Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 52
Filter
1.
Expert Opin Biol Ther ; 17(1): 15-30, 2017 01.
Article in English | MEDLINE | ID: mdl-27805416

ABSTRACT

INTRODUCTION: Antisense nucleic acid analogues can interact with pre-mRNA motifs and influence exon or splice site selection and thereby alter gene expression. Design of antisense molecules to target specific motifs can result in either exon exclusion or exon inclusion during splicing. Novel drugs exploiting the antisense concept are targeting rare, life-limiting diseases; however, the potential exists to treat a wide range of conditions by antisense-mediated splice intervention. Areas covered: In this review, the authors discuss the clinical translation of novel molecular therapeutics to address the fatal neuromuscular disorders Duchenne muscular dystrophy and spinal muscular atrophy. The review also highlights difficulties posed by issues pertaining to restricted participant numbers, variable phenotype and disease progression, and the identification and validation of study endpoints. Expert opinion: Translation of novel therapeutics for Duchenne muscular dystrophy and spinal muscular atrophy has been greatly advanced by multidisciplinary research, academic-industry partnerships and in particular, the engagement and support of the patient community. Sponsors, supporters and regulators are cooperating to deliver new drugs and identify and define meaningful outcome measures. Non-conventional and adaptive trial design could be particularly suited to clinical evaluation of novel therapeutics and strategies to treat serious, rare diseases that may be problematic to study using more conventional clinical trial structures.


Subject(s)
Exons/genetics , Genetic Therapy/trends , Muscular Dystrophy, Duchenne/drug therapy , Oligonucleotides, Antisense/therapeutic use , RNA Splicing/genetics , Translational Research, Biomedical/methods , Animals , Biological Therapy/methods , Biological Therapy/trends , Dystrophin/genetics , Exons/drug effects , Gene Expression , Gene Expression Regulation , Genetic Therapy/methods , Humans , Muscular Atrophy, Spinal/drug therapy , Muscular Atrophy, Spinal/genetics , Muscular Dystrophy, Duchenne/genetics , Oligonucleotides, Antisense/genetics , Oligonucleotides, Antisense/pharmacology , RNA Splicing/drug effects , Translational Research, Biomedical/trends
2.
Oncogenesis ; 4: e174, 2015 Nov 09.
Article in English | MEDLINE | ID: mdl-26551281

ABSTRACT

NUT midline carcinoma (NMC) is a fatal cancer that arises in various tissues along the upper midline of the body. The defining molecular feature of NMC is a chromosomal translocation that joins (in the majority of cases) the nuclear testis gene NUT (NUTM1) to the bromodomain protein family member 4 (BRD4) and thereby creating a fusion oncogene that disrupts cellular differentiation and drives the disease. In this study, we report the case of an adolescent NMC patient presenting with severe facial pain, proptosis and visual impairment due to a mass arising from the ethmoid sinus that invaded the right orbit and frontal lobe. Treatment involved radical resection, including exenteration of the affected eye with the view to consolidate treatment with radiation therapy; however, the patient experienced rapid tumor progression and passed away 79 days post resection. Molecular analysis of the tumor tissue identified a novel in-frame BRD4-NUT transcript, with BRD4 exon 15 fused to the last 124 nucleotides of NUT exon 2 (BRD4-NUT ex15:ex2Δnt1-585). The partial deletion of NUT exon 2 was attributed to a mid-exonic genomic breakpoint and the subsequent activation of a cryptic splice site further downstream within the exon. Inhibition of the canonical 3' acceptor splice site of NUT intron 1 in cell lines expressing the most common NMC fusion transcripts (PER-403, BRD4-NUT ex11:ex2; PER-624, BRD4-NUT ex15:ex2) induced alternative splicing from the same cryptic splice site as identified in the patient. Detection of low levels of an in-frame BRD4-NUT ex11:ex2Δnt1-585 transcript in PER-403 confirmed endogenous splicing from this alternative exon 2 splice site. Although further studies are necessary to assess the clinical relevance of the increasing number of variant fusions described in NMC, the findings presented in this case identify alternative splicing as a mechanism that contributes to this pathogenic complexity.

3.
Trends Mol Med ; 21(7): 417-26, 2015 Jul.
Article in English | MEDLINE | ID: mdl-26051381

ABSTRACT

Targeted dystrophin exon removal is a promising therapy for Duchenne muscular dystrophy (DMD); however, dystrophin expression in some reports is not supported by the associated data. As in the account of 'The Emperor's New Clothes', the validity of such claims must be questioned, with critical re-evaluation of available data. Is it appropriate to report clinical benefit and induction of dystrophin as dose dependent when the baseline is unclear? The inability to induce meaningful levels of dystrophin does not mean that dystrophin expression as an end point is irrelevant, nor that induced exon skipping as a strategy is flawed, but demands that drug safety and efficacy, and study parameters be addressed, rather than questioning the strategy or the validity of dystrophin as a biomarker.


Subject(s)
Dystrophin/metabolism , Animals , Dystrophin/genetics , Exons/genetics , Humans , Muscular Dystrophy, Duchenne/genetics , Muscular Dystrophy, Duchenne/metabolism
4.
Neuroscience ; 220: 228-36, 2012 Sep 18.
Article in English | MEDLINE | ID: mdl-22732506

ABSTRACT

Spinal muscular atrophy (SMA), a fatal genetic motor disorder of infants, is caused by diminished full-length survival of motor neuron (SMN) protein levels. Normally involved in small nuclear ribonucleoprotein (snRNP) assembly and pre-mRNA splicing, recent studies suggest that SMN plays a critical role in regulating apoptosis. Interestingly, the anti-apoptotic Bcl-x isoform, Bcl-xL, is reduced in SMA. In a related finding, Sam68, an RNA-binding protein, was found to modulate splicing of SMN and Bcl-xL transcripts, promoting SMNΔ7 and pro-apoptotic Bcl-xS transcripts. Here we demonstrate that Bcl-xL expression increases SMN protein by ∼2-fold in SH-SY5Y cells. Conversely, SMN expression increases Bcl-xL protein levels by ∼6-fold in SH-SY5Y cells, and ∼2.5-fold in the brains of transgenic mice over-expressing SMN (PrP-SMN). Moreover, Sam68 protein levels were markedly reduced following SMN and Bcl-xL expression in SH-SY5Y cells, suggesting a feedback mechanism co-regulating levels of both proteins. We also found that exogenous SMN expression increased full-length SMN transcripts, possibly by promoting exon 7 inclusion. Finally, co-expression of SMN and Bcl-xL produced an additive anti-apoptotic effect following PI3-kinase inhibition in SH-SY5Y cells. Our findings implicate Bcl-xL as another potential target in SMA therapeutics, and indicate that therapeutic increases in SMN may arise from modest increases in total SMN.


Subject(s)
Gene Expression Regulation , Motor Neurons/metabolism , Muscular Atrophy, Spinal/metabolism , Survival of Motor Neuron 1 Protein/metabolism , bcl-X Protein/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Animals , Blotting, Western , Cell Line , Humans , Mice , Mice, Transgenic , RNA-Binding Proteins/metabolism , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Transduction, Genetic
5.
Hum Gene Ther ; 18(9): 798-810, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17767400

ABSTRACT

Duchenne muscular dystrophy (DMD) is caused by mutations in the dystrophin gene that result in the absence of functional protein. In the majority of cases these are out-of-frame deletions that disrupt the reading frame. Several attempts have been made to restore the dystrophin mRNA reading frame by modulation of pre-mRNA splicing with antisense oligonucleotides (AOs), demonstrating success in cultured cells, muscle explants, and animal models. We are preparing for a phase I/IIa clinical trial aimed at assessing the safety and effect of locally administered AOs designed to inhibit inclusion of exon 51 into the mature mRNA by the splicing machinery, a process known as exon skipping. Here, we describe a series of systematic experiments to validate the sequence and chemistry of the exon 51 AO reagent selected to go forward into the clinical trial planned in the United Kingdom. Eight specific AO sequences targeting exon 51 were tested in two different chemical forms and in three different preclinical models: cultured human muscle cells and explants (wild type and DMD), and local in vivo administration in transgenic mice harboring the entire human DMD locus. Data have been validated independently in the different model systems used, and the studies describe a rational collaborative path for the preclinical selection of AOs for evaluation in future clinical trials.


Subject(s)
Alternative Splicing , Dystrophin/genetics , Exons , Muscle, Skeletal , Oligonucleotides, Antisense/analysis , RNA Precursors/metabolism , Animals , Base Sequence , Blotting, Western , Cells, Cultured , Dystrophin/chemistry , Gene Targeting , Humans , Mice , Mice, Transgenic , Molecular Sequence Data , Muscle, Skeletal/cytology , Muscle, Skeletal/metabolism , Muscular Dystrophy, Duchenne/genetics , Oligonucleotides, Antisense/chemistry , Oligonucleotides, Antisense/genetics , Organ Culture Techniques , RNA, Messenger/metabolism , Reproducibility of Results , Reverse Transcriptase Polymerase Chain Reaction
6.
Biochem Soc Trans ; 35(Pt 4): 826-8, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17635157

ABSTRACT

The cellular uptake of PMOs (phosphorodiamidate morpholino oligomers) can be enhanced by their conjugation to arginine-rich CPPs (cell-penetrating peptides). Here, we discuss our recent findings regarding (R-Ahx-R)(4)AhxB (Ahx is 6-aminohexanoic acid and B is beta-alanine) CPP-PMO conjugates in DMD (Duchenne muscular dystrophy) and murine coronavirus research. An (R-Ahx-R)(4)AhxB-PMO conjugate was the most effective compound in inducing the correction of mutant dystrophin transcripts in myoblasts derived from a canine model of DMD. Similarly, normal levels of dystrophin expression were restored in the diaphragms of mdx mice, with treatment starting at the neonatal stage, and protein was still detecTable 22 weeks after the last dose of an (R-Ahx-R)(4)AhxB-PMO conjugate. Effects of length, linkage and carbohydrate modification of this CPP on the delivery of a PMO were investigated in a coronavirus mouse model. An (R-Ahx-R)(4)AhxB-PMO conjugate effectively inhibited viral replication, in comparison with other peptides conjugated to the same PMO. Shortening the CPP length, modifying it with a mannosylated serine moiety or replacing it with the R(9)F(2) CPP significantly decreased the efficacy of the resulting PPMO (CPP-PMO conjugate). We attribute the success of this CPP to its stability in serum and its capacity to transport PMO to RNA targets in a manner superior to that of poly-arginine CPPs.


Subject(s)
Antiviral Agents/pharmacology , Coronavirus/drug effects , Morpholines/pharmacology , Muscular Dystrophy, Duchenne/drug therapy , Peptides/therapeutic use , RNA Splicing/drug effects , Virus Replication/drug effects , Animals , Drug Delivery Systems , Dystrophin/biosynthesis , Dystrophin/genetics , Humans , Mice , Morpholines/administration & dosage , Muscular Dystrophy, Duchenne/genetics , Protein Sorting Signals/physiology , Protein Transport/physiology , RNA Precursors/metabolism
7.
Mol Ther ; 15(1): 157-66, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17164787

ABSTRACT

Antisense oligonucleotides (AOs) can be used to redirect dystrophin pre-messenger RNA (mRNA) processing, to remove selected exons from the mature dystrophin mRNA, to overcome nonsense mutations, and/or restore the reading frame. Redundancy within the dystrophin protein allows some domains to be removed without seriously compromising function. One of the challenges for splicing blockade is to design AOs that efficiently remove targeted exons across the dystrophin pre-mRNA. AOs are initially designed to anneal to the more obvious motifs implicated in the splicing process, such as acceptor or donor splice sites and in silico predicted exonic splicing enhancers. The AOs are evaluated for their ability to induce targeted exon skipping after transfection into cultured myoblasts. Although no single motif has been implicated in the consistent induction of exon skipping, the length of the AO has emerged as an important parameter in designing compounds that redirect dystrophin pre-mRNA processing. We present data from in vitro studies in murine and human cells showing that appropriately designed AOs of 25-31 nucleotides are generally more effective at inducing exon skipping than shorter counterparts. However, there appears to be an upper limit in optimal length, which may have to be established on a case-by-case basis.


Subject(s)
Dystrophin/genetics , Exons/genetics , Oligonucleotides, Antisense/genetics , Animals , Base Sequence , Cell Line , Humans , Mice , Molecular Sequence Data , RNA, Messenger/genetics , Transcription, Genetic/genetics
8.
Neuromuscul Disord ; 16(9-10): 583-90, 2006 Oct.
Article in English | MEDLINE | ID: mdl-16919955

ABSTRACT

Antisense oligonucleotide (AO) manipulation of pre-mRNA splicing of the dystrophin gene is showing promise in overcoming Duchenne muscular dystrophy (DMD)-causing mutations. To date, this approach has been limited to studies using animal models or cultured human muscle cells, and evidence that AOs can induce exon skipping in human muscle has yet to be shown. In this study, we used different AO analogues to induce exon skipping in muscle explants derived from normal and DMD human tissue. We propose that inducing exon skipping in human muscle explants is closer to in vivo conditions than cells in monolayer cultures, and may minimize the numbers of participants in Phase I clinical studies to demonstrate proof of principle of exon skipping in human muscle.


Subject(s)
Dystrophin/genetics , Exons/genetics , Muscle, Skeletal/metabolism , Muscular Dystrophy, Duchenne/genetics , Muscular Dystrophy, Duchenne/metabolism , Mutation/genetics , Animals , Cells, Cultured , DNA Mutational Analysis , Dystrophin/biosynthesis , Genetic Predisposition to Disease/genetics , Genetic Testing , Genetic Therapy/methods , Genetic Therapy/trends , Humans , Mice , Mice, Inbred mdx , Muscle, Skeletal/pathology , Muscle, Skeletal/physiopathology , Muscular Dystrophy, Duchenne/physiopathology , Oligonucleotides, Antisense/genetics , Oligonucleotides, Antisense/pharmacology , Oligonucleotides, Antisense/therapeutic use , RNA Precursors/genetics , RNA Splicing/genetics
9.
Gene Ther ; 13(19): 1373-81, 2006 Oct.
Article in English | MEDLINE | ID: mdl-16724091

ABSTRACT

Manipulation of pre-mRNA splicing by antisense oligonucleotides (AOs) offers considerable potential for a number of genetic disorders. One of these is Duchenne muscular dystrophy (DMD), where mutations in the dystrophin gene typically result in premature termination of translation that causes a loss of functional protein. AOs can induce exon skipping such that the mutation is by-passed and the reading frame restored, producing an internally deleted protein similar to that found in the milder Becker muscular dystrophy. To date, this approach has been applied to the mdx mouse model in vitro and in vivo and in human myoblast cultures. Here, we report the application of AO-directed exon skipping to induce dystrophin expression in vitro in a canine model of DMD, golden retriever muscular dystrophy (GRMD). The efficacy of 2'-O-methyl phosphorothioate (2OMe), phosphorodiamidate morpholino oligomers (PMOs) and peptide-linked PMOs (PMO-Pep) to induce dystrophin expression was assessed. The 2OMe chemistry was only effective for short-term induction of corrected transcript and could not induce detectable dystrophin protein. The PMO chemistry generally induced limited exon skipping at only high concentrations; however, a low level of dystrophin protein was produced in treated cells. Use of the PMO-Pep, applied here for the first time to a DMD model, was able to induce high and sustained levels of exon skipping and induced the highest level of dystrophin expression with no apparent adverse effects upon the cells. The induction of dystrophin in the GRMD model offers the potential for further testing of AO delivery regimens in a larger animal model of DMD, in preparation for application in human clinical trials.


Subject(s)
Alternative Splicing , Dystrophin/genetics , Exons , Genetic Therapy/methods , Muscular Dystrophy, Duchenne/therapy , Oligonucleotides, Antisense/pharmacology , Animals , Blotting, Western/methods , Cells, Cultured , Dogs , Dystrophin/analysis , Dystrophin/metabolism , Gene Expression , Humans , Muscular Dystrophy, Duchenne/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Transfection/methods
10.
Acta Myol ; 24(3): 222-9, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16629057

ABSTRACT

Antisense oligonucleotide induced exon skipping has recently emerged as a potential therapy to by-pass the consequences of many, but not all dystrophin mutations that lead to Duchenne muscular dystrophy. Targeted removal of one or more exons, to restore a disrupted reading frame, or omit a nonsense mutation, could lessen the consequences of an estimated 80% of dystrophin gene mutations. Promising in vitro and in vivo experiments in animal models of dystrophinopathies, as well as demonstration of induced exon skipping in cultured human myogenic cells have prompted considerable enthusiasm. Furthermore, advances in antisense oligonucleotide chemistries have resulted in the development of more stable and less toxic compounds, some of which are currently in Phase III clinical trials for selected antiviral applications. This review will summarize developments in induced exon skipping that have paved the way to clinical trials and some of the challenges and possible limitations.


Subject(s)
Dystrophin/genetics , Exons/genetics , Genetic Therapy , Muscular Dystrophy, Duchenne/genetics , Muscular Dystrophy, Duchenne/therapy , Oligoribonucleotides, Antisense , Humans , RNA Splicing/genetics
11.
FEBS Lett ; 552(2-3): 145-9, 2003 Sep 25.
Article in English | MEDLINE | ID: mdl-14527677

ABSTRACT

The use of antisense oligonucleotides (AOs) to induce exon skipping leading to generation of an in-frame dystrophin protein product could be of benefit in around 70% of Duchenne muscular dystrophy patients. We describe the use of hyaluronidase enhanced electrotransfer to deliver uncomplexed 2'-O-methyl modified phosphorothioate AO to adult dystrophic mouse muscle, resulting in dystrophin expression in 20-30% of fibres in tibialis anterior muscle after a single injection. Although expression was transient, many of the corrected fibres initially showed levels of dystrophin expression well above the 20% of endogenous previously shown to be necessary for phenotypic correction of the dystrophic phenotype.


Subject(s)
Dystrophin/genetics , Muscular Dystrophy, Animal/genetics , Muscular Dystrophy, Animal/therapy , Oligodeoxyribonucleotides, Antisense/administration & dosage , Oligodeoxyribonucleotides, Antisense/genetics , Animals , Base Sequence , Dystrophin/chemistry , Dystrophin/metabolism , Electroporation/methods , Genetic Therapy , Humans , Hyaluronoglucosaminidase , Male , Mice , Mice, Inbred mdx , Muscle, Skeletal/metabolism , Muscular Dystrophy, Animal/metabolism , Muscular Dystrophy, Duchenne/genetics , Muscular Dystrophy, Duchenne/metabolism , Muscular Dystrophy, Duchenne/therapy , Oligodeoxyribonucleotides, Antisense/chemistry , Transduction, Genetic
12.
Exp Neurol ; 170(1): 72-84, 2001 Jul.
Article in English | MEDLINE | ID: mdl-11421585

ABSTRACT

In this study we demonstrate the potential for combining biocompatible polymers with genetically engineered cells to elicit axon regrowth across tissue defects in the injured CNS. Eighteen- to 21-day-old rats received implants of poly N-(2-hydroxypropyl)-methacrylamide (HPMA) hydrogels containing RGD peptide sequences that had been infiltrated with control (untransfected) fibroblasts (n = 8), fibroblasts engineered to express brain-derived neurotrophic factor (BDNF) (n = 5), ciliary neurotrophic factor (CNTF) (n = 5), or a mixture of BDNF and CNTF expressing fibroblasts (n = 11). Fibroblasts were prelabeled with Hoechst 33342. Cell/polymer constructs were inserted into cavities made in the left optic tract, between thalamus and superior colliculus. After 4-8 weeks, retinal projections were analyzed by injecting right eyes with cholera toxin (B-subunit). Rats were perfused 24 h later and sections were immunoreacted to visualize retinal axons, other axons (RT97 antibody), host astrocytes and macrophages, donor fibroblasts, and extracellular matrix molecules. The volume fraction (VF) of each gel that was occupied by RT97(+) axons was quantified. RT-PCR confirmed expression of the transgenes prior to, and 5 weeks after, transplantation. Compared to control rats (mean VF = 0.02 +/- 0.01% SEM) there was increased ingrowth of RT97(+) axons into implants in CNTF (mean VF = 0.33 +/- 0.19%) and BDNF (mean VF = 0.62 +/-0.19%) groups. Axon growth into hydrogels in the mixed BDNF/CNTF group (mean VF = 3.58 +/- 0.92%) was significantly greater (P < 0.05) than in the BDNF or CNTF fibroblast groups. Retinal axons exhibited a complex branching pattern within gels containing BDNF or BDNF/CNTF fibroblasts; however, they regrew the greatest distances within implants containing both BDNF and CNTF expressing cells.


Subject(s)
Axons/metabolism , Brain Injuries/therapy , Brain-Derived Neurotrophic Factor/biosynthesis , Ciliary Neurotrophic Factor/biosynthesis , Fibroblasts/metabolism , Animals , Antigens, Differentiation/metabolism , Axons/drug effects , Brain Injuries/pathology , Brain-Derived Neurotrophic Factor/genetics , Brain-Derived Neurotrophic Factor/pharmacology , Cell Division/drug effects , Cells, Cultured , Ciliary Neurotrophic Factor/genetics , Ciliary Neurotrophic Factor/pharmacology , Disease Models, Animal , Drug Implants , Extracellular Matrix/metabolism , Female , Fibroblasts/cytology , Fibroblasts/transplantation , Fibronectins/metabolism , Hydrogel, Polyethylene Glycol Dimethacrylate/metabolism , Hydrogel, Polyethylene Glycol Dimethacrylate/pharmacology , Rats , Rats, Inbred F344 , Retina/cytology , Superior Colliculi/cytology , Thalamus/cytology , Transgenes , Visual Pathways/drug effects , Visual Pathways/pathology
13.
Neuromuscul Disord ; 11(3): 239-43, 2001 Apr.
Article in English | MEDLINE | ID: mdl-11297938

ABSTRACT

Golden retriever muscular dystrophy arises from a mutation in the acceptor splice site of intron 6 of the dystrophin gene. Skipping of exon 7 disrupts the mRNA reading frame and results in premature termination of translation. We are using this animal model to evaluate treatments for Duchenne muscular dystrophy, including gene repair induced by chimeric oligonucleotides. After injection of golden retriever muscular dystrophy (GRMD) muscle with a chimeric oligonucleotide to repair the lesion, immunostaining revealed a modest increase in the number of dystrophin-positive fibres at the injection sites. Dystrophin gene transcripts containing exon 7 were detected by reverse transcription-polymerase chain reaction, suggesting that low levels of splice site correction may have occurred. However, DNA sequencing of these apparently normal dystrophin gene transcripts revealed that the first five bases of exon 7 were missing. It will be important to be aware of this phenomenon with respect to further gene correction studies in the canine model.


Subject(s)
Alternative Splicing/genetics , Muscular Dystrophy, Duchenne/genetics , Mutagenesis, Site-Directed/genetics , RNA Splice Sites/genetics , Animals , Chimera/genetics , Disease Models, Animal , Dogs , Dystrophin/genetics , Exons/genetics , Female , Frameshift Mutation/genetics , Immunohistochemistry , Male , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Muscle, Skeletal/physiopathology , Muscular Dystrophy, Duchenne/metabolism , Muscular Dystrophy, Duchenne/physiopathology , Oligonucleotides/pharmacology , RNA, Messenger/genetics , Reading Frames/genetics , Reverse Transcriptase Polymerase Chain Reaction , Transcription, Genetic/genetics
14.
Proc Natl Acad Sci U S A ; 98(1): 42-7, 2001 Jan 02.
Article in English | MEDLINE | ID: mdl-11120883

ABSTRACT

Duchenne muscular dystrophy (DMD) is a severe muscle wasting disease arising from defects in the dystrophin gene, typically nonsense or frameshift mutations, that preclude the synthesis of a functional protein. A milder, allelic version of the disease, Becker muscular dystrophy, generally arises from in-frame deletions that allow synthesis of a shorter but still semifunctional protein. Therapies to introduce functional dystrophin into dystrophic tissue through either cell or gene replacement have not been successful to date. We report an alternative approach where 2'-O-methyl antisense oligoribonucleotides have been used to modify processing of the dystrophin pre-mRNA in the mdx mouse model of DMD. By targeting 2'-O-methyl antisense oligoribonucleotides to block motifs involved in normal dystrophin pre-mRNA splicing, we induced excision of exon 23, and the mdx nonsense mutation, without disrupting the reading frame. Exon 23 skipping was first optimized in vitro in transfected H-2K(b)-tsA58 mdx myoblasts and then induced in vivo. Immunohistochemical staining demonstrated the synthesis and correct subsarcolemmal localization of dystrophin and gamma-sarcoglycan in the mdx mouse after intramuscular delivery of antisense oligoribonucleotide:liposome complexes. This approach should reduce the severity of DMD by allowing a dystrophic gene transcript to be modified, such that it can be translated into a Becker-dystrophin-like protein.


Subject(s)
Dystrophin/biosynthesis , Dystrophin/genetics , Exons/genetics , Muscular Dystrophy, Duchenne/genetics , Oligoribonucleotides, Antisense/genetics , RNA Splicing/genetics , Animals , Base Sequence , Cells, Cultured , Cytoskeletal Proteins/metabolism , Disease Models, Animal , Fluorescein , Immunohistochemistry , Injections, Intramuscular , Introns/genetics , Membrane Glycoproteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Inbred mdx , Microscopy, Fluorescence , Molecular Sequence Data , Muscles/metabolism , Muscular Dystrophy, Duchenne/therapy , Oligoribonucleotides, Antisense/administration & dosage , Oligoribonucleotides, Antisense/therapeutic use , Open Reading Frames/genetics , Phosphatidylethanolamines/metabolism , RNA Precursors/genetics , RNA Precursors/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Sarcoglycans
15.
Am J Vet Res ; 62(12): 1964-8, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11763189

ABSTRACT

OBJECTIVES: To determine the distribution of a 231-base pair (bp) element in the dystrophin gene 3' untranslated region (UTR) in a colony of Golden Retrievers with muscular dystrophy and other unrelated dogs and to estimate the frequency of recombination for the canine dystrophin gene. ANIMALS: 77 dogs from the Golden Retriever Muscular Dystrophy (GRMD) colony at the Murdoch Veterinary School and 30 unrelated dogs from the Murdoch University Veterinary Clinic. PROCEDURE: Samples of blood or hair from dogs were used for amplification of DNA, using primers to the canine dystrophin 3' UTR. RESULTS: The DNA from affected dogs generated a larger PCR product than that obtained from clinically normal dogs. Products were cloned and sequenced, and the difference in size was found to be attributable to a 231-bp short interspersed nucleotide element (SINE). The SINE was found in all affected dogs in the colony but not in most unaffected puppies in the colony. Eighteen of 19 dogs in the colony were heterozygous for the GRMD mutation, and 7 of 30 unrelated dogs also were heterozygous for the SINE. CONCLUSION AND CLINICAL RELEVANCE: Evidence of recombination between the GRMD mutation and the SINE was observed in only 4 dogs (2 sets of littermates) in the GRMD colony. Incidence of this SINE in a few unrelated dogs suggests that this particular insertion into the dystrophin gene may have been a recent event. The SINE in the dystrophin 3' UTR did not have an apparent influence on dystrophin mRNA concentrations.


Subject(s)
3' Untranslated Regions/genetics , Dog Diseases/genetics , Dystrophin/genetics , Muscular Dystrophy, Animal/genetics , Short Interspersed Nucleotide Elements/genetics , Animals , Base Sequence , DNA/chemistry , DNA/genetics , DNA/isolation & purification , Dogs , Female , Male , Molecular Sequence Data , Mutation , RNA/chemistry , RNA/genetics , RNA/isolation & purification , Reverse Transcriptase Polymerase Chain Reaction/veterinary
16.
Curr Opin Neurol ; 13(5): 553-60, 2000 Oct.
Article in English | MEDLINE | ID: mdl-11073362

ABSTRACT

Gene therapy for inherited muscle disease is an active area of research and development. Initial emphasis has been on gene replacement but alternative approaches are increasingly being considered in order to overcome difficulties, such as the immune rejection of transduced cells, the need for appropriate and tissue-specific control of expression, and the requirement for systemic spread in some conditions. However, the most significant obstacles to the clinical success of gene therapy are still the lack of efficiency and accuracy of gene medicine delivery.


Subject(s)
Genetic Therapy , Muscular Diseases/genetics , Muscular Diseases/therapy , Animals , Genetic Vectors , Humans , Point Mutation , RNA/genetics , RNA, Messenger/genetics , Viruses/genetics
17.
J Cell Biol ; 148(5): 985-96, 2000 Mar 06.
Article in English | MEDLINE | ID: mdl-10704448

ABSTRACT

Conventionally, nonsense mutations within a gene preclude synthesis of a full-length functional protein. Obviation of such a blockage is seen in the mdx mouse, where despite a nonsense mutation in exon 23 of the dystrophin gene, occasional so-called revertant muscle fibers are seen to contain near-normal levels of its protein product. Here, we show that reversion of dystrophin expression in mdx mice muscle involves unprecedented massive loss of up to 30 exons. We detected several alternatively processed transcripts that could account for some of the revertant dystrophins and could not detect genomic deletion from the region commonly skipped in revertant dystrophin. This, together with exon skipping in two noncontiguous regions, favors aberrant splicing as the mechanism for the restoration of dystrophin, but is hard to reconcile with the clonal idiosyncrasy of revertant dystrophins. Revertant dystrophins retain functional domains and mediate plasmalemmal assembly of the dystrophin-associated glycoprotein complex. Physiological function of revertant fibers is demonstrated by the clonal growth of revertant clusters with age, suggesting that revertant dystrophin could be used as a guide to the construction of dystrophin expression vectors for individual gene therapy. The dystrophin gene in the mdx mouse provides a favored system for study of exon skipping associated with nonsense mutations.


Subject(s)
Alternative Splicing/genetics , Codon, Nonsense/genetics , Dystrophin/genetics , Exons/genetics , Muscle Fibers, Skeletal/metabolism , Muscular Dystrophy, Animal/genetics , Aging/genetics , Animals , Antibodies/metabolism , Cell Nucleus/metabolism , Dystrophin/biosynthesis , Dystrophin/immunology , Epitopes/genetics , Epitopes/immunology , Immunohistochemistry , Male , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Inbred mdx , Muscle, Skeletal/metabolism , Protein Structure, Tertiary/genetics , RNA, Messenger/biosynthesis
18.
Neuromuscul Disord ; 10(2): 100-7, 2000 Feb.
Article in English | MEDLINE | ID: mdl-10714584

ABSTRACT

We report two siblings with a relatively severe limb-girdle muscular dystrophy. The elder sister presented at 8 years of age with inability to climb and abnormal gait. At 12 years she was barely ambulant. Her sister followed a similar course. Serum creatine kinase was 8500-10000 IU (N 25-200) in the elder sister and 17000-19000 IU in the younger sister. Muscle biopsy of the elder sister at 8 years showed chronic myopathic changes with loss of muscle fibres, active necrosis and regeneration. Immunocytochemistry demonstrated normal spectrin and dystrophin, reduced alpha-sarcoglycan and absent gamma-sarcoglycan--indicating a gamma-sarcoglycanopathy. Haplotype analysis for the markers D13S115, D13S232, D13S292, D13S787, D13S1243 and D13S283 internal to and flanking the gamma-sarcoglycan gene showed the affected sisters shared haplotypes, indicating it was possible they were suffering from a gamma-sarcoglycanopathy. Non-inheritance of paternal alleles for D13S232, D13S292 and D13S1243 suggested the inheritance of a deletion, which was confirmed by FISH, using a genomic probe from the gamma-sarcoglycan gene. The gamma-sarcoglycan cDNA was amplified by reverse transcriptase PCR from the muscle biopsy of the elder sister and sequenced. A missense mutation changing codon 69 from GGC glycine to CGC arginine was identified. HhaI digestion of exon 3 genomic PCR products showed the two affected sisters were hemizygous for the mutation, while the mother and grandmother were heterozygotes. The mutation, identified by SSCP analysis, was not observed in 116 unrelated, unaffected individuals. Previously, only two other missense mutations, the Cys283Tyr missense mutation in Gypsies and the Leu193Ser mutation in a Dutch family, have been described in the gamma-sarcoglycan gene. The fact that the affected individuals in the current and Gypsy families are gamma-sarcoglycan negative may indicate that codons 69 and 283 are important in gamma-sarcoglycan function.


Subject(s)
Gene Deletion , Muscular Dystrophies/genetics , Mutation, Missense/genetics , Adolescent , Child , Female , Humans , In Situ Hybridization, Fluorescence , Muscles/pathology , Muscular Dystrophies/pathology , Pedigree , Polymorphism, Single-Stranded Conformational
19.
Neuromuscul Disord ; 9(5): 289-95, 1999 Jul.
Article in English | MEDLINE | ID: mdl-10407848

ABSTRACT

We have determined the molecular basis for skeletal myopathy and dilated cardiomyopathy in two male German short-haired pointer (GSHP) littermates. Analysis of skeletal muscle demonstrated a complete absence of dystrophin on Western blot analysis. PCR analysis of genomic DNA revealed a deletion encompassing the entire dystrophin gene. Molecular cytogenetic analysis of lymphocytes from the dam and both dystrophic pups confirmed a visible deletion in the p21 region of the affected canine X chromosome. Utrophin is up-regulated in the skeletal muscle, but does not appear to ameliorate the dystrophic canine phenotype. This new canine model should further our understanding of the physiological and biochemical processes in Duchenne muscular dystrophy.


Subject(s)
Dog Diseases/genetics , Dystrophin/genetics , Muscular Dystrophy, Animal/genetics , Animals , Biopsy , Blotting, Western , Chromosome Deletion , Creatine Kinase/blood , DNA/genetics , Disease Models, Animal , Dog Diseases/pathology , Dogs , In Situ Hybridization, Fluorescence , Male , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Muscular Dystrophy, Animal/pathology , Mutation , Polymerase Chain Reaction , X Chromosome/genetics
20.
Neuromuscul Disord ; 9(5): 330-8, 1999 Jul.
Article in English | MEDLINE | ID: mdl-10407856

ABSTRACT

The mdx mouse, which carries a nonsense mutation in exon 23 of the dystrophin gene, has been used as an animal model of Duchenne muscular dystrophy to evaluate cell or gene replacement therapies. Despite the mdx mutation, which should preclude the synthesis of a functional dystrophin protein, rare, naturally occurring dystrophin-positive fibres have been observed in mdx muscle tissue. These dystrophin-positive fibres are thought to have arisen from an exon-skipping mechanism, either somatic mutations or alternative splicing. Increasing the frequency of these fibres may offer another therapeutic approach to reduce the severity of Duchenne muscular dystrophy. Antisense oligonucleotides have been shown to block aberrant splicing in the human beta-globin gene. We wished to use a similar approach to re-direct normal processing of the dystrophin pre-mRNA and induce specific exon skipping. Antisense 2'-O-methyl-oligoribonucleotides, directed to the 3' and 5' splice sites of introns 22 and 23, respectively in the mdx pre-mRNA, were used to transfect myoblast cultures. The 5' antisense oligonucleotide appeared to efficiently displace factors normally involved in the removal of intron 23 so that exon 23 was also removed during the splicing of the dystrophin pre-mRNA. Approximately 50% of the dystrophin gene mRNAs were missing this exon 6 h after transfection of primary mdx myotubes, with all transcripts showing skipping of exon 23 after 24 h. Deletion of exon 23 does not disrupt the reading frame and should allow the synthesis of a shorter but presumably functional Becker-like dystrophin. Molecular intervention at dystrophin pre-mRNA splicing has the potential to reduce the severity of a Duchenne mutation to the milder Becker phenotype.


Subject(s)
Dystrophin/genetics , Muscular Dystrophy, Animal/genetics , Oligonucleotides, Antisense/genetics , RNA, Messenger/genetics , Alternative Splicing , Amino Acid Sequence , Animals , Base Sequence , Cell Line , Exons , Mice , Mice, Inbred C57BL , Mice, Inbred mdx , Muscular Dystrophy, Animal/therapy , Mutation , Oligonucleotides, Antisense/therapeutic use , RNA Precursors/genetics , RNA Processing, Post-Transcriptional
SELECTION OF CITATIONS
SEARCH DETAIL
...