Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Cells ; 12(20)2023 10 12.
Artículo en Inglés | MEDLINE | ID: mdl-37887288

RESUMEN

Limb-Girdle Muscular Dystrophy R9 (LGMDR9) is a dystroglycanopathy caused by Fukutin-related protein (FKRP) defects leading to the deficiency of α-DG glycosylation, essential to membrane integrity. Recombinant adeno-associated viral vector (rAAV) gene therapy offers great therapeutic promise for such neuromuscular disorders. Pre-clinical studies have paved the way for a phase 1/2 clinical trial aiming to evaluate the safety and efficacy of FKRP gene therapy in LGMDR9 patients. To demonstrate product activity, quality, and consistency throughout product and clinical development, regulatory authorities request several quality controls, including a potency assay aiming to demonstrate and quantify the intended biological effect of the gene therapy product. In the present study, we generated FKRP knock-out (KO) cells fully depleted of α-DG glycosylation using CRISPR-Cas9 to assess the functional activity of a rAAV-FKRP gene therapy. We then developed a high-throughput On-Cell-Western methodology to evaluate the restoration of α-DG glycosylation in KO-FKRP cells and determine the biological activity of the FKRP transgene. The determination of the half maximal effective concentration (EC50) provides a method to compare the rAAV-FKRP batch using a reference standard. The generation of KO-FKRP muscle cells associated with the high-throughput On-Cell-Western technique may serve as a cell-based potency assay to assess rAAV-FKRP gene therapy products.


Asunto(s)
Distrofia Muscular de Cinturas , Pentosiltransferasa , Humanos , Línea Celular , Sistemas CRISPR-Cas/genética , Distroglicanos/metabolismo , Terapia Genética/métodos , Músculo Esquelético/metabolismo , Distrofia Muscular de Cinturas/metabolismo , Pentosiltransferasa/genética
2.
Life Sci Alliance ; 6(1)2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-36265896

RESUMEN

Duchenne muscular dystrophy (DMD) is a severe muscle disease caused by impaired expression of dystrophin. Whereas mitochondrial dysfunction is thought to play an important role in DMD, the mechanism of this dysfunction remains to be clarified. Here we demonstrate that in DMD and other muscular dystrophies, a large number of Dlk1-Dio3 clustered miRNAs (DD-miRNAs) are coordinately up-regulated in regenerating myofibers and in the serum. To characterize the biological effect of this dysregulation, 14 DD-miRNAs were simultaneously overexpressed in vivo in mouse muscle. Transcriptomic analysis revealed highly similar changes between the muscle ectopically overexpressing 14 DD-miRNAs and the mdx diaphragm, with naturally up-regulated DD-miRNAs. Among the commonly dysregulated pathway we found repressed mitochondrial metabolism, and oxidative phosphorylation (OxPhos) in particular. Knocking down the DD-miRNAs in iPS-derived skeletal myotubes resulted in increased OxPhos activities. The data suggest that (1) DD-miRNAs are important mediators of dystrophic changes in DMD muscle, (2) mitochondrial metabolism and OxPhos in particular are targeted in DMD by coordinately up-regulated DD-miRNAs. These findings provide insight into the mechanism of mitochondrial dysfunction in muscular dystrophy.


Asunto(s)
MicroARNs , Distrofia Muscular de Duchenne , Animales , Ratones , Proteínas de Unión al Calcio/metabolismo , Distrofina , Ratones Endogámicos mdx , MicroARNs/genética , MicroARNs/metabolismo , Mitocondrias/genética , Mitocondrias/metabolismo , Músculo Esquelético/metabolismo , Distrofia Muscular de Duchenne/metabolismo
3.
Biomedicines ; 10(6)2022 Jun 16.
Artículo en Inglés | MEDLINE | ID: mdl-35740450

RESUMEN

Limb girdle muscular dystrophies (LGMD), caused by mutations in 29 different genes, are the fourth most prevalent group of genetic muscle diseases. Although the link between LGMD and its genetic origins has been determined, LGMD still represent an unmet medical need. Here, we describe a platform for modeling LGMD based on the use of human induced pluripotent stem cells (hiPSC). Thanks to the self-renewing and pluripotency properties of hiPSC, this platform provides a renewable and an alternative source of skeletal muscle cells (skMC) to primary, immortalized, or overexpressing cells. We report that skMC derived from hiPSC express the majority of the genes and proteins that cause LGMD. As a proof of concept, we demonstrate the importance of this cellular model for studying LGMDR9 by evaluating disease-specific phenotypes in skMC derived from hiPSC obtained from four patients.

4.
J Neuromuscul Dis ; 8(5): 785-793, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-32925084

RESUMEN

BACKGROUND: Muscular dystrophies (MD) are a large group of genetic diseases characterized by a progressive loss of muscle. The Latent TGFß Binding Protein 4 (LTBP4) in the DBA/2 background and the Cytidine Monophosphate-sialic Acid Hydroxylase (CMAH) proteins were previously identified as genetic modifiers in severe MD. OBJECTIVE: We investigated whether these modifiers could also influence a mild phenotype such as the one observed in a mouse model of Limb-Girdle MD2I (LGMD2I). METHODS: The FKRPL276I mouse model was backcrossed onto the DBA/2 background, and in separate experiments the Cmah gene was inactivated in FKRPL276I mice by crossing with a Cmah-/- mouse and selecting the double-mutants. The mdx mouse was used as control for these two genome modifications. Consequences at the histological level as well as quantification of expression level by RT-qPCR of genes relevant for muscular dystrophy were then performed. RESULTS: We observed minimal to no effect of the DBA/2 background on the mild FKRPL276I mouse phenotype, while this same background was previously shown to increase inflammation and fibrosis in the mdx mouse. Similarly, the Cmah-/- deletion had no observable effect on the FKRPL276I mouse phenotype whereas it was seen to increase features of regeneration in mdx mice. CONCLUSIONS: These modifiers were not observed to impact the severity of the presentation of the mild FKRPL276I model. An interesting association of the CMAH modifier with the regeneration process in the mdx model was seen and sheds new light on the influence of this protein on the dystrophic phenotype.


Asunto(s)
Oxigenasas de Función Mixta/genética , Distrofia Muscular de Cinturas/genética , Pentosiltransferasa , Animales , Modelos Animales de Enfermedad , Ratones , Ratones Endogámicos mdx , Fenotipo
5.
Hum Mutat ; 40(10): 1874-1885, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31268217

RESUMEN

Genetic variants in Fukutin-related protein (FKRP), an essential enzyme of the glycosylation pathway of α-dystroglycan, can lead to pathologies with different severities affecting the eye, brain, and muscle tissues. Here, we generate an in vitro cellular system to characterize the cellular localization as well as the functional potential of the most common FKRP patient missense mutations. We observe a differential retention in the endoplasmic reticulum (ER), the indication of misfolded proteins. We find data supporting that mutant protein able to overcome this ER-retention through overexpression present functional levels comparable to the wild-type. We also identify a specific region in FKRP protein localized between residues 300 and 321 in which genetic variants found in patients lead to correctly localized proteins but which are nevertheless functionally impaired or catalytically dead in our model, indicating that this particular region might be important for the enzymatic activity of FKRP within the Golgi. Our system thus allows the functional testing of patient-specific mutant proteins and the identification of candidate mutants to be further explored with the aim of finding pharmacological treatments targeting the protein quality control system.


Asunto(s)
Variación Genética , Pentosiltransferasa/genética , Pentosiltransferasa/metabolismo , Alelos , Línea Celular , Distroglicanos/metabolismo , Retículo Endoplásmico/metabolismo , Técnica del Anticuerpo Fluorescente , Expresión Génica , Estudios de Asociación Genética , Predisposición Genética a la Enfermedad , Glicosilación , Humanos , Modelos Biológicos , Mutación , Transporte de Proteínas
6.
Hum Mol Genet ; 26(10): 1952-1965, 2017 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-28334834

RESUMEN

Limb Girdle Muscular Dystrophies type 2I (LGMD2I), a recessive autosomal muscular dystrophy, is caused by mutations in the Fukutin Related Protein (FKRP) gene. It has been proposed that FKRP, a ribitol-5-phosphate transferase, is a participant in α-dystroglycan (αDG) glycosylation, which is important to ensure the cell/matrix anchor of muscle fibers. A LGMD2I knock-in mouse model was generated to express the most frequent mutation (L276I) encountered in patients. The expression of FKRP was not altered neither at transcriptional nor at translational levels, but its function was impacted since abnormal glycosylation of αDG was observed. Skeletal muscles were functionally impaired from 2 months of age and a moderate dystrophic pattern was evident starting from 6 months of age. Gene transfer with a rAAV2/9 vector expressing Fkrp restored biochemical defects, corrected the histological abnormalities and improved the resistance to eccentric stress in the mouse model. However, injection of high doses of the vector induced a decrease of αDG glycosylation and laminin binding, even in WT animals. Finally, intravenous injection of the rAAV-Fkrp vector into a dystroglycanopathy mouse model due to Fukutin (Fktn) knock-out indicated a dose-dependent toxicity. These data suggest requirement for a control of FKRP expression in muscles.


Asunto(s)
Distrofia Muscular de Cinturas/terapia , Proteínas/genética , Proteínas/uso terapéutico , Animales , Modelos Animales de Enfermedad , Distroglicanos/metabolismo , Expresión Génica , Regulación de la Expresión Génica/genética , Terapia Genética/métodos , Glicosilación , Ratones , Ratones Noqueados , Músculo Esquelético/metabolismo , Distrofias Musculares/genética , Distrofia Muscular de Cinturas/genética , Mutación , Pentosafosfatos/metabolismo , Pentosiltransferasa , Unión Proteica , Procesamiento Proteico-Postraduccional , Proteínas/metabolismo , Transferasas
7.
Hum Gene Ther Clin Dev ; 24(2): 65-76, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23721401

RESUMEN

Mutations in dysferlin and anoctamin 5 are the cause of muscular disorders, with the main presentations as limb-girdle muscular dystrophy or Miyoshi type of distal myopathy. Both these proteins have been implicated in sarcolemmal resealing. On the basis of similarities in associated phenotypes and protein functions, we tested the hypothesis that ANO5 protein could compensate for dysferlin absence. We first defined that the main transcript of ANO5 expressed in skeletal muscle is the 22-exon full-length isoform, and we demonstrated that dysferlin-deficient (Dysf (prmd)) mice have lower Ano5 expression levels, an observation that further enhanced the rational of the tested hypothesis. We then showed that AAV-mediated transfer of human ANO5 (hANO5) did not lead to apparent toxicity in wild-type mice. Finally, we demonstrated that AAV-hANO5 injection was not able to compensate for dysferlin deficiency in the Dysf (prmd) mouse model or improve the membrane repair defect seen in the absence of dysferlin. Consequently, overexpressing hANO5 does not seem to provide a valuable therapeutic strategy for dysferlin deficiency.


Asunto(s)
Canales de Cloruro/metabolismo , Dependovirus/genética , Proteínas de la Membrana/metabolismo , Distrofia Muscular de Cinturas/terapia , Animales , Anoctaminas , Línea Celular , Canales de Cloruro/genética , Regulación hacia Abajo , Disferlina , Técnicas de Transferencia de Gen , Vectores Genéticos/genética , Vectores Genéticos/metabolismo , Humanos , Proteínas de la Membrana/deficiencia , Proteínas de la Membrana/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Músculo Esquelético/metabolismo , Distrofia Muscular de Cinturas/patología , Fenotipo , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo
8.
Skelet Muscle ; 3(1): 3, 2013 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-23414517

RESUMEN

BACKGROUND: The complexity of the skeletal muscle and the identification of numerous human disease-causing mutations in its constitutive proteins make it an interesting tissue for proteomic studies aimed at understanding functional relationships of interacting proteins in both health and diseases. METHOD: We undertook a large-scale study using two-hybrid screens and a human skeletal-muscle cDNA library to establish a proteome-scale map of protein-protein interactions centered on proteins involved in limb-girdle muscular dystrophies (LGMD). LGMD is a group of more than 20 different neuromuscular disorders that principally affect the proximal pelvic and shoulder girdle muscles. RESULTS AND CONCLUSION: The interaction network we unraveled incorporates 1018 proteins connected by 1492 direct binary interactions and includes 1420 novel protein-protein interactions. Computational, experimental and literature-based analyses were performed to assess the overall quality of this network. Interestingly, LGMD proteins were shown to be highly interconnected, in particular indirectly through sarcomeric proteins. In-depth mining of the LGMD-centered interactome identified new candidate genes for orphan LGMDs and other neuromuscular disorders. The data also suggest the existence of functional links between LGMD2B/dysferlin and gene regulation, between LGMD2C/γ-sarcoglycan and energy control and between LGMD2G/telethonin and maintenance of genome integrity. This dataset represents a valuable resource for future functional investigations.

9.
Hum Mutat ; 33(2): 429-39, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22095924

RESUMEN

Sarcoglycanopathies (SGP) are a group of autosomal recessive muscle disorders caused by primary mutations in one of the four sarcoglycan genes. The sarcoglycans (α-, ß-, γ-, and δ-sarcoglycan) form a tetrameric complex at the muscle membrane that is part of the dystrophin-glycoprotein complex and plays an essential role for membrane integrity during muscle contractions. We previously showed that the most frequent missense mutation in α-sarcoglycan (p.R77C) leads to the absence of the protein at the cell membrane due to its blockade by the endoplasmic reticulum (ER) quality control. Moreover, we demonstrated that inhibition of the ER α-mannosidase I activity using kifunensine could rescue the mutant protein localization at the cell membrane. Here, we investigate 25 additional disease-causing missense mutations in the sarcoglycan genes with respect to intracellular fate and localization rescue of the mutated proteins by kifunensine. Our studies demonstrate that, similarly to p.R77C, 22 of 25 of the selected mutations lead to defective intracellular trafficking of the SGs proteins. Six of these were saved from ER retention upon kifunensine treatment. The trafficking of SGs mutants rescued by kifunensine was associated with mutations that have moderate structural impact on the protein.


Asunto(s)
Retículo Endoplásmico/metabolismo , Mutación , Sarcoglicanos/química , Sarcoglicanos/genética , Alcaloides/farmacología , Retículo Endoplásmico/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Células HEK293 , Células HeLa , Humanos , Transporte de Proteínas/efectos de los fármacos , Sarcoglicanopatías/genética , Sarcoglicanos/antagonistas & inhibidores , Sarcolema/metabolismo
10.
Hum Mol Genet ; 19(23): 4608-24, 2010 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-20855473

RESUMEN

The dominant tibial muscular dystrophy (TMD) and recessive limb-girdle muscular dystrophy 2J are allelic disorders caused by mutations in the C-terminus of titin, a giant sarcomeric protein. Both clinical presentations were initially identified in a large Finnish family and linked to a founder mutation (FINmaj). To further understand the physiopathology of these two diseases, we generated a mouse model carrying the FINmaj mutation. In heterozygous mice, dystrophic myopathology appears late at 9 months of age in few distal muscles. In homozygous (HO) mice, the first signs appear in the Soleus at 1 month of age and extend to most muscles at 6 months of age. Interestingly, the heart is also severely affected in HO mice. The mutation leads to the loss of the very C-terminal end of titin and to a secondary deficiency of calpain 3, a partner of titin. By crossing the FINmaj model with a calpain 3-deficient model, the TMD phenotype was corrected, demonstrating a participation of calpain 3 in the pathogenesis of this disease.


Asunto(s)
Calpaína/metabolismo , Modelos Animales de Enfermedad , Miopatías Distales , Proteínas Musculares/metabolismo , Distrofia Muscular de Cinturas , Animales , Western Blotting , Calpaína/deficiencia , Calpaína/genética , Conectina , Análisis Mutacional de ADN , Miopatías Distales/genética , Miopatías Distales/metabolismo , Miopatías Distales/patología , Ecocardiografía , Ligamiento Genético , Predisposición Genética a la Enfermedad , Heterocigoto , Ratones , Microscopía Electrónica , Proteínas Musculares/deficiencia , Proteínas Musculares/genética , Distrofia Muscular de Cinturas/genética , Distrofia Muscular de Cinturas/metabolismo , Distrofia Muscular de Cinturas/patología , Mutación , Reacción en Cadena de la Polimerasa , Proteínas Quinasas/genética , Sarcómeros/genética , Sarcómeros/ultraestructura
11.
Hum Mol Genet ; 17(9): 1214-21, 2008 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-18252745

RESUMEN

Limb girdle muscular dystrophy type 2D (LGMD2D, OMIM600119) is a genetic progressive myopathy that is caused by mutations in the human alpha-sarcoglycan gene (SGCA). Here, we have introduced in mice the most prevalent LGMD2D mutation, R77C. It should be noted that the natural murine residue at this position is a histidine. The model is, therefore, referred as Sgca(H77C/H77C). Unexpectedly, we observed an absence of LGMD2D-like phenotype at histological or physiological level. Using a heterologous cellular model of the sarcoglycan complex formation, we showed that the R77C allele encodes a protein that fails to be delivered to its proper cellular localization in the plasma membrane, and consequently to the disappearance of a positively charged residue. Subsequently, we transferred an AAV vector coding for the human R77C protein in the muscle of Sgca-null mice and were able to pharmacologically rescue the R77C protein from endoplasmic reticulum-retention using proteasome or mannosidase I inhibitors. This suggests a therapeutic approach for LGMD2D patients carrying mutations that impair alpha-sarcoglycan trafficking.


Asunto(s)
Manosidasas/metabolismo , Mutación Missense , Sarcoglicanos/genética , Sarcoglicanos/metabolismo , Alcaloides/administración & dosificación , Animales , Línea Celular Tumoral , Cisteína/genética , Femenino , Humanos , Leupeptinas/farmacología , Manosidasas/antagonistas & inhibidores , Ratones , Ratones Noqueados , Músculos/patología , Músculos/virología , Distrofia Muscular de Cinturas/genética , Distrofia Muscular Animal/metabolismo , Distrofia Muscular Animal/virología , Fenotipo , Transporte de Proteínas/efectos de los fármacos , Sarcoglicanos/análisis
12.
FASEB J ; 22(5): 1521-9, 2008 May.
Artículo en Inglés | MEDLINE | ID: mdl-18073330

RESUMEN

Limb-girdle muscular dystrophy type 2A (LGMD2A) is a recessive genetic disorder caused by mutations in the cysteine protease calpain 3 (CAPN3) that leads to selective muscle wasting. We previously showed that CAPN3 deficiency is associated with a profound perturbation of the NF-kappaB/IkappaB alpha survival pathway. In this study, the consequences of altered NF-kappaB/IkappaB alpha pathway were investigated using biological materials from LGMD2A patients. We first show that the antiapoptotic factor cellular-FLICE inhibitory protein (c-FLIP), which is dependent on the NF-kappaB pathway in normal muscle cells, is down-regulated in LGMD2A biopsies. In muscle cells isolated from LGMD2A patients, NF-kappaB is readily activated on cytokine induction as shown by an increase in its DNA binding activity. However, we observed discrepant transcriptional responses depending on the NF-kappaB target genes. IkappaB alpha is expressed following NF-kappaB activation independent of the CAPN3 status, whereas expression of c-FLIP is obtained only when CAPN3 is present. These data lead us to postulate that CAPN3 intervenes in the regulation of the expression of NF-kappaB-dependent survival genes to prevent apoptosis in skeletal muscle. Deregulations in the NF-kappaB pathway could be part of the mechanism responsible for the muscle wasting resulting from CAPN3 deficiency.


Asunto(s)
Proteína Reguladora de Apoptosis Similar a CASP8 y FADD/biosíntesis , Calpaína/fisiología , Proteínas Musculares/fisiología , Distrofia Muscular de Cinturas/fisiopatología , FN-kappa B/fisiología , Apoptosis/fisiología , Calpaína/deficiencia , Células Cultivadas , Regulación hacia Abajo , Humanos , Proteínas I-kappa B/biosíntesis , Interleucina-1beta/fisiología , Modelos Biológicos , Proteínas Musculares/deficiencia , Músculo Esquelético/metabolismo , Factor de Necrosis Tumoral alfa/fisiología , Proteína X Asociada a bcl-2/biosíntesis , Proteína bcl-X/biosíntesis
13.
Mol Ther ; 15(1): 53-61, 2007 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-17164775

RESUMEN

alpha-Sarcoglycanopathy (limb-girdle muscular dystrophy type 2D, LGMD2D) is a recessive muscular disorder caused by deficiency in alpha-sarcoglycan, a transmembrane protein part of the dystrophin-associated complex. To date, no treatment exists for this disease. We constructed recombinant pseudotype-1 adeno-associated virus (rAAV) vectors expressing the human alpha-sarcoglycan cDNA from a ubiquitous or a muscle-specific promoter. Evidence of specific immune response leading to disappearance of the vector was observed with the ubiquitous promoter. In contrast, efficient and sustained transgene expression with correct sarcolemmal localization and without evident toxicity was obtained with the muscle-specific promoter after intra-arterial injection into the limbs of an LGMD2D murine model. Transgene expression resulted in restoration of the sarcoglycan complex, histological improvement, membrane stabilization, and correction of pseudohypertrophy. More importantly, alpha-sarcoglycan transfer produced full rescue of the contractile force deficits and stretch sensibility and led to an increase of the global activity of the animals when both posterior limbs are injected. Our results establish the feasibility for AAV-mediated alpha-sarcoglycan gene transfer as a therapeutic approach.


Asunto(s)
Dependovirus/genética , Vectores Genéticos/administración & dosificación , Vectores Genéticos/genética , Músculos/metabolismo , Sarcoglicanos/deficiencia , Sarcoglicanos/metabolismo , Animales , Permeabilidad de la Membrana Celular , Dependovirus/clasificación , Distrofina/metabolismo , Expresión Génica , Regulación de la Expresión Génica , Hipertrofia/genética , Hipertrofia/metabolismo , Hipertrofia/patología , Inyecciones Intraarteriales , Cinética , Ratones , Especificidad de Órganos , Fenotipo , Regiones Promotoras Genéticas/genética , Unión Proteica , Sarcoglicanos/genética
14.
Mol Ther ; 15(1): 53-61, 2007 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-28182933

RESUMEN

α-Sarcoglycanopathy (limb-girdle muscular dystrophy type 2D, LGMD2D) is a recessive muscular disorder caused by deficiency in α-sarcoglycan, a transmembrane protein part of the dystrophin-associated complex. To date, no treatment exists for this disease. We constructed recombinant pseudotype-1 adeno-associated virus (rAAV) vectors expressing the human α-sarcoglycan cDNA from a ubiquitous or a muscle-specific promoter. Evidence of specific immune response leading to disappearance of the vector was observed with the ubiquitous promoter. In contrast, efficient and sustained transgene expression with correct sarcolemmal localization and without evident toxicity was obtained with the muscle-specific promoter after intra-arterial injection into the limbs of an LGMD2D murine model. Transgene expression resulted in restoration of the sarcoglycan complex, histological improvement, membrane stabilization, and correction of pseudohypertrophy. More importantly, α-sarcoglycan transfer produced full rescue of the contractile force deficits and stretch sensibility and led to an increase of the global activity of the animals when both posterior limbs are injected. Our results establish the feasibility for AAV-mediated α-sarcoglycan gene transfer as a therapeutic approach.

15.
Mol Ther ; 13(2): 250-9, 2006 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-16290124

RESUMEN

Calpainopathy (limb-girdle muscular dystrophy type 2A, LGMD2A) is a recessive muscular disorder caused by deficiency in the calcium-dependent cysteine protease calpain 3. To date, no treatment exists for this disease. We evaluated the potential of recombinant adeno-associated virus (rAAV) vectors for gene therapy in a murine model for LGMD2A. To drive the expression of calpain 3, we used rAAV2/1 pseudotyped vectors and muscle-specific promoters to avoid calpain 3 cell toxicity. We report efficient and stable transgene expression in muscle with restoration of the proteolytic activity and without evident toxicity. In addition, calpain 3 was correctly targeted to the sarcomere. Moreover, its presence resulted in improvement of the histological features and in therapeutic efficacy at the physiological levels, including correction of atrophy and full rescue of the contractile force deficits. Our results establish the feasibility of AAV-mediated calpain 3 gene transfer as a therapeutic approach.


Asunto(s)
Calpaína/genética , Calpaína/uso terapéutico , Dependovirus/genética , Técnicas de Transferencia de Gen , Terapia Genética/métodos , Proteínas Musculares/genética , Proteínas Musculares/uso terapéutico , Distrofia Muscular de Cinturas/terapia , Animales , Calpaína/biosíntesis , Calpaína/deficiencia , Modelos Animales de Enfermedad , Activación Enzimática/genética , Estabilidad de Enzimas/genética , Vectores Genéticos/uso terapéutico , Inyecciones Intramusculares , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Proteínas Musculares/biosíntesis , Proteínas Musculares/deficiencia , Músculo Esquelético/enzimología , Distrofia Muscular de Cinturas/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...