Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
Hum Mol Genet ; 25(16): 3555-3563, 2016 08 15.
Article in English | MEDLINE | ID: mdl-27378686

ABSTRACT

In preclinical models for Duchenne muscular dystrophy, dystrophin restoration during adeno-associated virus (AAV)-U7-mediated exon-skipping therapy was shown to decrease drastically after six months in treated muscles. This decline in efficacy is strongly correlated with the loss of the therapeutic AAV genomes, probably due to alterations of the dystrophic myofiber membranes. To improve the membrane integrity of the dystrophic myofibers at the time of AAV-U7 injection, mdx muscles were pre-treated with a single dose of the peptide-phosphorodiamidate morpholino (PPMO) antisense oligonucleotides that induced temporary dystrophin expression at the sarcolemma. The PPMO pre-treatment allowed efficient maintenance of AAV genomes in mdx muscles and enhanced the AAV-U7 therapy effect with a ten-fold increase of the protein level after 6 months. PPMO pre-treatment was also beneficial to AAV-mediated gene therapy with transfer of micro-dystrophin cDNA into muscles. Therefore, avoiding vector genome loss after AAV injection by PPMO pre-treatment would allow efficient long-term restoration of dystrophin and the use of lower and thus safer vector doses for Duchenne patients.


Subject(s)
Dystrophin/genetics , Genetic Therapy , Morpholinos/administration & dosage , Muscular Dystrophy, Animal/therapy , Muscular Dystrophy, Duchenne/therapy , Oligonucleotides, Antisense/administration & dosage , Animals , Dependovirus/genetics , Exons/genetics , Gene Transfer Techniques , Genetic Vectors/administration & dosage , Humans , Mice, Inbred mdx , Muscle, Skeletal/drug effects , Muscle, Skeletal/pathology , Muscular Dystrophy, Animal/genetics , Muscular Dystrophy, Duchenne/genetics , Sarcolemma/drug effects , Sarcolemma/pathology
2.
Nucleic Acids Res ; 41(17): 8391-402, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23861443

ABSTRACT

RNA-based therapeutic approaches using splice-switching oligonucleotides have been successfully applied to rescue dystrophin in Duchenne muscular dystrophy (DMD) preclinical models and are currently being evaluated in DMD patients. Although the modular structure of dystrophin protein tolerates internal deletions, many mutations that affect nondispensable domains of the protein require further strategies. Among these, trans-splicing technology is particularly attractive, as it allows the replacement of any mutated exon by its normal version as well as introducing missing exons or correcting duplication mutations. We have applied such a strategy in vitro by using cotransfection of pre-trans-splicing molecule (PTM) constructs along with a reporter minigene containing part of the dystrophin gene harboring the stop-codon mutation found in the mdx mouse model of DMD. Optimization of the different functional domains of the PTMs allowed achieving accurate and efficient trans-splicing of up to 30% of the transcript encoded by the cotransfected minigene. Optimized parameters included mRNA stabilization, choice of splice site sequence, inclusion of exon splice enhancers and artificial intronic sequence. Intramuscular delivery of adeno-associated virus vectors expressing PTMs allowed detectable levels of dystrophin in mdx and mdx4Cv, illustrating that a given PTM can be suitable for a variety of mutations.


Subject(s)
Dystrophin/genetics , Trans-Splicing , Animals , Dependovirus/genetics , Dystrophin/analysis , Exons , Genetic Vectors , Genotype , Humans , Introns , Mice , Mice, Inbred mdx , Muscle Fibers, Skeletal/chemistry , Muscles/chemistry , Muscular Dystrophy, Duchenne/genetics , NIH 3T3 Cells , RNA Splice Sites , RNA, Messenger/analysis
3.
Mol Ther ; 21(8): 1551-8, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23752313

ABSTRACT

In the context of future adeno-associated viral (AAV)-based clinical trials for Duchenne myopathy, AAV genome fate in dystrophic muscles is of importance considering the viral capsid immunogenicity that prohibits recurring treatments. We showed that AAV genomes encoding non-therapeutic U7 were lost from mdx dystrophic muscles within 3 weeks after intramuscular injection. In contrast, AAV genomes encoding U7ex23 restoring expression of a slightly shortened dystrophin were maintained endorsing that the arrest of the dystrophic process is crucial for maintaining viral genomes in transduced fibers. Indeed, muscles treated with low doses of AAV-U7ex23, resulting in sub-optimal exon skipping, displayed much lower titers of viral genomes, showing that sub-optimal dystrophin restoration does not prevent AAV genome loss. We also followed therapeutic viral genomes in severe dystrophic dKO mice over time after systemic treatment with scAAV9-U7ex23. Dystrophin restoration decreased significantly between 3 and 12 months in various skeletal muscles, which was correlated with important viral genome loss, except in the heart. Altogether, these data show that the success of future AAV-U7 therapy for Duchenne patients would require optimal doses of AAV-U7 to induce substantial levels of dystrophin to stabilize the treated fibers and maintain the long lasting effect of the treatment.


Subject(s)
Alternative Splicing , Dependovirus/genetics , Genetic Vectors/genetics , Genome, Viral , Muscular Dystrophy, Animal/genetics , Muscular Dystrophy, Duchenne/genetics , RNA, Small Nuclear/genetics , Animals , Cardiotoxins/pharmacology , Dependovirus/metabolism , Dystrophin/genetics , Dystrophin/metabolism , Exons , Gene Expression , Genetic Therapy , Genetic Vectors/administration & dosage , Genetic Vectors/metabolism , Humans , Injections, Intramuscular , Mice , Mice, Inbred mdx , Muscle, Skeletal/drug effects , Muscle, Skeletal/metabolism , Muscular Dystrophy, Animal/therapy , Muscular Dystrophy, Duchenne/therapy
4.
Mol Ther ; 20(11): 2120-33, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22968479

ABSTRACT

Duchenne muscular dystrophy (DMD) is an X-linked recessive disorder resulting from lesions of the gene encoding dystrophin. These usually consist of large genomic deletions, the extents of which are not correlated with the severity of the phenotype. Out-of-frame deletions give rise to dystrophin deficiency and severe DMD phenotypes, while internal deletions that produce in-frame mRNAs encoding truncated proteins can lead to a milder myopathy known as Becker muscular dystrophy (BMD). Widespread restoration of dystrophin expression via adeno-associated virus (AAV)-mediated exon skipping has been successfully demonstrated in the mdx mouse model and in cardiac muscle after percutaneous transendocardial delivery in the golden retriever muscular dystrophy dog (GRMD) model. Here, a set of optimized U7snRNAs carrying antisense sequences designed to rescue dystrophin were delivered into GRMD skeletal muscles by AAV1 gene transfer using intramuscular injection or forelimb perfusion. We show sustained correction of the dystrophic phenotype in extended muscle areas and partial recovery of muscle strength. Muscle architecture was improved and fibers displayed the hallmarks of mature and functional units. A 5-year follow-up ruled out immune rejection drawbacks but showed a progressive decline in the number of corrected muscle fibers, likely due to the persistence of a mild dystrophic process such as occurs in BMD phenotypes. Although AAV-mediated exon skipping was shown safe and efficient to rescue a truncated dystrophin, it appears that recurrent treatments would be required to maintain therapeutic benefit ahead of the progression of the disease.


Subject(s)
Alternative Splicing , Dependovirus/genetics , Dystrophin/genetics , Muscular Dystrophy, Animal/therapy , Oligoribonucleotides, Antisense/genetics , RNA, Small Nuclear/genetics , Animals , Base Sequence , Calcium/metabolism , Dogs , Exons , Forelimb/physiopathology , Genetic Therapy , Genetic Vectors/administration & dosage , Injections, Intramuscular , Molecular Sequence Data , Muscle Contraction , Muscle Fibers, Skeletal/metabolism , Muscle Fibers, Skeletal/pathology , Muscle Strength , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Muscle, Skeletal/physiopathology , Muscular Dystrophy, Animal/physiopathology , Transcription, Genetic , Utrophin/genetics , Utrophin/metabolism
5.
PLoS One ; 5(5): e10894, 2010 May 28.
Article in English | MEDLINE | ID: mdl-20531943

ABSTRACT

BACKGROUND: Trans-splicing strategies for mRNA repair involve engineered transcripts designed to anneal target mRNAs in order to interfere with their natural splicing, giving rise to mRNA chimeras where endogenous mutated exons have been replaced by exogenous replacement sequences. A number of trans-splicing molecules have already been proposed for replacing either the 5' or the 3' part of transcripts to be repaired. Here, we show the feasibility of RNA surgery by using a double trans-splicing approach allowing the specific substitution of a given mutated exon. METHODOLOGY/PRINCIPAL FINDINGS: As a target we used a minigene encoding a fragment of the mdx dystrophin gene enclosing the mutated exon (exon 23). This minigene was cotransfected with a variety of exon exchange constructions, differing in their annealing domains. We obtained accurate and efficient replacement of exon 23 in the mRNA target. Adding up a downstream intronic splice enhancer DISE in the exon exchange molecule enhanced drastically its efficiency up to 25-45% of repair depending on the construction in use. CONCLUSIONS/SIGNIFICANCE: These results demonstrate the possibility to fix up mutated exons, refurbish deleted exons and introduce protein motifs, while keeping natural untranslated sequences, which are essential for mRNA stability and translation regulation. Conversely to the well-known exon skipping, exon exchange has the advantage to be compatible with almost any type of mutations and more generally to a wide range of genetic conditions. In particular, it allows addressing disorders caused by dominant mutations.


Subject(s)
Dystrophin/genetics , Exons/genetics , Muscular Dystrophy, Animal/genetics , Animals , Base Sequence , DNA Mutational Analysis , Enhancer Elements, Genetic/genetics , Introns/genetics , Mice , Molecular Sequence Data , NIH 3T3 Cells , RNA, Messenger/genetics , Trans-Splicing/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...