Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
1.
Int J Mol Sci ; 24(14)2023 Jul 13.
Artículo en Inglés | MEDLINE | ID: mdl-37511179

RESUMEN

Duchenne muscular dystrophy (DMD) is a yet incurable rare genetic disease that affects the skeletal and cardiac muscles, leading to progressive muscle wasting and premature death. DMD is caused by the lack of dystrophin, a muscle protein essential for the biochemical support and integrity of muscle fibers. Gene replacement strategies for Duchenne muscular dystrophy (DMD) employing the adeno-associated virus (AAV) face the challenge imposed by the limited packaging capacity of AAV, only allowing the accommodation of a short version of dystrophin (µDys) that is still far removed from correcting human disease. The need to develop strategies leading to the expression of a best performing dystrophin variant led to only few studies reporting on the use of dual vectors, but none reported on a method to assess in vivo transgene reconstitution efficiency, the degree of which directly affects the use of safe AAV dosing. We report here on the generation of a dual AAV vector approach for the expression of a larger dystrophin version (quasidystrophin) based on homologous recombination, and the development of a methodology employing a strategic droplet digital PCR design, to determine the recombination efficiency as well as the occurrence of unwanted concatemerization events or aberrant expression from the single vectors. We demonstrated that, upon systemic delivery in the dystrophic D2.B10-Dmdmdx/J (DBA2mdx) mice, our dual AAV approach led to high transgene reconstitution efficiency and negligible Inverted Terminal Repeats (ITR)-dependent concatemerization, with consequent remarkable protein restoration in muscles and improvement of muscle pathology. This evidence supports the suitability of our system for gene therapy application and the potential of this methodology to assess and improve the feasibility for therapeutic translation of multiple vector approaches.


Asunto(s)
Distrofia Muscular de Duchenne , Ratones , Humanos , Animales , Distrofia Muscular de Duchenne/genética , Distrofia Muscular de Duchenne/terapia , Distrofia Muscular de Duchenne/metabolismo , Distrofina/genética , Distrofina/metabolismo , Dependovirus/genética , Dependovirus/metabolismo , Músculo Esquelético/metabolismo , Ratones Endogámicos mdx , Vectores Genéticos/genética
2.
Int J Mol Sci ; 23(4)2022 Feb 11.
Artículo en Inglés | MEDLINE | ID: mdl-35216132

RESUMEN

Duchenne muscular dystrophy (DMD) is the most common and cureless muscle pediatric genetic disease, which is caused by the lack or the drastically reduced expression of dystrophin. Experimental therapeutic approaches for DMD have been mainly focused in recent years on attempts to restore the expression of dystrophin. While significant progress was achieved, the therapeutic benefit of treated patients is still unsatisfactory. Efficiency in gene therapy for DMD is hampered not only by incompletely resolved technical issues, but likely also due to the progressive nature of DMD. It is indeed suspected that some of the secondary pathologies, which are evolving over time in DMD patients, are not fully corrected by the restoration of dystrophin expression. We recently identified perturbations of the mevalonate pathway and of cholesterol metabolism in DMD patients. Taking advantage of the mdx model for DMD, we then demonstrated that some of these perturbations are improved by treatment with the cholesterol-lowering drug, simvastatin. In the present investigation, we tested whether the combination of the restoration of dystrophin expression with simvastatin treatment could have an additive beneficial effect in the mdx model. We confirmed the positive effects of microdystrophin, and of simvastatin, when administrated separately, but detected no additive effect by their combination. Thus, the present study does not support an additive beneficial effect by combining dystrophin restoration with a metabolic normalization by simvastatin.


Asunto(s)
Distrofia Muscular de Duchenne/tratamiento farmacológico , Distrofia Muscular de Duchenne/terapia , Simvastatina/administración & dosificación , Animales , Modelos Animales de Enfermedad , Terapia Genética/métodos , Masculino , Ratones , Ratones Endogámicos mdx , Músculo Esquelético/efectos de los fármacos
3.
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
4.
Mol Ther ; 22(6): 1176-1187, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24618805

RESUMEN

Muscular dystrophies are a group of genetically distinct diseases for which no treatment exists. While gene transfer approach is being tested for several of these diseases, such strategies can be hampered when the size of the corresponding complementary DNA (cDNA) exceeds the packaging capacity of adeno-associated virus vectors. This issue concerns, in particular, dysferlinopathies and titinopathies that are due to mutations in the dysferlin (DYSF) and titin (TTN) genes. We investigated the efficacy of RNA trans-splicing as a mode of RNA therapy for these two types of diseases. Results obtained with RNA trans-splicing molecules designed to target the 3' end of mouse titin and human dysferlin pre-mRNA transcripts indicated that trans-splicing of pre-mRNA generated from minigene constructs or from the endogenous genes was achieved. Collectively, these results provide the first demonstration of DYSF and TTN trans-splicing reprogramming in vitro and in vivo. However, in addition to these positive results, we uncovered a possible issue of the technique in the form of undesirable translation of RNA pre-trans-splicing molecules, directly from open reading frames present on the molecule or associated with internal alternative cis-splicing. These events may hamper the efficiency of the trans-splicing process and/or lead to toxicity.


Asunto(s)
Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Distrofias Musculares/terapia , Proteínas Quinasas/genética , Proteínas Quinasas/metabolismo , Precursores del ARN/genética , ARN Mensajero/metabolismo , Empalme Alternativo , Animales , Línea Celular , Disferlina , Humanos , Ratones , Ratones Endogámicos C57BL , Terapia Molecular Dirigida , Distrofias Musculares/genética , Sistemas de Lectura Abierta , Trans-Empalme
5.
Am J Pathol ; 182(6): 2298-309, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23624156

RESUMEN

Dysferlinopathy refers to a group of autosomal recessive muscular dystrophies due to mutations in the dysferlin gene causing deficiency of a membrane-bound protein crucially involved in plasma membrane repair. The condition is characterized by marked clinical heterogeneity, the different phenotypes/modes of presentation being unrelated to the genotype. For unknown reasons, patients are often remarkably active before the onset of symptoms. Dysferlin deficiency-related persistence of mechanically induced sarcolemma disruptions causes myofiber damage and necrosis. We postulate that limited myodamage may initially remain hidden with well-preserved resistance to physical strains. By subjecting dysferlin-deficient B6.A/J-Dysf(prmd) mice to long-term swimming exercise, we observed that concentric/isometric strain improved muscle strength and alleviated muscular dystrophy by limiting the accumulation of membrane lesions. By contrast, eccentric strain induced by long-term running in a wheel worsened the dystrophic process. Myofiber damage induced by eccentric strain increased with age, reflecting the accumulation of non-necrotic membrane lesions up to a critical threshold. This phenomenon was modulated by daily spontaneous activity. Transposed to humans, our results may suggest that the past activity profile shapes the clinical phenotype of the myopathy and that patients with dysferlinopathy should likely benefit from concentric exercise-based physiotherapy.


Asunto(s)
Distrofia Muscular de Cinturas/rehabilitación , Condicionamiento Físico Animal/fisiología , Envejecimiento/patología , Envejecimiento/fisiología , Animales , Membrana Celular/ultraestructura , Disferlina , Locomoción/fisiología , Proteínas de la Membrana/deficiencia , Ratones , Ratones Mutantes , Microscopía Electrónica , Contracción Muscular/fisiología , Fibras Musculares Esqueléticas/patología , Fuerza Muscular/fisiología , Músculo Esquelético/fisiopatología , Distrofia Muscular de Cinturas/etiología , Distrofia Muscular de Cinturas/patología , Distrofia Muscular de Cinturas/fisiopatología , Necrosis , Músculo Cuádriceps/ultraestructura , Carrera/fisiología , Natación/fisiología
6.
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
7.
Hum Mol Genet ; 19(10): 1897-907, 2010 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-20154340

RESUMEN

Deficiency of the dysferlin protein presents as two major clinical phenotypes: limb-girdle muscular dystrophy type 2B and Miyoshi myopathy. Dysferlin is known to participate in membrane repair, providing a potential hypothesis to the underlying pathophysiology of these diseases. The size of the dysferlin cDNA prevents its direct incorporation into an adeno-associated virus (AAV) vector for therapeutic gene transfer into muscle. To bypass this limitation, we split the dysferlin cDNA at the exon 28/29 junction and cloned it into two independent AAV vectors carrying the appropriate splicing sequences. Intramuscular injection of the corresponding vectors into a dysferlin-deficient mouse model led to the expression of full-length dysferlin for at least 1 year. Importantly, systemic injection in the tail vein of the two vectors led to a widespread although weak expression of the full-length protein. Injections were associated with an improvement of the histological aspect of the muscle, a reduction in the number of necrotic fibers, restoration of membrane repair capacity and a global improvement in locomotor activity. Altogether, these data support the use of such a strategy for the treatment of dysferlin deficiency.


Asunto(s)
Dependovirus/genética , Técnicas de Transferencia de Gen , Terapia Genética , Vectores Genéticos/genética , Proteínas de la Membrana/deficiencia , Proteínas de la Membrana/uso terapéutico , Proteínas Musculares/deficiencia , Proteínas Musculares/uso terapéutico , Distrofia Muscular de Cinturas/genética , Animales , Cruzamientos Genéticos , Disferlina , Femenino , Inyecciones Intramusculares , Masculino , Proteínas de la Membrana/genética , Membranas/patología , Ratones , Ratones Endogámicos C57BL , Proteínas Musculares/genética , Músculo Esquelético/patología , Distrofia Muscular de Cinturas/terapia , Mutación , Fenotipo , Transgenes , Cicatrización de Heridas
8.
Front Pharmacol ; 13: 856804, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35571097

RESUMEN

Limb-girdle muscular dystrophy type R3 (LGMD R3) is a rare genetic disorder characterized by a progressive proximal muscle weakness and caused by mutations in the SGCA gene encoding alpha-sarcoglycan (α-SG). Here, we report the results of a mechanistic screening ascertaining the molecular mechanisms involved in the degradation of the most prevalent misfolded R77C-α-SG protein. We performed a combinatorial study to identify drugs potentializing the effect of a low dose of the proteasome inhibitor bortezomib on the R77C-α-SG degradation inhibition. Analysis of the screening associated to artificial intelligence-based predictive ADMET characterization of the hits led to identification of the HDAC inhibitor givinostat as potential therapeutical candidate. Functional characterization revealed that givinostat effect was related to autophagic pathway inhibition, unveiling new theories concerning degradation pathways of misfolded SG proteins. Beyond the identification of a new therapeutic option for LGMD R3 patients, our results shed light on the potential repurposing of givinostat for the treatment of other genetic diseases sharing similar protein degradation defects such as LGMD R5 and cystic fibrosis.

9.
Biochim Biophys Acta ; 1793(12): 1886-93, 2009 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19781581

RESUMEN

Mechanically damaged plasma membrane undergoes rapid calcium-dependent resealing that appears to depend, at least in part, on calpain-mediated cortical cytoskeletal remodeling. Cells null for Capns1, the non-catalytic small subunit present in both m- and mu-calpains, do not undergo calcium-mediated resealing. However, it is not known which of these calpains is needed for repair, or whether other major cytosolic proteinases may participate. Utilizing isozyme-selective siRNAs to decrease expression of Capn1 or Capn2, catalytic subunits of mu- and m-calpains, respectively, in a mouse embryonic fibroblast cell line, we now show that substantial loss of both activities is required to compromise calcium-mediated survival after cell scrape-damage. Using skeletal myotubes derived from Capn3-null mice, we were unable to demonstrate loss of sarcolemma resealing after needle scratch or laser damage. Isolated muscle fibers from Capn3 knockout mice also efficiently repaired laser damage. Employing either a cell line expressing a temperature sensitive E1 ubiquitin ligase, or lactacystin, a specific proteasome inhibitor, it was not possible to demonstrate an effect of the proteasome on calcium-mediated survival after injury. Moreover, several cell-permeant caspase inhibitors were incapable of significantly decreasing survival or inhibiting membrane repair. Taken together with previous studies, the results show that m- or mu-calpain can facilitate repair of damaged plasma membrane. While there was no evidence for the involvement of calpain-3, the proteasome or caspases in early events of plasma membrane repair, our studies do not rule out their participation in downstream events that may link plasma membrane repair to adaptive remodeling after injury.


Asunto(s)
Calcio/metabolismo , Calpaína/metabolismo , Caspasas/metabolismo , Membrana Celular/enzimología , Complejo de la Endopetidasa Proteasomal/metabolismo , Animales , Calpaína/genética , Caspasas/genética , Línea Celular , Membrana Celular/genética , Ratones , Ratones Noqueados , Proteínas Musculares , Complejo de la Endopetidasa Proteasomal/genética
10.
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
11.
Sci Rep ; 9(1): 6915, 2019 05 06.
Artículo en Inglés | MEDLINE | ID: mdl-31061434

RESUMEN

Limb-girdle muscular dystrophy type 2D (LGMD2D) is characterized by a progressive proximal muscle weakness. LGMD2D is caused by mutations in the gene encoding α-sarcoglycan (α-SG), a dystrophin-associated glycoprotein that plays a key role in the maintenance of sarcolemma integrity in striated muscles. We report here on the development of a new in vitro high-throughput screening assay that allows the monitoring of the proper localization of the most prevalent mutant form of α-SG (R77C substitution). Using this assay, we screened a library of 2560 FDA-approved drugs and bioactive compounds and identified thiostrepton, a cyclic antibiotic, as a potential drug to repurpose for LGMD2D treatment. Characterization of the thiostrepton effect revealed a positive impact on R77C-α-SG and other missense mutant protein localization (R34H, I124T, V247M) in fibroblasts overexpressing these proteins. Finally, further investigations of the molecular mechanisms of action of the compound revealed an inhibition of the chymotrypsin-like activity of the proteasome 24 h after thiostrepton treatment and a synergistic effect with bortezomib, an FDA-approved proteasome inhibitor. This study reports on the first in vitro model for LGMD2D that is compatible with high-throughput screening and proposes a new therapeutic option for LGMD2D caused by missense mutations of α-SG.


Asunto(s)
Proteínas Mutantes/química , Proteínas Mutantes/metabolismo , Pliegue de Proteína/efectos de los fármacos , Proteolisis/efectos de los fármacos , Sarcoglicanos/química , Sarcoglicanos/metabolismo , Tioestreptona/farmacología , Línea Celular , Membrana Celular/efectos de los fármacos , Membrana Celular/metabolismo , Evaluación Preclínica de Medicamentos , Humanos , Células Madre Pluripotentes Inducidas/citología , Proteínas Mutantes/genética , Mioblastos/citología , Mioblastos/efectos de los fármacos , Sarcoglicanos/genética
12.
Sci Transl Med ; 11(520)2019 11 27.
Artículo en Inglés | MEDLINE | ID: mdl-31776291

RESUMEN

Limb-girdle muscular dystrophy type 2A (LGMD2A or LGMDR1) is a neuromuscular disorder caused by mutations in the calpain 3 gene (CAPN3). Previous experiments using adeno-associated viral (AAV) vector-mediated calpain 3 gene transfer in mice indicated cardiac toxicity associated with the ectopic expression of the calpain 3 transgene. Here, we performed a preliminary dose study in a severe double-knockout mouse model deficient in calpain 3 and dysferlin. We evaluated safety and biodistribution of AAV9-desmin-hCAPN3 vector administration to nonhuman primates (NHPs) with a dose of 3 × 1013 viral genomes/kg. Vector administration did not lead to observable adverse effects or to detectable toxicity in NHP. Of note, the transgene expression did not produce any abnormal changes in cardiac morphology or function of injected animals while reaching therapeutic expression in skeletal muscle. Additional investigation on the underlying causes of cardiac toxicity observed after gene transfer in mice and the role of titin in this phenomenon suggest species-specific titin splicing. Mice have a reduced capacity for buffering calpain 3 activity compared to NHPs and humans. Our studies highlight a complex interplay between calpain 3 and titin binding sites and demonstrate an effective and safe profile for systemic calpain 3 vector delivery in NHP, providing critical support for the clinical potential of calpain 3 gene therapy in humans.


Asunto(s)
Calpaína/genética , Calpaína/uso terapéutico , Cardiotoxicidad/etiología , Conectina/genética , Terapia Genética/efectos adversos , Proteínas Musculares/genética , Proteínas Musculares/uso terapéutico , Distrofia Muscular de Cinturas/genética , Distrofia Muscular de Cinturas/terapia , Empalme del ARN/genética , Animales , Sitios de Unión , Biomarcadores/sangre , Cardiotoxicidad/sangre , Conectina/química , Dependovirus/genética , Disferlina/deficiencia , Disferlina/metabolismo , Estabilidad de Enzimas , Regulación de la Expresión Génica , Ratones Noqueados , MicroARNs/genética , MicroARNs/metabolismo , Músculo Esquelético/metabolismo , Distrofia Muscular de Cinturas/sangre , Distrofia Muscular de Cinturas/patología , Miocardio/metabolismo , Miocardio/patología , Primates , Dominios Proteicos , Proteolisis , Especificidad de la Especie , Distribución Tisular , Transgenes
14.
PLoS One ; 13(1): e0191274, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29360879

RESUMEN

Sarcoglycanopathies are rare autosomic limb girdle muscular dystrophies caused by mutations in one of the genes coding for sarcoglycan (α, ß, δ, and γ-sarcoglycans). Sarcoglycans form a complex, which is an important part of the dystrophin-associated glycoprotein complex that protects sarcolemma against muscle contraction-induced damages. Absence of one of the sarcoglycan at the plasma membrane induces the disappearance of the whole complex and perturbs muscle fiber membrane integrity. We previously demonstrated that point mutations in the human sarcoglycan genes affects the folding of the corresponding protein, which is then retained in the endoplasmic reticulum by the protein quality control and prematurely degraded by the proteasome. Interestingly, modulation of the quality control using pharmacological compounds allowed the rescue of the membrane localization of the mutated sarcoglycan. Two previously generated mouse models, knock-in for the most common sarcoglycan mutant, R77C α-sarcoglycan, failed in reproducing the dystrophic phenotype observed in human patients. Based on these results and the need to test therapies for these fatal diseases, we decided to generate a new knock-in mouse model carrying the missense mutation T151R in the ß-sarcoglycan gene since this is the second sarcoglycan protein with the most frequently reported missense mutations. Muscle analysis, performed at the age of 4 and 9-months, showed the presence of the mutated ß-sarcoglycan protein and of the other components of the dystrophin-associated glycoprotein complex at the muscle membrane. In addition, these mice did not develop a dystrophic phenotype, even at a late stage or in condition of stress-inducing exercise. We can speculate that the absence of phenotype in mouse may be due to a higher tolerance of the endoplasmic reticulum quality control for amino-acid changes in mice compared to human.


Asunto(s)
Distrofia Muscular de Cinturas/genética , Mutación Missense , Sarcoglicanos/genética , Secuencia de Aminoácidos , Sustitución de Aminoácidos , Animales , Modelos Animales de Enfermedad , Femenino , Humanos , Masculino , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Transgénicos , Músculo Esquelético/metabolismo , Músculo Esquelético/patología , Distrofia Muscular de Cinturas/metabolismo , Distrofia Muscular de Cinturas/patología , Proteolisis , Sarcoglicanos/metabolismo , Especificidad de la Especie
15.
Mol Cell Biol ; 23(24): 9127-35, 2003 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-14645524

RESUMEN

Calpain 3 (Capn3) is known as the skeletal muscle-specific member of the calpains, a family of intracellular nonlysosomal cysteine proteases. This enigmatic protease has many unique features among the calpain family and, importantly, mutations in Capn3 have been shown to be responsible for limb girdle muscular dystrophy type 2A. Here we demonstrate that the Capn3 activation mechanism is similar to the universal activation of caspases and corresponds to an autolysis within the active site of the protease. We undertook a search for substrates in immature muscle cells, as several lines of evidence suggest that Capn3 is mostly in an inactive state in muscle and needs a signal to be activated. In this model, Capn3 proteolytic activity leads to disruption of the actin cytoskeleton and disorganization of focal adhesions through cleavage of several endogenous proteins. In addition, we show that titin, a previously identified Capn3 partner, and filamin C are further substrates of Capn3. Finally, we report that Capn3 colocalizes in vivo with its substrates at various sites along cytoskeletal structures. We propose that Capn3-mediated cleavage produces an adaptive response of muscle cells to external and/or internal stimuli, establishing Capn3 as a muscle cytoskeleton regulator.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales , Calpaína/metabolismo , Músculo Esquelético/enzimología , Actinas/metabolismo , Animales , Autólisis , Calpaína/química , Calpaína/genética , Dominio Catalítico , Células Cultivadas , Conectina , Proteínas Contráctiles/metabolismo , Proteínas del Citoesqueleto , Citoesqueleto/enzimología , Activación Enzimática , Filaminas , Técnicas In Vitro , Ratones , Proteínas de Microfilamentos/metabolismo , Modelos Biológicos , Proteínas Musculares/metabolismo , Células 3T3 NIH , Fosfoproteínas/metabolismo , Proteínas Quinasas/metabolismo , Estructura Terciaria de Proteína , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Sarcolema/enzimología , Sarcómeros/enzimología , Especificidad por Sustrato , Talina/metabolismo
16.
J Mol Biol ; 346(1): 215-22, 2005 Feb 11.
Artículo en Inglés | MEDLINE | ID: mdl-15663939

RESUMEN

Constant efforts are ongoing for the development of new imaging methods that allow the investigation of molecular processes in vivo. Protein-protein interactions, enzymatic activities and intracellular Ca2+ fluxes, have been resolved in cultured cells using a variety of fluorescence resonance energy transfer (FRET) detection methods. However, FRET has not been used so far in conjunction with 3D intravital imaging. We evaluated here a combination of multiphoton microscopy (MPM), method of choice for non-destructive living tissue investigation, and FRET imaging to monitor calpain proteolytic activity in living mice muscle. We show that kinetics of ubiquitous calpains activation can be efficiently and quantitatively monitored in living mouse tissues at cellular level with a FRET-based indicator upon calcium influx. The ability to visualize calpain activity in living tissue offers a unique opportunity to challenge remaining questions on the biological functions of calpains and to evaluate the therapeutic potential of calpain inhibitors in many degenerative conditions.


Asunto(s)
Calpaína/metabolismo , Transferencia Resonante de Energía de Fluorescencia/instrumentación , Transferencia Resonante de Energía de Fluorescencia/métodos , Animales , Calcio/farmacología , Calibración , Ratones , Músculos/efectos de los fármacos , Músculos/enzimología , Fotones
17.
Mol Ther Methods Clin Dev ; 2: 15009, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26029720

RESUMEN

Recombinant adeno-associated virus (rAAV) is currently the best vector for gene delivery into the skeletal muscle. However, the 5-kb packaging size of this virus is a major obstacle for large gene transfer. This past decade, many different strategies were developed to circumvent this issue (concatemerization-splicing, overlapping vectors, hybrid dual or fragmented AAV). Loss of function mutations in the DYSF gene whose coding sequence is 6.2kb lead to progressive muscular dystrophies (LGMD2B: OMIM_253601; MM: OMIM_254130; DMAT: OMIM_606768). In this study, we compared large gene transfer techniques to deliver the DYSF gene into the skeletal muscle. After rAAV8s intramuscular injection into dysferlin deficient mice, we showed that the overlap strategy is the most effective approach to reconstitute a full-length messenger. After systemic administration, the level of dysferlin obtained on different muscles corresponded to 0.5- to 2-fold compared to the normal level. We further demonstrated that the overlapping vector set was efficient to correct the histopathology, resistance to eccentric contractions and whole body force in the dysferlin deficient mice. Altogether, these data indicate that using overlapping vectors could be a promising approach for a potential clinical treatment of dysferlinopathies.

18.
PLoS One ; 7(5): e38036, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22666441

RESUMEN

Mutations in the dysferlin gene are the cause of Limb-girdle Muscular Dystrophy type 2B and Miyoshi Myopathy. The dysferlin protein has been implicated in sarcolemmal resealing, leading to the idea that the pathophysiology of dysferlin deficiencies is due to a deficit in membrane repair. Here, we show using two different approaches that fulfilling membrane repair as asseyed by laser wounding assay is not sufficient for alleviating the dysferlin deficient pathology. First, we generated a transgenic mouse overexpressing myoferlin to test the hypothesis that myoferlin, which is homologous to dysferlin, can compensate for the absence of dysferlin. The myoferlin overexpressors show no skeletal muscle abnormalities, and crossing them with a dysferlin-deficient model rescues the membrane fusion defect present in dysferlin-deficient mice in vitro. However, myoferlin overexpression does not correct muscle histology in vivo. Second, we report that AAV-mediated transfer of a minidysferlin, previously shown to correct the membrane repair deficit in vitro, also fails to improve muscle histology. Furthermore, neither myoferlin nor the minidysferlin prevented myofiber degeneration following eccentric exercise. Our data suggest that the pathogenicity of dysferlin deficiency is not solely related to impairment in sarcolemmal repair and highlight the care needed in selecting assays to assess potential therapies for dysferlinopathies.


Asunto(s)
Membrana Celular/metabolismo , Membrana Celular/patología , Terapia Genética/métodos , Distrofia Muscular de Cinturas/genética , Distrofia Muscular de Cinturas/patología , Animales , Efecto Espectador/genética , Dependovirus/genética , Disferlina , Femenino , Eliminación de Gen , Regulación de la Expresión Génica/genética , Humanos , Masculino , Fusión de Membrana/genética , Proteínas de la Membrana/deficiencia , Proteínas de la Membrana/genética , Ratones , Ratones Transgénicos , Proteínas Musculares/genética , Músculos/metabolismo , Músculos/patología , Músculos/fisiopatología , Distrofia Muscular de Cinturas/terapia , Fenotipo , Sarcolema/metabolismo , Sarcolema/patología , Resultado del Tratamiento
19.
FEBS J ; 277(20): 4322-37, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20860623

RESUMEN

A multiprotein complex encompassing a transcription regulator, cardiac ankyrin repeat protein (CARP), and the calpain 3 protease was identified in the N2A elastic region of the giant sarcomeric protein titin. The present study aimed to investigate the function(s) of this complex in the skeletal muscle. We demonstrate that CARP subcellular localization is controlled by the activity of calpain 3: the higher the calpain 3, the more important the sarcomeric retention of CARP. This regulation would occur through cleavage of the N-terminal end of CARP by the protease. We show that, upon CARP over-expression, the transcription factor nuclear factor NF-κB p65 DNA-binding activity decreases. Taken as a whole, CARP and its regulator calpain 3 appear to occupy a central position in the important cell fate-governing NF-κB pathway. Interestingly, the expression of the atrophying protein MURF1, one of NF-κB main targets, remains unchanged in presence of CARP, suggesting that the pathway encompassing calpain 3/CARP/NF-κB does not play a role in muscle atrophy. With NF-κB also having anti-apoptotic effects, the inability of calpain 3 to lower CARP-driven inhibition of NF-κB could reduce muscle cell survival, hence partly accounting for the dystrophic pattern observed in limb girdle muscular dystrophy 2A, a pathology resulting from the protease deficiency.


Asunto(s)
Calpaína/metabolismo , Proteínas Musculares/metabolismo , Músculo Esquelético/metabolismo , FN-kappa B/metabolismo , Proteínas Nucleares/metabolismo , Proteínas Represoras/metabolismo , Transducción de Señal/fisiología , Humanos , Distrofia Muscular de Cinturas/patología , FN-kappa B/antagonistas & inhibidores
20.
FEBS J ; 276(3): 669-84, 2009 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19143834

RESUMEN

In an attempt to identify potential therapeutic targets for the correction of muscle wasting, the gene expression of several pivotal proteins involved in protein metabolism was investigated in experimental atrophy induced by transient or definitive denervation, as well as in four animal models of muscular dystrophies (deficient for calpain 3, dysferlin, alpha-sarcoglycan and dystrophin, respectively). The results showed that: (a) the components of the ubiquitin-proteasome pathway are upregulated during the very early phases of atrophy but do not greatly increase in the muscular dystrophy models; (b) forkhead box protein O1 mRNA expression is augmented in the muscles of a limb girdle muscular dystrophy 2A murine model; and (c) the expression of cardiac ankyrin repeat protein (CARP), a regulator of transcription factors, appears to be persistently upregulated in every condition, suggesting that CARP could be a hub protein participating in common pathological molecular pathway(s). Interestingly, the mRNA level of a cell cycle inhibitor known to be upregulated by CARP in other tissues, p21(WAF1/CIP1), is consistently increased whenever CARP is upregulated. CARP overexpression in muscle fibres fails to affect their calibre, indicating that CARP per se cannot initiate atrophy. However, a switch towards fast-twitch fibres is observed, suggesting that CARP plays a role in skeletal muscle plasticity. The observation that p21(WAF1/CIP1) is upregulated, put in perspective with the effects of CARP on the fibre type, fits well with the idea that the mechanisms at stake might be required to oppose muscle remodelling in skeletal muscle.


Asunto(s)
Músculo Esquelético/metabolismo , Atrofia Muscular/metabolismo , Atrofia Muscular/patología , Distrofias Musculares/metabolismo , Distrofias Musculares/patología , Proteínas Nucleares/metabolismo , Proteínas Represoras/metabolismo , Animales , Biomarcadores/metabolismo , Calpaína/deficiencia , Calpaína/genética , Calpaína/metabolismo , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/genética , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Modelos Animales de Enfermedad , Proteína Forkhead Box O1 , Factores de Transcripción Forkhead/metabolismo , Perfilación de la Expresión Génica , Masculino , Ratones , Proteínas Musculares/deficiencia , Proteínas Musculares/genética , Proteínas Musculares/metabolismo , Atrofia Muscular/genética , Distrofias Musculares/genética , Proteínas Nucleares/genética , Complejo de la Endopetidasa Proteasomal/metabolismo , Proteínas Represoras/genética , Transducción de Señal , Regulación hacia Arriba
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA