Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
Nat Commun ; 10(1): 2788, 2019 06 26.
Article in English | MEDLINE | ID: mdl-31243277

ABSTRACT

Many potentially therapeutic molecules have been identified for treating Duchenne muscular dystrophy. However, targeting those molecules only to sites of active pathology is an obstacle to their clinical use. Because dystrophic muscles become extensively inflamed, we tested whether expressing a therapeutic transgene in leukocyte progenitors that invade muscle would provide selective, timely delivery to diseased muscle. We designed a transgene in which leukemia inhibitory factor (LIF) is under control of a leukocyte-specific promoter and transplanted transgenic cells into dystrophic mice. Transplantation diminishes pathology, reduces Th2 cytokines in muscle and biases macrophages away from a CD163+/CD206+ phenotype that promotes fibrosis. Transgenic cells also abrogate TGFß signaling, reduce fibro/adipogenic progenitor cells and reduce fibrogenesis of muscle cells. These findings indicate that leukocytes expressing a LIF transgene reduce fibrosis by suppressing type 2 immunity and highlight a novel application by which immune cells can be genetically modified as potential therapeutics to treat muscle disease.


Subject(s)
Genetic Therapy , Leukemia Inhibitory Factor/metabolism , Muscular Dystrophy, Animal/therapy , Animals , Bone Marrow Cells/metabolism , Gene Expression Regulation , Leukemia Inhibitory Factor/genetics , Male , Mice , Mice, Inbred mdx , Muscle, Skeletal/pathology , Random Allocation , Specific Pathogen-Free Organisms , Transgenes
2.
Article in English | MEDLINE | ID: mdl-30050367

ABSTRACT

Duchenne muscular dystrophy (DMD) is an autosomal dominant, X-linked neuromuscular disorder caused by mutations in dystrophin, one of the largest genes known to date. Dystrophin gene mutations are generally transmitted from the mother to male offspring and can occur throughout the coding length of the gene. The majority of the methodologies aimed at treating the disorder have focused on restoring a shorter, although partially functional, dystrophin protein. The approach has the potential of converting a severe DMD phenotype into a milder form of the disease known as Becker muscular dystrophy. Others have focused on ameliorating the disease by targeting secondary pathologies such as inflammation or loss of regeneration. Of great potential is the development of strategies that are capable of restoring full-length dystrophin expression due to their ability to produce a normal, fully functional protein. Among these strategies, the use of read-through compounds (RTCs) that could be administered orally represents an ideal option. Gentamicin has been previously tested in clinical trials for DMD with limited or no success, and its use in the clinic has been dismissed due to issues of toxicity and lack of clear benefits to patients. More recently, new RTCs have been identified and tested in animal models for DMD. This review will focus on one of those RTCs known as ataluren that has now completed Phase III clinical studies for DMD and at providing an overview of the different stages that have led to its clinical development for the disease. The impact that this new drug may have on DMD and its future perspectives will also be described, with an emphasis on the importance of further assessing the clinical benefits of this molecule in patients as it becomes available on the market in different countries.

3.
Cell Metab ; 20(6): 927-9, 2014 Dec 02.
Article in English | MEDLINE | ID: mdl-25470540

ABSTRACT

Myriad strategies have been explored to compensate for the lack of dystrophin or to skip mutations that cause the lethal disease Duchenne muscular dystrophy (DMD). A new study shows that gene editing strategies used by bacteria can be applied in zygotes of a mouse model of DMD to correct the genetic defect that causes muscular dystrophy (Long et al., 2014).


Subject(s)
Bacteria/genetics , Dystrophin/genetics , Muscular Dystrophy, Duchenne/genetics , Animals , Disease Models, Animal , Mice , Muscular Dystrophy, Duchenne/therapy , Mutation
4.
Biology (Basel) ; 3(4): 752-80, 2014 Nov 14.
Article in English | MEDLINE | ID: mdl-25405319

ABSTRACT

Centers for the screening of biologically active compounds and genomic libraries are becoming common in the academic setting and have enabled researchers devoted to developing strategies for the treatment of diseases or interested in studying a biological phenomenon to have unprecedented access to libraries that, until few years ago, were accessible only by pharmaceutical companies. As a result, new drugs and genetic targets have now been identified for the treatment of Duchenne muscular dystrophy (DMD), the most prominent of the neuromuscular disorders affecting children. Although the work is still at an early stage, the results obtained to date are encouraging and demonstrate the importance that these centers may have in advancing therapeutic strategies for DMD as well as other diseases. This review will provide a summary of the status and progress made toward the development of a cure for this disorder and implementing high-throughput screening (HTS) technologies as the main source of discovery. As more academic institutions are gaining access to HTS as a valuable discovery tool, the identification of new biologically active molecules is likely to grow larger. In addition, the presence in the academic setting of experts in different aspects of the disease will offer the opportunity to develop novel assays capable of identifying new targets to be pursued as potential therapeutic options. These assays will represent an excellent source to be used by pharmaceutical companies for the screening of larger libraries providing the opportunity to establish strong collaborations between the private and academic sectors and maximizing the chances of bringing into the clinic new drugs for the treatment of DMD.

5.
Front Physiol ; 5: 148, 2014.
Article in English | MEDLINE | ID: mdl-24795643

ABSTRACT

The progressive loss of muscle mass characteristic of many muscular dystrophies impairs the efficacy of most of the gene and molecular therapies currently being pursued for the treatment of those disorders. It is becoming increasingly evident that a therapeutic application, to be effective, needs to target not only mature myofibers, but also muscle progenitors cells or muscle stem cells able to form new muscle tissue and to restore myofibers lost as the result of the diseases or during normal homeostasis so as to guarantee effective and lost lasting effects. Correction of the genetic defect using oligodeoxynucleotides (ODNs) or engineered nucleases holds great potential for the treatment of many of the musculoskeletal disorders. The encouraging results obtained by studying in vitro systems and model organisms have set the groundwork for what is likely to become an emerging field in the area of molecular and regenerative medicine. Furthermore, the ability to isolate and expand from patients various types of muscle progenitor cells capable of committing to the myogenic lineage provides the opportunity to establish cell lines that can be used for transplantation following ex vivo manipulation and expansion. The purpose of this article is to provide a perspective on approaches aimed at correcting the genetic defect using gene editing strategies and currently under development for the treatment of Duchenne muscular dystrophy (DMD), the most sever of the neuromuscular disorders. Emphasis will be placed on describing the potential of using the patient own stem cell as source of transplantation and the challenges that gene editing technologies face in the field of regenerative biology.

6.
Stem Cells ; 32(7): 1817-30, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24753122

ABSTRACT

Duchenne muscular dystrophy (DMD) is a fatal disease caused by mutations in the dystrophin gene, which result in the complete absence of dystrophin protein throughout the body. Gene correction strategies hold promise to treating DMD. Our laboratory has previously demonstrated the ability of peptide nucleic acid single-stranded oligodeoxynucleotides (PNA-ssODNs) to permanently correct single-point mutations at the genomic level. In this study, we show that PNA-ssODNs can target and correct muscle satellite cells (SCs), a population of stem cells capable of self-renewing and differentiating into muscle fibers. When transplanted into skeletal muscles, SCs transfected with correcting PNA-ssODNs were able to engraft and to restore dystrophin expression. The number of dystrophin-positive fibers was shown to significantly increase over time. Expression was confirmed to be the result of the activation of a subpopulation of SCs that had undergone repair as demonstrated by immunofluorescence analyses of engrafted muscles using antibodies specific to full-length dystrophin transcripts and by genomic DNA analysis of dystrophin-positive fibers. Furthermore, the increase in dystrophin expression detected over time resulted in a significant improvement in muscle morphology. The ability of transplanted cells to return into quiescence and to activate upon demand was confirmed in all engrafted muscles following injury. These results demonstrate the feasibility of using gene editing strategies to target and correct SCs and further establish the therapeutic potential of this approach to permanently restore dystrophin expression into muscle of DMD patients.


Subject(s)
Dystrophin/genetics , Muscular Dystrophy, Duchenne/pathology , Myoblasts, Skeletal/metabolism , Peptide Nucleic Acids/genetics , Animals , Cells, Cultured , Dystrophin/metabolism , Gene Expression , Genetic Engineering , Genetic Therapy , Humans , Mice, Inbred C57BL , Mice, Inbred mdx , Mice, Nude , Muscle Fibers, Skeletal/metabolism , Muscle Fibers, Skeletal/pathology , Muscular Dystrophy, Duchenne/metabolism , Myoblasts, Skeletal/transplantation , Peptide Nucleic Acids/metabolism , Regenerative Medicine , Transfection
7.
Methods Mol Biol ; 1114: 59-83, 2014.
Article in English | MEDLINE | ID: mdl-24557897

ABSTRACT

The past decade has seen the development of new technologies capable of editing the genome that have naturally led to exploring their therapeutic application for the treatment of many disorders. Among those, Duchenne muscular dystrophy (DMD) represents an ideal candidate for gene editing primarily due to the large size of dystrophin, the gene responsible for the disease, which limits the use of gene replacement approaches. Critical in the evaluation of the efficacy of the treatment is the development of a method that can accurately quantitate the frequencies of gene repair obtained in the dystrophin gene at both the genomic level as well as the mRNA level. The mdx (5cv) mouse model of DMD offers an ideal system to precisely determine the frequencies of gene repair. Here we describe the methods used for determining those frequencies and the limitations associated with the use of gene correction for the treatment of DMD. Clinical approaches to muscle disorders using ssODNs will heavily rely on the optimization of the technology and will have to take into consideration the safety, efficacy and cost of the procedure in vision of systemic delivery of the therapeutic treatment.


Subject(s)
DNA, Single-Stranded/genetics , Gene Expression , Muscle Cells/metabolism , Oligodeoxyribonucleotides/genetics , Animals , Cell Culture Techniques , DNA, Single-Stranded/chemical synthesis , Mice , Mice, Inbred mdx , Muscle Cells/cytology , Muscular Dystrophy, Duchenne/genetics , Oligodeoxyribonucleotides/chemical synthesis , Transfection
8.
Methods Mol Biol ; 1114: 399-411, 2014.
Article in English | MEDLINE | ID: mdl-24557918

ABSTRACT

Gene correction is attractive for single gene mutation disorders, such as Duchenne muscular dystrophy (DMD). The mdx mouse model of DMD is dystrophin deficient due to a premature chain-terminating point mutation in exon 23 of the dystrophin gene. Gene editing of genomic DNA using single-stranded oligodeoxynucleotides (ssODNs) offers the potential to change the DNA sequence to alter mRNA and protein expression in defined ways. When applied to fetal skeletal muscle of mdx mice in utero, this technology leads to restoration of dystrophin protein expression, thus providing a valid gene-based therapeutic application at the earliest developmental stage. Here, we describe detailed methods for gene editing using muscle delivery of ssODNs to the fetal mdx mouse in utero at embryonic day 16 and to test correction of dystrophin deficiency at different ages after birth.


Subject(s)
Gene Transfer Techniques , Oligodeoxyribonucleotides/genetics , Animals , DNA, Single-Stranded/administration & dosage , DNA, Single-Stranded/genetics , Dystrophin/genetics , Gene Expression , Genetic Therapy , Mice , Mice, Inbred mdx , Muscle, Skeletal/metabolism , Muscular Dystrophy, Duchenne/genetics , Muscular Dystrophy, Duchenne/therapy , Oligodeoxyribonucleotides/administration & dosage
9.
Future Neurol ; 8(4)2013 Jul 01.
Article in English | MEDLINE | ID: mdl-24273459

ABSTRACT

The Second Muscular Dystrophy Association Scientific Meeting was held on 21-24 April 2013 in Washington (DC, USA). The meeting provided an opportunity for research scientists, clinicians, government agencies and industry experts to highlight and discuss different aspects of therapy development for neuromuscular diseases, including novel targets, biomarkers, therapeutic approaches, animal models and clinical trials. With 500 participants, 66 presentations and 200 abstracts, the 3-day conference has become a central focus for scientists interested in translational research and moving potential therapies forward from the bench to the bedside. Key issues covered by the meeting included the need to identify new drugs to treat patients with neuromuscular diseases and the importance of establishing collaborations between government, academic and industry sectors toward an efficient and rigorous translational path for neuromuscular diseases.

10.
Hum Mol Genet ; 22(16): 3283-95, 2013 Aug 15.
Article in English | MEDLINE | ID: mdl-23612904

ABSTRACT

Cell-mediated regenerative approaches using muscle progenitor cells hold promises for the treatment of many forms of muscle disorders. Their applicability in the clinic, however, is hindered by the low levels of regeneration obtained after transplantation and the large number of cells required to achieve an effect. To better understand the mechanisms that regulate the temporal switch of replicating muscle progenitor cells into terminally differentiated cells and to develop new strategies that could enhance muscle regeneration, we have developed and performed a high-throughput screening (HTS) capable of identifying genes that play active roles during myogenesis. Secondary and tertiary screens were used to confirm the effects of RNAi in vitro and in vivo and to select for candidate hits that significantly increase regeneration into skeletal muscles. Downregulation of cyclin D2 (CCND2) was shown to dramatically enhance myogenic differentiation of muscle progenitor cells and to induce a robust regeneration after cell transplantation into skeletal muscles of dystrophin-deficient mice. Protein interaction network and pathway analysis revealed that CCND2 directly interacts with the cyclin-dependent kinase Cdk4 to inhibit phosphorylation of the retinoblastoma protein (pRb), thus blocking the activation of the myogenic switch during fusion. These studies identify CCND2 as a new key regulator of terminal differentiation in muscle progenitor cells and open new possibilities for the treatment of many forms of muscle disorders characterized by impaired regeneration and loss of muscle mass.


Subject(s)
Cyclin D2/genetics , Cyclin D2/metabolism , Muscle Development/genetics , Myoblasts, Skeletal/physiology , Regeneration , Animals , Cell Fusion , Cells, Cultured , Cyclin-Dependent Kinase 4/genetics , Cyclin-Dependent Kinase 4/metabolism , Mice , Mice, Transgenic , Muscular Dystrophy, Duchenne/genetics , Muscular Dystrophy, Duchenne/metabolism , MyoD Protein/genetics , MyoD Protein/metabolism , Myoblasts, Skeletal/transplantation , Myogenic Regulatory Factor 5/genetics , Myogenic Regulatory Factor 5/metabolism , Myogenin/genetics , Myogenin/metabolism , Protein Interaction Mapping , RNA, Small Interfering , Retinoblastoma Protein/metabolism
11.
Hum Mol Genet ; 21(18): 4007-20, 2012 Sep 15.
Article in English | MEDLINE | ID: mdl-22692682

ABSTRACT

Molecules that induce ribosomal read-through of nonsense mutations in mRNA and allow production of a full-length functional protein hold great therapeutic potential for the treatment of many genetic disorders. Two such read-through compounds, RTC13 and RTC14, were recently identified by a luciferase-independent high-throughput screening assay and were shown to have potential therapeutic functions in the treatment of nonsense mutations in the ATM and the dystrophin genes. We have now tested the ability of RTC13 and RTC14 to restore dystrophin expression into skeletal muscles of the mdx mouse model for Duchenne muscular dystrophy (DMD). Direct intramuscular injection of compound RTC14 did not result in significant read-through activity in vivo and demonstrated the levels of dystrophin protein similar to those detected using gentamicin. In contrast, significant higher amounts of dystrophin were detected after intramuscular injection of RTC13. When administered systemically, RTC13 was shown to partially restore dystrophin protein in different muscle groups, including diaphragm and heart, and improved muscle function. An increase in muscle strength was detected in all treated animals and was accompanied by a significant decrease in creatine kinase levels. These studies establish the therapeutic potential of RTC13 in vivo and advance this newly identified compound into preclinical application for DMD.


Subject(s)
Dystrophin/genetics , Furans/pharmacology , Muscle, Skeletal/physiopathology , Muscular Dystrophy, Duchenne/drug therapy , Phenols/pharmacology , Schiff Bases/pharmacology , Thiazolidines/pharmacology , Transcription, Genetic/drug effects , Animals , Codon, Nonsense , Disease Models, Animal , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical , Dystrophin/metabolism , Furans/administration & dosage , Furans/pharmacokinetics , Gentamicins/administration & dosage , Gentamicins/pharmacology , Injections, Intramuscular , Injections, Intraperitoneal , Male , Mice , Mice, Inbred C57BL , Mice, Inbred mdx , Muscle Fibers, Skeletal/drug effects , Muscle Fibers, Skeletal/metabolism , Muscle Strength/drug effects , Muscle, Skeletal/drug effects , Muscle, Skeletal/metabolism , Muscular Dystrophy, Duchenne/metabolism , Muscular Dystrophy, Duchenne/physiopathology , Oxadiazoles/administration & dosage , Oxadiazoles/pharmacokinetics , Oxadiazoles/pharmacology , Phenols/administration & dosage , Protein Synthesis Inhibitors/administration & dosage , Protein Synthesis Inhibitors/pharmacology , Reading Frames , Schiff Bases/administration & dosage , Thiazolidines/administration & dosage , Thiazolidines/pharmacokinetics
12.
Hum Mol Genet ; 20(16): 3151-60, 2011 Aug 15.
Article in English | MEDLINE | ID: mdl-21576124

ABSTRACT

Antisense morpholino oligonucleotides (AMOs) can reprogram pre-mRNA splicing by complementary binding to a target site and regulating splice site selection, thereby offering a potential therapeutic tool for genetic disorders. However, the application of this technology into a clinical scenario has been limited by the low correction efficiency in vivo and inability of AMOs to efficiently cross the blood brain barrier and target brain cells when applied to neurogenetic disorders such as ataxia-telangiecatasia (A-T). We previously used AMOs to correct subtypes of ATM splicing mutations in A-T cells; AMOs restored up to 20% of the ATM protein and corrected the A-T cellular phenotype. In this study, we demonstrate that an arginine-rich cell-penetrating peptide, (RXRRBR)(2)XB, dramatically improved ATM splicing correction efficiency when conjugated with AMOs, and almost fully corrected aberrant splicing. The restored ATM protein was close to normal levels in cells with homozygous splicing mutations, and a gene dose effect was observed in cells with heterozygous mutations. A significant amount of the ATM protein was still detected 21 days after a single 5 µm treatment. Systemic administration of an fluorescein isothiocyanate-labeled (RXRRBR)(2)XB-AMO in mice showed efficient uptake in the brain. Fluorescence was evident in Purkinje cells after a single intravenous injection of 60 mg/kg. Furthermore, multiple injections significantly increased uptake in all areas of the brain, notably in cerebellum and Purkinje cells, and showed no apparent signs of toxicity. Taken together, these results highlight the therapeutic potential of (RXRRBR)(2)XB-AMOs in A-T and other neurogenetic disorders.


Subject(s)
Arginine/chemistry , Cell Cycle Proteins/genetics , Cell-Penetrating Peptides/pharmacology , Cerebellum/metabolism , DNA-Binding Proteins/genetics , Gene Transfer Techniques , Oligonucleotides, Antisense/pharmacology , Protein Serine-Threonine Kinases/genetics , RNA Splicing/genetics , Tumor Suppressor Proteins/genetics , Amino Acid Sequence , Animals , Ataxia Telangiectasia/enzymology , Ataxia Telangiectasia/pathology , Ataxia Telangiectasia Mutated Proteins , Cell-Penetrating Peptides/chemistry , Cerebellum/drug effects , Fluorescein-5-isothiocyanate/metabolism , Mice , Molecular Sequence Data , Protein Transport/drug effects , Purkinje Cells/drug effects , Purkinje Cells/metabolism , RNA Splicing/drug effects , Radiation Tolerance/drug effects
13.
Hum Mol Genet ; 19(16): 3266-81, 2010 Aug 15.
Article in English | MEDLINE | ID: mdl-20542988

ABSTRACT

Permanent correction of gene defects is an appealing approach to the treatment of genetic disorders. The use of single-stranded oligodeoxynucleotides (ssODNs) has been demonstrated to induce single-point mutations in the dystrophin gene and to restore dystrophin expression in the skeletal muscle of models of Duchenne muscular dystrophy (DMD). Here we show that ssODNs made of peptide nucleic acids (PNA-ssODNs) can achieve gene repair frequencies more than 10-fold higher than those obtained using an older generation of targeting oligonucleotides. Correction was demonstrated in muscles cells isolated from mdx(5cv) mice and was stably inherited over time. Direct intramuscular injection of PNA-ssODNs targeting the mdx(5cv) mutation resulted in a significant increase in dystrophin-positive fibers when compared with muscles that received the ssODNs designed to correct the dystrophin gene but made of unmodified bases. Correction was demonstrated at both the mRNA and the DNA levels using quantitative PCR and was confirmed by direct sequencing of amplification products. Analysis at the protein level demonstrated expression of full-length dystrophin in vitro as well as in vivo. These results demonstrate that oligonucleotides promoting strand invasion in the DNA double helix can significantly enhance gene repair frequencies of the dystrophin gene. The use of PNA-ssODNs has important implications in terms of both efficacy and duration of the repair process in muscles and may have a role in advancing the treatment of DMD.


Subject(s)
DNA Repair , Dystrophin/genetics , Oligodeoxyribonucleotides/genetics , Peptide Nucleic Acids/genetics , Animals , Animals, Newborn , Base Sequence , Blotting, Western , Cells, Cultured , DNA, Single-Stranded/genetics , Gene Expression , Injections, Intramuscular , Mice , Mice, Inbred C57BL , Mice, Inbred mdx , Muscles/cytology , Muscles/metabolism , Mutation , Myoblasts/cytology , Myoblasts/metabolism , Oligodeoxyribonucleotides/administration & dosage , Reverse Transcriptase Polymerase Chain Reaction , Transfection
14.
Nucleic Acids Res ; 37(22): 7468-82, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19854937

ABSTRACT

Gene editing mediated by oligonucleotides has been shown to induce stable single base alterations in genomic DNA in both prokaryotic and eukaryotic organisms. However, the low frequencies of gene repair have limited its applicability for both basic manipulation of genomic sequences and for the development of therapeutic approaches for genetic disorders. Here, we show that single-stranded oligodeoxynucleotides (ssODNs) containing a methyl-CpG modification and capable of binding to the methyl-CpG binding domain protein 4 (MBD4) are able to induce >10-fold higher levels of gene correction than ssODNs lacking the specific modification. Correction was stably inherited through cell division and was confirmed at the protein, transcript and genomic levels. Downregulation of MBD4 expression using RNAi prevented the enhancement of gene correction efficacy obtained using the methyl-CpG-modified ssODN, demonstrating the specificity of the repair mechanism being recruited. Our data demonstrate that efficient manipulation of genomic targets can be achieved and controlled by the type of ssODN used and by modulation of the repair mechanism involved in the correction process. This new generation of ssODNs represents an important technological advance that is likely to have an impact on multiple applications, especially for gene therapy where permanent correction of the genetic defect has clear advantages over viral and other nonviral approaches currently being tested.


Subject(s)
Base Pair Mismatch , DNA Repair , Endodeoxyribonucleases/metabolism , Oligodeoxyribonucleotides/chemistry , Animals , Binding Sites , Cell Survival , Cells, Cultured , CpG Islands , Endodeoxyribonucleases/antagonists & inhibitors , Endodeoxyribonucleases/genetics , Genes, Reporter , Green Fluorescent Proteins/analysis , Green Fluorescent Proteins/genetics , Mice , Oligodeoxyribonucleotides/metabolism , RNA Interference , Transfection
15.
J Exp Med ; 206(10): 2285-97, 2009 Sep 28.
Article in English | MEDLINE | ID: mdl-19770270

ABSTRACT

Large numbers of genetic disorders are caused by nonsense mutations for which compound-induced readthrough of premature termination codons (PTCs) might be exploited as a potential treatment strategy. We have successfully developed a sensitive and quantitative high-throughput screening (HTS) assay, protein transcription/translation (PTT)-enzyme-linked immunosorbent assay (ELISA), for identifying novel PTC-readthrough compounds using ataxia-telangiectasia (A-T) as a genetic disease model. This HTS PTT-ELISA assay is based on a coupled PTT that uses plasmid templates containing prototypic A-T mutated (ATM) mutations for HTS. The assay is luciferase independent. We screened approximately 34,000 compounds and identified 12 low-molecular-mass nonaminoglycosides with potential PTC-readthrough activity. From these, two leading compounds consistently induced functional ATM protein in ATM-deficient cells containing disease-causing nonsense mutations, as demonstrated by direct measurement of ATM protein, restored ATM kinase activity, and colony survival assays for cellular radiosensitivity. The two compounds also demonstrated readthrough activity in mdx mouse myotube cells carrying a nonsense mutation and induced significant amounts of dystrophin protein.


Subject(s)
Aminoglycosides/pharmacology , Cell Cycle Proteins/genetics , Codon, Nonsense , DNA-Binding Proteins/genetics , Protein Serine-Threonine Kinases/genetics , Tumor Suppressor Proteins/genetics , Animals , Ataxia Telangiectasia Mutated Proteins , Cell Cycle Proteins/metabolism , Cell Line , Cell Survival , Chromosomal Proteins, Non-Histone/metabolism , Enzyme-Linked Immunosorbent Assay , Mice , Mice, Inbred C57BL , Muscle Fibers, Skeletal/metabolism , Phosphorylation
16.
Front Biosci ; 13: 517-27, 2008 Jan 01.
Article in English | MEDLINE | ID: mdl-17981565

ABSTRACT

Duchenne Muscular dystrophy (DMD) is one of the most severe forms of hereditary diseases in muscles. The identification and characterization of dystrophin, the gene responsible for the disease has lead to the development of potential gene therapy treatments for this disorder. The complex structure and size of the dystrophin gene represent a challenge for some gene therapy approaches such as gene replacement mediated by viral vectors. Others, including oligonucleotide-mediated gene therapies have allowed forms of manipulation in the dystrophin gene not possible with other disorders. The use of oligonucleotides to modulate gene expression has shown to be a feasible alternative treatment to DMD. Antisense-mediated technologies have made outstanding progress in the last decade and two phase I clinical trials for exon skipping in DMD are already in progress. Gene correction mediated by oligonucleotides faces much greater obstacles, but the outcome of the approach, permanent correction of the gene defect, represents an ideal treatment to the disease. Gene therapy mediated by antisense oligonucleotides or oligonucleotide mediated gene editing have the potential to have a primary role in gene therapy applications to muscles, but they are still far from representing an effective cure. Factors like safety and sustained beneficial effects in patients will have to be considered in detail before this technology can become applicable to the treatment of muscles disorders. Ultimately the need for production of oligonucleotides in large scale and the cost of treatment for each individual patient will play a big role in the feasibility of these approaches in DMD.


Subject(s)
Muscular Dystrophy, Duchenne/genetics , Muscular Dystrophy, Duchenne/therapy , Oligonucleotides/chemistry , Animals , DNA/metabolism , Dogs , Dystrophin/metabolism , Exons , Gene Expression Regulation , Gene Transfer Techniques , Genetic Therapy/methods , Humans , Mice , Oligonucleotides, Antisense/chemistry , RNA, Messenger/metabolism
17.
Hum Gene Ther ; 17(8): 871-6, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16942446

ABSTRACT

Peripheral vascular disease (PVD), characterized by insufficient blood supply to extremities, can be a devastating illness. Although many gene therapy strategies for PVD using vascular endothelial growth factor (VEGF) have resulted in increased blood vessel formation, the vessels are often impermanent and regress after therapy, probably because of the short-lived VEGF expression mediated by gene therapy vectors (14 days or less). phiC31 integrase is a recombinase originally isolated from a bacteriophage of Streptomyces. This integrase performs efficient chromosomal integration of plasmid DNA into mammalian genomes that results in long-term transgene expression. In this study, gene transfer was achieved by intramuscular injection of VEGF and integrase plasmid DNAs into the tibialis anterior muscle in the mouse hindlimb, followed by electroporation of the muscle with needle electrodes. We observed VEGF levels significantly above background 40 days after injection in animals that received phiC31 integrase and the VEGF plasmid. Site-specific integration of plasmid DNA in the chromosomes of muscle tissue was verified by polymerase chain reaction at a common integration site. These results suggest the possible utility of the phiC31 integrase system to treat ischemic disease.


Subject(s)
Bacteriophages , Hindlimb/blood supply , Integrases , Ischemia/therapy , Plasmids , Vascular Endothelial Growth Factor A , Viral Proteins , Animals , Bacteriophages/enzymology , Bacteriophages/genetics , Electroporation/methods , Female , Genetic Therapy/methods , Integrases/genetics , Ischemia/genetics , Mice , Recombination, Genetic/genetics , Time Factors , Vascular Endothelial Growth Factor A/genetics , Viral Proteins/genetics
18.
Proc Natl Acad Sci U S A ; 103(2): 419-24, 2006 Jan 10.
Article in English | MEDLINE | ID: mdl-16387861

ABSTRACT

Plasmid-mediated gene therapy can restore dystrophin expression in skeletal muscle in the mdx mouse, a model of Duchenne muscular dystrophy. However, sufficient long-term expression and distribution of dystrophin remain a hurdle for translating this technology into a viable treatment for Duchenne muscular dystrophy. To improve plasmid-mediated gene therapy for muscle diseases, we studied the effects of targeted plasmid integration using a phage integrase (phiC31) that can mediate the integration of suitably modified plasmids into the mammalian genome. Using a luciferase expression plasmid, we monitored plasmid gene expression noninvasively in living mice by bioluminescence imaging. Coinjection of an integrase plasmid resulted in 5- to 10-fold higher levels of sustained luciferase expression. Likewise, plasmid-mediated dystrophin expression in mdx muscle was enhanced by integration. Using a combination of dystrophin and luciferase plasmids, we analyzed the functional benefit of dystrophin expression in the dystrophic muscle. The expression of dystrophin slowed the loss of luciferase expression associated with muscle degeneration, and that protection was enhanced by targeted integration of the dystrophin plasmid. In the presence of integrase, dystrophin expression was distributed along a much greater length of individual fibers, and this was associated with increased protection against degenerative changes. These data demonstrate the importance of both the level and distribution of dystrophin expression to achieve therapeutic efficacy, and that the efficacy can be enhanced by targeted plasmid integration.


Subject(s)
Gene Targeting , Genetic Therapy/methods , Muscular Dystrophies/genetics , Muscular Dystrophies/therapy , Plasmids/genetics , Plasmids/metabolism , Recombination, Genetic/genetics , Animals , DNA/genetics , Dystrophin/biosynthesis , Dystrophin/genetics , Dystrophin/metabolism , Gene Expression Regulation , Integrases/genetics , Integrases/metabolism , Mice , Mice, Inbred C57BL , Muscular Dystrophies/metabolism , Time Factors
19.
Hum Mol Genet ; 14(2): 221-33, 2005 Jan 15.
Article in English | MEDLINE | ID: mdl-15563511

ABSTRACT

Defects in the dystrophin gene cause the severe degenerative muscle disorder, Duchenne muscular dystrophy (DMD). Among the gene therapy approaches to DMD under investigation, a gene editing approach using oligonucleotide vectors has yielded encouraging results. Here, we extend our studies of gene editing with self-pairing, chimeric RNA/DNA oligonucleotides (RDOs) to the use of oligodeoxynucleotides (ODNs) to correct point mutations in the dystrophin gene. The ODN vectors offer many advantages over the RDO vectors, and we compare the targeting efficiencies in the mdx(5cv) mouse model of DMD. We found that ODNs targeted to either the transcribed or the non-transcribed strand of the dystrophin gene were capable of inducing gene repair, with efficiencies comparable to that seen with RDO vectors. Oligonucleotide-mediated repair was demonstrated at the genomic, mRNA and protein levels in muscle cells both in vitro and in vivo, and the correction was stable over time. Interestingly, there was a strand bias observed with the ODNs, with more efficient correction of the non-transcribed strand even though the dystrophin gene is not transcribed in proliferating myoblasts. This finding demonstrates that strand bias of ODN-mediated gene repair is likely to be due to the specific sequence of the target gene in addition to any effects of transcription. A better understanding of how the efficiency of gene editing relates to the target sequence will offer the opportunity for rational oligonucleotide design for further development of this elegant approach to gene therapy for DMD and other genetic diseases.


Subject(s)
Dystrophin/genetics , Muscle Cells/metabolism , Muscular Dystrophy, Duchenne/genetics , Animals , Base Sequence , Dystrophin/metabolism , Gene Transfer Techniques , In Situ Hybridization, Fluorescence , Mice , Mice, Inbred mdx , Molecular Sequence Data , Muscular Dystrophy, Duchenne/metabolism , Mutation , RNA, Messenger/metabolism , Sequence Analysis, DNA
20.
Hum Mol Genet ; 12(10): 1087-99, 2003 May 15.
Article in English | MEDLINE | ID: mdl-12719373

ABSTRACT

The most common types of dystrophin gene mutations that cause Duchenne muscular dystrophy (DMD) are large deletions that result in a shift of the translational reading frame. Such mutations generally lead to a complete absence of dystrophin protein in the muscle cells of affected individuals. Any therapeutic modality that could restore the reading frame would have the potential to substantially reduce the severity of the disease by allowing the production of an internally deleted, but partially functional, dystrophin protein as occurs in Becker muscular dystrophy (BMD). One approach to restoring the reading frame would be to alter the splicing of the pre-mRNA to produce an in-frame transcript. We have tested the ability of chimeric RNA/DNA oligonucleotides (chimeraplasts) to alter key bases in specific splice sequences in the dystrophin gene to induce exon skipping. Using cells from the mdx mouse as a model system, we show that chimeraplast-mediated base conversion in the intron 22/exon 23 splice junction induces alternative splicing and the production of in-frame transcripts. Interestingly, multiple alternative transcripts were induced by this targeted splice site mutation. Direct sequencing indicated that several of these were predicted to produce in-frame dystrophin transcripts with internal deletions. Indeed, multiple forms of dystrophin protein were observed by western blot analysis, and the functionality of the products was demonstrated by the restoration of expression and localization of a dystrophin-associated protein, alpha-dystroglycan, in differentiated cells. These data demonstrate that chimeraplasts can induce exon skipping by altering splice site sequences at the genomic level. As such, chimeraplast-mediated exon skipping has the potential to be used to transform a severe DMD phenotype into a much milder BMD phenotype.


Subject(s)
Dystrophin/genetics , Exons/genetics , Muscle Cells/metabolism , Oligoribonucleotides/genetics , Alternative Splicing , Animals , Base Sequence , Cytoskeletal Proteins/biosynthesis , Cytoskeletal Proteins/genetics , Dystroglycans , Dystrophin/biosynthesis , Gene Expression Regulation , Membrane Glycoproteins/biosynthesis , Membrane Glycoproteins/genetics , Mice , Mice, Inbred mdx , Molecular Sequence Data , Muscular Dystrophy, Duchenne/genetics , Muscular Dystrophy, Duchenne/therapy , Oligodeoxyribonucleotides/metabolism , Oligoribonucleotides/metabolism , Oligoribonucleotides/therapeutic use , Point Mutation
SELECTION OF CITATIONS
SEARCH DETAIL
...