Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
1.
Development ; 144(15): 2737-2747, 2017 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-28694257

RESUMEN

Postnatal growth of skeletal muscle largely depends on the expansion and differentiation of resident stem cells, the so-called satellite cells. Here, we demonstrate that postnatal satellite cells express components of the bone morphogenetic protein (BMP) signaling machinery. Overexpression of noggin in postnatal mice (to antagonize BMP ligands), satellite cell-specific knockout of Alk3 (the gene encoding the BMP transmembrane receptor) or overexpression of inhibitory SMAD6 decreased satellite cell proliferation and accretion during myofiber growth, and ultimately retarded muscle growth. Moreover, reduced BMP signaling diminished the adult satellite cell pool. Abrogation of BMP signaling in satellite cell-derived primary myoblasts strongly diminished cell proliferation and upregulated the expression of cell cycle inhibitors p21 and p57 In conclusion, these results show that BMP signaling defines postnatal muscle development by regulating satellite cell-dependent myofiber growth and the generation of the adult muscle stem cell pool.


Asunto(s)
Proteínas Morfogenéticas Óseas/metabolismo , Células Satélite del Músculo Esquelético/citología , Células Satélite del Músculo Esquelético/metabolismo , Animales , Western Blotting , Proteínas Morfogenéticas Óseas/genética , Proliferación Celular/genética , Proliferación Celular/fisiología , Células Cultivadas , Femenino , Inmunohistoquímica , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes , Fibras Musculares Esqueléticas/citología , Fibras Musculares Esqueléticas/metabolismo , Músculo Esquelético/citología , Músculo Esquelético/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa , Transducción de Señal/genética , Transducción de Señal/fisiología
2.
Hum Mol Genet ; 24(14): 4049-60, 2015 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-25904108

RESUMEN

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.


Asunto(s)
Proteínas de la Membrana/genética , Proteínas Musculares/genética , Precursores del ARN/genética , Empalmosomas/genética , Trans-Empalme , Animales , Células Cultivadas , Biología Computacional , Disferlina , Humanos , Intrones , Proteínas de la Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Musculares/metabolismo , Músculo Esquelético/metabolismo , Distrofias Musculares/genética , Distrofia Muscular de Cinturas/genética , Mioblastos/citología , Mioblastos/metabolismo , Precursores del ARN/metabolismo , Sitios de Empalme de ARN , Empalmosomas/metabolismo
3.
Hum Mutat ; 34(10): 1387-95, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23864287

RESUMEN

Limb-girdle muscular dystrophy type 2A (LGMD2A) is the most frequent autosomal recessive muscular dystrophy. It is caused by mutations in the calpain-3 (CAPN3) gene. The majority of the mutations described to date are located in the coding sequence of the gene. However, it is estimated that 25% of the mutations are present at exon-intron boundaries and modify the pre-mRNA splicing of the CAPN3 transcript. We have previously described the first deep intronic mutation in the CAPN3 gene: c.1782+1072G>C mutation. This mutation causes the pseudoexonization of an intronic sequence of the CAPN3 gene in the mature mRNA. In the present work, we show that the point mutation generates the inclusion of the pseudoexon in the mRNA using a minigene assay. In search of a treatment that restores normal splicing, splicing modulation was induced by RNA-based strategies, which included antisense oligonucleotides and modified small-nuclear RNAs. The best effect was observed with antisense sequences, which induced pseudoexon skipping in both HeLa cells cotransfected with mutant minigene and in fibroblasts from patients. Finally, transfection of antisense sequences and siRNA downregulation of serine/arginine-rich splicing factor 1 (SRSF1) indicate that binding of this factor to splicing enhancer sequences is involved in pseudoexon activation.


Asunto(s)
Exones , Intrones , Distrofia Muscular de Cinturas/genética , Mutación , Oligonucleótidos Antisentido/genética , ARN Nuclear Pequeño/genética , Empalme Alternativo , Calpaína/genética , Línea Celular , Femenino , Fibroblastos/metabolismo , Expresión Génica , Regulación de la Expresión Génica , Orden Génico , Humanos , Persona de Mediana Edad , Proteínas Musculares/genética , Proteínas Nucleares/metabolismo , Oligonucleótidos Antisentido/metabolismo , Proteínas de Unión al ARN/metabolismo , Secuencias Reguladoras de Ácidos Nucleicos , Factores de Empalme Serina-Arginina
4.
J Virol ; 86(12): 6620-31, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22496229

RESUMEN

Recombinant adeno-associated viruses (rAAVs) hold enormous potential for human gene therapy. Despite the well-established safety and efficacy of rAAVs for in vivo gene transfer, there is still little information concerning the fate of vectors in blood following systemic delivery. We screened for serum proteins interacting with different AAV serotypes in humans, macaques, dogs, and mice. We report that serotypes rAAV-1, -5, and -6 but not serotypes rAAV-2, -7, -8, -9, and -10 interact in human sera with galectin 3 binding protein (hu-G3BP), a soluble scavenger receptor. Among the three serotypes, rAAV-6 has the most important capacities for binding to G3BP. rAAV-6 also bound G3BP in dog sera but not in macaque and mouse sera. In mice, rAAV-6 interacted with another protein of the innate immune system, C-reactive protein (CRP). Furthermore, interaction of hu-G3BP with rAAV-6 led to the formation of aggregates and hampered transduction when the two were codelivered into the mouse. Based on these data, we propose that species-specific interactions of AAVs with blood proteins may differentially impact vector distribution and efficacy in different animal models.


Asunto(s)
Antígenos de Neoplasias/metabolismo , Biomarcadores de Tumor/metabolismo , Proteínas Portadoras/metabolismo , Dependovirus/fisiología , Glicoproteínas/metabolismo , Animales , Antígenos de Neoplasias/sangre , Antígenos de Neoplasias/genética , Biomarcadores de Tumor/sangre , Biomarcadores de Tumor/genética , Proteínas Portadoras/sangre , Proteínas Portadoras/genética , Dependovirus/clasificación , Dependovirus/genética , Perros , Terapia Genética/instrumentación , Vectores Genéticos/clasificación , Vectores Genéticos/genética , Vectores Genéticos/fisiología , Glicoproteínas/sangre , Glicoproteínas/genética , Humanos , Macaca , Ratones , Ratones Endogámicos C57BL , Unión Proteica , Especificidad de la Especie , Transducción Genética
5.
Mol Ther ; 20(11): 2120-33, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22968479

RESUMEN

Duchenne muscular dystrophy (DMD) is an X-linked recessive disorder resulting from lesions of the gene encoding dystrophin. These usually consist of large genomic deletions, the extents of which are not correlated with the severity of the phenotype. Out-of-frame deletions give rise to dystrophin deficiency and severe DMD phenotypes, while internal deletions that produce in-frame mRNAs encoding truncated proteins can lead to a milder myopathy known as Becker muscular dystrophy (BMD). Widespread restoration of dystrophin expression via adeno-associated virus (AAV)-mediated exon skipping has been successfully demonstrated in the mdx mouse model and in cardiac muscle after percutaneous transendocardial delivery in the golden retriever muscular dystrophy dog (GRMD) model. Here, a set of optimized U7snRNAs carrying antisense sequences designed to rescue dystrophin were delivered into GRMD skeletal muscles by AAV1 gene transfer using intramuscular injection or forelimb perfusion. We show sustained correction of the dystrophic phenotype in extended muscle areas and partial recovery of muscle strength. Muscle architecture was improved and fibers displayed the hallmarks of mature and functional units. A 5-year follow-up ruled out immune rejection drawbacks but showed a progressive decline in the number of corrected muscle fibers, likely due to the persistence of a mild dystrophic process such as occurs in BMD phenotypes. Although AAV-mediated exon skipping was shown safe and efficient to rescue a truncated dystrophin, it appears that recurrent treatments would be required to maintain therapeutic benefit ahead of the progression of the disease.


Asunto(s)
Empalme Alternativo , Dependovirus/genética , Distrofina/genética , Distrofia Muscular Animal/terapia , Oligorribonucleótidos Antisentido/genética , ARN Nuclear Pequeño/genética , Animales , Secuencia de Bases , Calcio/metabolismo , Perros , Exones , Miembro Anterior/fisiopatología , Terapia Genética , Vectores Genéticos/administración & dosificación , Inyecciones Intramusculares , Datos de Secuencia Molecular , Contracción Muscular , Fibras Musculares Esqueléticas/metabolismo , Fibras Musculares Esqueléticas/patología , Fuerza Muscular , Músculo Esquelético/metabolismo , Músculo Esquelético/patología , Músculo Esquelético/fisiopatología , Distrofia Muscular Animal/fisiopatología , Transcripción Genética , Utrofina/genética , Utrofina/metabolismo
6.
Mol Ther ; 20(3): 580-9, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22146342

RESUMEN

Although restoration of dystrophin expression via exon skipping in both cardiac and skeletal muscle has been successfully demonstrated in the mdx mouse, restoration of cardiac dystrophin expression in large animal models of Duchenne muscular dystrophy (DMD) has proven to be a challenge. In large animals, investigators have focused on using intravenous injection of antisense oligonucleotides (AO) to mediate exon skipping. In this study, we sought to optimize restoration of cardiac dystrophin expression in the golden retriever muscular dystrophy (GRMD) model using percutaneous transendocardial delivery of recombinant AAV6 (rAAV6) to deliver a modified U7 small nuclear RNA (snRNA) carrying antisense sequence to target the exon splicing enhancers of exons 6 and 8 and correct the disrupted reading frame. We demonstrate restoration of cardiac dystrophin expression at 13 months confirmed by reverse transcription-PCR (RT-PCR) and immunoblot as well as membrane localization by immunohistochemistry. This was accompanied by improved cardiac function as assessed by cardiac magnetic resonance imaging (MRI). Percutaneous transendocardial delivery of rAAV6 expressing a modified U7 exon skipping construct is a safe, effective method for restoration of dystrophin expression and improvement of cardiac function in the GRMD canine and may be easily translatable to human DMD patients.


Asunto(s)
Empalme Alternativo , Dependovirus/genética , Distrofina/genética , Vectores Genéticos/genética , Distrofia Muscular de Duchenne/genética , Distrofia Muscular de Duchenne/terapia , Animales , Línea Celular , Modelos Animales de Enfermedad , Perros , Distrofina/metabolismo , Ecocardiografía , Exones , Fibrosis , Expresión Génica , Orden Génico , Técnicas de Transferencia de Gen , Vectores Genéticos/farmacocinética , Genoma Viral , Humanos , Imagen por Resonancia Magnética , Distrofia Muscular de Duchenne/diagnóstico , Miocardio/patología , ARN Mensajero/metabolismo
7.
Mol Ther ; 18(5): 881-7, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-20104211

RESUMEN

Duchenne muscular dystrophy is characterized by muscular atrophy, fibrosis, and fat accumulation. Several groups have demonstrated that in the mdx mouse, the exon-skipping strategy can restore a quasi-dystrophin in almost 100% of the muscle fibers. On the other hand, inhibition of the myostatin pathway in adult mice has been described to enhance muscle growth and improve muscle force. Our aim was to combine these two strategies to evaluate a possible additive effect. We have chosen to inhibit the myostatin pathway using the technique of RNA interference directed against the myostatin receptor AcvRIIb mRNA (sh-AcvRIIb). The restoration of a quasi-dystrophin was mediated by the vectorized U7 exon-skipping technique (U7-DYS). Adeno-associated vectors carrying either the sh-AcvrIIb construct alone, the U7-DYS construct alone, or a combination of both constructs were injected in the tibialis anterior (TA) muscle of dystrophic mdx mice. We show that even if each separate approach has some effects on muscle physiology, the combination of the dystrophin rescue and the downregulation of the myostatin receptor is required to massively improve both the tetanic force and the specific force. This study provides a novel pharmacogenetic strategy for treatment of certain neuromuscular diseases associated with muscle wasting.


Asunto(s)
Distrofina/metabolismo , Distrofia Muscular de Duchenne/terapia , Miostatina/metabolismo , Receptores de Activinas Tipo II/genética , Receptores de Activinas Tipo II/metabolismo , Animales , Línea Celular , Dependovirus/genética , Distrofina/genética , Femenino , Vectores Genéticos , Humanos , Ratones , Ratones Endogámicos mdx , Interferencia de ARN , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal/genética , Transducción de Señal/fisiología
8.
Hum Mutat ; 31(2): 136-42, 2010 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19953532

RESUMEN

Mutations in DYSF encoding dysferlin cause primary dysferlinopathies, autosomal recessive diseases that mainly present clinically as Limb Girdle Muscular Dystrophy type 2B and Miyoshi myopathy. More than 350 different sequence variants have been reported in DYSF. Like dystrophin, the size of the dysferlin mRNA is above the limited packaging size of AAV vectors. Alternative strategies to AAV gene transfer in muscle cells must then be addressed for patients. A gene therapy approach for Duchenne muscular dystrophy was recently developed, based on exon-skipping strategy. Numerous sequences are recognized by splicing protein complexes and, when specifically blocked by antisense oligoucleotides (AON), the corresponding exon is skipped. We hypothesized that this approach could be useful for patients affected with dysferlinopathies. To confirm this assumption, exon 32 was selected as a prioritary target for exon skipping strategy. This option was initially driven by the report from Sinnreich and colleagues of a patient with a very mild and late-onset phenotype associated to a natural skipping of exon 32. Three different antisense oligonucleotides were tested in myoblasts generated from control and patient MyoD transduced fibroblasts, either as oligonucleotides or after incorporation into lentiviral vectors. These approaches led to a high efficiency of exon 32 skipping. Therefore, these results seem promising, and could be applied to several other exons in the DYSF gene. Patients carrying mutations in exons whose the in-frame suppression has been proven to have no major consequences on the protein function, might benefit of exon-skipping based gene correction.


Asunto(s)
Exones/genética , Fibroblastos/metabolismo , Fibroblastos/patología , Proteínas de la Membrana/deficiencia , Proteínas de la Membrana/genética , Proteínas Musculares/deficiencia , Proteínas Musculares/genética , Mutación/genética , Oligonucleótidos Antisentido/metabolismo , Animales , Secuencia de Bases , Células Cultivadas , Codón sin Sentido/genética , Análisis Mutacional de ADN , Disferlina , Mutación del Sistema de Lectura/genética , Humanos , Lentivirus/genética , Ratones , Datos de Secuencia Molecular
9.
Mol Ther Nucleic Acids ; 13: 198-207, 2018 Dec 07.
Artículo en Inglés | MEDLINE | ID: mdl-30292141

RESUMEN

Limb girdle muscular dystrophy 2B (LGMD2B) is without treatment and caused by mutations in the dysferlin gene (DYSF). One-third is missense mutations leading to dysferlin aggregation and amyloid formation, in addition to defects in sarcolemmal repair and progressive muscle wasting. Dysferlin-null mouse models do not allow study of the consequences of missense mutations. We generated a new mouse model (MMex38) carrying a missense mutation in exon 38 in analogy to a clinically relevant human DYSF variant (DYSF p.Leu1341Pro). The targeted mutation induces all characteristics of missense mutant dysferlinopathy, including a progressive dystrophic pattern, amyloid formation, and defects in membrane repair. We chose U7 small nuclear RNA (snRNA)-based splice switching to demonstrate a possible exon-skipping strategy in this new animal model. We show that Dysf exons 37 and 38 can successfully be skipped in vivo. Overall, the MMex38 mouse model provides an ideal tool for preclinical development of treatment strategies for dysferlinopathy.

10.
Mol Ther Methods Clin Dev ; 3: 16035, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27226971

RESUMEN

Recombinant adeno-associated viruses (rAAV) are largely used for gene transfer in research, preclinical developments, and clinical trials. Their broad in vivo biodistribution and long-term efficacy in postmitotic tissues make them good candidates for numerous gene transfer applications. Upstream processes able to produce large amounts of rAAV were developed, particularly those using baculovirus expression vector system. In parallel, downstream processes present a large panel of purification methods, often including multiple and time consuming steps. Here, we show that simple tangential flow filtration, coupled with an optimized iodixanol-based isopycnic density gradient, is sufficient to purify several liters of crude lysate produced by baculovirus expression vector system in only one working day, leading to high titers and good purity of rAAV products. Moreover, we show that the viral vectors retain their in vitro and in vivo functionalities. Our results demonstrate that simple, rapid, and relatively low-cost methods can easily be implemented for obtaining a high-quality grade of gene therapy products based on rAAV technology.

11.
Mol Ther Methods Clin Dev ; 2: 15024, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26207258

RESUMEN

Recombinant adeno-associated viruses (rAAV) are promising candidates for gene therapy approaches. The last two decades were particularly fruitful in terms of processes applied in the production and purification of this type of gene transfer vectors. This rapid technological evolution led to better yields and higher levels of vector purity. Recently, some reports showed that rAAV produced by transient tri-transfection method in adherent human embryonic kidney 293 cells can be harvested directly from supernatant, leading to easier and faster purification compared to classical virus extraction from cell pellets. Here, we compare these approaches with new vector recovery method using small quantity of detergent at the initial clarification step to treat the whole transfected cell culture. Coupled with tangential flow filtration and iodixanol-based isopycnic density gradient, this new method significantly increases rAAV yields and conserves high vector purity. Moreover, this approach leads to the reduction of the total process duration. Finally, the vectors maintain their functionality, showing unexpected higher in vitro and in vivo transduction efficacies. This new development in rAAV downstream process once more demonstrates the great capacity of these vectors to easily accommodate to large panel of methods, able to furthermore ameliorate their safety, functionality, and scalability.

12.
Elife ; 42015 Oct 13.
Artículo en Inglés | MEDLINE | ID: mdl-26459831

RESUMEN

Dystrophin forms an essential link between sarcolemma and cytoskeleton, perturbation of which causes muscular dystrophy. We analysed Dystrophin binding dynamics in vivo for the first time. Within maturing fibres of host zebrafish embryos, our analysis reveals a pool of diffusible Dystrophin and complexes bound at the fibre membrane. Combining modelling, an improved FRAP methodology and direct semi-quantitative analysis of bleaching suggests the existence of two membrane-bound Dystrophin populations with widely differing bound lifetimes: a stable, tightly bound pool, and a dynamic bound pool with high turnover rate that exchanges with the cytoplasmic pool. The three populations were found consistently in human and zebrafish Dystrophins overexpressed in wild-type or dmd(ta222a/ta222a) zebrafish embryos, which lack Dystrophin, and in Gt(dmd-Citrine)(ct90a) that express endogenously-driven tagged zebrafish Dystrophin. These results lead to a new model for Dystrophin membrane association in developing muscle, and highlight our methodology as a valuable strategy for in vivo analysis of complex protein dynamics.


Asunto(s)
Distrofina/análisis , Músculo Esquelético/química , Músculo Esquelético/crecimiento & desarrollo , Pez Cebra/embriología , Animales , Recuperación de Fluorescencia tras Fotoblanqueo , Humanos
13.
Curr Gene Ther ; 15(6): 563-71, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26415573

RESUMEN

Duchenne muscular dystrophy (DMD) is characterized by the loss of a functional dystrophin protein; the muscles of DMD patients progressively degenerate as a result of mechanical stress during contractions, and the condition eventually leads to premature death. By means antisense oligonucleotides (AONs), it is possible to modulate pre-mRNA splicing eliminating mutated exons and restoring dystrophin open reading frame. To overcome the hurdles in using AONs for therapeutic interventions, we exerted engineered human DMD stem cells with a lentivirus, which permanently and efficiently delivered the cloned AONs. Here we describe for the first time the exosome-mediated release of AONs from engineered human DMD CD133+ stem cells allowing the rescue of murine dystrophin expression. Finally, upon release, AONs could be internalized by host cells suggesting a potential role of exosomes acting as vesicular carriers for DMD gene therapy.


Asunto(s)
Distrofina/genética , Terapia Genética/métodos , Distrofia Muscular de Duchenne/genética , Distrofia Muscular de Duchenne/terapia , Células Madre/citología , Animales , Efecto Espectador/fisiología , Células Cultivadas , Distrofina/biosíntesis , Exones/genética , Humanos , Ratones , Ratones SCID , Músculo Esquelético/patología , Oligonucleótidos Antisentido/genética , Empalme del ARN/genética
14.
PLoS One ; 8(8): e69879, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23936358

RESUMEN

Conventional non-viral gene transfer uses bacterial plasmid DNA containing antibiotic resistance genes, cis-acting bacterial sequence elements, and prokaryotic methylation patterns that may adversely affect transgene expression and vector stability in vivo. Here, we describe novel replicative forms of a eukaryotic vector DNA that consist solely of an expression cassette flanked by adeno-associated virus (AAV) inverted terminal repeats. Extensive structural analyses revealed that this AAV-derived vector DNA consists of linear, duplex molecules with covalently closed ends (termed closed-ended, linear duplex, or "CELiD", DNA). CELiD vectors, produced in Sf9 insect cells, require AAV rep gene expression for amplification. Amounts of CELiD DNA produced from insect cell lines stably transfected with an ITR-flanked transgene exceeded 60 mg per 5 × 10(9) Sf9 cells, and 1-15 mg from a comparable number of parental Sf9 cells in which the transgene was introduced via recombinant baculovirus infection. In mice, systemically delivered CELiD DNA resulted in long-term, stable transgene expression in the liver. CELiD vectors represent a novel eukaryotic alternative to bacterial plasmid DNA.


Asunto(s)
ADN Recombinante/genética , ADN/genética , Dependovirus/genética , Dependovirus/fisiología , Genoma Viral/genética , Transfección/métodos , Replicación Viral , Animales , Ingeniería Genética , Vectores Genéticos/genética , Masculino , Ratones , Células Sf9 , Spodoptera , Factores de Tiempo
15.
FEBS J ; 280(23): 6045-60, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24028392

RESUMEN

The protein dysferlin is abundantly expressed in skeletal and cardiac muscles, where its main function is membrane repair. Mutations in the dysferlin gene are involved in two autosomal recessive muscular dystrophies: Miyoshi myopathy and limb-girdle muscular dystrophy type 2B. Development of effective therapies remains a great challenge. Strategies to repair the dysferlin gene by skipping mutated exons, using antisense oligonucleotides (AONs), may be suitable only for a subset of mutations, while cell and gene therapy can be extended to all mutations. AON-treated blood-derived CD133+ stem cells isolated from patients with Miyoshi myopathy led to partial dysferlin reconstitution in vitro but failed to express dysferlin after intramuscular transplantation into scid/blAJ dysferlin null mice. We thus extended these experiments producing the full-length dysferlin mediated by a lentiviral vector in blood-derived CD133+ stem cells isolated from the same patients. Transplantation of engineered blood-derived CD133+ stem cells into scid/blAJ mice resulted in sufficient dysferlin expression to correct functional deficits in skeletal muscle membrane repair. Our data suggest for the first time that lentivirus-mediated delivery of full-length dysferlin in stem cells isolated from Miyoshi myopathy patients could represent an alternative therapeutic approach for treatment of dysferlinopathies.


Asunto(s)
Antígenos CD/metabolismo , Miopatías Distales/terapia , Glicoproteínas/metabolismo , Proteínas de la Membrana/metabolismo , Proteínas Musculares/metabolismo , Músculo Esquelético/patología , Atrofia Muscular/terapia , Oligonucleótidos Antisentido/farmacología , Péptidos/metabolismo , Trasplante de Células Madre , Células Madre/citología , Antígeno AC133 , Adulto , Animales , Western Blotting , Células Cultivadas , Miopatías Distales/genética , Miopatías Distales/patología , Disferlina , Técnica del Anticuerpo Fluorescente , Humanos , Técnicas para Inmunoenzimas , Hibridación Fluorescente in Situ , Inyecciones Intramusculares , Lentivirus/genética , Masculino , Proteínas de la Membrana/genética , Ratones , Ratones Endogámicos A , Ratones SCID , Proteínas Musculares/genética , Músculo Esquelético/lesiones , Músculo Esquelético/metabolismo , Atrofia Muscular/genética , Atrofia Muscular/patología , Mutación/genética , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Células Madre/metabolismo
16.
Hum Gene Ther ; 24(7): 702-13, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23805838

RESUMEN

Central core disease is a myopathy often arising from mutations in the type 1 ryanodine receptor (RYR1) gene, encoding the sarcoplasmic reticulum calcium release channel RyR1. No treatment is currently available for this disease. We studied the pathological situation of a severely affected child with two recessive mutations, which resulted in a massive reduction in the amount of RyR1. The paternal mutation induced the inclusion of a new in-frame pseudo-exon in RyR1 mRNA that resulted in the insertion of additional amino acids leading to the instability of the protein. We hypothesized that skipping this additional exon would be sufficient to restore RyR1 expression and to normalize calcium releases. We therefore developed U7-AON lentiviral vectors to force exon skipping on affected primary muscle cells. The efficiency of the exon skipping was evaluated at the mRNA level, at the protein level, and at the functional level using calcium imaging. In these affected cells, we observed a decreased inclusion of the pseudo-exon, an increased RyR1 protein expression, and a restoration of calcium releases of normal amplitude either upon direct RyR1 stimulation or in response to membrane depolarization. This study is the first demonstration of the potential of exon-skipping strategy for the therapy of central core disease, from the molecular to the functional level.


Asunto(s)
Exones/genética , Regulación de la Expresión Génica/genética , Terapia Genética/métodos , Miopatía del Núcleo Central/terapia , Canal Liberador de Calcio Receptor de Rianodina/genética , Western Blotting , Calcio/metabolismo , Cartilla de ADN/genética , Vectores Genéticos/genética , Células HEK293 , Humanos , Lentivirus , Microscopía Fluorescente , Mutación/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Canal Liberador de Calcio Receptor de Rianodina/metabolismo
17.
Nat Struct Mol Biol ; 18(1): 85-7, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21186365

RESUMEN

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.


Asunto(s)
Silenciador del Gen , Proteínas Serina-Treonina Quinasas/genética , ARN Nuclear Pequeño/fisiología , Empalme Alternativo , Células Cultivadas , Expansión de las Repeticiones de ADN , Ingeniería Genética , Humanos , Distrofia Miotónica/genética , Proteína Quinasa de Distrofia Miotónica , ARN Mensajero/genética , ARN Mensajero/metabolismo , ARN Nuclear Pequeño/química , ARN Nuclear Pequeño/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA