Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Hum Gene Ther ; 32(19-20): 1138-1146, 2021 10.
Article in English | MEDLINE | ID: mdl-33765840

ABSTRACT

Duchenne muscular dystrophy (DMD) is an X-linked recessive disease that affects 1:5,000 live male births and is characterized by muscle wasting. By the age of 13 years, affected individuals are often wheelchair bound and suffer from respiratory and cardiac failure, which results in premature death. Although the administration of corticosteroids and ventilation can relieve the symptoms and extend the patients' lifespan, currently no cure exists for DMD. Among the different approaches under preclinical and clinical testing, gene therapy, using adeno-associated viral (AAV) vectors, is one of the most promising. In this study, we delivered intravenously AAV9 vectors expressing the microdystrophin MD1 (ΔR4-R23/ΔCT) under control of the synthetic muscle-specific promoter Spc5-12 and assessed the effect of adding a cardiac-specific cis-regulatory module (designated as CS-CRM4) on its expression profile in skeletal and cardiac muscles. Results show that Spc5-12 promoter, in combination with an AAV serotype that has high tropism for the heart, drives high MD1 expression levels in cardiac muscle in mdx mice. The additional regulatory element CS-CRM4 can further improve MD1 expression in cardiac muscles, but its effect is dose dependent and enhancement becomes evident only at lower vector doses.


Subject(s)
Dystrophin , Muscular Dystrophy, Duchenne , Animals , Dependovirus/genetics , Dystrophin/genetics , Genetic Vectors/genetics , Humans , Male , Mice , Mice, Inbred mdx , Muscle, Skeletal , Muscular Dystrophy, Duchenne/genetics , Muscular Dystrophy, Duchenne/therapy , Myocardium
2.
J Cachexia Sarcopenia Muscle ; 11(3): 768-782, 2020 06.
Article in English | MEDLINE | ID: mdl-32031328

ABSTRACT

BACKGROUND: Spinal muscular atrophy (SMA) is caused by genetic defects in the survival motor neuron 1 (SMN1) gene that lead to SMN deficiency. Different SMN-restoring therapies substantially prolong survival and function in transgenic mice of SMA. However, these therapies do not entirely prevent muscle atrophy and restore function completely. To further improve the outcome, we explored the potential of a combinatorial therapy by modulating SMN production and muscle-enhancing approach as a novel therapeutic strategy for SMA. METHODS: The experiments were performed in a mouse model of severe SMA. A previously reported 25-mer morpholino antisense oligomer PMO25 was used to restore SMN expression. The adeno-associated virus-mediated expression of myostatin propeptide was used to block the myostatin pathway. Newborn SMA mice were treated with a single subcutaneous injection of 40 µg/g (therapeutic dose) or 10 µg/g (low-dose) PMO25 on its own or together with systemic delivery of a single dose of adeno-associated virus-mediated expression of myostatin propeptide. The multiple effects of myostatin inhibition on survival, skeletal muscle phenotype, motor function, neuromuscular junction maturation, and proprioceptive afferences were evaluated. RESULTS: We show that myostatin inhibition acts synergistically with SMN-restoring antisense therapy in SMA mice treated with the higher therapeutic dose PMO25 (40 µg/g), by increasing not only body weight (21% increase in male mice at Day 40), muscle mass (38% increase), and fibre size (35% increase in tibialis anterior muscle in 3 month female SMA mice), but also motor function and physical performance as measured in hanging wire test (two-fold increase in time score) and treadmill exercise test (two-fold increase in running distance). In SMA mice treated with low-dose PMO25 (10 µg/g), the early application of myostatin inhibition prolongs survival (40% increase), improves neuromuscular junction maturation (50% increase) and innervation (30% increase), and increases both the size of sensory neurons in dorsal root ganglia (60% increase) and the preservation of proprioceptive synapses in the spinal cord (30% increase). CONCLUSIONS: These data suggest that myostatin inhibition, in addition to the well-known effect on muscle mass, can also positively influence the sensory neural circuits that may enhance motor neurons function. While the availability of the antisense drug Spinraza for SMA and other SMN-enhancing therapies has provided unprecedented improvement in SMA patients, there are still unmet needs in these patients. Our study provides further rationale for considering myostatin inhibitors as a therapeutic intervention in SMA patients, in combination with SMN-restoring drugs.


Subject(s)
Muscular Atrophy, Spinal/drug therapy , Myostatin/antagonists & inhibitors , Oligonucleotides, Antisense/therapeutic use , Animals , Disease Models, Animal , Female , Humans , Mice , Mice, Transgenic , Muscular Atrophy, Spinal/mortality , Oligonucleotides, Antisense/pharmacology , Survival Analysis , Treatment Outcome
3.
Hum Mol Genet ; 28(10): 1694-1708, 2019 05 15.
Article in English | MEDLINE | ID: mdl-30649389

ABSTRACT

Oculopharyngeal muscular dystrophy (OPMD) is a rare late onset genetic disease leading to ptosis, dysphagia and proximal limb muscles at later stages. A short abnormal (GCN) triplet expansion in the polyA-binding protein nuclear 1 (PABPN1) gene leads to PABPN1-containing aggregates in the muscles of OPMD patients. Here we demonstrate that treating mice with guanabenz acetate (GA), an FDA-approved antihypertensive drug, reduces the size and number of nuclear aggregates, improves muscle force, protects myofibers from the pathology-derived turnover and decreases fibrosis. GA targets various cell processes, including the unfolded protein response (UPR), which acts to attenuate endoplasmic reticulum (ER) stress. We demonstrate that GA increases both the phosphorylation of the eukaryotic translation initiation factor 2α subunit and the splicing of Xbp1, key components of the UPR. Altogether these data show that modulation of protein folding regulation is beneficial for OPMD and promote the further development of GA or its derivatives for treatment of OPMD in humans. Furthermore, they support the recent evidences that treating ER stress could be therapeutically relevant in other more common proteinopathies.


Subject(s)
Guanabenz/pharmacology , Muscular Dystrophy, Oculopharyngeal/drug therapy , Poly(A)-Binding Protein I/genetics , X-Box Binding Protein 1/genetics , Alternative Splicing/drug effects , Alternative Splicing/genetics , Animals , Disease Models, Animal , Endoplasmic Reticulum Stress/drug effects , Fibrosis/drug therapy , Fibrosis/genetics , Fibrosis/pathology , Humans , Mice , Muscular Dystrophy, Oculopharyngeal/genetics , Muscular Dystrophy, Oculopharyngeal/pathology , Phosphorylation/drug effects , Protein Aggregates/drug effects , Protein Aggregates/genetics , Protein Folding , Unfolded Protein Response/drug effects
4.
Nucleic Acids Res ; 44(22): 10929-10945, 2016 12 15.
Article in English | MEDLINE | ID: mdl-27507886

ABSTRACT

A short abnormal polyalanine expansion in the polyadenylate-binding protein nuclear-1 (PABPN1) protein causes oculopharyngeal muscular dystrophy (OPMD). Mutated PABPN1 proteins accumulate as insoluble intranuclear aggregates in muscles of OPMD patients. While the roles of PABPN1 in nuclear polyadenylation and regulation of alternative poly(A) site choice have been established, the molecular mechanisms which trigger pathological defects in OPMD and the role of aggregates remain to be determined. Using exon array, for the first time we have identified several splicing defects in OPMD. In particular, we have demonstrated a defect in the splicing regulation of the muscle-specific Troponin T3 (TNNT3) mutually exclusive exons 16 and 17 in OPMD samples compared to controls. This splicing defect is directly linked to the SC35 (SRSF2) splicing factor and to the presence of nuclear aggregates. As reported here, PABPN1 aggregates are able to trap TNNT3 pre-mRNA, driving it outside nuclear speckles, leading to an altered SC35-mediated splicing. This results in a decreased calcium sensitivity of muscle fibers, which could in turn plays a role in muscle pathology. We thus report a novel mechanism of alternative splicing deregulation that may play a role in various other diseases with nuclear inclusions or foci containing an RNA binding protein.


Subject(s)
Muscular Dystrophy, Oculopharyngeal/metabolism , Poly(A)-Binding Protein I/metabolism , RNA Precursors/metabolism , Troponin T/genetics , Adult , Aged , Aged, 80 and over , Alternative Splicing , Animals , Case-Control Studies , Female , HEK293 Cells , Humans , Male , Mice , Mice, Transgenic , Middle Aged , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Muscular Dystrophy, Oculopharyngeal/genetics , Muscular Dystrophy, Oculopharyngeal/pathology , Poly(A)-Binding Protein I/genetics , Protein Aggregates , RNA Precursors/genetics , RNA Transport , Serine-Arginine Splicing Factors/metabolism , Troponin T/metabolism
5.
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
6.
Nucleic Acids Res ; 44(2): 744-60, 2016 Jan 29.
Article in English | MEDLINE | ID: mdl-26682797

ABSTRACT

Duchenne muscular dystrophy (DMD) is a genetic neuromuscular disorder caused by the absence of dystrophin. We developed a novel gene therapy approach based on the use of the piggyBac (PB) transposon system to deliver the coding DNA sequence (CDS) of either full-length human dystrophin (DYS: 11.1 kb) or truncated microdystrophins (MD1: 3.6 kb; MD2: 4 kb). PB transposons encoding microdystrophins were transfected in C2C12 myoblasts, yielding 65±2% MD1 and 66±2% MD2 expression in differentiated multinucleated myotubes. A hyperactive PB (hyPB) transposase was then deployed to enable transposition of the large-size PB transposon (17 kb) encoding the full-length DYS and green fluorescence protein (GFP). Stable GFP expression attaining 78±3% could be achieved in the C2C12 myoblasts that had undergone transposition. Western blot analysis demonstrated expression of the full-length human DYS protein in myotubes. Subsequently, dystrophic mesoangioblasts from a Golden Retriever muscular dystrophy dog were transfected with the large-size PB transposon resulting in 50±5% GFP-expressing cells after stable transposition. This was consistent with correction of the differentiated dystrophic mesoangioblasts following expression of full-length human DYS. These results pave the way toward a novel non-viral gene therapy approach for DMD using PB transposons underscoring their potential to deliver large therapeutic genes.


Subject(s)
DNA Transposable Elements/genetics , Dystrophin/genetics , Genetic Therapy/methods , Muscular Dystrophy, Duchenne/pathology , Animals , Cell Differentiation , Cells, Cultured , Dogs , Dystrophin/metabolism , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Humans , Male , Muscular Dystrophy, Duchenne/metabolism , Muscular Dystrophy, Duchenne/therapy , Myoblasts, Skeletal/cytology , Myoblasts, Skeletal/metabolism , Stem Cells/cytology , Stem Cells/metabolism , Transfection
7.
Curr Gene Ther ; 15(4): 395-415, 2015.
Article in English | MEDLINE | ID: mdl-26159373

ABSTRACT

Duchenne muscular dystrophy (DMD), an X-linked inherited musclewasting disease primarily affecting young boys with prevalence of between1:3,500- 1:5,000, is a rare genetic disease caused by defects in the gene for dystrophin. Dystrophin protein is critical to the stability of myofibers in skeletal and cardiac muscle. There is currently no cure available to ameliorate DMD and/or its patho-physiology. A number of therapeutic strategies including molecular-based therapeutics that replace or correct the missing or nonfunctional dystrophin protein have been devised to correct the patho-physiological consequences induced by dystrophin absence. We will review the current in vivo experimentation status (including preclinical models and clinical trials) for two of these approaches, namely: 1) Adeno-associated virus (AAV) mediated (micro) dystrophin gene augmentation/ supplementation and 2) Antisense oligonucleotide (AON)-mediated exon skipping strategies.


Subject(s)
Dependovirus/genetics , Dystrophin/genetics , Genetic Therapy/methods , Muscular Dystrophy, Duchenne/etiology , Muscular Dystrophy, Duchenne/therapy , Oligonucleotides, Antisense/pharmacology , Animals , Clinical Trials as Topic , Dependovirus/immunology , Disease Models, Animal , Dystrophin/deficiency , Exons , Gene Transfer Techniques , Genetic Vectors/administration & dosage , Genetic Vectors/immunology , Humans , Muscular Dystrophy, Duchenne/genetics , Oligonucleotides, Antisense/genetics , Oxadiazoles/pharmacology , RNA Editing
8.
Mol Ther ; 23(8): 1341-1348, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25959011

ABSTRACT

The fatal X-linked Duchenne muscular dystrophy (DMD), characterized by progressive muscle wasting and muscle weakness, is caused by mutations within the DMD gene. The use of antisense oligonucleotides (AOs) modulating pre-mRNA splicing to restore the disrupted dystrophin reading frame, subsequently generating a shortened but functional protein has emerged as a potential strategy in DMD treatment. AO therapy has recently been applied to induce out-of-frame exon skipping of myostatin pre-mRNA, knocking-down expression of myostatin protein, and such an approach is suggested to enhance muscle hypertrophy/hyperplasia and to reduce muscle necrosis. Within this study, we investigated dual exon skipping of dystrophin and myostatin pre-mRNAs using phosphorodiamidate morpholino oligomers conjugated with an arginine-rich peptide (B-PMOs). Intraperitoneal administration of B-PMOs was performed in neonatal mdx males on the day of birth, and at weeks 3 and 6. At week 9, we observed in treated mice (as compared to age-matched, saline-injected controls) normalization of muscle mass, a recovery in dystrophin expression, and a decrease in muscle necrosis, particularly in the diaphragm. Our data provide a proof of concept for antisense therapy combining dystrophin restoration and myostatin inhibition for the treatment of DMD.


Subject(s)
Dystrophin/genetics , Exons , Myostatin/genetics , Oligonucleotides, Antisense/chemistry , Open Reading Frames , Alternative Splicing , Animals , Animals, Newborn , Arginine/chemistry , Diaphragm/metabolism , Disease Models, Animal , Dystrophin/metabolism , Genetic Therapy , Male , Mice , Mice, Inbred C57BL , Mice, Inbred mdx , Morpholinos/genetics , Muscle, Skeletal/metabolism , Muscular Dystrophy, Duchenne/genetics , Myostatin/metabolism , Necrosis , Peptides/chemistry , Reading Frames
9.
Expert Opin Biol Ther ; 14(2): 209-30, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24308293

ABSTRACT

INTRODUCTION: Duchenne muscular dystrophy (DMD) is a lethal X-linked inherited disorder characterised by progressive muscle weakness, wasting and degeneration. Although the gene affected in DMD was identified over 25 years ago, there is still no effective treatment. AREAS COVERED: Here we review some of the genetic-based strategies aimed at amelioration of the DMD phenotype. A number of Phase II/III clinical trials of antisense oligonucleotide-induced exon skipping for restoration of the open reading frame (ORF) of the DMD gene have recently been completed. The potential strategies for overcoming the hurdles that appear to prevent exon skipping becoming an effective treatment for DMD currently are discussed. EXPERT OPINION: The applicability of exon skipping as a therapy to DMD is restricted and the development of alternative strategies that are more encompassing is needed. The rapid pre-clinical advances that are being made in the field of adeno-associated virus (AAV)-based delivery of micro-dystrophin would address this. The obstacles to be faced with gene replacement strategies would include the need for high viral titres, efficient muscle targeting and avoidance of immune response to vector and transgene. The new emerging field of gene editing could potentially provide permanent correction of the DMD gene and the feasibility of such an approach to DMD is discussed.


Subject(s)
Dystrophin/genetics , Genetic Therapy/methods , Muscular Dystrophy, Duchenne/genetics , Muscular Dystrophy, Duchenne/therapy , Animals , Clinical Trials, Phase II as Topic , Clinical Trials, Phase III as Topic , Codon, Terminator , Dependovirus , Exons , Gene Transfer Techniques , Genetic Vectors , Humans , Mice , Mutation , Oligonucleotides, Antisense/genetics , Open Reading Frames , Phenotype , RNA Splicing , Transgenes
10.
J Gen Virol ; 87(Pt 12): 3545-3549, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17098969

ABSTRACT

The emergence of variant fowlpox viruses (FWPVs) and increasing field use of recombinants against avian influenza H5N1 emphasize the need to monitor vaccines and to distinguish them from field strains. Five commercial vaccines, two laboratory viruses and two European field isolates were characterized by PCR and sequencing at 18 loci differing between attenuated FP9 and its pathogenic progenitor. PCR failed to discriminate between the viruses and sequence determination revealed no significant differences at any locus, except for a polymorphic locus encompassed by deletion 24 (9.3 kbp) in FP9. Surprisingly, 'novel' previously unreported sequence (spanning 1.2 kbp) was found in both European field isolates and three of the vaccines. It was absent from the other two vaccines, removed by a 1.2 kbp deletion identical to that surprisingly also observed in the completely sequenced genome of FPV USDA. This locus (H9) adds a potentially useful tool for discriminating between FWPV field isolates and vaccines.


Subject(s)
Fowlpox virus/genetics , Open Reading Frames/genetics , Viral Proteins/genetics , Viral Vaccines/genetics , Amino Acid Sequence , DNA, Viral/chemistry , DNA, Viral/genetics , Fowlpox virus/immunology , Fowlpox virus/isolation & purification , Molecular Sequence Data , Polymerase Chain Reaction , Sequence Analysis, DNA , Vaccines, Synthetic/genetics
11.
J Gen Virol ; 87(Pt 8): 2191-2201, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16847115

ABSTRACT

Avipoxvirus infections have been observed in an extensive range of wild, captive and domesticated avian hosts, yet little is known about the genome diversity and host-range specificity of the causative agent(s). Genome-sequence data are largely restricted to Fowlpox virus (FWPV) and Canarypox virus (CNPV), which have been sequenced completely, showing considerable divergence between them. It is therefore proving difficult, by empirical approaches, to identify pan-genus, avipoxvirus-specific oligonucleotide probes for PCR and sequencing to support phylogenetic studies. A previous preliminary study used the fpv167 locus, which encodes orthologues of vaccinia virus core protein P4b (A3). PCR per se did not discriminate between viruses, but restriction-enzyme or sequence analysis indicated that the avipoxviruses clustered either with FWPV or with CNPV. Here, further study of the P4b locus demonstrated a third cluster, from psittacine birds. A newly identified locus, flanking fpv140 (orthologue of vaccinia virus H3L), confirms the taxonomic structure. This locus is particularly useful in that viruses from the fowlpox-like and canarypox-like clusters can be discriminated by PCR on the basis of fragment size, whilst sequence comparison allows discrimination for the first time between Pigeonpox virus and Turkeypox virus. Except within the psittacines, virus and avian host taxonomies do not show tight correlation, with viruses from the same species located in very different clades. Nor are all the existing recognized avipoxvirus species, defined primarily by avian host species (such as CNPV and Sparrowpox virus), resolved within the present structure.


Subject(s)
Avipoxvirus/classification , Avipoxvirus/genetics , Phylogeny , Polymerase Chain Reaction/methods , Polymorphism, Genetic , Amino Acid Sequence , Animals , Canarypox virus/genetics , DNA, Viral/genetics , Fowlpox virus/genetics , Gene Order , Genes, Viral , Molecular Sequence Data , Sequence Analysis, DNA
SELECTION OF CITATIONS
SEARCH DETAIL
...