↓ Skip to main content

PLOS

Homologous Recombination Mediates Functional Recovery of Dysferlin Deficiency following AAV5 Gene Transfer

Overview of attention for article published in PLOS ONE, June 2012
Altmetric Badge

Mentioned by

twitter
3 X users
patent
6 patents
facebook
2 Facebook pages

Citations

dimensions_citation
63 Dimensions

Readers on

mendeley
42 Mendeley
Title
Homologous Recombination Mediates Functional Recovery of Dysferlin Deficiency following AAV5 Gene Transfer
Published in
PLOS ONE, June 2012
DOI 10.1371/journal.pone.0039233
Pubmed ID
Authors

William E. Grose, K. Reed Clark, Danielle Griffin, Vinod Malik, Kimberly M. Shontz, Chrystal L. Montgomery, Sarah Lewis, Robert H. Brown, Paul M. L. Janssen, Jerry R. Mendell, Louise R. Rodino-Klapac

Abstract

The dysferlinopathies comprise a group of untreatable muscle disorders including limb girdle muscular dystrophy type 2B, Miyoshi myopathy, distal anterior compartment syndrome, and rigid spine syndrome. As with other forms of muscular dystrophy, adeno-associated virus (AAV) gene transfer is a particularly auspicious treatment strategy, however the size of the DYSF cDNA (6.5 kb) negates packaging into traditional AAV serotypes known to express well in muscle (i.e. rAAV1, 2, 6, 8, 9). Potential advantages of a full cDNA versus a mini-gene include: maintaining structural-functional protein domains, evading protein misfolding, and avoiding novel epitopes that could be immunogenic. AAV5 has demonstrated unique plasticity with regards to packaging capacity and recombination of virions containing homologous regions of cDNA inserts has been implicated in the generation of full-length transcripts. Herein we show for the first time in vivo that homologous recombination following AAV5.DYSF gene transfer leads to the production of full length transcript and protein. Moreover, gene transfer of full-length dysferlin protein in dysferlin deficient mice resulted in expression levels sufficient to correct functional deficits in the diaphragm and importantly in skeletal muscle membrane repair. Intravascular regional gene transfer through the femoral artery produced high levels of transduction and enabled targeting of specific muscle groups affected by the dysferlinopathies setting the stage for potential translation to clinical trials. We provide proof of principle that AAV5 mediated delivery of dysferlin is a highly promising strategy for treatment of dysferlinopathies and has far-reaching implications for the therapeutic delivery of other large genes.

X Demographics

X Demographics

The data shown below were collected from the profiles of 3 X users who shared this research output. Click here to find out more about how the information was compiled.
Mendeley readers

Mendeley readers

The data shown below were compiled from readership statistics for 42 Mendeley readers of this research output. Click here to see the associated Mendeley record.

Geographical breakdown

Country Count As %
Korea, Republic of 1 2%
United States 1 2%
Unknown 40 95%

Demographic breakdown

Readers by professional status Count As %
Student > Ph. D. Student 10 24%
Researcher 9 21%
Student > Master 6 14%
Student > Bachelor 5 12%
Other 5 12%
Other 4 10%
Unknown 3 7%
Readers by discipline Count As %
Agricultural and Biological Sciences 13 31%
Biochemistry, Genetics and Molecular Biology 8 19%
Medicine and Dentistry 6 14%
Neuroscience 4 10%
Psychology 1 2%
Other 2 5%
Unknown 8 19%