Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Tetrahedron ; 76(2): 130819, 2020 Jan 10.
Article in English | MEDLINE | ID: mdl-32713969

ABSTRACT

Following on from ezutromid, the first-in-class benzoxazole utrophin modulator that progressed to Phase 2 clinical trials for the treatment of Duchenne muscular dystrophy, a new chemotype was designed to optimise its physicochemical and ADME profile. Herein we report the synthesis of SMT022357, a second generation utrophin modulator preclinical candidate, and an asymmetric synthesis of its constituent enantiomers. The pharmacological properties of both enantiomers were evaluated in vitro and in vivo. No significant difference in the activity or efficacy was observed between the two enantiomers; activity was found to be comparable to the racemic mixture.

2.
Biochem Soc Trans ; 48(3): 813-821, 2020 06 30.
Article in English | MEDLINE | ID: mdl-32597486

ABSTRACT

Genetic approaches for the diagnosis and treatment of inherited muscle diseases have advanced rapidly in recent years. Many of the advances have occurred in the treatment of Duchenne muscular dystrophy (DMD), a muscle wasting disease where affected boys are typically wheelchair bound by age 12 years and generally die in their twenties from respiratory failure or cardiomyopathy. Dystrophin is a 421 kD protein which links F-actin to the extracellular matrix via the dystrophin-associated protein complex (DAPC) at the muscle membrane. In the absence of dystrophin, the DAPC is lost, making the muscle membrane more susceptible to contraction-induced injury. The identification of the gene causing DMD in 1986 resulted in improved diagnosis of the disease and the identification of hotspots for mutation. There is currently no effective treatment. However, there are several promising genetic therapeutic approaches at the preclinical stage or in clinical trials including read-through of stop codons, exon skipping, delivery of dystrophin minigenes and the modulation of expression of the dystrophin related protein, utrophin. In spite of significant progress, the problem of targeting all muscles, including diaphragm and heart at sufficiently high levels, remains a challenge. Any therapy also needs to consider the immune response and some treatments are mutation specific and therefore limited to a subgroup of patients. This short review provides a summary of the current status of DMD therapy with a particular focus on those genetic strategies that have been taken to the clinic.


Subject(s)
Muscular Dystrophy, Duchenne/diagnosis , Muscular Dystrophy, Duchenne/therapy , CRISPR-Cas Systems , Codon, Terminator , Dystrophin/genetics , Exons , Genetic Therapy/methods , Humans , Male , Muscular Dystrophy, Duchenne/genetics
3.
J Med Chem ; 63(14): 7880-7891, 2020 07 23.
Article in English | MEDLINE | ID: mdl-32551645

ABSTRACT

Utrophin modulation is a promising therapeutic strategy for Duchenne muscular dystrophy (DMD), which should be applicable to all patient populations. Following on from ezutromid, the first-generation utrophin modulator, we describe the development of a second generation of utrophin modulators, based on the bioisosteric replacement of the sulfone group with a phosphinate ester and substitution of the metabolically labile naphthalene with a haloaryl substituent. The improved physicochemical and absorption, distribution, metabolism, and excretion (ADME) properties, further reflected in the enhanced pharmacokinetic profile of the most advanced compounds, 30 and 27, led to significantly better in vivo exposure compared to ezutromid and alleviation of the dystrophic phenotype in mdx mice. While 30 was found to have dose-limiting hepatotoxicity, 27 and its enantiomers exhibited limited off-target effects, resulting in a safe profile and highlighting their potential utility as next-generation utrophin modulators suitable for progression toward a future DMD therapy.


Subject(s)
Benzoxazoles/therapeutic use , Muscular Dystrophy, Duchenne/drug therapy , Utrophin/metabolism , Animals , Benzoxazoles/chemical synthesis , Benzoxazoles/pharmacokinetics , Benzoxazoles/toxicity , Escherichia coli/drug effects , Mice, Inbred mdx , Molecular Structure , Muscular Dystrophy, Duchenne/metabolism , Mutagenicity Tests , Rats , Salmonella typhimurium/drug effects , Stereoisomerism , Structure-Activity Relationship , Up-Regulation/drug effects
4.
Hum Mol Genet ; 28(13): 2189-2200, 2019 07 01.
Article in English | MEDLINE | ID: mdl-30990876

ABSTRACT

Duchenne muscular dystrophy (DMD) is a lethal neuromuscular disorder caused by loss of dystrophin. Several therapeutic modalities are currently in clinical trials but none will achieve maximum functional rescue and full disease correction. Therefore, we explored the potential of combining the benefits of dystrophin with increases of utrophin, an autosomal paralogue of dystrophin. Utrophin and dystrophin can be co-expressed and co-localized at the same muscle membrane. Wild-type (wt) levels of dystrophin are not significantly affected by a moderate increase of utrophin whereas higher levels of utrophin reduce wt dystrophin, suggesting a finite number of actin binding sites at the sarcolemma. Thus, utrophin upregulation strategies may be applied to the more mildly affected Becker patients with lower dystrophin levels. Whereas increased dystrophin in wt animals does not offer functional improvement, overexpression of utrophin in wt mice results in a significant supra-functional benefit over wt. These findings highlight an additive benefit of the combined therapy and potential new unique roles of utrophin. Finally, we show a 30% restoration of wt dystrophin levels, using exon-skipping, together with increased utrophin levels restores dystrophic muscle function to wt levels offering greater therapeutic benefit than either single approach alone. Thus, this combination therapy results in additive functional benefit and paves the way for potential future combinations of dystrophin- and utrophin-based strategies.


Subject(s)
Dystrophin/genetics , Muscular Dystrophy, Duchenne/therapy , Utrophin/genetics , Animals , Dystrophin/metabolism , Exons , Mice , Mice, Inbred C57BL , Mice, Inbred mdx , Morpholinos/chemical synthesis , Morpholinos/therapeutic use , Muscle, Skeletal/metabolism , Muscular Dystrophy, Duchenne/genetics , Myofibrils/metabolism , Sarcolemma/metabolism , Up-Regulation , Utrophin/metabolism
5.
Hum Mol Genet ; 28(2): 307-319, 2019 01 15.
Article in English | MEDLINE | ID: mdl-30304405

ABSTRACT

Duchenne muscular dystrophy (DMD) is a lethal, X-linked muscle-wasting disease caused by lack of the cytoskeletal protein dystrophin. Constitutive utrophin expression, a structural and functional paralogue of dystrophin, can successfully prevent the dystrophic pathology in the dystrophin-deficient mdx mouse model. In dystrophic muscles, utrophin is increased as part of the repair process and localized at the sarcolemma of regenerating myofibers. The presence of developmental myosin such as embryonic myosin (MyHC-emb) and neonatal represents a useful marker of muscle regeneration and a meaningful indicator of muscle damage, which correlates with the clinical severity of milder Becker muscular dystrophy and DMD patients. In the present study, we demonstrate that MyHC-emb is a robust marker of regeneration at different ages and in different skeletal muscles. We also evaluate the correlation between utrophin, dystrophin and MyHC-emb in wild-type (wt) and regenerating dystrophic muscles. Restoration of dystrophin significantly reduced MyHC-emb levels. Similarly, overexpression of utrophin in the transgenic mdx-Fiona mice reduced the number of MyHC-emb positive fibers to wt level, prevented the regenerative process and rescued the muscle function. In contrast, the absence of utrophin in the dystrophin-deficient double-knockout mice resulted in a higher MyHC-emb content and in a more severe dystrophic pathophysiology than in mdx mice. These data illustrate the importance of monitoring utrophin and MyHC-emb levels in the preclinical evaluation of therapies and provide translational support for the use of developmental myosin as a disease biomarker in DMD clinical trials.


Subject(s)
Dystrophin/metabolism , Muscle, Skeletal/metabolism , Muscular Dystrophy, Duchenne/metabolism , Myosins/metabolism , Regeneration , Utrophin/metabolism , Animals , Biomarkers/metabolism , Clinical Trials as Topic , Disease Models, Animal , Embryo, Mammalian/metabolism , Male , Mice, Inbred C57BL , Mice, Inbred mdx , Mice, Transgenic , Muscle, Skeletal/embryology , Muscle, Skeletal/physiology , Muscular Dystrophy, Animal , Muscular Dystrophy, Duchenne/drug therapy , Muscular Dystrophy, Duchenne/embryology , Muscular Dystrophy, Duchenne/pathology , Sarcolemma/metabolism
6.
Mol Ther Methods Clin Dev ; 11: 92-105, 2018 Dec 14.
Article in English | MEDLINE | ID: mdl-30417024

ABSTRACT

Duchenne muscular dystrophy (DMD) is an X-linked muscle-wasting disease caused by mutations in the dystrophin gene. DMD boys are wheelchair-bound around 12 years and generally survive into their twenties. There is currently no effective treatment except palliative care, although personalized treatments such as exon skipping, stop codon read-through, and viral-based gene therapies are making progress. Patients present with skeletal muscle pathology, but most also show cardiomyopathy by the age of 10. A systemic therapeutic approach is needed that treats the heart and skeletal muscle defects in all patients. The dystrophin-related protein utrophin has been shown to compensate for the lack of dystrophin in the mildly affected BL10/mdx mouse. The purpose of this investigation was to demonstrate that AAV9-mediated micro-utrophin transgene delivery can not only functionally replace dystrophin in the heart, but also attenuate the skeletal muscle phenotype in severely affected D2/mdx mice. The data presented here show that utrophin can indeed alleviate the pathology in skeletal and cardiac muscle in D2/mdx mice. These results endorse the view that utrophin modulation has the potential to increase the quality life of all DMD patients whatever their mutation.

7.
Sci Rep ; 7: 43697, 2017 03 02.
Article in English | MEDLINE | ID: mdl-28252048

ABSTRACT

Despite promising therapeutic avenues, there is currently no effective treatment for Duchenne muscular dystrophy (DMD), a lethal monogenic disorder caused by the loss of the large cytoskeletal protein, dystrophin. A highly promising approach to therapy, applicable to all DMD patients irrespective to their genetic defect, is to modulate utrophin, a functional paralogue of dystrophin, able to compensate for the primary defects of DMD restoring sarcolemmal stability. One of the major difficulties in assessing the effectiveness of therapeutic strategies is to define appropriate outcome measures. In the present study, we utilised an aptamer based proteomics approach to profile 1,310 proteins in plasma of wild-type, mdx and Fiona (mdx overexpressing utrophin) mice. Comparison of the C57 and mdx sera revealed 83 proteins with statistically significant >2 fold changes in dystrophic serum abundance. A large majority of previously described biomarkers (ANP32B, THBS4, CAMK2A/B/D, CYCS, CAPNI) were normalised towards wild-type levels in Fiona animals. This work also identified potential mdx markers specific to increased utrophin (DUS3, TPI1) and highlights novel mdx biomarkers (GITR, MYBPC1, HSP60, SIRT2, SMAD3, CNTN1). We define a panel of putative protein mdx biomarkers to evaluate utrophin based strategies which may help to accelerate their translation to the clinic.


Subject(s)
Biomarkers , Blood Proteins , Utrophin/blood , Animals , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Humans , Mice , Mice, Transgenic , Muscular Dystrophy, Animal , Muscular Dystrophy, Duchenne/blood , Muscular Dystrophy, Duchenne/genetics , Muscular Dystrophy, Duchenne/therapy , Proteome , Proteomics/methods , Translational Research, Biomedical , Utrophin/therapeutic use
8.
Elife ; 42015 Nov 02.
Article in English | MEDLINE | ID: mdl-26523393

ABSTRACT

The mammalian suprachiasmatic nucleus (SCN) drives daily rhythmic behavior and physiology, yet a detailed understanding of its coordinated transcriptional programmes is lacking. To reveal the finer details of circadian variation in the mammalian SCN transcriptome we combined laser-capture microdissection (LCM) and RNA-seq over a 24 hr light / dark cycle. We show that 7-times more genes exhibited a classic sinusoidal expression signature than previously observed in the SCN. Another group of 766 genes unexpectedly peaked twice, near both the start and end of the dark phase; this twin-peaking group is significantly enriched for synaptic transmission genes that are crucial for light-induced phase shifting of the circadian clock. 341 intergenic non-coding RNAs, together with novel exons of annotated protein-coding genes, including Cry1, also show specific circadian expression variation. Overall, our data provide an important chronobiological resource (www.wgpembroke.com/shiny/SCNseq/) and allow us to propose that transcriptional timing in the SCN is gating clock resetting mechanisms.


Subject(s)
Biological Clocks , Gene Expression Regulation , Suprachiasmatic Nucleus/physiology , Transcription, Genetic , Animals , Gene Expression Profiling , Laser Capture Microdissection , Mice, Inbred C3H , Sequence Analysis, RNA
9.
Hum Mol Genet ; 24(15): 4212-24, 2015 Aug 01.
Article in English | MEDLINE | ID: mdl-25935002

ABSTRACT

Duchenne muscular dystrophy (DMD) is a lethal, X-linked muscle-wasting disease caused by lack of the cytoskeletal protein dystrophin. There is currently no cure for DMD although various promising approaches are progressing through human clinical trials. By pharmacologically modulating the expression of the dystrophin-related protein utrophin, we have previously demonstrated in dystrophin-deficient mdx studies, daily SMT C1100 treatment significantly reduced muscle degeneration leading to improved muscle function. This manuscript describes the significant disease modifying benefits associated with daily dosing of SMT022357, a second-generation compound in this drug series with improved physicochemical properties and a more robust metabolism profile. These studies in the mdx mouse demonstrate that oral administration of SMT022357 leads to increased utrophin expression in skeletal, respiratory and cardiac muscles. Significantly, utrophin expression is localized along the length of the muscle fibre, not just at the synapse, and is fibre-type independent, suggesting that drug treatment is modulating utrophin transcription in extra-synaptic myonuclei. This results in improved sarcolemmal stability and prevents dystrophic pathology through a significant reduction of regeneration, necrosis and fibrosis. All these improvements combine to protect the mdx muscle from contraction induced damage and enhance physiological function. This detailed evaluation of the SMT C1100 drug series strongly endorses the therapeutic potential of utrophin modulation as a disease modifying therapeutic strategy for all DMD patients irrespective of their dystrophin mutation.


Subject(s)
Dystrophin/biosynthesis , Muscle Fibers, Skeletal/drug effects , Muscular Dystrophy, Duchenne/drug therapy , Utrophin/biosynthesis , Animals , Dystrophin/genetics , Gene Expression Regulation/drug effects , Humans , Mice , Mice, Inbred mdx , Muscle Contraction/drug effects , Muscle Contraction/genetics , Muscle Fibers, Skeletal/pathology , Muscles/drug effects , Muscles/pathology , Muscular Dystrophy, Animal/genetics , Muscular Dystrophy, Duchenne/genetics , Muscular Dystrophy, Duchenne/pathology , Sarcolemma/drug effects , Sarcolemma/genetics , Utrophin/genetics
10.
Nat Med ; 21(3): 270-5, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25642938

ABSTRACT

Antisense oligonucleotides (AONs) hold promise for therapeutic correction of many genetic diseases via exon skipping, and the first AON-based drugs have entered clinical trials for neuromuscular disorders. However, despite advances in AON chemistry and design, systemic use of AONs is limited because of poor tissue uptake, and recent clinical reports confirm that sufficient therapeutic efficacy has not yet been achieved. Here we present a new class of AONs made of tricyclo-DNA (tcDNA), which displays unique pharmacological properties and unprecedented uptake by many tissues after systemic administration. We demonstrate these properties in two mouse models of Duchenne muscular dystrophy (DMD), a neurogenetic disease typically caused by frame-shifting deletions or nonsense mutations in the gene encoding dystrophin and characterized by progressive muscle weakness, cardiomyopathy, respiratory failure and neurocognitive impairment. Although current naked AONs do not enter the heart or cross the blood-brain barrier to any substantial extent, we show that systemic delivery of tcDNA-AONs promotes a high degree of rescue of dystrophin expression in skeletal muscles, the heart and, to a lesser extent, the brain. Our results demonstrate for the first time a physiological improvement of cardio-respiratory functions and a correction of behavioral features in DMD model mice. This makes tcDNA-AON chemistry particularly attractive as a potential future therapy for patients with DMD and other neuromuscular disorders or with other diseases that are eligible for exon-skipping approaches requiring whole-body treatment.


Subject(s)
Dystrophin/drug effects , Heart/drug effects , Muscle, Skeletal/drug effects , Muscular Dystrophy, Duchenne , Nanoparticles , Oligodeoxyribonucleotides, Antisense/pharmacology , RNA, Messenger/analysis , Animals , Blood-Brain Barrier/metabolism , Codon, Nonsense , Disease Models, Animal , Dystrophin/genetics , Exons , Genetic Therapy , Mice , Microscopy, Electron, Transmission , Muscle, Skeletal/metabolism , Myocardium/metabolism , Oligodeoxyribonucleotides, Antisense/metabolism , Transcriptome/drug effects
11.
Hum Mol Genet ; 21(11): 2559-71, 2012 Jun 01.
Article in English | MEDLINE | ID: mdl-22388933

ABSTRACT

Duchenne muscular dystrophy (DMD) is a severe neuromuscular disorder caused by mutations in the dystrophin gene that result in the absence of functional protein. Antisense-mediated exon skipping is one of the most promising approaches for the treatment of DMD and recent clinical trials have demonstrated encouraging results. However, antisense oligonucleotide-mediated exon skipping for DMD still faces major hurdles such as extremely low efficacy in the cardiac muscle, poor cellular uptake and relatively rapid clearance from circulation, which means that repeated administrations are required to achieve some therapeutic efficacy. To overcome these limitations, we previously proposed the use of small nuclear RNAs (snRNAs), especially U7snRNA to shuttle the antisense sequences after vectorization into adeno-associated virus (AAV) vectors. In this study, we report for the first time the efficiency of the AAV-mediated exon skipping approach in the utrophin/dystrophin double-knockout (dKO) mouse which is a very severe and progressive mouse model of DMD. Following a single intravenous injection of scAAV9-U7ex23 in dKO mice, near-normal levels of dystrophin expression were restored in all muscles examined, including the heart. This resulted in a considerable improvement of their muscle function and dystrophic pathology as well as a remarkable extension of the dKO mice lifespan. These findings suggest great potential for AAV-U7 in systemic treatment of the DMD phenotype.


Subject(s)
Dependovirus/genetics , Dystrophin/genetics , Exons , Muscular Dystrophy, Duchenne/genetics , Muscular Dystrophy, Duchenne/therapy , RNA, Small Nuclear/genetics , Utrophin/genetics , Animals , Dependovirus/metabolism , Disease Models, Animal , Dystrophin/metabolism , Genetic Therapy , Mice , Muscular Dystrophy, Duchenne/pathology , Phenotype , RNA, Small Nuclear/therapeutic use , Utrophin/metabolism
12.
Mol Ther ; 20(6): 1212-21, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22354379

ABSTRACT

Duchenne muscular dystrophy (DMD) is a fatal muscle wasting disorder caused by mutations in the dystrophin gene. Antisense-mediated exon skipping is one of the most promising approaches for the treatment of DMD but still faces personalized medicine challenges as different mutations found in DMD patients require skipping of different exons. However, 70% of DMD patients harbor dystrophin gene deletions in a mutation-rich area or "hot-spot" in the central genomic region. In this study, we have developed 11 different U7 small-nuclear RNA, to shuttle antisense sequences designed to mask key elements involved in the splicing of exons 45 to 55. We demonstrate that these constructs induce efficient exon skipping both in vitro in DMD patients' myoblasts and in vivo in human DMD (hDMD) mice and that they can be combined into a single vector to achieve a multi skipping of at least 3 exons. These very encouraging results provide proof of principle that efficient multiexon-skipping can be achieved using adeno-associated viral (AAV) vectors encoding multiple U7 small-nuclear RNAs (U7snRNAs), offering therefore very promising tools for clinical treatment of DMD.


Subject(s)
Alternative Splicing , Dystrophin/genetics , Exons , Genetic Vectors , Muscular Dystrophy, Duchenne/genetics , RNA, Small Nuclear , Animals , Cell Line , DNA, Antisense , Dependovirus/genetics , Disease Models, Animal , Gene Order , Humans , Lentivirus/genetics , Mice , Mice, Transgenic , Muscular Dystrophy, Duchenne/therapy , Myoblasts/metabolism , Transduction, Genetic
13.
Mol Ther Nucleic Acids ; 1: e31, 2012 Jun 26.
Article in English | MEDLINE | ID: mdl-23344083

ABSTRACT

Many genetic diseases are induced by mutations disturbing the maturation of pre-mRNAs, often affecting splicing. Antisense oligoribonucleotides (AONs) have been used to modulate splicing thereby circumventing the deleterious effects of mutations. Stable delivery of antisense sequences is achieved by linking them to small nuclear RNA (snRNAs) delivered by viral vectors, as illustrated by studies where therapeutic exon skipping was obtained in animal models of Duchenne muscular dystrophy (DMD). Yet, clinical translation of these approaches is limited by the amounts of vector to be administered. In this respect, maximizing the amount of snRNA antisense shuttle delivered by the vector is essential. Here, we have used a muscle- and heart-specific enhancer (MHCK) to drive the expression of U7 snRNA shuttles carrying antisense sequences against the human or murine DMD pre-mRNAs. Although antisense delivery and subsequent exon skipping were improved both in tissue culture and in vivo, we observed the formation of additional U7 snRNA by-products following gene transfer. These included aberrantly 3' processed as well as unprocessed species that may arise because of the saturation of the cellular processing capacity. Future efforts to increase the amounts of functional U7 shuttles delivered into a cell will have to take this limitation into account.

14.
Hum Mol Genet ; 20(3): 413-21, 2011 Feb 01.
Article in English | MEDLINE | ID: mdl-21062902

ABSTRACT

Duchenne muscular dystrophy (DMD) is an X-linked recessive disease caused, in most cases, by the complete absence of the 427 kDa cytoskeletal protein, dystrophin. There is no effective treatment, and affected individuals die from respiratory failure and cardiomyopathy by age 30. Here, we investigated whether cardiomyopathy could be prevented in animal models of DMD by increasing diaphragm utrophin or dystrophin expression and thereby restoring diaphragm function. In a transgenic mdx mouse, where utrophin was over expressed in the skeletal muscle and the diaphragm, but not in the heart, we found cardiac function, specifically right and left ventricular ejection fraction as measured using in vivo magnetic resonance imaging, was restored to wild-type levels. In mdx mice treated with a peptide-conjugated phosphorodiamidate morpholino oligomer (PPMO) that resulted in high levels of dystrophin restoration in the skeletal muscle and the diaphragm only, cardiac function was also restored to wild-type levels. In dystrophin/utrophin-deficient double-knockout (dKO) mice, a more severely affected animal model of DMD, treatment with a PPMO again produced high levels of dystrophin only in the skeletal muscle and the diaphragm, and once more restored cardiac function to wild-type levels. In the dKO mouse, there was no difference in heart function between treatment of the diaphragm plus the heart and treatment of the diaphragm alone. Restoration of diaphragm and other respiratory muscle function, irrespective of the method used, was sufficient to prevent cardiomyopathy in dystrophic mice. This novel mechanism of treating respiratory muscles to prevent cardiomyopathy in dystrophic mice warrants further investigation for its implications on the need to directly treat the heart in DMD.


Subject(s)
Cardiomyopathies/prevention & control , Diaphragm/physiopathology , Dystrophin/metabolism , Morpholines/pharmacology , Muscular Dystrophy, Animal/drug therapy , Utrophin/metabolism , Animals , Cytoskeletal Proteins/metabolism , Diaphragm/drug effects , Diaphragm/metabolism , Dystrophin/genetics , Heart/physiopathology , Magnetic Resonance Imaging , Mice , Mice, Inbred mdx , Mice, Knockout , Mice, Transgenic , Morpholinos , Muscle, Skeletal/drug effects , Muscle, Skeletal/metabolism , Muscular Dystrophy, Animal/physiopathology , Stroke Volume , Utrophin/genetics
15.
Mol Ther ; 18(1): 198-205, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19844193

ABSTRACT

Duchenne muscular dystrophy (DMD) is a severe neuromuscular disorder caused by mutations in the dystrophin gene that result in the absence of functional protein. Antisense-mediated exon-skipping is one of the most promising approaches for the treatment of DMD because of its capacity to correct the reading frame and restore dystrophin expression, which has been demonstrated in vitro and in vivo. In particular, peptide-conjugated phosphorodiamidate morpholino oligomers (PPMOs) have recently been shown to induce widespread high levels of dystrophin expression in the mdx mouse model. Here, we report the efficiency of the PPMO-mediated exon-skipping approach in the utrophin/dystrophin double-knockout mouse (dKO) mouse, which is a much more severe and progressive mouse model of DMD. Repeated intraperitoneal (i.p.) injections of a PPMO targeted to exon 23 of dystrophin pre-mRNA in dKO mice induce a near-normal level of dystrophin expression in all muscles examined, except for the cardiac muscle, resulting in a considerable improvement of their muscle function and dystrophic pathology. These findings suggest great potential for PPMOs in systemic treatment of the DMD phenotype.


Subject(s)
Dystrophin/metabolism , Exons/genetics , Morpholines/therapeutic use , Muscular Dystrophy, Animal/drug therapy , Muscular Dystrophy, Animal/metabolism , Utrophin/metabolism , Animals , Immunohistochemistry , Injections, Intraperitoneal , Mice , Mice, Inbred mdx , Mice, Knockout , Morpholines/administration & dosage , Morpholines/chemistry , Morpholinos , Muscular Dystrophy, Animal/pathology , Peptides/chemistry , Reverse Transcriptase Polymerase Chain Reaction
16.
Mol Ther ; 17(7): 1234-40, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19455105

ABSTRACT

Duchenne muscular dystrophy (DMD) is a fatal muscle wasting disorder caused by mutations in the dystrophin gene. In most cases, the open-reading frame is disrupted which results in the absence of functional protein. Antisense-mediated exon skipping is one of the most promising approaches for the treatment of DMD and has recently been shown to correct the reading frame and restore dystrophin expression in vitro and in vivo. Specific exon skipping can be achieved using synthetic oligonucleotides or viral vectors encoding modified small nuclear RNAs (snRNAs), by masking important splicing sites. In this study, we demonstrate that enhanced exon skipping can be induced by a U7 snRNA carrying binding sites for the heterogeneous ribonucleoprotein A1 (hnRNPA1). In DMD patient cells, bifunctional U7 snRNAs harboring silencer motifs induce complete skipping of exon 51, and thus restore dystrophin expression to near wild-type levels. Furthermore, we show the efficacy of these constructs in vivo in transgenic mice carrying the entire human DMD locus after intramuscular injection of adeno-associated virus (AAV) vectors encoding the bifunctional U7 snRNA. These new constructs are very promising for the optimization of therapeutic exon skipping for DMD, but also offer powerful and versatile tools to modulate pre-mRNA splicing in a wide range of applications.


Subject(s)
RNA, Small Nuclear/genetics , Animals , Blotting, Western , Cell Line , Cells, Cultured , Dystrophin , Exons/genetics , Genetic Therapy/methods , Genetic Vectors/genetics , Heterogeneous-Nuclear Ribonucleoprotein Group A-B/genetics , Humans , Mice , Mice, Transgenic , Models, Biological , Muscular Dystrophy, Duchenne/therapy , Myoblasts/metabolism , NIH 3T3 Cells , Reverse Transcriptase Polymerase Chain Reaction
17.
Proc Natl Acad Sci U S A ; 104(7): 2431-6, 2007 Feb 13.
Article in English | MEDLINE | ID: mdl-17283335

ABSTRACT

The neuronal soluble N-ethylmaleimide-sensitive factor attachment protein receptor (SNARE) complex is essential for synaptic vesicle exocytosis, but its study has been limited by the neonatal lethality of murine SNARE knockouts. Here, we describe a viable mouse line carrying a mutation in the b-isoform of neuronal SNARE synaptosomal-associated protein of 25 kDa (SNAP-25). The causative I67T missense mutation results in increased binding affinities within the SNARE complex, impaired exocytotic vesicle recycling and granule exocytosis in pancreatic beta-cells, and a reduction in the amplitude of evoked cortical excitatory postsynaptic potentials. The mice also display ataxia and impaired sensorimotor gating, a phenotype which has been associated with psychiatric disorders in humans. These studies therefore provide insights into the role of the SNARE complex in both diabetes and psychiatric disease.


Subject(s)
Ataxia/genetics , Exocytosis/genetics , Gait Disorders, Neurologic/genetics , Mutation, Missense , Synaptic Vesicles/genetics , Synaptosomal-Associated Protein 25/genetics , Alcoholic Intoxication , Animals , Diabetes Mellitus/etiology , Genes, Dominant , Insulin-Secreting Cells , Mental Disorders/etiology , Mice , Mice, Mutant Strains , Models, Animal , SNARE Proteins/physiology , Synaptosomal-Associated Protein 25/physiology
SELECTION OF CITATIONS
SEARCH DETAIL