Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Mol Ther Nucleic Acids ; 10: 376-386, 2018 Mar 02.
Article in English | MEDLINE | ID: mdl-29499949

ABSTRACT

We assessed the potential of Lmna-mRNA repair by spliceosome-mediated RNA trans-splicing as a therapeutic approach for LMNA-related congenital muscular dystrophy. This gene therapy strategy leads to reduction of mutated transcript expression for the benefit of corresponding wild-type (WT) transcripts. We developed 5'-RNA pre-trans-splicing molecules containing the first five exons of Lmna and targeting intron 5 of Lmna pre-mRNA. Among nine pre-trans-splicing molecules, differing in the targeted sequence in intron 5 and tested in C2C12 myoblasts, three induced trans-splicing events on endogenous Lmna mRNA and confirmed at protein level. Further analyses performed in primary myotubes derived from an LMNA-related congenital muscular dystrophy (L-CMD) mouse model led to a partial rescue of the mutant phenotype. Finally, we tested this approach in vivo using adeno-associated virus (AAV) delivery in newborn mice and showed that trans-splicing events occurred in WT mice 50 days after AAV delivery, although at a low rate. Altogether, while these results provide the first evidence for reprogramming LMNA mRNA in vitro, strategies to improve the rate of trans-splicing events still need to be developed for efficient application of this therapeutic approach in vivo.

2.
Dis Model Mech ; 10(4): 487-497, 2017 04 01.
Article in English | MEDLINE | ID: mdl-28188264

ABSTRACT

Myotonic dystrophy type 1 (DM1) and type 2 (DM2) are autosomal dominant neuromuscular diseases caused by microsatellite expansions and belong to the family of RNA-dominant disorders. Availability of cellular models in which the DM mutation is expressed within its natural context is essential to facilitate efforts to identify new therapeutic compounds. Here, we generated immortalized DM1 and DM2 human muscle cell lines that display nuclear RNA aggregates of expanded repeats, a hallmark of myotonic dystrophy. Selected clones of DM1 and DM2 immortalized myoblasts behave as parental primary myoblasts with a reduced fusion capacity of immortalized DM1 myoblasts when compared with control and DM2 cells. Alternative splicing defects were observed in differentiated DM1 muscle cell lines, but not in DM2 lines. Splicing alterations did not result from differentiation delay because similar changes were found in immortalized DM1 transdifferentiated fibroblasts in which myogenic differentiation has been forced by overexpression of MYOD1. As a proof-of-concept, we show that antisense approaches alleviate disease-associated defects, and an RNA-seq analysis confirmed that the vast majority of mis-spliced events in immortalized DM1 muscle cells were affected by antisense treatment, with half of them significantly rescued in treated DM1 cells. Immortalized DM1 muscle cell lines displaying characteristic disease-associated molecular features such as nuclear RNA aggregates and splicing defects can be used as robust readouts for the screening of therapeutic compounds. Therefore, immortalized DM1 and DM2 muscle cell lines represent new models and tools to investigate molecular pathophysiological mechanisms and evaluate the in vitro effects of compounds on RNA toxicity associated with myotonic dystrophy mutations.


Subject(s)
Drug Evaluation, Preclinical , Muscle, Skeletal/pathology , Myotonic Dystrophy/drug therapy , Myotonic Dystrophy/pathology , Adult , Alternative Splicing/drug effects , Alternative Splicing/genetics , Cell Line, Transformed , Child , Female , Fibroblasts/drug effects , Fibroblasts/metabolism , Fibroblasts/pathology , Humans , Male , Middle Aged , Muscle Fibers, Skeletal/drug effects , Muscle Fibers, Skeletal/metabolism , Muscle Fibers, Skeletal/pathology , MyoD Protein/metabolism , Oligonucleotides, Antisense/pharmacology , Oligonucleotides, Antisense/therapeutic use , RNA/metabolism
3.
Mol Ther Nucleic Acids ; 5(9): e362, 2016 Sep 13.
Article in English | MEDLINE | ID: mdl-27623444

ABSTRACT

Dynamin 2 (DNM2) is a large GTPase, ubiquitously expressed, involved in membrane trafficking and regulation of actin and microtubule cytoskeletons. DNM2 mutations cause autosomal dominant centronuclear myopathy which is a rare congenital myopathy characterized by skeletal muscle weakness and histopathological features including nuclear centralization in absence of regeneration. No curative treatment is currently available for the DNM2-related autosomal dominant centronuclear myopathy. In order to develop therapeutic strategy, we evaluated here the potential of Spliceosome-Mediated RNA Trans-splicing technology to reprogram the Dnm2-mRNA in vitro and in vivo in mice. We show that classical 3'-trans-splicing strategy cannot be considered as accurate therapeutic strategy regarding toxicity of the pre-trans-splicing molecules leading to low rate of trans-splicing in vivo. Thus, we tested alternative strategies devoted to prevent this toxicity and enhance frequency of trans-splicing events. We succeeded to overcome the toxicity through a 5'-trans-splicing strategy which also allows detection of trans-splicing events at mRNA and protein levels in vitro and in vivo. These results suggest that the Spliceosome-Mediated RNA Trans-splicing strategy may be used to reprogram mutated Dnm2-mRNA but highlight the potential toxicity linked to the molecular tools which have to be carefully investigated during preclinical development.

4.
Nat Commun ; 7: 11067, 2016 Apr 11.
Article in English | MEDLINE | ID: mdl-27063795

ABSTRACT

Myotonic dystrophy (DM) is caused by the expression of mutant RNAs containing expanded CUG repeats that sequester muscleblind-like (MBNL) proteins, leading to alternative splicing changes. Cardiac alterations, characterized by conduction delays and arrhythmia, are the second most common cause of death in DM. Using RNA sequencing, here we identify novel splicing alterations in DM heart samples, including a switch from adult exon 6B towards fetal exon 6A in the cardiac sodium channel, SCN5A. We find that MBNL1 regulates alternative splicing of SCN5A mRNA and that the splicing variant of SCN5A produced in DM presents a reduced excitability compared with the control adult isoform. Importantly, reproducing splicing alteration of Scn5a in mice is sufficient to promote heart arrhythmia and cardiac-conduction delay, two predominant features of myotonic dystrophy. In conclusion, misregulation of the alternative splicing of SCN5A may contribute to a subset of the cardiac dysfunctions observed in myotonic dystrophy.


Subject(s)
Alternative Splicing/genetics , Arrhythmias, Cardiac/complications , Arrhythmias, Cardiac/genetics , Heart Conduction System/physiopathology , Myotonic Dystrophy/complications , Myotonic Dystrophy/genetics , NAV1.5 Voltage-Gated Sodium Channel/genetics , Adult , Aged , Animals , Base Sequence , Binding Sites , Computer Simulation , Electrophysiological Phenomena , Exons/genetics , Female , HEK293 Cells , Heart Conduction System/pathology , Humans , Male , Middle Aged , Molecular Sequence Data , NAV1.5 Voltage-Gated Sodium Channel/metabolism , Nucleotide Motifs/genetics , RNA-Binding Proteins/metabolism , Sodium Channels/metabolism , Xenopus
5.
Nat Commun ; 6: 7205, 2015 May 28.
Article in English | MEDLINE | ID: mdl-26018658

ABSTRACT

Myotonic Dystrophy type 1 (DM1) is a dominant neuromuscular disease caused by nuclear-retained RNAs containing expanded CUG repeats. These toxic RNAs alter the activities of RNA splicing factors resulting in alternative splicing misregulation and muscular dysfunction. Here we show that the abnormal splicing of DMD exon 78 found in dystrophic muscles of DM1 patients is due to the functional loss of MBNL1 and leads to the re-expression of an embryonic dystrophin in place of the adult isoform. Forced expression of embryonic dystrophin in zebrafish using an exon-skipping approach severely impairs the mobility and muscle architecture. Moreover, reproducing Dmd exon 78 missplicing switch in mice induces muscle fibre remodelling and ultrastructural abnormalities including ringed fibres, sarcoplasmic masses or Z-band disorganization, which are characteristic features of dystrophic DM1 skeletal muscles. Thus, we propose that splicing misregulation of DMD exon 78 compromises muscle fibre maintenance and contributes to the progressive dystrophic process in DM1.


Subject(s)
Dystrophin/genetics , Gene Expression Regulation, Developmental , Membrane Proteins/genetics , Muscle Fibers, Skeletal/metabolism , Muscle Proteins/genetics , Myotonic Dystrophy/genetics , RNA Splicing/genetics , RNA-Binding Proteins/genetics , Zebrafish Proteins/genetics , Animals , Chromatography, Liquid , Dystrophin/metabolism , Exons , Homeostasis , Humans , Immunohistochemistry , Immunoprecipitation , Membrane Proteins/metabolism , Mice , Microscopy, Electron , Muscle Fibers, Skeletal/ultrastructure , Muscle Proteins/metabolism , Myotonic Dystrophy/pathology , Real-Time Polymerase Chain Reaction , Sarcoplasmic Reticulum/ultrastructure , Tandem Mass Spectrometry , Zebrafish Proteins/metabolism
6.
J Allergy Clin Immunol ; 133(4): 1116-23, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24332219

ABSTRACT

BACKGROUND: Recombination-activating gene 1 (RAG1) deficiency results in severe combined immunodeficiency (SCID) caused by a complete lack of T and B lymphocytes. If untreated, patients succumb to recurrent infections. OBJECTIVES: We sought to develop lentiviral gene therapy for RAG1-induced SCID and to test its safety. METHODS: Constructs containing the viral spleen-focus-forming virus (SF), ubiquitous promoters, or cell type-restricted promoters driving sequence-optimized RAG1 were compared for efficacy and safety in sublethally preconditioned Rag1(-/-) mice undergoing transplantation with transduced bone marrow progenitors. RESULTS: Peripheral blood CD3(+) T-cell reconstitution was achieved with SF, ubiquitous promoters, and cell type-restricted promoters but 3- to 18-fold lower than that seen in wild-type mice, and with a compromised CD4(+)/CD8(+) ratio. Mitogen-mediated T-cell responses and T cell-dependent and T cell-independent B-cell responses were not restored, and T-cell receptor patterns were skewed. Reconstitution of mature peripheral blood B cells was approximately 20-fold less for the SF vector than in wild-type mice and often not detectable with the other promoters, and plasma immunoglobulin levels were abnormal. Two months after transplantation, gene therapy-treated mice had rashes with cellular tissue infiltrates, activated peripheral blood CD44(+)CD69(+) T cells, high plasma IgE levels, antibodies against double-stranded DNA, and increased B cell-activating factor levels. Only rather high SF vector copy numbers could boost T- and B-cell reconstitution, but mRNA expression levels during T- and B-cell progenitor stages consistently remained less than wild-type levels. CONCLUSIONS: These results underline that further development is required for improved expression to successfully treat patients with RAG1-induced SCID while maintaining low vector copy numbers and minimizing potential risks, including autoimmune reactions resembling Omenn syndrome.


Subject(s)
Genetic Therapy , Genetic Vectors/genetics , Homeodomain Proteins/genetics , Lentivirus/genetics , Severe Combined Immunodeficiency/genetics , Severe Combined Immunodeficiency/therapy , Animals , Autoimmunity/genetics , Bone Marrow Cells/metabolism , Disease Models, Animal , Female , Gene Dosage , Gene Expression , Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells/metabolism , Humans , Immunophenotyping , Male , Mice , Mice, Knockout , Phenotype , Severe Combined Immunodeficiency/immunology , Spleen/immunology , T-Lymphocytes/metabolism , Thymus Gland/immunology , Transduction, Genetic , Transplantation Chimera
7.
Nucleic Acids Res ; 41(17): 8391-402, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23861443

ABSTRACT

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


Subject(s)
Dystrophin/genetics , Trans-Splicing , Animals , Dependovirus/genetics , Dystrophin/analysis , Exons , Genetic Vectors , Genotype , Humans , Introns , Mice , Mice, Inbred mdx , Muscle Fibers, Skeletal/chemistry , Muscles/chemistry , Muscular Dystrophy, Duchenne/genetics , NIH 3T3 Cells , RNA Splice Sites , RNA, Messenger/analysis
8.
Nat Struct Mol Biol ; 18(1): 85-7, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21186365

ABSTRACT

We describe a function for modified human U7 small nuclear RNAs (hU7-snRNAs) distinct from modification of pre-mRNA splicing events. Engineered hU7-snRNAs containing a poly-CAG antisense sequence targeting the expanded CUG repeats of mutant DMPK transcripts in myotonic dystrophy caused specific degradation of pathogenic DMPK mRNAs without affecting the products of wild-type DMPK alleles. Abolition of the RNA gain-of-function toxicity that is responsible for pathogenesis supports the use of hU7-snRNAs for gene silencing in RNA-dominant disorders in which expanded repeats are expressed.


Subject(s)
Gene Silencing , Protein Serine-Threonine Kinases/genetics , RNA, Small Nuclear/physiology , Alternative Splicing , Cells, Cultured , DNA Repeat Expansion , Genetic Engineering , Humans , Myotonic Dystrophy/genetics , Myotonin-Protein Kinase , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Small Nuclear/chemistry , RNA, Small Nuclear/metabolism
9.
Drug Deliv ; 11(6): 351-63, 2004.
Article in English | MEDLINE | ID: mdl-15736830

ABSTRACT

Nonviral gene delivery systems are a promising approach for gene therapy applications, despite their low in vivo gene transfer efficiency. One approach to enhance this efficiency is to incorporate targeting elements into cationic lipid/DNA complexes (lipoplexes). Ligand-containing lipoplexes have to retain their efficiency while exposing accessible ligand on their surface. Physicochemical properties (particle size, surface charge, and efficacy of DNA complexation) of the lipoplexes largely determine their gene transfer efficiency. We synthesized glycolipids with various galactosylated head ligand and incorporated them into lipoplexes. We showed that incorporation of up to 33% mol of glycolipid did not change the physicochemical properties of lipoplexes. Some of our glycolipids yielded lipoplexes whose galactosyl heads were well exposed on the surface as demonstrated by a strong interaction with Ricinus communis agglutinin. Glycolipid-containing lipoplexes gave an efficient gene transfer on hepatocytes, although no ligand-targeted transfection could be observed.


Subject(s)
Gene Targeting/methods , Glycolipids/chemical synthesis , Lectins/chemical synthesis , Cell Line, Tumor , Chemistry, Pharmaceutical , Gene Transfer Techniques , Glycolipids/administration & dosage , Glycolipids/genetics , Humans , Lectins/administration & dosage , Lectins/genetics , Liposomes
SELECTION OF CITATIONS
SEARCH DETAIL
...