Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 85
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Am J Pathol ; 192(11): 1604-1618, 2022 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-36113555

RESUMEN

Duchenne muscular dystrophy (DMD) is a severe neuromuscular disease caused by Dmd mutations, resulting in the absence of dystrophin in skeletal muscle, and a greater susceptibility to damage during contraction (exercise). The current study evaluated whether voluntary exercise impacts a Dmd exon skipping and muscle physiology in a severe DMD murine model. D2-mdx mice were intramuscularly injected with an adeno-associated virus (AAV) U7 snRNA to correct Dmd reading frame, and allowed to voluntary run on a wheel for 1 month. Voluntary running did not induce muscle fiber regeneration, as indicated by the percentage of centronucleated fibers, Myh3 and Myh4 expression, and maximal force production, and thus possibly did not compromise the gene therapy approach. Voluntary running did not impact the number of viral genomes and the expression of U7 and Dmd 1 month after injection of AAV-U7 injected just before exercise initiation, but reduced the amount of dystrophin in dystrophin-expressing fibers from 80% to 65% of the muscle cross-sectional area. In conclusion, voluntary running did not induce muscle damage and had no drastic detrimental effect on the AAV gene therapy exon skipping approach in a severe murine DMD model. Moreover, these results suggest considering exercise as an additional element in the design and conception of future therapeutic approaches for DMD.

2.
Clin Exp Pharmacol Physiol ; 50(9): 749-756, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37381823

RESUMEN

Skeletal muscles in animal models of Duchenne muscular dystrophy (DMD) are more susceptible to contraction-induced functional loss, which is not related to fatigue. Valproic acid (VPA) reportedly improves serological and histological markers of damage in dystrophin-deficient murine muscle. Here, we tested whether VPA would reduce the susceptibility to contraction-induced functional loss in two murine DMD models. Adult female mdx (mild) and D2-mdx (severe) DMD murine models were administered VPA (240 mg/kg) or saline for 7 days. Some VPA-treated mdx mice also performed voluntary running in a wheel, which is known to reduce the susceptibility to contraction-induced functional loss; that is, isometric force drop following eccentric contractions. In situ muscle function was assessed before, during and after eccentric contractions. Muscle utrophin and desmin expression were also evaluated using immunoblotting. Interestingly, VPA reduced the isometric force drop following eccentric contractions in both murine models, without change in the relative eccentric maximal force and in the expression of utrophin and desmin. VPA for 7 days combined with voluntary running had no additive effect compared to VPA alone. Furthermore, VPA reduced the absolute isometric maximal force before eccentric contractions in both murine models. The results of our study indicated that VPA in both murine DMD models reduced the susceptibility to contraction-induced functional loss but increased muscle weakness.


Asunto(s)
Distrofia Muscular de Duchenne , Femenino , Animales , Ratones , Distrofia Muscular de Duchenne/tratamiento farmacológico , Distrofia Muscular de Duchenne/metabolismo , Distrofia Muscular de Duchenne/patología , Ácido Valproico/farmacología , Ácido Valproico/metabolismo , Ratones Endogámicos mdx , Utrofina/metabolismo , Modelos Animales de Enfermedad , Desmina/metabolismo , Contracción Muscular/fisiología , Músculo Esquelético/metabolismo
3.
Nucleic Acids Res ; 49(8): 4472-4492, 2021 05 07.
Artículo en Inglés | MEDLINE | ID: mdl-33836079

RESUMEN

Skeletal muscle is a dynamic tissue the size of which can be remodeled through the concerted actions of various cues. Here, we investigated the skeletal muscle transcriptional program and identified key tissue-specific regulatory genetic elements. Our results show that Myod1 is bound to numerous skeletal muscle enhancers in collaboration with the glucocorticoid receptor (GR) to control gene expression. Remarkably, transcriptional activation controlled by these factors occurs through direct contacts with the promoter region of target genes, via the CpG-bound transcription factor Nrf1, and the formation of Ctcf-anchored chromatin loops, in a myofiber-specific manner. Moreover, we demonstrate that GR negatively controls muscle mass and strength in mice by down-regulating anabolic pathways. Taken together, our data establish Myod1, GR and Nrf1 as key players of muscle-specific enhancer-promoter communication that orchestrate myofiber size regulation.


Asunto(s)
Cromatina/metabolismo , Elementos de Facilitación Genéticos , Músculo Esquelético/metabolismo , Proteína MioD/metabolismo , Factor Nuclear 1 de Respiración/metabolismo , Receptores de Glucocorticoides/metabolismo , Animales , Línea Celular , Cromatina/genética , Secuenciación de Inmunoprecipitación de Cromatina , Regulación de la Expresión Génica/genética , Histonas/genética , Histonas/metabolismo , Masculino , Redes y Vías Metabólicas/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Fuerza Muscular/genética , Músculo Esquelético/fisiología , Proteína MioD/genética , Mioblastos/metabolismo , Factor Nuclear 1 de Respiración/genética , Receptores de Glucocorticoides/genética , Proteínas Recombinantes
4.
Hum Mol Genet ; 28(18): 3091-3100, 2019 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-31179493

RESUMEN

Duchenne muscular dystrophy (DMD) is a rare genetic disease affecting 1 in 3500-5000 newborn boys. It is due to mutations in the DMD gene with a consequent lack of dystrophin protein that leads to deterioration of myofibres and their replacement with fibro-adipogenic tissue. Out-of-frame mutations in the DMD gene can be modified by using antisense oligonucleotides (AONs) to promote skipping of specific exons such that the reading frame is restored and the resulting protein produced, though truncated, is functional. We have shown that AONs can also be used to knock down myostatin, a negative regulator of muscle growth and differentiation, through disruption of the transcript reading frame, and thereby enhance muscle strength. In young mdx mice, combined dystrophin and myostatin exon skipping therapy greatly improved DMD pathology, compared to the single dystrophin skipping approach. Here we show that in aged (>15-month-old) mdx mice, when the pathology is significantly more severe and more similar to the one observed in DMD patients, the effect of the combined therapy is slightly attenuated but still beneficial in improving the disease phenotype. These results confirm the beneficial outcome of the combination approach and support its translation into DMD clinical trials.


Asunto(s)
Distrofina/genética , Distrofina/metabolismo , Exones , Regulación de la Expresión Génica , Músculos/metabolismo , Miostatina/genética , Miostatina/metabolismo , Empalme del ARN , Animales , Modelos Animales de Enfermedad , Inmunohistoquímica , Ratones , Ratones Endogámicos mdx , Músculos/patología , Músculos/fisiopatología , Distrofia Muscular de Duchenne/genética , Distrofia Muscular de Duchenne/metabolismo , Distrofia Muscular de Duchenne/patología , Distrofia Muscular de Duchenne/fisiopatología , ARN Mensajero/genética
5.
J Physiol ; 598(17): 3667-3689, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32515007

RESUMEN

KEY POINTS: Desmin, similar to dystrophin, is associated with costameric structures bridging sarcomeres to the extracellular matrix. Deletion of the desmin gene in mdx mice [double knockout (DKO) mice] induces marked muscle weakness and fatigue resistance compared to mdx mice. Muscle fragility (higher susceptibility to contraction-induced injury) was also aggravated in DKO mice compared to mdx mice. By contrast to mdx mice, the DKO mice did not undergo muscle hypertrophy. Desmin cDNA transfer with adeno-associated virus in newborn mdx mice reduced muscle weakness. Overall, desmin plays important and beneficial roles in muscle wasting, performance and fragility in dystrophic muscle. ABSTRACT: Duchenne muscular dystrophy (DMD) is a severe neuromuscular disease caused by dystrophin deficiency. Desmin, similar to dystrophin, is associated with costameric structures bridging sarcomeres to the extracellular matrix that contributes to muscle function. In the present study, we attempted to provide further insight into the roles of desmin, for which the expression is increased in the muscle from the mouse mdx DMD model. We show that a deletion of the desmin gene (Des) in mdx mice [double knockout (DKO) mice, mdx:desmin-/-] induces a marked muscle weakness; namely, a reduced absolute maximal force production and increased fatigue compared to that in mdx mice. Fragility (i.e. higher susceptibility to contraction-induced injury) was also aggravated in DKO mice compared to mdx mice, despite the promotion of supposedly less fragile muscle fibres in DKO mice, and this worsening of fragility was related to a decreased muscle excitability. Moreover, in contrast to mdx mice, the DKO mice did not undergo muscle hypertrophy, as indicated by smaller and fewer fibres, with a reduced percentage of centronucleated fibres, potentially explaining the severe muscle weakness. Notably, Desmin cDNA transfer with adeno-associated virus in newborn mdx mice improved specific maximal force normalized to muscle weight. Overall, desmin plays important and beneficial roles in muscle wasting, performance and fragility in dystrophic mdx mice, which differ, at least in part, from those observed in healthy muscle.


Asunto(s)
Músculo Esquelético , Distrofia Muscular de Duchenne , Animales , Desmina/genética , Modelos Animales de Enfermedad , Distrofina/genética , Ratones , Ratones Endogámicos mdx , Distrofia Muscular de Duchenne/genética
6.
Int J Mol Sci ; 22(1)2020 Dec 30.
Artículo en Inglés | MEDLINE | ID: mdl-33396724

RESUMEN

Laminopathies are a clinically heterogeneous group of disorders caused by mutations in the LMNA gene, which encodes the nuclear envelope proteins lamins A and C. The most frequent diseases associated with LMNA mutations are characterized by skeletal and cardiac involvement, and include autosomal dominant Emery-Dreifuss muscular dystrophy (EDMD), limb-girdle muscular dystrophy type 1B, and LMNA-related congenital muscular dystrophy (LMNA-CMD). Although the exact pathophysiological mechanisms responsible for LMNA-CMD are not yet understood, severe contracture and muscle atrophy suggest that mutations may impair skeletal muscle growth. Using human muscle stem cells (MuSCs) carrying LMNA-CMD mutations, we observe impaired myogenic fusion with disorganized cadherin/ß catenin adhesion complexes. We show that skeletal muscle from Lmna-CMD mice is unable to hypertrophy in response to functional overload, due to defective fusion of activated MuSCs, defective protein synthesis and defective remodeling of the neuromuscular junction. Moreover, stretched myotubes and overloaded muscle fibers with LMNA-CMD mutations display aberrant mechanical regulation of the yes-associated protein (YAP). We also observe defects in MuSC activation and YAP signaling in muscle biopsies from LMNA-CMD patients. These phenotypes are not recapitulated in closely related but less severe EDMD models. In conclusion, combining studies in vitro, in vivo, and patient samples, we find that LMNA-CMD mutations interfere with mechanosignaling pathways in skeletal muscle, implicating A-type lamins in the regulation of skeletal muscle growth.


Asunto(s)
Lamina Tipo A/genética , Músculo Esquelético/metabolismo , Músculo Esquelético/patología , Distrofia Muscular de Cinturas/etiología , Distrofia Muscular de Cinturas/metabolismo , Mutación , Transducción de Señal , Animales , Biopsia , Comunicación Celular , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Modelos Animales de Enfermedad , Técnica del Anticuerpo Fluorescente , Expresión Génica , Estudios de Asociación Genética , Predisposición Genética a la Enfermedad , Genotipo , Humanos , Lamina Tipo A/metabolismo , Ratones , Fibras Musculares Esqueléticas/metabolismo , Músculo Esquelético/crecimiento & desarrollo , Distrofia Muscular de Cinturas/patología , Unión Neuromuscular/metabolismo , Fenotipo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
7.
Am J Pathol ; 188(11): 2662-2673, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30142334

RESUMEN

Dystrophin deficiency in mdx mice, a model for Duchenne muscular dystrophy, leads to muscle weakness revealed by a reduced specific maximal force as well as fragility (ie, higher susceptibility to contraction-induced injury, as shown by a greater force decrease after lengthening contractions). Both symptoms could be improved with dystrophin restoration-based therapies and long-term (months) voluntary exercise. Herein, we evaluated the effect of short-term (1-week) voluntary wheel running. We found that running improved fragility of tibialis anterior muscle (TA), but not plantaris muscle, independently of utrophin up-regulation, without affecting weakness. Moreover, TA muscle excitability was also preserved by running, as shown by compound muscle action potential measurements after lengthening contractions. Of interest, the calcineurin inhibitor cyclosporin A prevented the effect of running on both muscle fragility and excitability. Cyclosporin also prevented the running-induced changes in expression of genes involved in excitability (Scn4a and Cacna1s) and slower contractile phenotype (Myh2 and Tnni1) in TA muscle. In conclusion, short-term voluntary exercise improves TA muscle fragility in mdx mice, without worsening weakness. Its effect was related to preserved excitability, calcineurin pathway activation, and changes in the program of genes involved in excitability and slower contractile phenotype. Thus, remediation of muscle fragility of Duchenne muscular dystrophy patients through appropriate exercise training deserves to be explored in more detail.


Asunto(s)
Calcineurina/metabolismo , Distrofia Muscular Animal/prevención & control , Condicionamiento Físico Animal , Animales , Ratones , Ratones Endogámicos mdx , Actividad Motora , Contracción Muscular , Distrofia Muscular Animal/metabolismo , Distrofia Muscular Animal/patología
8.
Am J Pathol ; 187(3): 505-516, 2017 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-28056338

RESUMEN

Collagen IV is a major component of basement membranes (BMs). The α1(IV) chain, encoded by the COL4A1 gene, is expressed ubiquitously and associates with the α2(IV) chain to form the α1α1α2(IV) heterotrimer. Several COL4A1 mutations affecting a conformational domain containing integrin-binding sites are responsible for the systemic syndrome of hereditary angiopathy, nephropathy, aneurysms, and cramps (HANAC). To analyze the pathophysiology of HANAC, Col4a1 mutant mice bearing the p.Gly498Val mutation were generated. Analysis of the skeletal muscles of Col4a1G498V mutant animals showed morphologic characteristics of a muscular dystrophy phenotype with myofiber atrophy, centronucleation, focal inflammatory infiltrates, and fibrosis. Abnormal ultrastructural aspects of muscle BMs was associated with reduced extracellular secretion of the mutant α1α1α2(IV) trimer. In addition to muscular dystrophic features, endothelial cell defects of the muscle capillaries were observed, with intracytoplasmic accumulation of the mutant α1α1α2(IV) molecules, endoplasmic reticulum cisternae dilation, and up-regulation of endoplasmic reticulum stress markers. Induction of the unfolded protein response in Col4a1 mutant muscle tissue resulted in an excess of apoptosis in endothelial cells. HANAC mutant animals also presented with a muscular functional impairment and increased serum creatine kinase levels reflecting altered muscle fiber sarcolemma. This extensive description of the muscular phenotype of the Col4a1 HANAC murine model suggests a potential contribution of primary endothelial cell defects, together with muscle BM alterations, to the development of COL4A1-related myopathy.


Asunto(s)
Vasos Sanguíneos/anomalías , Colágeno Tipo IV/genética , Calambre Muscular/genética , Músculo Esquelético/irrigación sanguínea , Músculo Esquelético/patología , Mutación/genética , Enfermedad de Raynaud/genética , Animales , Apoptosis , Vasos Sanguíneos/patología , Peso Corporal , Creatina Quinasa/sangre , Distrofina/metabolismo , Estrés del Retículo Endoplásmico , Células Endoteliales/patología , Células Endoteliales/ultraestructura , Matriz Extracelular/metabolismo , Integrina beta1/metabolismo , Ratones , Ratones Mutantes , Músculo Esquelético/ultraestructura , Tamaño de los Órganos , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/metabolismo
9.
Mol Ther ; 25(9): 2038-2052, 2017 09 06.
Artículo en Inglés | MEDLINE | ID: mdl-28663100

RESUMEN

One of the most promising therapeutic approaches for familial amyotrophic lateral sclerosis linked to superoxide dismutase 1 (SOD1) is the suppression of toxic mutant SOD1 in the affected tissues. Here, we report an innovative molecular strategy for inducing substantial, widespread, and sustained reduction of mutant human SOD1 (hSOD1) levels throughout the body of SOD1G93A mice, leading to therapeutic effects in animals. Adeno-associated virus serotype rh10 vectors (AAV10) were used to mediate exon skipping of the hSOD1 pre-mRNA by expression of exon-2-targeted antisense sequences embedded in a modified U7 small-nuclear RNA (AAV10-U7-hSOD). Skipping of hSOD1 exon 2 led to the generation of a premature termination codon, inducing production of a deleted transcript that was subsequently degraded by the activation of nonsense-mediated decay. Combined intravenous and intracerebroventricular delivery of AAV10-U7-hSOD increased the survival of SOD1G93A mice injected either at birth or at 50 days of age (by 92% and 58%, respectively) and prevented weight loss and the decline of neuromuscular function. This study reports the effectiveness of an exon-skipping approach in SOD1-ALS mice, supporting the translation of this technology to the treatment of this as yet incurable disease.


Asunto(s)
Esclerosis Amiotrófica Lateral/genética , Dependovirus/genética , Terapia Genética , Vectores Genéticos/genética , Superóxido Dismutasa-1/genética , Edad de Inicio , Esclerosis Amiotrófica Lateral/mortalidad , Esclerosis Amiotrófica Lateral/fisiopatología , Esclerosis Amiotrófica Lateral/terapia , Animales , Modelos Animales de Enfermedad , Exones , Orden Génico , Técnicas de Transferencia de Gen , Terapia Genética/métodos , Vectores Genéticos/administración & dosificación , Humanos , Ratones , Ratones Transgénicos , Actividad Motora/genética , Oligonucleótidos Antisentido , Sitios de Empalme de ARN , ARN Mensajero/genética , ARN Mensajero/metabolismo , Recuperación de la Función , Superóxido Dismutasa-1/metabolismo , Tasa de Supervivencia , Transducción Genética
10.
Am J Physiol Endocrinol Metab ; 313(1): E12-E25, 2017 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-28351832

RESUMEN

To better define the role of male and female gonad-related factors (MGRF, presumably testosterone, and FGRF, presumably estradiol, respectively) on mouse hindlimb skeletal muscle contractile performance/function gain during postnatal development, we analyzed the effect of castration initiated before puberty in male and female mice. We found that muscle absolute and specific (normalized to muscle weight) maximal forces were decreased in 6-mo-old male and female castrated mice compared with age- and sex-matched intact mice, without alteration in neuromuscular transmission. Moreover, castration decreased absolute and specific maximal powers, another important aspect of muscle performance, in 6-mo-old males, but not in females. Absolute maximal force was similarly reduced by castration in 3-mo-old muscle fiber androgen receptor (AR)-deficient and wild-type male mice, indicating that the effect of MGRF was muscle fiber AR independent. Castration reduced the muscle weight gain in 3-mo mice of both sexes and in 6-mo females but not in males. We also found that bone morphogenetic protein signaling through Smad1/5/9 was not altered by castration in atrophic muscle of 3-mo-old mice of both sexes. Moreover, castration decreased the sexual dimorphism regarding muscle performance. Together, these results demonstrated that in the long term, MGRF and FGRF promote muscle performance gain in mice during postnatal development, independently of muscle growth in males, largely via improving muscle contractile quality (force and power normalized), and that MGFR and FGRF also contribute to sexual dimorphism. However, the mechanisms underlying MGFR and FGRF actions remain to be determined.


Asunto(s)
Envejecimiento/fisiología , Hormonas Esteroides Gonadales/metabolismo , Contracción Muscular/fisiología , Fuerza Muscular/fisiología , Músculo Esquelético/crecimiento & desarrollo , Animales , Animales Recién Nacidos , Peso Corporal/fisiología , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Fatiga Muscular/fisiología , Músculo Esquelético/metabolismo , Factores Sexuales
11.
Muscle Nerve ; 55(2): 254-261, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-27312354

RESUMEN

INTRODUCTION: The effect of constitutive inactivation of the gene encoding myostatin on the gain in muscle performance during postnatal growth has not been well characterized. METHODS: We analyzed 2 murine myostatin knockout (KO) models, (i) the Lee model (KOLee ) and (ii) the Grobet model (KOGrobet ), and measured the contraction of tibialis anterior muscle in situ. RESULTS: Absolute maximal isometric force was increased in 6-month-old KOLee and KOGrobet mice, as compared to wild-type mice. Similarly, absolute maximal power was increased in 6-month-old KOLee mice. In contrast, specific maximal force (relative maximal force per unit of muscle mass was decreased in all 6-month-old male and female KO mice, except in 6-month-old female KOGrobet mice, whereas specific maximal power was reduced only in male KOLee mice. CONCLUSIONS: Genetic inactivation of myostatin increases maximal force and power, but in return it reduces muscle quality, particularly in male mice. Muscle Nerve 55: 254-261, 2017.


Asunto(s)
Contracción Muscular/genética , Fuerza Muscular/genética , Músculo Esquelético/fisiología , Enfermedades Musculares/patología , Miostatina/deficiencia , Animales , Animales Recién Nacidos , Modelos Animales de Enfermedad , Femenino , Masculino , Ratones , Ratones Noqueados , Enfermedades Musculares/genética , Miostatina/genética , Factores Sexuales
12.
PLoS Genet ; 10(5): e1004386, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24852826

RESUMEN

Thousands of long intergenic non-coding RNAs (lincRNAs) are encoded by the mammalian genome. However, the function of most of these lincRNAs has not been identified in vivo. Here, we demonstrate a role for a novel lincRNA, linc-MYH, in adult fast-type myofiber specialization. Fast myosin heavy chain (MYH) genes and linc-MYH share a common enhancer, located in the fast MYH gene locus and regulated by Six1 homeoproteins. linc-MYH in nuclei of fast-type myofibers prevents slow-type and enhances fast-type gene expression. Functional fast-sarcomeric unit formation is achieved by the coordinate expression of fast MYHs and linc-MYH, under the control of a common Six-bound enhancer.


Asunto(s)
Proteínas de Homeodominio/genética , Contracción Muscular/genética , Cadenas Pesadas de Miosina/genética , ARN Largo no Codificante/genética , Animales , Clonación Molecular , Elementos de Facilitación Genéticos , Proteínas de la Matriz Extracelular/genética , Regulación del Desarrollo de la Expresión Génica , Proteínas de Homeodominio/metabolismo , Ratones Noqueados , Músculo Esquelético/crecimiento & desarrollo , Proteína-Lisina 6-Oxidasa/genética
13.
J Cell Sci ; 127(Pt 21): 4589-601, 2014 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-25179606

RESUMEN

Synemin, a type IV intermediate filament (IF) protein, forms a bridge between IFs and cellular membranes. As an A-kinase-anchoring protein, it also provides temporal and spatial targeting of protein kinase A (PKA). However, little is known about its functional roles in either process. To better understand its functions in muscle tissue, we generated synemin-deficient (Synm(-) (/-)) mice. Synm(-) (/-) mice displayed normal development and fertility but showed a mild degeneration and regeneration phenotype in myofibres and defects in sarcolemma membranes. Following mechanical overload, Synm(-) (/-) mice muscles showed a higher hypertrophic capacity with increased maximal force and fatigue resistance compared with control mice. At the molecular level, increased remodelling capacity was accompanied by decreased myostatin (also known as GDF8) and atrogin (also known as FBXO32) expression, and increased follistatin expression. Furthermore, the activity of muscle-mass control molecules (the PKA RIIα subunit, p70S6K and CREB1) was increased in mutant mice. Finally, analysis of muscle satellite cell behaviour suggested that the absence of synemin could affect the balance between self-renewal and differentiation of these cells. Taken together, our results show that synemin is necessary to maintain membrane integrity and regulates signalling molecules during muscle hypertrophy.


Asunto(s)
Hipertrofia/metabolismo , Proteínas de Filamentos Intermediarios/metabolismo , Músculo Esquelético/metabolismo , Músculo Esquelético/patología , Enfermedades Musculares/metabolismo , Animales , Diferenciación Celular/genética , Diferenciación Celular/fisiología , Desmina/genética , Desmina/metabolismo , Hipertrofia/patología , Proteínas de Filamentos Intermediarios/genética , Masculino , Ratones , Ratones Noqueados , Músculo Esquelético/ultraestructura , Enfermedades Musculares/genética
14.
Am J Pathol ; 185(7): 2012-24, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-26009153

RESUMEN

There is fear that mechanical overloading (OVL; ie, high-force contractions) accelerates Duchenne muscular dystrophy. Herein, we determined whether short-term OVL combined with wheel running, short-term OVL combined with irradiation, and long-term OVL are detrimental for hind limb mdx mouse muscle, a murine model of Duchene muscular dystrophy exhibiting milder dystrophic features. OVL was induced by the surgical ablation of the synergic muscles of the plantaris muscle, a fast muscle susceptible to contraction-induced muscle damage in mdx mice. We found that short-term OVL combined with wheel and long-term OVL did not worsen the deficit in specific maximal force (ie, absolute maximal force normalized to muscle size) and histological markers of muscle damage (percentage of regenerating fibers and fibrosis) in mdx mice. Moreover, long-term OVL did not increase the alteration in calcium homeostasis and did not deplete muscle cell progenitors expressing Pax 7 in mdx mice. Irradiation before short-term OVL, which is believed to inhibit muscle regeneration, was not more detrimental to mdx than control mice. Interestingly, short-term OVL combined with wheel and long-term OVL markedly improved the susceptibility to contraction-induced damage, increased absolute maximal force, induced hypertrophy, and promoted a slower, more oxidative phenotype. Together, these findings indicate that OVL is beneficial to mdx muscle, and muscle regeneration does not mask the potentially detrimental effect of OVL.


Asunto(s)
Músculo Esquelético/fisiopatología , Distrofia Muscular Animal/fisiopatología , Distrofia Muscular de Duchenne/fisiopatología , Animales , Modelos Animales de Enfermedad , Femenino , Hipertrofia , Extremidad Inferior , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos mdx , Actividad Motora , Contracción Muscular , Músculo Esquelético/efectos de la radiación , Mutación , Regeneración , Células Satélite del Músculo Esquelético/fisiología , Células Satélite del Músculo Esquelético/efectos de la radiación
15.
FASEB J ; 29(6): 2473-83, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25713059

RESUMEN

Adult skeletal muscle is a dynamic, remarkably plastic tissue, which allows myofibers to switch from fast/glycolytic to slow/oxidative types and to increase mitochondrial fatty acid oxidation (mFAO) capacity and vascularization in response to exercise training. mFAO is the main muscle energy source during endurance exercise, with carnitine palmitoyltransferase 1 (CPT1) being the key regulatory enzyme. Whether increasing muscle mFAO affects skeletal muscle physiology in adulthood actually remains unknown. To investigate this, we used in vivo electrotransfer technology to express in mouse tibialis anterior (TA), a fast/glycolytic muscle, a mutated CPT1 form (CPT1mt) that is active but insensitive to malonyl-CoA, its physiologic inhibitor. In young (2-mo-old) adult mice, muscle CPT1mt expression enhanced mFAO (+40%), but also increased the percentage of oxidative fibers (+28%), glycogen content, and capillary-to-fiber density (+45%). This CPT1mt-induced muscle remodeling, which mimicked exercise-induced oxidative phenotype, led to a greater resistance to muscle fatigue. In the context of aging, characterized by sarcopenia and reduced oxidative capacity, CPT1mt expression in TAs from aged (20-mo-old) mice partially reversed aging-associated sarcopenia and fiber-type transition, and increased muscle capillarity. These findings provide evidence that mFAO regulates muscle phenotype and may be a potential target to combat age-related decline in muscle function.


Asunto(s)
Carnitina O-Palmitoiltransferasa/metabolismo , Ácidos Grasos/metabolismo , Mitocondrias Musculares/metabolismo , Músculo Esquelético/metabolismo , Factores de Edad , Animales , Western Blotting , Carnitina O-Palmitoiltransferasa/genética , Expresión Génica , Glucógeno/metabolismo , Masculino , Ratones Endogámicos C57BL , Mitocondrias Musculares/fisiología , Fatiga Muscular/genética , Fatiga Muscular/fisiología , Músculo Esquelético/irrigación sanguínea , Músculo Esquelético/fisiología , Mutación , Oxidación-Reducción , Fenotipo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Sarcopenia/genética , Sarcopenia/fisiopatología , Transfección
16.
J Cell Sci ; 126(Pt 8): 1806-19, 2013 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-23444364

RESUMEN

The sarcoplasmic reticulum (SR) is a specialized form of endoplasmic reticulum (ER) in skeletal muscle and is essential for calcium homeostasis. The mechanisms involved in SR remodeling and maintenance of SR subdomains are elusive. In this study, we identified myotubularin (MTM1), a phosphoinositide phosphatase mutated in X-linked centronuclear myopathy (XLCNM, or myotubular myopathy), as a key regulator of phosphatidylinositol 3-monophosphate (PtdIns3P) levels at the SR. MTM1 is predominantly located at the SR cisternae of the muscle triads, and Mtm1-deficient mouse muscles and myoblasts from XLCNM patients exhibit abnormal SR/ER networks. In vivo modulation of MTM1 enzymatic activity in skeletal muscle using ectopic expression of wild-type or a dead-phosphatase MTM1 protein leads to differential SR remodeling. Active MTM1 is associated with flat membrane stacks, whereas dead-phosphatase MTM1 mutant promotes highly curved cubic membranes originating from the SR and enriched in PtdIns3P. Overexpression of a tandem FYVE domain with high affinity for PtdIns3P alters the shape of the SR cisternae at the triad. Our findings, supported by the parallel analysis of the Mtm1-null mouse and an in vivo study, reveal a direct function of MTM1 enzymatic activity in SR remodeling and a key role for PtdIns3P in promoting SR membrane curvature in skeletal muscle. We propose that alteration in SR remodeling is a primary cause of X-linked centronuclear myopathy. The tight regulation of PtdIns3P on specific membrane subdomains may be a general mechanism to control membrane curvature.


Asunto(s)
Músculo Esquelético/metabolismo , Fosfatos de Fosfatidilinositol/metabolismo , Proteínas Tirosina Fosfatasas no Receptoras/metabolismo , Retículo Sarcoplasmático/metabolismo , Animales , Western Blotting , Línea Celular , Inmunoprecipitación , Masculino , Ratones , Microscopía Electrónica de Transmisión , Músculo Esquelético/ultraestructura , Unión Proteica , Proteínas Tirosina Fosfatasas no Receptoras/genética
17.
J Nutr ; 145(7): 1429-37, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-26019250

RESUMEN

BACKGROUND: Aging is associated with profound metabolic disturbances, and citrulline may be of use to limit them. OBJECTIVE: The aim of this work was to evaluate the long-term effect of citrulline supplementation on metabolism in healthy aged rats. METHODS: Twenty-month-old male rats were randomly assigned to be fed (ad libitum) for 12 wk with either a citrulline-enriched diet (1 g ⋅ kg(-1) ⋅ d(-1)) or a standard diet [rendered isonitrogenous by addition of nonessential amino acids (NEAAs)]. Motor activity and muscle strength were measured, body composition was assessed, and muscle metabolism (protein structure, mitochondrial exploration, and transductional factors) and lipid metabolism (lipoprotein composition and sensitivity to oxidative stress) were explored. RESULTS: Compared with the NEAA-treated group, citrulline supplementation was associated with lower mortality (0% vs. 20%; P = 0.05), 9% higher lean body mass (P < 0.05), and 13% lower fat mass (P < 0.05). Compared with the NEAA-treated group, citrulline-treated rats had greater muscle mass (+14-48% depending on type of muscle; P < 0.05 for tibialis, gastrocnemius, and plantaris). Susceptibility to oxidation of lipoproteins, as measured by the maximal concentration of 7-ketocholesterol after copper-induced VLDL and LDL oxidation, was lower in citrulline-treated rats than in NEAA-treated rats (187 ± 8 µmol/L vs. 243 ± 7 µmol/L; P = 0.0005). CONCLUSIONS: Citrulline treatment in male aged rats favorably modulates body composition and protects against lipid oxidation and, thus, emerges as an interesting candidate to help prevent the aging process.


Asunto(s)
Composición Corporal/efectos de los fármacos , Citrulina/farmacología , Suplementos Dietéticos , Envejecimiento/efectos de los fármacos , Aminoácidos/sangre , Animales , HDL-Colesterol/sangre , Cetocolesteroles , Metabolismo de los Lípidos , Masculino , Diana Mecanicista del Complejo 1 de la Rapamicina , Complejos Multiproteicos/genética , Complejos Multiproteicos/metabolismo , Músculo Esquelético/efectos de los fármacos , Músculo Esquelético/metabolismo , Tamaño de los Órganos/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Serina-Treonina Quinasas TOR/genética , Serina-Treonina Quinasas TOR/metabolismo , Triglicéridos/sangre
18.
FASEB J ; 28(7): 3211-24, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24652947

RESUMEN

AMP-activated protein kinase (AMPK) is a sensor of cellular energy status that plays a central role in skeletal muscle metabolism. We used skeletal muscle-specific AMPKα1α2 double-knockout (mdKO) mice to provide direct genetic evidence of the physiological importance of AMPK in regulating muscle exercise capacity, mitochondrial function, and contraction-stimulated glucose uptake. Exercise performance was significantly reduced in the mdKO mice, with a reduction in maximal force production and fatigue resistance. An increase in the proportion of myofibers with centralized nuclei was noted, as well as an elevated expression of interleukin 6 (IL-6) mRNA, possibly consistent with mild skeletal muscle injury. Notably, we found that AMPKα1 and AMPKα2 isoforms are dispensable for contraction-induced skeletal muscle glucose transport, except for male soleus muscle. However, the lack of skeletal muscle AMPK diminished maximal ADP-stimulated mitochondrial respiration, showing an impairment at complex I. This effect was not accompanied by changes in mitochondrial number, indicating that AMPK regulates muscle metabolic adaptation through the regulation of muscle mitochondrial oxidative capacity and mitochondrial substrate utilization but not baseline mitochondrial muscle content. Together, these results demonstrate that skeletal muscle AMPK has an unexpected role in the regulation of mitochondrial oxidative phosphorylation that contributes to the energy demands of the exercising muscle.-Lantier, L., Fentz, J., Mounier, R., Leclerc, J., Treebak, J. T., Pehmøller, C., Sanz, N., Sakakibara, I., Saint-Amand, E., Rimbaud, S., Maire, P., Marette, A., Ventura-Clapier, R., Ferry, A., Wojtaszewski, J. F. P., Foretz, M., Viollet, B. AMPK controls exercise endurance, mitochondrial oxidative capacity, and skeletal muscle integrity.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Mitocondrias/metabolismo , Fibras Musculares Esqueléticas/metabolismo , Resistencia Física/fisiología , Animales , Glucosa/metabolismo , Interleucina-6/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Contracción Muscular/fisiología , Fatiga Muscular/fisiología , Oxidación-Reducción , Fosforilación/fisiología , Condicionamiento Físico Animal
19.
Muscle Nerve ; 52(5): 788-94, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25704632

RESUMEN

INTRODUCTION: The effects of voluntary activity initiated in adult mdx (C57BL/10ScSc-DMD(mdx) /J) mice on skeletal and cardiac muscle function have not been studied extensively. METHODS: We studied the effects of 3 months of voluntary wheel running initiated at age 7 months on hindlimb muscle weakness, increased susceptibility to muscle contraction-induced injury, and left ventricular function in mdx mice. RESULTS: We found that voluntary wheel running did not worsen the deficit in force-generating capacity and the force drop after lengthening contractions in either mdx mouse gender. It increased the absolute maximal force of skeletal muscle in female mdx mice. Moreover, it did not affect left ventricular function, structural heart dimensions, cardiac gene expression of inflammation, fibrosis, or remodeling markers. CONCLUSION: These results indicate that voluntary activity initiated at age 7 months had no detrimental effects on skeletal or cardiac muscles in either mdx mouse gender.


Asunto(s)
Miembro Posterior/fisiología , Miocardio , Condicionamiento Físico Animal/métodos , Condicionamiento Físico Animal/fisiología , Factores de Edad , Animales , Femenino , Corazón/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos mdx , Actividad Motora/fisiología , Contracción Muscular/fisiología
20.
Mol Ther ; 22(8): 1423-1433, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24861054

RESUMEN

Myostatin regulates skeletal muscle size via the activin receptor IIB (ActRIIB). However, its effect on muscle energy metabolism and energy-dependent muscle function remains largely unexplored. This question needs to be solved urgently since various therapies for neuromuscular diseases based on blockade of ActRIIB signaling are being developed. Here, we show in mice, that 4-month pharmacological abrogation of ActRIIB signaling by treatment with soluble ActRIIB-Fc triggers extreme muscle fatigability. This is associated with elevated serum lactate levels and a severe metabolic myopathy in the mdx mouse, an animal model of Duchenne muscular dystrophy. Blockade of ActRIIB signaling downregulates porin, a crucial ADP/ATP shuttle between cytosol and mitochondrial matrix leading to a consecutive deficiency of oxidative phosphorylation as measured by in vivo Phosphorus Magnetic Resonance Spectroscopy ((31)P-MRS). Further, ActRIIB blockade reduces muscle capillarization, which further compounds the metabolic stress. We show that ActRIIB regulates key determinants of muscle metabolism, such as Pparß, Pgc1α, and Pdk4 thereby optimizing different components of muscle energy metabolism. In conclusion, ActRIIB signaling endows skeletal muscle with high oxidative capacity and low fatigability. The severe metabolic side effects following ActRIIB blockade caution against deploying this strategy, at least in isolation, for treatment of neuromuscular disorders.


Asunto(s)
Receptores de Activinas Tipo II/antagonistas & inhibidores , Fragmentos Fc de Inmunoglobulinas/farmacología , Músculos/fisiopatología , Distrofia Muscular Animal/fisiopatología , Animales , Línea Celular , Metabolismo Energético/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana , Humanos , Masculino , Ratones , Ratones Endogámicos mdx , Porinas/metabolismo , Transducción de Señal/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA