37 results on '"Peccate, C"'
Search Results
2. MicroRNAs involved in nNOS regulation in dystrophic context
- Author
-
Guilbaud, M., primary, Gentil, C., additional, Holtzmann, I., additional, Gruszczynski, C., additional, Falcone, S., additional, Peccate, C., additional, Benkhelifa-Ziyyat, S., additional, Lorain, S., additional, Aurade, F., additional, Jeanson-Leh, L., additional, and Piétri-Rouxel, F., additional
- Published
- 2017
- Full Text
- View/download PDF
3. DMD TREATMENT: P.334PPMO pre-treatment is beneficial for AAV-based gene therapy in Duchenne muscular dystrophy
- Author
-
Forand, A., Muchir, A., Mougenot, N., Sevoz-Couche, C., Peccate, C., Lemaitre, M., Wood, M., Lorain, S., and Piétri-Rouxel, F.
- Published
- 2019
- Full Text
- View/download PDF
4. P.237 - MicroRNAs involved in nNOS regulation in dystrophic context
- Author
-
Guilbaud, M., Gentil, C., Holtzmann, I., Gruszczynski, C., Falcone, S., Peccate, C., Benkhelifa-Ziyyat, S., Lorain, S., Aurade, F., Jeanson-Leh, L., and Piétri-Rouxel, F.
- Published
- 2017
- Full Text
- View/download PDF
5. Expression clinique des porteuses symptomatiques de mutations du gè ; ne EMD. A propos de 4 cas
- Author
-
Ben Yaou, R, Llense, S, Deburgrave, N, Peccate, C, Canki-Klain, Nina, Amsallem, D : Journel, H, Laforet, P, Kaplan, JC, Leturcq, F, and Recan, D
- Subjects
clinique ,EMD gene ,mutation ,X-EDMD - Abstract
Objectifs : Les mutations du gè ; ne EMD, codant l’ émerine, sont responsables de la forme liée à ; l’ X de la Dystrophie musculaire d’ Emery-Dreifuss (X-EDMD), caractérisée par une faiblesse et amyotrophie huméro-péroniè ; res, des rétractions tendineuses et une cardiopathie dominée par des troubles de la conduction auriculo-ventriculaire avec risque accru de mort subite. Les femmes transmettrices sont le plus souvent asymptomatiques. Nous rapportons ici 4 femmes porteuses d'une mutation du gè ; ne EMD et présentant soit une atteinte cardiaque ou musculaire pures, soit un phénotype EDMD complet. Méthodes et résultats : Quatre femmes, mè ; res de patients EDMD, ont été cliniquement analysées sur les plans neurologique et cardiologique. L’ émerine a été quantifiée sur lymphoblastes par Western Blot et le gè ; ne EMD analysé par Southern blot/PCR-Séquençage. L’ inactivation du chromosome X a été testée par l’ analyse de la méthylation au locus du récepteur aux androgè ; nes. Deux patientes avaient une cardiopathie pure, la troisiè ; me une myopathie des ceintures isolée et la derniè ; re un EDMD complet associé à ; une hémiplégie gauche probablement d'origine thromboembolique. Chez cette patiente, l'émerine est trè ; s diminuée et l’ inactivation du chromosome X déséquilibrée. L'analyse du gè ; ne EMD a identifié quatre mutations nulles à ; l'état hétérozygote (un stop, une insertion et deux délétions). Conclusion : La transmettrices de mutations EMD expriment une symptomatologie variable non corrélée à ; la génétique, mais elle les expose au mê ; me risque cardiologique que les sujets masculins hémizygotes. Leur suivi cardiologique semble nécessaire pour détecter et prévenir les complications cardiaques.
- Published
- 2004
6. P.20.8 AAV genome loss from dystrophic mouse muscles during AAV-U7snRNA-mediated exon skipping therapy
- Author
-
Hir, M. Le, primary, Goyenvalle, A., additional, Peccate, C., additional, Précigout, G., additional, Davies, K.E., additional, Voit, T., additional, Garcia, L., additional, and Lorain, S., additional
- Published
- 2013
- Full Text
- View/download PDF
7. O.14 Exon exchange approach to repair Duchenne dystrophin transcripts
- Author
-
Lorain, S., primary, Peccate, C., additional, Le Hir, M., additional, Griffith, G., additional, Voit, T., additional, and Garcia, L., additional
- Published
- 2010
- Full Text
- View/download PDF
8. T.P.1.05 Trans-splicing approaches to repair Duchenne dystrophin transcripts
- Author
-
Lorain, S., primary, Peccate, C., additional, Griffith, G., additional, Voit, T., additional, and Garcia, L., additional
- Published
- 2009
- Full Text
- View/download PDF
9. Multitissular involvement in a family with LMNA and EMD mutations
- Author
-
Yaou, R. Ben, primary, Toutain, A., additional, Arimura, T., additional, Demay, L., additional, Massart, C., additional, Peccate, C., additional, Muchir, A., additional, Llense, S., additional, Deburgrave, N., additional, Leturcq, F., additional, Litim, K. E., additional, Rahmoun-Chiali, N., additional, Richard, P., additional, Babuty, D., additional, Récan-Budiartha, D., additional, and Bonne, G., additional
- Published
- 2007
- Full Text
- View/download PDF
10. Multitissular involvement in a family with LMNAand EMDmutations
- Author
-
Yaou, R Ben, Toutain, A, Arimura, T, Demay, L, Massart, C, Peccate, C, Muchir, A, Llense, S, Deburgrave, N, Leturcq, F, Litim, K E., Rahmoun-Chiali, N, Richard, P, Babuty, D, Récan-Budiartha, D, and Bonne, G
- Abstract
Mutations in the EMDand LMNAgenes, encoding emerin and lamins A and C, are responsible for the X-linked and autosomal dominant and recessive forms of Emery–Dreifuss muscular dystrophy (EDMD). LMNAmutations can also lead to several other disorders, collectively termed laminopathies, involving heart, fat, nerve, bone, and skin tissues, and some premature ageing syndromes.
- Published
- 2007
- Full Text
- View/download PDF
11. Heterogeneous clinical expression of LMNA and EMD mutations segregating in a single family. Pathophysiologic implications for nuclear envelope related disorders
- Author
-
Rabah BEN YAOU, Toutain, A., Babuty, D., Demay, L., Peccate, C., Muchir, A., Arimura, T., Massart, C., Deburgrave, N., Leturcq, F., Litim, K., Rahmouni-Chiali, N., Richard, P., Recan, D., and Bonne, G.
12. Inhibition of poly(ADP-Ribosyl)ation reduced vascular smooth muscle cells loss and improves aortic disease in a mouse model of human accelerated aging syndrome.
- Author
-
Cardoso D, Guilbert S, Guigue P, Carabalona A, Harhouri K, Peccate C, Tournois J, Guesmia Z, Ferreira L, Bartoli C, Levy N, Colleaux L, Nissan X, and Muchir A
- Subjects
- Animals, Mice, Humans, Aorta pathology, Aorta drug effects, Aorta metabolism, Poly ADP Ribosylation, Mice, Inbred C57BL, Poly(ADP-ribose) Polymerase Inhibitors pharmacology, Muscle, Smooth, Vascular metabolism, Muscle, Smooth, Vascular pathology, Muscle, Smooth, Vascular drug effects, Disease Models, Animal, Progeria pathology, Progeria genetics, Progeria metabolism, Myocytes, Smooth Muscle metabolism, Myocytes, Smooth Muscle drug effects, Myocytes, Smooth Muscle pathology, Lamin Type A metabolism, Lamin Type A genetics
- Abstract
Hutchinson-Gilford progeria syndrome (HGPS) is an extremely rare genetic disorder associated with features of accelerated aging. HGPS is an autosomal dominant disease caused by a de novo mutation of LMNA gene, encoding A-type lamins, resulting in the truncated form of pre-lamin A called progerin. While asymptomatic at birth, patients develop symptoms within the first year of life when they begin to display accelerated aging and suffer from growth retardation, and severe cardiovascular complications including loss of vascular smooth muscle cells (VSMCs). Recent works reported the loss of VSMCs as a major factor triggering atherosclerosis in HGPS. Here, we investigated the mechanisms by which progerin expression leads to massive VSMCs loss. Using aorta tissue and primary cultures of murine VSMCs from a mouse model of HGPS, we showed increased VSMCs death associated with increased poly(ADP-Ribosyl)ation. Poly(ADP-Ribosyl)ation is recognized as a post-translational protein modification that coordinates the repair at DNA damage sites. Poly-ADP-ribose polymerase (PARP) catalyzes protein poly(ADP-Ribosyl)ation by utilizing nicotinamide adenine dinucleotide (NAD
+ ). Our results provided the first demonstration linking progerin accumulation, augmented poly(ADP-Ribosyl)ation and decreased nicotinamide adenine dinucleotide (NAD+ ) level in VSMCs. Using high-throughput screening on VSMCs differentiated from iPSCs from HGPS patients, we identified a new compound, trifluridine able to increase NAD+ levels through decrease of PARP-1 activity. Lastly, we demonstrate that trifluridine treatment in vivo was able to alleviate aortic VSMCs loss and clinical sign of progeria, suggesting a novel therapeutic approach of cardiovascular disease in progeria., (© 2024. The Author(s).)- Published
- 2024
- Full Text
- View/download PDF
13. Setdb1 protects genome integrity in murine muscle stem cells to allow for regenerative myogenesis and inflammation.
- Author
-
Garcia P, Jarassier W, Brun C, Giordani L, Agostini F, Kung WH, Peccate C, Ravent J, Fall S, Petit V, Cheung TH, Ait-Si-Ali S, and Le Grand F
- Subjects
- Animals, Mice, Mice, Inbred C57BL, Muscle, Skeletal metabolism, Muscle, Skeletal pathology, Endogenous Retroviruses genetics, Stem Cells metabolism, Stem Cells cytology, Genome, Cell Differentiation genetics, Histone-Lysine N-Methyltransferase metabolism, Histone-Lysine N-Methyltransferase genetics, Inflammation pathology, Inflammation metabolism, Inflammation genetics, Regeneration genetics, Muscle Development genetics
- Abstract
The histone H3 lysine 9 methyltransferase SETDB1 controls transcriptional repression to direct stem cell fate. Here, we show that Setdb1 expression by adult muscle stem cells (MuSCs) is required for skeletal muscle regeneration. We find that SETDB1 represses the expression of endogenous retroviruses (ERVs) in MuSCs. ERV de-repression in Setdb1-null MuSCs prevents their amplification following exit from quiescence and promotes cell death. Multi-omics profiling shows that chromatin decompaction at ERV loci activates the DNA-sensing cGAS-STING pathway, entailing cytokine expression by Setdb1-null MuSCs. This is followed by aberrant infiltration of inflammatory cells, including pathological macrophages. The ensuing histiocytosis is accompanied by myofiber necrosis, which, in addition to progressive MuSCs depletion, completely abolishes tissue repair. In contrast, loss of Setdb1 in fibro-adipogenic progenitors (FAPs) does not impact immune cells. In conclusion, genome maintenance by SETDB1 in an adult somatic stem cell is necessary for both its regenerative potential and adequate reparative inflammation., Competing Interests: Declaration of interests The authors declare no competing interests., (Copyright © 2024 Elsevier Inc. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
14. Actin-microtubule cytoskeletal interplay mediated by MRTF-A/SRF signaling promotes dilated cardiomyopathy caused by LMNA mutations.
- Author
-
Le Dour C, Chatzifrangkeskou M, Macquart C, Magiera MM, Peccate C, Jouve C, Virtanen L, Heliö T, Aalto-Setälä K, Crasto S, Cadot B, Cardoso D, Mougenot N, Adesse D, Di Pasquale E, Hulot JS, Taimen P, Janke C, and Muchir A
- Subjects
- Male, Mice, Animals, Actins metabolism, Connexin 43 genetics, Tubulin genetics, Serum Response Factor genetics, Lamin Type A genetics, Lamin Type A metabolism, Microtubules metabolism, Myocytes, Cardiac metabolism, Mice, Knockout, Intermediate Filament Proteins genetics, Mutation, Actin Depolymerizing Factors genetics, Cardiomyopathy, Dilated metabolism
- Abstract
Mutations in the lamin A/C gene (LMNA) cause dilated cardiomyopathy associated with increased activity of ERK1/2 in the heart. We recently showed that ERK1/2 phosphorylates cofilin-1 on threonine 25 (phospho(T25)-cofilin-1) that in turn disassembles the actin cytoskeleton. Here, we show that in muscle cells carrying a cardiomyopathy-causing LMNA mutation, phospho(T25)-cofilin-1 binds to myocardin-related transcription factor A (MRTF-A) in the cytoplasm, thus preventing the stimulation of serum response factor (SRF) in the nucleus. Inhibiting the MRTF-A/SRF axis leads to decreased α-tubulin acetylation by reducing the expression of ATAT1 gene encoding α-tubulin acetyltransferase 1. Hence, tubulin acetylation is decreased in cardiomyocytes derived from male patients with LMNA mutations and in heart and isolated cardiomyocytes from Lmna
p.H222P/H222P male mice. In Atat1 knockout mice, deficient for acetylated α-tubulin, we observe left ventricular dilation and mislocalization of Connexin 43 (Cx43) in heart. Increasing α-tubulin acetylation levels in Lmnap.H222P/H222P mice with tubastatin A treatment restores the proper localization of Cx43 and improves cardiac function. In summary, we show for the first time an actin-microtubule cytoskeletal interplay mediated by cofilin-1 and MRTF-A/SRF, promoting the dilated cardiomyopathy caused by LMNA mutations. Our findings suggest that modulating α-tubulin acetylation levels is a feasible strategy for improving cardiac function., (© 2022. The Author(s).)- Published
- 2022
- Full Text
- View/download PDF
15. Muscle regeneration affects Adeno Associated Virus 1 mediated transgene transcription.
- Author
-
Mollard A, Peccate C, Forand A, Chassagne J, Julien L, Meunier P, Guesmia Z, Marais T, Bitoun M, Piétri-Rouxel F, Benkhelifa-Ziyyat S, and Lorain S
- Subjects
- Animals, Cardiotoxins pharmacology, Dependovirus genetics, Dependovirus metabolism, Dystrophin genetics, Dystrophin metabolism, Genetic Therapy, Genetic Vectors genetics, Mice, Mice, Inbred mdx, Muscle, Skeletal metabolism, Regeneration genetics, Transgenes, Muscular Dystrophy, Animal genetics, Muscular Dystrophy, Duchenne genetics, Muscular Dystrophy, Duchenne metabolism, Muscular Dystrophy, Duchenne therapy
- Abstract
Duchenne muscular dystrophy is a severe neuromuscular disease causing a progressive muscle wasting due to mutations in the DMD gene that lead to the absence of dystrophin protein. Adeno-associated virus (AAV)-based therapies aiming to restore dystrophin in muscles, by either exon skipping or microdystrophin expression, are very promising. However, the absence of dystrophin induces cellular perturbations that hinder AAV therapy efficiency. We focused here on the impact of the necrosis-regeneration process leading to nuclear centralization in myofiber, a common feature of human myopathies, on AAV transduction efficiency. We generated centronucleated myofibers by cardiotoxin injection in wild-type muscles prior to AAV injection. Intramuscular injections of AAV1 vectors show that transgene expression was drastically reduced in regenerated muscles, even when the AAV injection occurred 10 months post-regeneration. We show also that AAV genomes were not lost from cardiotoxin regenerated muscle and were properly localised in the myofiber nuclei but were less transcribed leading to muscle transduction defect. A similar defect was observed in muscles of the DMD mouse model mdx. Therefore, the regeneration process per se could participate to the AAV-mediated transduction defect observed in dystrophic muscles which may limit AAV-based therapies., (© 2022. The Author(s).)
- Published
- 2022
- Full Text
- View/download PDF
16. The non-muscle ADF/cofilin-1 controls sarcomeric actin filament integrity and force production in striated muscle laminopathies.
- Author
-
Vignier N, Chatzifrangkeskou M, Pinton L, Wioland H, Marais T, Lemaitre M, Le Dour C, Peccate C, Cardoso D, Schmitt A, Wu W, Biferi MG, Naouar N, Macquart C, Beuvin M, Decostre V, Bonne G, Romet-Lemonne G, Worman HJ, Tedesco FS, Jégou A, and Muchir A
- Subjects
- Adolescent, Adult, Animals, Cell Line, Child, Humans, Lamin Type A genetics, Laminopathies genetics, Male, Mice, Mice, Inbred C57BL, Mice, Knockout, Mitogen-Activated Protein Kinase 1 metabolism, Mitogen-Activated Protein Kinase 3 metabolism, Muscle, Striated pathology, Muscular Dystrophy, Emery-Dreifuss genetics, Muscular Dystrophy, Emery-Dreifuss metabolism, Mutation, Phosphorylation, Signal Transduction, Young Adult, Actin Cytoskeleton metabolism, Cofilin 1 metabolism, Destrin metabolism, Lamin Type A metabolism, Laminopathies metabolism, Muscle, Striated metabolism, Sarcomeres metabolism
- Abstract
Cofilins are important for the regulation of the actin cytoskeleton, sarcomere organization, and force production. The role of cofilin-1, the non-muscle-specific isoform, in muscle function remains unclear. Mutations in LMNA encoding A-type lamins, intermediate filament proteins of the nuclear envelope, cause autosomal Emery-Dreifuss muscular dystrophy (EDMD). Here, we report increased cofilin-1 expression in LMNA mutant muscle cells caused by the inability of proteasome degradation, suggesting a protective role by ERK1/2. It is known that phosphorylated ERK1/2 directly binds to and catalyzes phosphorylation of the actin-depolymerizing factor cofilin-1 on Thr25. In vivo ectopic expression of cofilin-1, as well as its phosphorylated form on Thr25, impairs sarcomere structure and force generation. These findings present a mechanism that provides insight into the molecular pathogenesis of muscular dystrophies caused by LMNA mutations., Competing Interests: Declaration of interests H.J.W. is on the scientific advisory board and owns equity in AlloMek Therapeutics. The remaining authors declare no competing interests., (Copyright © 2021 The Author(s). Published by Elsevier Inc. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
17. TGFβ signaling curbs cell fusion and muscle regeneration.
- Author
-
Girardi F, Taleb A, Ebrahimi M, Datye A, Gamage DG, Peccate C, Giordani L, Millay DP, Gilbert PM, Cadot B, and Le Grand F
- Subjects
- Adolescent, Adult, Animals, Blotting, Western, Cell Fusion, Cells, Cultured, Computational Biology, Fibroblasts cytology, Fibroblasts metabolism, Fluorescent Antibody Technique, Humans, In Situ Nick-End Labeling, Male, Mice, Real-Time Polymerase Chain Reaction, Regeneration genetics, Regeneration physiology, Stem Cells cytology, Stem Cells metabolism, Transforming Growth Factor beta genetics, Young Adult, Muscle, Skeletal cytology, Transforming Growth Factor beta metabolism
- Abstract
Muscle cell fusion is a multistep process involving cell migration, adhesion, membrane remodeling and actin-nucleation pathways to generate multinucleated myotubes. However, molecular brakes restraining cell-cell fusion events have remained elusive. Here we show that transforming growth factor beta (TGFβ) pathway is active in adult muscle cells throughout fusion. We find TGFβ signaling reduces cell fusion, regardless of the cells' ability to move and establish cell-cell contacts. In contrast, inhibition of TGFβ signaling enhances cell fusion and promotes branching between myotubes in mouse and human. Exogenous addition of TGFβ protein in vivo during muscle regeneration results in a loss of muscle function while inhibition of TGFβR2 induces the formation of giant myofibers. Transcriptome analyses and functional assays reveal that TGFβ controls the expression of actin-related genes to reduce cell spreading. TGFβ signaling is therefore requisite to limit mammalian myoblast fusion, determining myonuclei numbers and myofiber size.
- Published
- 2021
- Full Text
- View/download PDF
18. Self-assembly/condensation interplay in nano-to-microfibrillar silicified fibrin hydrogels.
- Author
-
Wang K, Albert K, Mosser G, Haye B, Percot A, Paris C, Peccate C, Trichet L, and Coradin T
- Subjects
- Animals, Cell Proliferation drug effects, Circular Dichroism, Kinetics, Mice, Microscopy, Electron, Scanning, Microscopy, Electron, Transmission, Myoblasts drug effects, Myofibroblasts metabolism, Nanoparticles chemistry, Nephelometry and Turbidimetry, Propylamines chemistry, Rheology, Silanes chemistry, Spectroscopy, Fourier Transform Infrared, Fibrin chemistry, Hydrogels chemistry, Silicon Dioxide chemistry, Tissue Engineering methods, Tissue Scaffolds
- Abstract
Fibrin-based gels are used in clinics as biological glues but their application as 3D cellularized scaffolds is hindered by processing and stability issues. Silicification of fibrin networks appears as a promising strategy not only to address these limitations but also to take advantage of the bioactivity of Si. However, it raises the question of the influence of silica sources on fibrin self-assembly. Here tetraethoxysilane, aminopropyltriethoxysilane and silica nanoparticles were used to design hybrid and nanocomposite fibrin-based hydrogels. By varying the concentration in silica source, we could evidence two regimes of interactions that depend on the extent of inorganic condensation. These interactions modulated the fibrillar structure of the fibrin network from more than 500 nm to less than 100 nm. These nanofibrillar hydrogels could exhibit higher mechanical properties than pure fibrin while preserving their capacity to support proliferation of myoblasts, opening promising perspectives for the use of fibrin-silica constructs in tissue engineering., Competing Interests: Declaration of competing interest There are no conflicts to declare., (Copyright © 2020 Elsevier B.V. All rights reserved.)
- Published
- 2020
- Full Text
- View/download PDF
19. X-linked muscular dystrophy in a Labrador Retriever strain: phenotypic and molecular characterisation.
- Author
-
Barthélémy I, Calmels N, Weiss RB, Tiret L, Vulin A, Wein N, Peccate C, Drougard C, Beroud C, Deburgrave N, Thibaud JL, Escriou C, Punzón I, Garcia L, Kaplan JC, Flanigan KM, Leturcq F, and Blot S
- Subjects
- Animals, Dogs, Genes, Modifier, Male, Muscle, Skeletal metabolism, Muscle, Skeletal pathology, Muscular Dystrophy, Duchenne pathology, Mutation, Disease Models, Animal, Dystrophin genetics, Muscular Dystrophy, Duchenne genetics, Phenotype
- Abstract
Background: Canine models of Duchenne muscular dystrophy (DMD) are a valuable tool to evaluate potential therapies because they faithfully reproduce the human disease. Several cases of dystrophinopathies have been described in canines, but the Golden Retriever muscular dystrophy (GRMD) model remains the most used in preclinical studies. Here, we report a new spontaneous dystrophinopathy in a Labrador Retriever strain, named Labrador Retriever muscular dystrophy (LRMD)., Methods: A colony of LRMD dogs was established from spontaneous cases. Fourteen LRMD dogs were followed-up and compared to the GRMD standard using several functional tests. The disease causing mutation was studied by several molecular techniques and identified using RNA-sequencing., Results: The main clinical features of the GRMD disease were found in LRMD dogs; the functional tests provided data roughly overlapping with those measured in GRMD dogs, with similar inter-individual heterogeneity. The LRMD causal mutation was shown to be a 2.2-Mb inversion disrupting the DMD gene within intron 20 and involving the TMEM47 gene. In skeletal muscle, the Dp71 isoform was ectopically expressed, probably as a consequence of the mutation. We found no evidence of polymorphism in either of the two described modifier genes LTBP4 and Jagged1. No differences were found in Pitpna mRNA expression levels that would explain the inter-individual variability., Conclusions: This study provides a full comparative description of a new spontaneous canine model of dystrophinopathy, found to be phenotypically equivalent to the GRMD model. We report a novel large DNA mutation within the DMD gene and provide evidence that LRMD is a relevant model to pinpoint additional DMD modifier genes.
- Published
- 2020
- Full Text
- View/download PDF
20. Combined Treatment with Peptide-Conjugated Phosphorodiamidate Morpholino Oligomer-PPMO and AAV-U7 Rescues the Severe DMD Phenotype in Mice.
- Author
-
Forand A, Muchir A, Mougenot N, Sevoz-Couche C, Peccate C, Lemaitre M, Izabelle C, Wood M, Lorain S, and Piétri-Rouxel F
- Abstract
Duchenne muscular dystrophy (DMD) is a devastating neuromuscular disease caused by an absence of the dystrophin protein, which is essential for muscle fiber integrity. Among the developed therapeutic strategies for DMD, the exon-skipping approach corrects the frameshift and partially restores dystrophin expression. It could be achieved through the use of antisense sequences, such as peptide-conjugated phosphorodiamidate morpholino oligomer (PPMO) or the small nuclear RNA-U7 carried by an adeno-associated virus (AAV) vector. AAV-based gene therapy approaches have potential for use in DMD treatment but are subject to a major limitation: loss of the AAV genome, necessitating readministration of the vector, which is not currently possible, due to the immunogenicity of the capsid. The PPMO approach requires repeated administrations and results in only weak cardiac dystrophin expression. Here, we evaluated a combination of PPMO- and AAV-based therapy in a mouse model of severe DMD. Striking benefits of this combined therapy were observed in striated muscles, with marked improvements in heart and diaphragm structure and function, with unrivalled extent of survival, opening novel therapeutic perspectives for patients., (© 2020 The Authors.)
- Published
- 2020
- Full Text
- View/download PDF
21. Interactions of Organosilanes with Fibrinogen and Their Influence on Muscle Cell Proliferation in 3D Fibrin Hydrogels.
- Author
-
Wang K, Trichet L, Rieu C, Peccate C, Pembouong G, Bouteiller L, and Coradin T
- Subjects
- Animals, Cell Line, Cell Proliferation, Hydrogels adverse effects, Mice, Myoblasts physiology, Protein Denaturation, Fibrin chemistry, Fibrinogen chemistry, Hydrogels chemistry, Myoblasts drug effects, Organosilicon Compounds chemistry
- Abstract
Silanization of biomacromolecules has emerged as a fruitful approach to prepare hybrid biohydrogels. However, very little is known about interactions between organosilanes and biopolymers in solution. Here we focused on fibrin, a protein of interest in the biomedical field, whose self-assembly process and resulting gel structure are highly sensitive to experimental conditions. Three main silanes were selected to decipher the relative influence of the silanol groups and organic functions. Whereas no protein denaturation was observed, silanes bearing hydrophobic groups had a surfactant-like behavior and could improve the dispersion of fibrinogen molecules, impacting gel formation kinetics and rheological properties. 3D cultures of myoblasts evidenced that organosilanes could promote or impede cell proliferation, suggesting interactions of silanols with fibrin. These results demonstrate that the two sides of the coin of organosilane reactivity are relevant at different stages of fibrin gel formation and must be considered for future development of hybrid biomaterials.
- Published
- 2019
- Full Text
- View/download PDF
22. Alteration of performance in a mouse model of Emery-Dreifuss muscular dystrophy caused by A-type lamins gene mutation.
- Author
-
Thomasson R, Vignier N, Peccate C, Mougenot N, Noirez P, and Muchir A
- Subjects
- Animals, Body Composition, Disease Models, Animal, Male, Mice, Mice, Transgenic, Muscular Dystrophy, Emery-Dreifuss metabolism, Muscular Dystrophy, Emery-Dreifuss physiopathology, Mutation, Ventricular Function, Left, Weight Loss, Lamin Type A genetics, Muscular Dystrophy, Emery-Dreifuss genetics
- Abstract
Autosomal Emery-Dreifuss muscular dystrophy (EDMD) is caused by mutations in the lamin A/C gene (LMNA) encoding A-type nuclear lamins, intermediate filament proteins of the nuclear envelope. Classically, the disease manifests as scapulo-humero-peroneal muscle wasting and weakness, early joint contractures and dilated cardiomyopathy with conduction blocks; however, variable skeletal muscle involvement can be present. Previously, we and other demonstrated altered activity of signaling pathways in hearts and striated muscles of LmnaH222P/H222P mice, a model of autosomal EDMD. We showed that blocking their activation improved cardiac function. However, the evaluation of the benefit of these treatments on the whole organism is suffering from a better knowledge of the performance in mouse models. We show in the present study that LmnaH222P/H222P mice display a significant loss of lean mass, consistent with the dystrophic process. This is associated with altered VO2 peak and respiratory exchange ratio. These results showed for the first time that LmnaH222P/H222P mice have decreased performance and provided a new useful means for future therapeutic interventions on this model of EDMD., (© The Author(s) 2019. Published by Oxford University Press. All rights reserved. For Permissions, please email: journals.permissions@oup.com.)
- Published
- 2019
- Full Text
- View/download PDF
23. miR-708-5p and miR-34c-5p are involved in nNOS regulation in dystrophic context.
- Author
-
Guilbaud M, Gentil C, Peccate C, Gargaun E, Holtzmann I, Gruszczynski C, Falcone S, Mamchaoui K, Ben Yaou R, Leturcq F, Jeanson-Leh L, and Piétri-Rouxel F
- Subjects
- Adolescent, Adult, Aged, Biopsy, Child, Computational Biology methods, Gene Expression Profiling methods, Gene Expression Regulation, Enzymologic, Humans, Male, MicroRNAs physiology, Muscle Proteins genetics, Muscle Proteins metabolism, Muscle, Skeletal enzymology, Muscle, Skeletal pathology, Muscular Dystrophy, Duchenne metabolism, Muscular Dystrophy, Duchenne pathology, Myoblasts enzymology, Nitric Oxide Synthase Type I metabolism, Real-Time Polymerase Chain Reaction methods, MicroRNAs genetics, Muscular Dystrophy, Duchenne genetics, Nitric Oxide Synthase Type I genetics
- Abstract
Background: Duchenne (DMD) and Becker (BMD) muscular dystrophies are caused by mutations in the DMD gene coding for dystrophin, a protein being part of a large sarcolemmal protein scaffold that includes the neuronal nitric oxide synthase (nNOS). The nNOS was shown to play critical roles in a variety of muscle functions and alterations of its expression and location in dystrophic muscle fiber leads to an increase of the muscle fatigability. We previously revealed a decrease of nNOS expression in BMD patients all presenting a deletion of exons 45 to 55 in the DMD gene (BMDd45-55), impacting the nNOS binding site of dystrophin. Since several studies showed deregulation of microRNAs (miRNAs) in dystrophinopathies, we focused on miRNAs that could target nNOS in dystrophic context., Methods: By a screening of 617 miRNAs in BMDd45-55 muscular biopsies using TLDA and an in silico study to determine which one could target nNOS, we selected four miRNAs. In order to select those that targeted a sequence of 3'UTR of NOS1, we performed luciferase gene reporter assay in HEK393T cells. Finally, expression of candidate miRNAs was modulated in control and DMD human myoblasts (DMDd45-52) to study their ability to target nNOS., Results: TLDA assay and the in silico study allowed us to select four miRNAs overexpressed in muscle biopsies of BMDd45-55 compared to controls. Among them, only the overexpression of miR-31, miR-708, and miR-34c led to a decrease of luciferase activity in an NOS1-3'UTR-luciferase assay, confirming their interaction with the NOS1-3'UTR. The effect of these three miRNAs was investigated on control and DMDd45-52 myoblasts. First, we showed a decrease of nNOS expression when miR-708 or miR-34c were overexpressed in control myoblasts. We then confirmed that DMDd45-52 cells displayed an endogenous increased of miR-31, miR-708, and miR-34c and a decreased of nNOS expression, the same characteristics observed in BMDd45-55 biopsies. In DMDd45-52 cells, we demonstrated that the inhibition of miR-708 and miR-34c increased nNOS expression, confirming that both miRNAs can modulate nNOS expression in human myoblasts., Conclusion: These results strongly suggest that miR-708 and miR-34c, overexpressed in dystrophic context, are new actors involved in the regulation of nNOS expression in dystrophic muscle.
- Published
- 2018
- Full Text
- View/download PDF
24. Allele-specific silencing therapy for Dynamin 2-related dominant centronuclear myopathy.
- Author
-
Trochet D, Prudhon B, Beuvin M, Peccate C, Lorain S, Julien L, Benkhelifa-Ziyyat S, Rabai A, Mamchaoui K, Ferry A, Laporte J, Guicheney P, Vassilopoulos S, and Bitoun M
- Subjects
- Alleles, Animals, Cells, Cultured, Humans, Mice, Mutation, Dynamin II genetics, Genetic Therapy, Myopathies, Structural, Congenital drug therapy, Myopathies, Structural, Congenital enzymology, Myopathies, Structural, Congenital physiopathology, RNA, Small Interfering therapeutic use
- Abstract
Rapid advances in allele-specific silencing by RNA interference established a strategy of choice to cure dominant inherited diseases by targeting mutant alleles. We used this strategy for autosomal-dominant centronuclear myopathy (CNM), a rare neuromuscular disorder without available treatment due to heterozygous mutations in the DNM2 gene encoding Dynamin 2. Allele-specific siRNA sequences were developed in order to specifically knock down the human and murine DNM2 -mRNA harbouring the p.R465W mutation without affecting the wild-type allele. Functional restoration was achieved in muscle from a knock-in mouse model and in patient-derived fibroblasts, both expressing the most frequently encountered mutation in patients. Restoring either muscle force in a CNM mouse model or DNM2 function in patient-derived cells is an essential breakthrough towards future gene-based therapy for dominant centronuclear myopathy., (© 2017 The Authors. Published under the terms of the CC BY 4.0 license.)
- Published
- 2018
- Full Text
- View/download PDF
25. RFX1 and RFX3 Transcription Factors Interact with the D Sequence of Adeno-Associated Virus Inverted Terminal Repeat and Regulate AAV Transduction.
- Author
-
Julien L, Chassagne J, Peccate C, Lorain S, Piétri-Rouxel F, Danos O, and Benkhelifa-Ziyyat S
- Subjects
- Dependovirus metabolism, HEK293 Cells, Humans, Protein Binding, Regulatory Factor X Transcription Factors genetics, Regulatory Factor X1 genetics, Terminal Repeat Sequences, Dependovirus genetics, Regulatory Factor X Transcription Factors metabolism, Regulatory Factor X1 metabolism, Transduction, Genetic
- Abstract
Adeno-associated virus (AAV) transduction efficiency depends on the way in which cellular proteins process viral genomes in the nucleus. In this study, we have investigated the binding of nuclear proteins to the double stranded D (dsD) sequence of the AAV inverted terminal repeat (ITRs) by electromobility shift assay. We present here several lines of evidence that transcription factors belonging to the RFX protein family bind specifically and selectively to AAV2 and AAV1 dsD sequences. Using supershift experiments, we characterize complexes containing RFX1 homodimers and RFX1/RFX3 heterodimers. Following transduction of HEK-293 cells, the AAV genome can be pulled-down by RFX1 and RFX3 antibodies. Moreover, our data suggest that RFX proteins which interact with transcriptional enhancers of several mammalian DNA viruses, can act as regulators of AAV mediated transgene expression.
- Published
- 2018
- Full Text
- View/download PDF
26. Dystrophin Threshold Level Necessary for Normalization of Neuronal Nitric Oxide Synthase, Inducible Nitric Oxide Synthase, and Ryanodine Receptor-Calcium Release Channel Type 1 Nitrosylation in Golden Retriever Muscular Dystrophy Dystrophinopathy.
- Author
-
Gentil C, Le Guiner C, Falcone S, Hogrel JY, Peccate C, Lorain S, Benkhelifa-Ziyyat S, Guigand L, Montus M, Servais L, Voit T, and Piétri-Rouxel F
- Subjects
- Animals, Dogs, Muscle, Skeletal cytology, Nitrosation, Dystrophin metabolism, Muscle Contraction physiology, Muscle, Skeletal metabolism, Nitric Oxide metabolism, Nitric Oxide Synthase Type I metabolism, Nitric Oxide Synthase Type II metabolism, Ryanodine Receptor Calcium Release Channel metabolism
- Abstract
At present, the clinically most advanced strategy to treat Duchenne muscular dystrophy (DMD) is the exon-skipping strategy. Whereas antisense oligonucleotide-based clinical trials are underway for DMD, it is essential to determine the dystrophin restoration threshold needed to ensure improvement of muscle physiology at the molecular level. A preclinical trial has been conducted in golden retriever muscular dystrophy (GRMD) dogs treated in a forelimb by locoregional delivery of rAAV8-U7snRNA to promote exon skipping on the canine dystrophin messenger. Here, we exploited rAAV8-U7snRNA-transduced GRMD muscle samples, well characterized for their percentage of dystrophin-positive fibers, with the aim of defining the threshold of dystrophin rescue necessary for normalization of the status of neuronal nitric oxide synthase mu (nNOSμ), inducible nitric oxide synthase (iNOS), and ryanodine receptor-calcium release channel type 1 (RyR1), crucial actors for efficient contractile function. Results showed that restoration of dystrophin in 40% of muscle fibers is needed to decrease abnormal cytosolic nNOSμ expression and to reduce overexpression of iNOS, these two parameters leading to a reduction in the NO level in the muscle fibers. Furthermore, the same percentage of dystrophin-positive fibers of 40% was associated with the normalization of RyR1 nitrosylation status and with stabilization of the RyR1-calstabin1 complex that is required to facilitate coupled gating. We concluded that a minimal threshold of 40% of dystrophin-positive fibers is necessary for the reinstatement of central proteins needed for proper muscle contractile function, and thus identified a rate of dystrophin expression significantly improving, at the molecular level, the dystrophic muscle physiology.
- Published
- 2016
- Full Text
- View/download PDF
27. Antisense pre-treatment increases gene therapy efficacy in dystrophic muscles.
- Author
-
Peccate C, Mollard A, Le Hir M, Julien L, McClorey G, Jarmin S, Le Heron A, Dickson G, Benkhelifa-Ziyyat S, Piétri-Rouxel F, Wood MJ, Voit T, and Lorain S
- Subjects
- Animals, Dependovirus genetics, Exons genetics, Gene Transfer Techniques, Genetic Vectors administration & dosage, Humans, Mice, Inbred mdx, Muscle, Skeletal drug effects, Muscle, Skeletal pathology, Muscular Dystrophy, Animal genetics, Muscular Dystrophy, Duchenne genetics, Sarcolemma drug effects, Sarcolemma pathology, Dystrophin genetics, Genetic Therapy, Morpholinos administration & dosage, Muscular Dystrophy, Animal therapy, Muscular Dystrophy, Duchenne therapy, Oligonucleotides, Antisense administration & dosage
- Abstract
In preclinical models for Duchenne muscular dystrophy, dystrophin restoration during adeno-associated virus (AAV)-U7-mediated exon-skipping therapy was shown to decrease drastically after six months in treated muscles. This decline in efficacy is strongly correlated with the loss of the therapeutic AAV genomes, probably due to alterations of the dystrophic myofiber membranes. To improve the membrane integrity of the dystrophic myofibers at the time of AAV-U7 injection, mdx muscles were pre-treated with a single dose of the peptide-phosphorodiamidate morpholino (PPMO) antisense oligonucleotides that induced temporary dystrophin expression at the sarcolemma. The PPMO pre-treatment allowed efficient maintenance of AAV genomes in mdx muscles and enhanced the AAV-U7 therapy effect with a ten-fold increase of the protein level after 6 months. PPMO pre-treatment was also beneficial to AAV-mediated gene therapy with transfer of micro-dystrophin cDNA into muscles. Therefore, avoiding vector genome loss after AAV injection by PPMO pre-treatment would allow efficient long-term restoration of dystrophin and the use of lower and thus safer vector doses for Duchenne patients., (© The Author 2016. Published by Oxford University Press. All rights reserved. For permissions, please e-mail: journals.permissions@oup.com.)
- Published
- 2016
- Full Text
- View/download PDF
28. Dysferlin rescue by spliceosome-mediated pre-mRNA trans-splicing targeting introns harbouring weakly defined 3' splice sites.
- Author
-
Philippi S, Lorain S, Beley C, Peccate C, Précigout G, Spuler S, and Garcia L
- Subjects
- Animals, Cells, Cultured, Computational Biology, Dysferlin, Humans, Introns, Membrane Proteins metabolism, Mice, Mice, Inbred C57BL, Mice, Knockout, Muscle Proteins metabolism, Muscle, Skeletal metabolism, Muscular Dystrophies genetics, Muscular Dystrophies, Limb-Girdle genetics, Myoblasts cytology, Myoblasts metabolism, RNA Precursors metabolism, RNA Splice Sites, Spliceosomes metabolism, Membrane Proteins genetics, Muscle Proteins genetics, RNA Precursors genetics, Spliceosomes genetics, Trans-Splicing
- Abstract
The modification of the pre-mRNA cis-splicing process employing a pre-mRNA trans-splicing molecule (PTM) is an attractive strategy for the in situ correction of genes whose careful transcription regulation and full-length expression is determinative for protein function, as it is the case for the dysferlin (DYSF, Dysf) gene. Loss-of-function mutations of DYSF result in different types of muscular dystrophy mainly manifesting as limb girdle muscular dystrophy 2B (LGMD2B) and Miyoshi muscular dystrophy 1 (MMD1). We established a 3' replacement strategy for mutated DYSF pre-mRNAs induced by spliceosome-mediated pre-mRNA trans-splicing (SmaRT) by the use of a PTM. In contrast to previously established SmaRT strategies, we particularly focused on the identification of a suitable pre-mRNA target intron other than the optimization of the PTM design. By targeting DYSF pre-mRNA introns harbouring differentially defined 3' splice sites (3' SS), we found that target introns encoding weakly defined 3' SSs were trans-spliced successfully in vitro in human LGMD2B myoblasts as well as in vivo in skeletal muscle of wild-type and Dysf(-/-) mice. For the first time, we demonstrate rescue of Dysf protein by SmaRT in vivo. Moreover, we identified concordant qualities among the successfully targeted Dysf introns and targeted endogenous introns in previously reported SmaRT approaches that might facilitate a selective choice of target introns in future SmaRT strategies., (© The Author 2015. Published by Oxford University Press. All rights reserved. For Permissions, please email: journals.permissions@oup.com.)
- Published
- 2015
- Full Text
- View/download PDF
29. Abnormal splicing switch of DMD's penultimate exon compromises muscle fibre maintenance in myotonic dystrophy.
- Author
-
Rau F, Lainé J, Ramanoudjame L, Ferry A, Arandel L, Delalande O, Jollet A, Dingli F, Lee KY, Peccate C, Lorain S, Kabashi E, Athanasopoulos T, Koo T, Loew D, Swanson MS, Le Rumeur E, Dickson G, Allamand V, Marie J, and Furling D
- Subjects
- 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, 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
- 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.
- Published
- 2015
- Full Text
- View/download PDF
30. Repair of rhodopsin mRNA by spliceosome-mediated RNA trans-splicing: a new approach for autosomal dominant retinitis pigmentosa.
- Author
-
Berger A, Lorain S, Joséphine C, Desrosiers M, Peccate C, Voit T, Garcia L, Sahel JA, and Bemelmans AP
- Subjects
- Animals, Binding Sites, Cell Line, Transformed, Gene Expression, Genes, Dominant, Genetic Vectors genetics, Humans, Introns, Mice, Mice, Transgenic, Phenotype, Photoreceptor Cells metabolism, Protein Interaction Domains and Motifs genetics, Protein Transport, RNA Precursors genetics, RNA Splicing, Retinitis Pigmentosa therapy, Rhodopsin chemistry, Rhodopsin metabolism, Transduction, Genetic, Genetic Therapy, RNA, Messenger genetics, Retinitis Pigmentosa genetics, Rhodopsin genetics, Trans-Splicing
- Abstract
The promising clinical results obtained for ocular gene therapy in recent years have paved the way for gene supplementation to treat recessively inherited forms of retinal degeneration. The situation is more complex for dominant mutations, as the toxic mutant gene product must be removed. We used spliceosome-mediated RNA trans-splicing as a strategy for repairing the transcript of the rhodopsin gene, the gene most frequently mutated in autosomal dominant retinitis pigmentosa. We tested 17 different molecules targeting the pre-mRNA intron 1, by transient transfection of HEK-293T cells, with subsequent trans-splicing quantification at the transcript level. We found that the targeting of some parts of the intron promoted trans-splicing more efficiently than the targeting of other areas, and that trans-splicing rate could be increased by modifying the replacement sequence. We then developed cell lines stably expressing the rhodopsin gene, for the assessment of phenotypic criteria relevant to the pathogenesis of retinitis pigmentosa. Using this model, we showed that trans-splicing restored the correct localization of the protein to the plasma membrane. Finally, we tested our best candidate by AAV gene transfer in a mouse model of retinitis pigmentosa that expresses a mutant allele of the human rhodopsin gene, and demonstrated the feasibility of trans-splicing in vivo. This work paves the way for trans-splicing gene therapy to treat retinitis pigmentosa due to rhodopsin gene mutation and, more generally, for the treatment of genetic diseases with dominant transmission.
- Published
- 2015
- Full Text
- View/download PDF
31. Intrinsic transgene immunogenicity gears CD8(+) T-cell priming after rAAV-mediated muscle gene transfer.
- Author
-
Carpentier M, Lorain S, Chappert P, Lalfer M, Hardet R, Urbain D, Peccate C, Adriouch S, Garcia L, Davoust J, and Gross DA
- Subjects
- Animals, Epitopes immunology, Mice, Mice, Inbred C57BL, Muscle, Skeletal metabolism, Antigen-Presenting Cells immunology, CD8-Positive T-Lymphocytes immunology, Dependovirus genetics, Gene Transfer Techniques, Genetic Vectors, Muscle, Skeletal immunology, Transgenes
- Abstract
Antitransgene CD8(+) T-cell responses are an important hurdle after recombinant adeno-associated virus (rAAV) vector-mediated gene transfer. Indeed, depending on the mutational genotype of the host, transgene amino-acid sequences of foreign origin can elicit deleterious cellular and humoral responses. We compared here two different major histocompatibility complex (MHC) class I epitopes of an engineered ovalbumin transgene delivered in muscle tissue by rAAV1 vector and found very different strength of CD8 responses, muscle destruction being correlated with the course of the immunodominant response. We further demonstrate that robust CD8(+) T-cell priming can occur through the cross-presentation pathway but requires the presence of either a strong MHC class II epitope or antibodies to the transgene product. Finally, manipulating transgene subcellular localization, we found that provided we avoid transgene expression in antigen presenting cells, the poorly accessible cytosolic form of ovalbumin transgene lacking strong MHC II epitope, evades CD8(+) T-cell priming and remains permanently expressed in muscle with no immune cell infiltration. Our results demonstrate that the intrinsic immunogenicity of transgenes delivered with rAAV vector in muscle can be manipulated in a rational manner to avoid adverse immune responses.
- Published
- 2015
- Full Text
- View/download PDF
32. Dystrophin rescue by trans-splicing: a strategy for DMD genotypes not eligible for exon skipping approaches.
- Author
-
Lorain S, Peccate C, Le Hir M, Griffith G, Philippi S, Précigout G, Mamchaoui K, Jollet A, Voit T, and Garcia L
- Subjects
- 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, Dystrophin genetics, Trans-Splicing
- 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.
- Published
- 2013
- Full Text
- View/download PDF
33. AAV genome loss from dystrophic mouse muscles during AAV-U7 snRNA-mediated exon-skipping therapy.
- Author
-
Le Hir M, Goyenvalle A, Peccate C, Précigout G, Davies KE, Voit T, Garcia L, and Lorain S
- Subjects
- Animals, Cardiotoxins pharmacology, Dependovirus metabolism, Dystrophin genetics, Dystrophin metabolism, Exons, Gene Expression, Genetic Therapy, Genetic Vectors administration & dosage, Genetic Vectors metabolism, Humans, Injections, Intramuscular, Mice, Mice, Inbred mdx, Muscle, Skeletal drug effects, Muscle, Skeletal metabolism, Muscular Dystrophy, Animal therapy, Muscular Dystrophy, Duchenne therapy, Alternative Splicing, Dependovirus genetics, Genetic Vectors genetics, Genome, Viral, Muscular Dystrophy, Animal genetics, Muscular Dystrophy, Duchenne genetics, RNA, Small Nuclear genetics
- Abstract
In the context of future adeno-associated viral (AAV)-based clinical trials for Duchenne myopathy, AAV genome fate in dystrophic muscles is of importance considering the viral capsid immunogenicity that prohibits recurring treatments. We showed that AAV genomes encoding non-therapeutic U7 were lost from mdx dystrophic muscles within 3 weeks after intramuscular injection. In contrast, AAV genomes encoding U7ex23 restoring expression of a slightly shortened dystrophin were maintained endorsing that the arrest of the dystrophic process is crucial for maintaining viral genomes in transduced fibers. Indeed, muscles treated with low doses of AAV-U7ex23, resulting in sub-optimal exon skipping, displayed much lower titers of viral genomes, showing that sub-optimal dystrophin restoration does not prevent AAV genome loss. We also followed therapeutic viral genomes in severe dystrophic dKO mice over time after systemic treatment with scAAV9-U7ex23. Dystrophin restoration decreased significantly between 3 and 12 months in various skeletal muscles, which was correlated with important viral genome loss, except in the heart. Altogether, these data show that the success of future AAV-U7 therapy for Duchenne patients would require optimal doses of AAV-U7 to induce substantial levels of dystrophin to stabilize the treated fibers and maintain the long lasting effect of the treatment.
- Published
- 2013
- Full Text
- View/download PDF
34. Exon exchange approach to repair Duchenne dystrophin transcripts.
- Author
-
Lorain S, Peccate C, Le Hir M, and Garcia L
- Subjects
- Animals, Base Sequence, DNA Mutational Analysis, Enhancer Elements, Genetic genetics, Introns genetics, Mice, Molecular Sequence Data, NIH 3T3 Cells, RNA, Messenger genetics, Trans-Splicing genetics, Dystrophin genetics, Exons genetics, Muscular Dystrophy, Animal genetics
- Abstract
Background: Trans-splicing strategies for mRNA repair involve engineered transcripts designed to anneal target mRNAs in order to interfere with their natural splicing, giving rise to mRNA chimeras where endogenous mutated exons have been replaced by exogenous replacement sequences. A number of trans-splicing molecules have already been proposed for replacing either the 5' or the 3' part of transcripts to be repaired. Here, we show the feasibility of RNA surgery by using a double trans-splicing approach allowing the specific substitution of a given mutated exon., Methodology/principal Findings: As a target we used a minigene encoding a fragment of the mdx dystrophin gene enclosing the mutated exon (exon 23). This minigene was cotransfected with a variety of exon exchange constructions, differing in their annealing domains. We obtained accurate and efficient replacement of exon 23 in the mRNA target. Adding up a downstream intronic splice enhancer DISE in the exon exchange molecule enhanced drastically its efficiency up to 25-45% of repair depending on the construction in use., Conclusions/significance: These results demonstrate the possibility to fix up mutated exons, refurbish deleted exons and introduce protein motifs, while keeping natural untranslated sequences, which are essential for mRNA stability and translation regulation. Conversely to the well-known exon skipping, exon exchange has the advantage to be compatible with almost any type of mutations and more generally to a wide range of genetic conditions. In particular, it allows addressing disorders caused by dominant mutations.
- Published
- 2010
- Full Text
- View/download PDF
35. Multitissular involvement in a family with LMNA and EMD mutations: Role of digenic mechanism?
- Author
-
Ben Yaou R, Toutain A, Arimura T, Demay L, Massart C, Peccate C, Muchir A, Llense S, Deburgrave N, Leturcq F, Litim KE, Rahmoun-Chiali N, Richard P, Babuty D, Récan-Budiartha D, and Bonne G
- Subjects
- Adolescent, Adult, Blotting, Western, Electromyography, Female, Fibroblasts metabolism, Fluorescent Antibody Technique, Genotype, Heart Diseases genetics, Humans, Lamin Type A metabolism, Male, Membrane Proteins metabolism, Middle Aged, Muscle, Skeletal pathology, Muscle, Skeletal physiopathology, Muscular Dystrophy, Emery-Dreifuss pathology, Mutation, Nuclear Proteins metabolism, Pedigree, Peripheral Nervous System Diseases genetics, Phenotype, Polymerase Chain Reaction, Lamin Type A genetics, Membrane Proteins genetics, Muscular Dystrophy, Emery-Dreifuss genetics, Muscular Dystrophy, Emery-Dreifuss physiopathology, Nuclear Proteins genetics
- Abstract
Background: Mutations in the EMD and LMNA genes, encoding emerin and lamins A and C, are responsible for the X-linked and autosomal dominant and recessive forms of Emery-Dreifuss muscular dystrophy (EDMD). LMNA mutations can also lead to several other disorders, collectively termed laminopathies, involving heart, fat, nerve, bone, and skin tissues, and some premature ageing syndromes., Methods: Fourteen members of a single family underwent neurologic, electromyographic, and cardiologic assessment. Gene mutation and protein expression analyses were performed for lamins A/C and emerin., Results: Clinical investigations showed various phenotypes, including isolated cardiac disease (seven patients), axonal neuropathy (one patient), and a combination of EDMD with axonal neuropathy (two patients), whereas five subjects remained asymptomatic. Genetic analyses identified the coincidence of a previously described homozygous LMNA mutation (c.892C-->T, p. R298C) and a new in-frame EMD deletion (c.110-112delAGA, p. delK37), which segregate independently. Analyses of the contribution of these mutations showed 1) the EMD codon deletion acts in X-linked dominant fashion and was sufficient to induce the cardiac disease, 2) the combination of both the hemizygous EMD and the homozygous LMNA mutations was necessary to induce the EDMD phenotype, 3) emerin was present in reduced amount in EMD-mutated cells, and 4) lamin A/C and emerin expression was most dramatically affected in the doubly mutated fibroblasts., Conclusions: This highlights the crucial role of lamin A/C-emerin interactions, with evidence for synergistic effects of these mutations that lead to Emery-Dreifuss muscular dystrophy as the worsened result of digenic mechanism in this family.
- Published
- 2007
- Full Text
- View/download PDF
36. Protein- and mRNA-based phenotype-genotype correlations in DMD/BMD with point mutations and molecular basis for BMD with nonsense and frameshift mutations in the DMD gene.
- Author
-
Deburgrave N, Daoud F, Llense S, Barbot JC, Récan D, Peccate C, Burghes AH, Béroud C, Garcia L, Kaplan JC, Chelly J, and Leturcq F
- Subjects
- Adolescent, Adult, Biopsy, Child, Child, Preschool, Codon, Nonsense, DNA Mutational Analysis methods, DNA, Complementary chemistry, Female, Frameshift Mutation, Genotype, Humans, Male, Muscular Dystrophy, Duchenne genetics, Phenotype, Point Mutation, RNA, Messenger chemistry, RNA, Messenger metabolism, Dystrophin genetics, Muscular Dystrophy, Duchenne diagnosis, Mutation
- Abstract
Straightforward detectable Duchenne muscular dystrophy (DMD) gene rearrangements, such as deletions or duplications involving an entire exon or more, are involved in about 70% of dystrophinopathies. In the remaining 30% a variety of point mutations or "small" mutations are suspected. Due to their diversity and to the large size and complexity of the DMD gene, these point mutations are difficult to detect. To overcome this diagnostic issue, we developed and optimized a routine muscle biopsy-based diagnostic strategy. The mutation detection rate is almost as high as 100% and mutations were identified in all patients for whom the diagnosis of DMD and Becker muscular dystrophy (BMD) was clinically suspected and further supported by the detection on Western blot of quantitative and/or qualitative dystrophin protein abnormalities. Here we report a total of 124 small mutations including 11 nonsense and frameshift mutations detected in BMD patients. In addition to a comprehensive assessment of muscular phenotypes that takes into account consequences of mutations on the expression of the dystrophin mRNA and protein, we provide and discuss genomic, mRNA, and protein data that pinpoint molecular mechanisms underlying BMD phenotypes associated with nonsense and frameshift mutations., ((c) 2006 Wiley-Liss, Inc.)
- Published
- 2007
- Full Text
- View/download PDF
37. Dystrophinopathy caused by mid-intronic substitutions activating cryptic exons in the DMD gene.
- Author
-
Béroud C, Carrié A, Beldjord C, Deburgrave N, Llense S, Carelle N, Peccate C, Cuisset JM, Pandit F, Carré-Pigeon F, Mayer M, Bellance R, Récan D, Chelly J, Kaplan JC, and Leturcq F
- Subjects
- Adolescent, Adult, Base Sequence genetics, DNA Mutational Analysis, Dystrophin genetics, Female, Genetic Testing, Humans, Male, Molecular Sequence Data, Muscular Dystrophy, Duchenne physiopathology, Open Reading Frames genetics, Pedigree, RNA Splice Sites genetics, RNA, Messenger genetics, Dystrophin deficiency, Exons genetics, Introns genetics, Muscular Dystrophy, Duchenne genetics, Point Mutation genetics
- Abstract
In the course of a mutation search performed by muscle dystrophin transcript analysis in 72 Duchenne and Becker Muscular Dystrophies (DMD/BMD) patients without gross gene defect, we encountered four unrelated cases with additional out-of-frame sequences precisely intercalated between two intact exons of the mature muscle dystrophin mRNA. An in silico search of the whole dystrophin genomic sequence revealed that these inserts correspond to cryptic exons flanked by one strong and one weak consensus splice site and located in the mid-part of large introns (introns 60, 9, 1M, and 62, respectively). In each case we identified an intronic point mutation activating the cryptic donor or acceptor splice site. The patients exhibited a BMD/intermediate phenotype consistent with the presence of reduced amounts of normally spliced transcript and normal dystrophin. The frequency of this new type of mutation is not negligible (6% of our series of 65 patients with 'small' mutations). It would be missed if the exploration of the DMD gene is exclusively performed on exons and flanking sequences of genomic DNA.
- Published
- 2004
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.