker
Kay E Davies
Nigel G Laing
Kristen J Nowak
Actin nemaline myopathy mouse reproduces disease, suggests other actin disease phenotypes and provides cautionary note on muscle transgene expression.
PLoS ONE
author_facet Gianina Ravenscroft
Connie Jackaman
Caroline A Sewry
Elyshia McNamara
Sarah E Squire
Allyson C Potter
John Papadimitriou
Lisa M Griffiths
Anthony J Bakker
Kay E Davies
Nigel G Laing
Kristen J Nowak
author_sort Gianina Ravenscroft
title Actin nemaline myopathy mouse reproduces disease, suggests other actin disease phenotypes and provides cautionary note on muscle transgene expression.
title_short Actin nemaline myopathy mouse reproduces disease, suggests other actin disease phenotypes and provides cautionary note on muscle transgene expression.
title_full Actin nemaline myopathy mouse reproduces disease, suggests other actin disease phenotypes and provides cautionary note on muscle transgene expression.
title_fullStr Actin nemaline myopathy mouse reproduces disease, suggests other actin disease phenotypes and provides cautionary note on muscle transgene expression.
title_full_unstemmed Actin nemaline myopathy mouse reproduces disease, suggests other actin disease phenotypes and provides cautionary note on muscle transgene expression.
title_sort actin nemaline myopathy mouse reproduces disease, suggests other actin disease phenotypes and provides cautionary note on muscle transgene expression.
publisher Public Library of Science (PLoS)
series PLoS ONE
issn 1932-6203
publishDate 2011-01-01
description 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.
url http://europepmc.org/articles/PMC3235150?pdf=render
work_keys_str_mv AT gianinaravenscroft actinnemalinemyopathymousereproducesdiseasesuggestsotheractindiseasephenotypesandprovidescautionarynoteonmuscletransgeneexpression
AT conniejackaman actinnemalinemyopathymousereproducesdiseasesuggestsotheractindiseasephenotypesandprovidescautionarynoteonmuscletransgeneexpression
AT carolineasewry actinnemalinemyopathymousereproducesdiseasesuggestsotheractindiseasephenotypesandprovidescautionarynoteonmuscletransgeneexpression
AT elyshiamcnamara actinnemalinemyopathymousereproducesdiseasesuggestsotheractindiseasephenotypesandprovidescautionarynoteonmuscletransgeneexpression
AT sarahesquire actinnemalinemyopathymousereproducesdiseasesuggestsotheractindiseasephenotypesandprovidescautionarynoteonmuscletransgeneexpression
AT allysoncpotter actinnemalinemyopathymousereproducesdiseasesuggestsotheractindiseasephenotypesandprovidescautionarynoteonmuscletransgeneexpression
AT johnpapadimitriou actinnemalinemyopathymousereproducesdiseasesuggestsotheractindiseasephenotypesandprovidescautionarynoteonmuscletransgeneexpression
AT lisamgriffiths actinnemalinemyopathymousereproducesdiseasesuggestsotheractindiseasephenotypesandprovidescautionarynoteonmuscletransgeneexpression
AT anthonyjbakker actinnemalinemyopathymousereproducesdiseasesuggestsotheractindiseasephenotypesandprovidescautionarynoteonmuscletransgeneexpression
AT kayedavies actinnemalinemyopathymousereproducesdiseasesuggestsotheractindiseasephenotypesandprovidescautionarynoteonmuscletransgeneexpression
AT nigelglaing actinnemalinemyopathymousereproducesdiseasesuggestsotheractindiseasephenotypesandprovidescautionarynoteonmuscletransgeneexpression
AT kristenjnowak actinnemalinemyopathymousereproducesdiseasesuggestsotheractindiseasephenotypesandprovidescautionarynoteonmuscletransgeneexpression
_version_ 1725414396205727744