Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 66
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Hum Mol Genet ; 29(13): 2162-2170, 2020 08 03.
Artículo en Inglés | MEDLINE | ID: mdl-32472139

RESUMEN

Laminin-α2 related congenital muscular dystrophy (LAMA2-CMD) is a fatal muscle disease caused by mutations in the LAMA2 gene. Laminin-α2 is critical for the formation of laminin-211 and -221 heterotrimers in the muscle basal lamina. LAMA2-CMD patients exhibit hypotonia from birth and progressive muscle loss that results in developmental delay, confinement to a wheelchair, respiratory insufficiency and premature death. There is currently no cure or effective treatment for LAMA2-CMD. Several studies have shown laminin-111 can serve as an effective protein-replacement therapy for LAMA2-CMD. Studies have demonstrated early treatment with laminin-111 protein results in an increase in life expectancy and improvements in muscle pathology and function. Since LAMA2-CMD patients are often diagnosed after advanced disease, it is unclear if laminin-111 protein therapy at an advanced stage of the disease can have beneficial outcomes. In this study, we tested the efficacy of laminin-111 protein therapy after disease onset in a mouse model of LAMA2-CMD. Our results showed laminin-111 treatment after muscle disease onset increased life expectancy, promoted muscle growth and increased muscle stiffness. Together these studies indicate laminin-111 protein therapy either early or late in the disease process could serve as an effective protein replacement therapy for LAMA2-CMD.


Asunto(s)
Laminina/farmacología , Enfermedades Musculares/genética , Distrofias Musculares/genética , Animales , Membrana Basal/efectos de los fármacos , Membrana Basal/crecimiento & desarrollo , Modelos Animales de Enfermedad , Humanos , Laminina/genética , Ratones , Músculo Esquelético/efectos de los fármacos , Músculo Esquelético/crecimiento & desarrollo , Enfermedades Musculares/patología , Distrofias Musculares/patología , Mutación/genética
2.
Connect Tissue Res ; 62(1): 40-52, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-32867551

RESUMEN

PURPOSE: To describe potential signaling (cross-talk) between dystrophic skeletal muscle and tendon in Duchenne muscular dystrophy. MATERIALS AND METHODS: Review of Duchenne muscular dystrophy and associated literature relevant to muscle-tendon cross-talk. RESULTS AND CONCLUSIONS: Duchenne muscular dystrophy results from the absence of the protein dystrophin and the associated dystrophin - glycoprotein complex, which are thought to provide both structural support and signaling functions for the muscle fiber. In addition, there are other potential signal pathways that could represent cross-talk between muscle and tendon, particularly at the myotendinous junction. Duchenne muscular dystrophy is characterized by multiple pathophysiologic mechanisms. Herein, we explore three of these: (1) the extracellular matrix, fibrosis, and fat deposition; (2) satellite cells; and (3) tensegrity. A key signaling protein that emerged in each was transforming growth factor - beta one (TGF-ß1).].


Asunto(s)
Distrofia Muscular de Duchenne , Distrofina , Humanos , Músculo Esquelético , Tendones , Factor de Crecimiento Transformador beta
3.
Mol Ther ; 28(2): 382-393, 2020 02 05.
Artículo en Inglés | MEDLINE | ID: mdl-31784415

RESUMEN

Multiple clinical trials employing recombinant adeno-associated viral (rAAV) vectors have been initiated for neuromuscular disorders, including Duchenne and limb-girdle muscular dystrophies, spinal muscular atrophy, and recently X-linked myotubular myopathy (XLMTM). Our previous work on a canine model of XLMTM showed that a single rAAV8-cMTM1 systemic infusion corrected structural abnormalities within the muscle and restored contractile function, with affected dogs surviving more than 4 years post injection. This remarkable therapeutic efficacy presents a unique opportunity to identify the downstream molecular drivers of XLMTM pathology and to what extent the whole muscle transcriptome is restored to normal after gene transfer. Herein, RNA-sequencing was used to examine the transcriptomes of the Biceps femoris and Vastus lateralis in a previously described canine cohort that showed dose-dependent clinical improvements after rAAV8-cMTM1 gene transfer. Our analysis confirmed several dysregulated genes previously observed in XLMTM mice but also identified transcripts linked to XLMTM pathology. We demonstrated XLMTM transcriptome remodeling and dose-dependent normalization of gene expression after gene transfer and created metrics to pinpoint potential biomarkers of disease progression and correction.


Asunto(s)
Dependovirus/genética , Técnicas de Transferencia de Gen , Terapia Genética , Vectores Genéticos/genética , Músculo Esquelético/metabolismo , Miopatías Estructurales Congénitas/genética , Transcriptoma , Animales , Biomarcadores , Modelos Animales de Enfermedad , Perros , Dosificación de Gen , Perfilación de la Expresión Génica , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Transducción Genética
4.
Exerc Sport Sci Rev ; 48(2): 74-82, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-32168170

RESUMEN

Hypothalamic pro-opiomelanocortin (POMC) neurons are key sensory neurons for energy balance. The basic helix-loop-helix transcription factor NHLH2 is expressed in POMC neurons, and Nhlh2 knockout mice show adult-onset obesity with low exercise behavior. Evidence is presented to explore the hypothesis that NHLH2 transcriptional activity within POMC neurons is crucial for maintaining motivated spontaneous activity and enforced exercise.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Ejercicio Físico/fisiología , Hipotálamo/metabolismo , Motivación/fisiología , Neuronas/metabolismo , Proopiomelanocortina/metabolismo , Transcripción Genética , Animales , Ejercicio Físico/psicología , Humanos , Modelos Animales
5.
J Mol Cell Cardiol ; 135: 160-171, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31445917

RESUMEN

Novel therapeutic strategies to treat mitochondrial deficiencies in acute coronary syndromes are needed. Complex I of the mitochondrial electron transport system is damaged following ischemia/reperfusion (I/R) injury. This disruption contributes to aberrant electron transport, diminished bioenergetics, an altered redox environment, and mitochondrial damage involved in tissue injury. In this study, we determined the cardiac and mitochondrial effects of idebenone, a benzoquinone currently in several clinical trials with purported 'antioxidant' effects. We employed complimentary models of ischemia/reperfusion injury in perfused hearts, permeabilized cardiac fibers, isolated mitochondria, and in cells to elucidate idebenone's cardioprotective mechanism(s). In ex vivo whole hearts, infarct size was markedly reduced with post-ischemic idebenone treatment (25 ±â€¯5% area at risk, AAR) compared to controls (56 ±â€¯6% AAR, P < .05). Several parameters of hemodynamic function were also significantly improved after idebenone treatment. Parallel studies of anoxia/reoxygenation were conducted using isolated mitochondria and permeabilized ventricular fibers. In isolated mitochondria, we simultaneously monitored respiration and ROS emission. Idebenone treatment modestly elevated succinate-derived H2O2 production when compared to vehicle control (1.34 ±â€¯0.05 vs 1.21 ±â€¯0.05%, H2O2/O2 respectively, P < .05). Isolated mitochondria subjected to anoxia/reoxygenation demonstrated higher rates of respiration with idebenone treatment (2360 ±â€¯69 pmol/s*mg) versus vehicle control (1995 ±â€¯101 pmol/s*mg). Both mitochondria and permeabilized cardiac fibers produced high rates of H2O2 after anoxia/reoxygenation, with idebenone showing no discernable attenuation on H2O2 production. These insights were further investigated with studies in mitochondria isolated from reperfused ventricle. The profound decrease in complex-I dependent respiration after ischemia/reperfusion (701 ±â€¯59 pmolO2/s*mg compared to 1816 ±â€¯105 pmol O2/s*mg in normoxic mitochondria) was attenuated with idebenone treatment (994 ±â€¯76 vs pmol O2/s*mg, P < .05). Finally, the effects of idebenone were determined using permeabilized cell models with chemical inhibition of complex I. ADP-dependent oxidative phosphorylation capacity was significantly higher in complex-I inhibited cells treated acutely with idebenone (89.0 ±â€¯4.2 pmol/s*million cells versus 70.1 ±â€¯8.2 pmol/s*million cells in untreated cells). Taken together, these data indicate that the cardioprotective effects of idebenone treatment do not involve ROS-scavenging but appear to involve augmentation of the quinone pool, thus providing reducing equivalents downstream of complex I. As this compound is already in clinical trials for other indications, it may provide a safe and useful approach to mitigate ischemia/reperfusion injury in patients.


Asunto(s)
Complejo I de Transporte de Electrón/efectos de los fármacos , Infarto del Miocardio/tratamiento farmacológico , Daño por Reperfusión/tratamiento farmacológico , Ubiquinona/análogos & derivados , Animales , Modelos Animales de Enfermedad , Complejo I de Transporte de Electrón/genética , Humanos , Mitocondrias Cardíacas/efectos de los fármacos , Mitocondrias Cardíacas/genética , Mitocondrias Cardíacas/patología , Infarto del Miocardio/genética , Infarto del Miocardio/patología , Miocardio/metabolismo , Miocardio/patología , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , Fosforilación Oxidativa/efectos de los fármacos , Consumo de Oxígeno/efectos de los fármacos , Ratas , Especies Reactivas de Oxígeno/metabolismo , Daño por Reperfusión/genética , Daño por Reperfusión/patología , Ubiquinona/farmacología
6.
Mol Ther ; 25(4): 839-854, 2017 04 05.
Artículo en Inglés | MEDLINE | ID: mdl-28237839

RESUMEN

X-linked myotubular myopathy (XLMTM) results from MTM1 gene mutations and myotubularin deficiency. Most XLMTM patients develop severe muscle weakness leading to respiratory failure and death, typically within 2 years of age. Our objective was to evaluate the efficacy and safety of systemic gene therapy in the p.N155K canine model of XLMTM by performing a dose escalation study. A recombinant adeno-associated virus serotype 8 (rAAV8) vector expressing canine myotubularin (cMTM1) under the muscle-specific desmin promoter (rAAV8-cMTM1) was administered by simple peripheral venous infusion in XLMTM dogs at 10 weeks of age, when signs of the disease are already present. A comprehensive analysis of survival, limb strength, gait, respiratory function, neurological assessment, histology, vector biodistribution, transgene expression, and immune response was performed over a 9-month study period. Results indicate that systemic gene therapy was well tolerated, prolonged lifespan, and corrected the skeletal musculature throughout the body in a dose-dependent manner, defining an efficacious dose in this large-animal model of the disease. These results support the development of gene therapy clinical trials for XLMTM.


Asunto(s)
Dependovirus/genética , Terapia Genética , Vectores Genéticos/genética , Músculo Esquelético/metabolismo , Miopatías Estructurales Congénitas/genética , Animales , Biopsia , Dependovirus/clasificación , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Perros , Marcha , Expresión Génica , Terapia Genética/efectos adversos , Terapia Genética/métodos , Vectores Genéticos/administración & dosificación , Vectores Genéticos/efectos adversos , Vectores Genéticos/farmacocinética , Inmunidad Celular , Inmunidad Humoral , Estimación de Kaplan-Meier , Fuerza Muscular , Músculo Esquelético/patología , Músculo Esquelético/fisiopatología , Músculo Esquelético/ultraestructura , Miopatías Estructurales Congénitas/diagnóstico , Miopatías Estructurales Congénitas/mortalidad , Miopatías Estructurales Congénitas/terapia , Proteínas Tirosina Fosfatasas no Receptoras/genética , Recuperación de la Función , Reflejo , Pruebas de Función Respiratoria , Distribución Tisular , Transgenes/genética , Transgenes/inmunología , Resultado del Tratamiento
7.
Muscle Nerve ; 56(5): 943-953, 2017 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-28370029

RESUMEN

INTRODUCTION: X-linked myotubular myopathy (XLMTM), a devastating pediatric disease caused by the absence of the protein myotubularin, results from mutations in the MTM1 gene. While there is no cure for XLMTM, we previously reported effects of MTM1 gene therapy using adeno-associated virus (AAV) vector on muscle weakness and pathology in MTM1-mutant dogs. Here, we followed 2 AAV-infused dogs over 4 years. METHODS: We evaluated gait, strength, respiration, neurological function, muscle pathology, AAV vector copy number (VCN), and transgene expression. RESULTS: Four years following AAV-mediated gene therapy, gait, respiratory performance, neurological function and pathology in AAV-infused XLMTM dogs remained comparable to their healthy littermate controls despite a decline in VCN and muscle strength. CONCLUSIONS: AAV-mediated gene transfer of MTM1 in young XLMTM dogs results in long-term expression of myotubularin transgene with normal muscular performance and neurological function in the absence of muscle pathology. These findings support a clinical trial in patients. Muscle Nerve 56: 943-953, 2017.


Asunto(s)
Terapia Genética , Miopatías Estructurales Congénitas/terapia , Proteínas Tirosina Fosfatasas no Receptoras/uso terapéutico , Adenosina Trifosfatasas/metabolismo , Animales , Dependovirus/genética , Modelos Animales de Enfermedad , Perros , Femenino , Trastornos Neurológicos de la Marcha/etiología , Glucuronidasa/genética , Glucuronidasa/metabolismo , Humanos , Estudios Longitudinales , Microscopía Electrónica , Músculo Esquelético/patología , Músculo Esquelético/ultraestructura , Mutación/genética , Miopatías Estructurales Congénitas/complicaciones , Miopatías Estructurales Congénitas/genética , Miopatías Estructurales Congénitas/veterinaria , NAD/metabolismo , Examen Neurológico , Proteínas Tirosina Fosfatasas no Receptoras/genética , Trastornos Respiratorios/etiología , Transducción Genética
8.
Ann Neurol ; 78(2): 222-34, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25925726

RESUMEN

OBJECTIVE: We have recently reported on the pathology of the neuromuscular junction (NMJ) in Pompe disease, reflecting disruption of neuronal and muscle homeostasis as a result of glycogen accumulation. The aim of this study was to examine how the alteration of NMJ physiology contributes to Pompe disease pathology; we performed molecular, physiological, and histochemical analyses of NMJ-related measures of the tibialis anterior muscles of young-, mid-, and late-stage alpha-glucosidase (GAA)-deficient mice. METHODS: We performed intramuscular injection of an adeno-associated virus (AAV)9 vector expressing GAA (AAV9-hGAA) into the tibialis anterior muscle of Gaa(-/-) mice at early, mid, and severe pathological time points. We analyzed expression of NMJ-related genes, in situ muscle force production, and clearance of glycogen in conjunction with histological assessment of the NMJ. RESULTS: Our data demonstrate that AAV9-hGAA is able to replace GAA to the affected tissue and modify AChR mRNA expression, muscle force production, motor endplate area, and innervation status. Importantly, the degree of restoration for these outcomes is limited by severity of disease. Early restoration of GAA activity was most effective, whereas late correction of GAA expression was not effective in modifying parameters reflecting NMJ structure and function nor in force restoration despite resolution of glycogen storage in muscle. INTERPRETATION: Our data provide new mechanistic insight into the pathology of Pompe disease and suggest that early systemic correction to both neural and muscle tissues may be essential for successful correction of neuromuscular function in Pompe disease. Ann Neurol 2015;78:222-234.


Asunto(s)
Terapia Genética , Enfermedad del Almacenamiento de Glucógeno Tipo II/genética , Fuerza Muscular/fisiología , Músculo Esquelético/metabolismo , Unión Neuromuscular/metabolismo , ARN Mensajero/metabolismo , Receptores Colinérgicos/genética , alfa-Glucosidasas/genética , Animales , Dependovirus , Modelos Animales de Enfermedad , Vectores Genéticos , Glucógeno/metabolismo , Enfermedad del Almacenamiento de Glucógeno Tipo II/metabolismo , Enfermedad del Almacenamiento de Glucógeno Tipo II/fisiopatología , Miembro Posterior , Inyecciones Intramusculares , Contracción Isométrica , Ratones , Ratones Noqueados , Músculo Esquelético/patología , Músculo Esquelético/fisiopatología , Unión Neuromuscular/patología , Unión Neuromuscular/fisiopatología , Receptores Colinérgicos/metabolismo , Factores de Tiempo
9.
Am J Physiol Cell Physiol ; 308(1): C33-40, 2015 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-25298424

RESUMEN

Duchenne muscular dystrophy (DMD) is one of the most frequent types of muscular dystrophy. Alterations in intracellular calcium (Ca(2+)) handling are thought to contribute to the disease severity in DMD, possibly due to the activation of Ca(2+)-activated proteases. The purpose of this study was twofold: 1) to determine whether prolonged excitation-contraction (E-C) coupling disruption following repeated contractions is greater in animals lacking both dystrophin and utrophin (mdx/Utr(-/-)) compared with mice lacking only dystrophin (mdx); and 2) to assess whether protease inhibition can prevent E-C coupling failure following repeated tetani in these dystrophic mouse models. Excitation-contraction coupling was assessed using Fura-2 ratio, as an index of intracellular free Ca(2+) concentration, in response to electrical stimulation of single muscle fibers from the flexor digitorum brevis muscle. Resting Fura-2 ratio was higher in dystrophic compared with control (Con) fibers, but peak Fura-2 ratios during stimulation were similar in dystrophic and Con fibers. One hour after a series of repeated tetani, peak Fura-2 ratios were reduced by 30 ± 5.6%, 23 ± 2%, and 36 ± 3.1% in mdx, mdx/Utr(+/-), and mdx/Utr(-/-), respectively, with the greatest reduction in mdx/Utr(-/-) fibers (P < 0.05). Protease inhibition attenuated this decrease in peak Fura-2 ratio. These data indicate that E-C coupling impairment after repeated contractions is greatest in fibers lacking both dystrophin and utrophin and that prevention of protease activation can mitigate the prolonged E-C coupling impairment. These data further suggest that acute protease inhibition may be useful in reducing muscle weakness in DMD.


Asunto(s)
Calcio/metabolismo , Acoplamiento Excitación-Contracción , Fibras Musculares Esqueléticas/enzimología , Músculo Esquelético/enzimología , Distrofia Muscular de Duchenne/enzimología , Péptido Hidrolasas/metabolismo , Animales , Modelos Animales de Enfermedad , Estimulación Eléctrica , Acoplamiento Excitación-Contracción/efectos de los fármacos , Ratones Endogámicos mdx , Ratones Noqueados , Fibras Musculares Esqueléticas/efectos de los fármacos , Fuerza Muscular , Debilidad Muscular , Músculo Esquelético/efectos de los fármacos , Músculo Esquelético/fisiopatología , Distrofia Muscular de Duchenne/tratamiento farmacológico , Distrofia Muscular de Duchenne/genética , Distrofia Muscular de Duchenne/fisiopatología , Inhibidores de Proteasas/farmacología , Factores de Tiempo , Utrofina/deficiencia , Utrofina/genética
10.
Hum Mol Genet ; 22(8): 1525-38, 2013 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-23307925

RESUMEN

No effective treatment exists for patients with X-linked myotubular myopathy (XLMTM), a fatal congenital muscle disease caused by deficiency of the lipid phosphatase, myotubularin. The Mtm1δ4 and Mtm1 p.R69C mice model severely and moderately symptomatic XLMTM, respectively, due to differences in the degree of myotubularin deficiency. Contractile function of intact extensor digitorum longus (EDL) and soleus muscles from Mtm1δ4 mice, which produce no myotubularin, is markedly impaired. Contractile forces generated by chemically skinned single fiber preparations from Mtm1δ4 muscle were largely preserved, indicating that weakness was largely due to impaired excitation contraction coupling. Mtm1 p.R69C mice, which produce small amounts of myotubularin, showed impaired contractile function only in EDL muscles. Short-term replacement of myotubularin with a prototypical targeted protein replacement agent (3E10Fv-MTM1) in Mtm1δ4 mice improved contractile function and muscle pathology. These promising findings suggest that even low levels of myotubularin protein replacement can improve the muscle weakness and reverse the pathology that characterizes XLMTM.


Asunto(s)
Terapia de Reemplazo Enzimático , Miopatías Estructurales Congénitas/patología , Miopatías Estructurales Congénitas/terapia , Proteínas Tirosina Fosfatasas no Receptoras/genética , Animales , Modelos Animales de Enfermedad , Fatiga/metabolismo , Fatiga/fisiopatología , Femenino , Humanos , Ratones , Debilidad Muscular/genética , Debilidad Muscular/terapia , Músculo Esquelético/fisiopatología , Músculos/enzimología , Músculos/metabolismo , Músculos/patología , Miopatías Estructurales Congénitas/enzimología , Miopatías Estructurales Congénitas/genética , Proteínas Tirosina Fosfatasas no Receptoras/biosíntesis , Proteínas Tirosina Fosfatasas no Receptoras/deficiencia
11.
J Cell Sci ; 125(Pt 22): 5329-37, 2012 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-22956541

RESUMEN

In response to severe injury, adult skeletal muscle exhibits a remarkable regenerative capacity due to a resident muscle stem/progenitor cell population. While a number of factors are expressed in the muscle progenitor cell (MPC) population, the molecular networks that govern this cell population remain an area of active investigation. In this study, utilizing knockdown techniques and overexpression of Foxk1 in the myogenic lineage, we observed dysregulation of Foxo and Mef2 downstream targets. Utilizing an array of technologies, we establish that Foxk1 represses the transcriptional activity of Foxo4 and Mef2 and physically interacts with Foxo4 and Mef2, thus promoting MPC proliferation and antagonizing the myogenic lineage differentiation program, respectively. Correspondingly, knockdown of Foxk1 in C2C12 myoblasts results in cell cycle arrest, and Foxk1 overexpression in C2C12CAR myoblasts retards muscle differentiation. Collectively, we have established that Foxk1 promotes MPC proliferation by repressing Foxo4 transcriptional activity and inhibits myogenic differentiation by repressing Mef2 activity. These studies enhance our understanding of the transcriptional networks that regulate the MPC population and muscle regeneration.


Asunto(s)
Diferenciación Celular , Factores de Transcripción Forkhead/metabolismo , Desarrollo de Músculos , Factores Reguladores Miogénicos/metabolismo , Animales , Ciclo Celular , Proteínas de Ciclo Celular , Proliferación Celular , ADN/metabolismo , Factores de Transcripción MEF2 , Masculino , Ratones , Ratones Noqueados , Músculo Esquelético/citología , Músculo Esquelético/fisiología , Unión Proteica , Regeneración , Proteínas Represoras/metabolismo , Transcripción Genética
12.
FASEB J ; 27(4): 1585-99, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23325319

RESUMEN

Mutations in the human SEPN1 gene, encoding selenoprotein N (SepN), cause SEPN1-related myopathy (SEPN1-RM) characterized by muscle weakness, spinal rigidity, and respiratory insufficiency. As with other members of the selenoprotein family, selenoprotein N incorporates selenium in the form of selenocysteine (Sec). Most selenoproteins that have been functionally characterized are involved in oxidation-reduction (redox) reactions, with the Sec residue located at their catalytic site. To model SEPN1-RM, we generated a Sepn1-knockout (Sepn1(-/-)) mouse line. Homozygous Sepn1(-/-) mice are fertile, and their weight and lifespan are comparable to wild-type (WT) animals. Under baseline conditions, the muscle histology of Sepn1(-/-) mice remains normal, but subtle core lesions could be detected in skeletal muscle after inducing oxidative stress. Ryanodine receptor (RyR) calcium release channels showed lower sensitivity to caffeine in SepN deficient myofibers, suggesting a possible role of SepN in RyR regulation. SepN deficiency also leads to abnormal lung development characterized by enlarged alveoli, which is associated with decreased tissue elastance and increased quasi-static compliance of Sepn1(-/-) lungs. This finding raises the possibility that the respiratory syndrome observed in patients with SEPN1 mutations may have a primary pulmonary component in addition to the weakness of respiratory muscles.


Asunto(s)
Pulmón/crecimiento & desarrollo , Pulmón/metabolismo , Enfermedades Musculares/genética , Selenoproteínas/deficiencia , Animales , Humanos , Pulmón/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Musculares/genética , Proteínas Musculares/metabolismo , Músculo Esquelético/metabolismo , Músculo Esquelético/patología , Mutación/genética , Estrés Oxidativo/genética , Canal Liberador de Calcio Receptor de Rianodina/genética , Canal Liberador de Calcio Receptor de Rianodina/metabolismo , Selenocisteína/genética , Selenoproteínas/metabolismo
13.
Br J Nutr ; 111(10): 1791-800, 2014 May 28.
Artículo en Inglés | MEDLINE | ID: mdl-24524266

RESUMEN

Duchenne muscular dystrophy (DMD) is a severe muscle disease that affects afflicted males from a young age, and the mdx mouse is an animal model of this disease. Although new drugs are in development, it is also essential to assess potential dietary therapies that could assist in the management of DMD. In the present study, we compared two diets, high-MUFA diet v. high-PUFA diet, in mdx mice. To generate the high-PUFA diet, a portion of dietary MUFA (oleic acid) was replaced with the dietary essential n-3 PUFA α-linolenic acid (ALA). We sought to determine whether ALA, compared with oleic acid, was beneficial in mdx mice. Consumption of the high-PUFA diet resulted in significantly higher n-3 PUFA content and reduced arachidonic acid content in skeletal muscle phospholipids (PL), while the high-MUFA diet led to higher oleate content in PL. Mdx mice on the high-MUFA diet exhibited 2-fold lower serum creatine kinase activity than those on the high-PUFA diet (P< 0·05) as well as a lower body fat percentage (P< 0·05), but no significant difference in skeletal muscle histopathology results. There was no significant difference between the dietary groups with regard to phosphorylated p65 (an inflammatory marker) in skeletal muscle. In conclusion, alteration of PL fatty acid (FA) composition by the high-PUFA diet made mdx muscle more susceptible to sarcolemmal leakiness, while the high-MUFA diet exhibited a more favourable impact. These results may be important for designing dietary treatments for DMD patients, and future work on dietary FA profiles, such as comparing other FA classes and dose effects, is needed.


Asunto(s)
Creatina Quinasa/sangre , Grasas de la Dieta/metabolismo , Ácidos Grasos Monoinsaturados/metabolismo , Ácidos Grasos Omega-3/metabolismo , Músculo Esquelético/metabolismo , Distrofia Muscular de Duchenne/patología , Fosfolípidos/aislamiento & purificación , Análisis de Varianza , Animales , Ácido Araquidónico/metabolismo , Biomarcadores/metabolismo , Cromatografía Liquida , Modelos Animales de Enfermedad , Inflamación/metabolismo , Masculino , Ratones , Ratones Endogámicos mdx , Músculo Esquelético/patología , FN-kappa B/análisis , Ácido Oléico/metabolismo , Fosforilación , Aceites de Plantas/metabolismo
14.
Proc Natl Acad Sci U S A ; 108(25): 10196-201, 2011 Jun 21.
Artículo en Inglés | MEDLINE | ID: mdl-21633012

RESUMEN

In response to physiological stimuli, skeletal muscle alters its myofiber composition to significantly affect muscle performance and metabolism. This process requires concerted regulation of myofiber-specific isoforms of sarcomeric and calcium regulatory proteins that couple action potentials to the generation of contractile force. Here, we identify Sox6 as a fast myofiber-enriched repressor of slow muscle gene expression in vivo. Mice lacking Sox6 specifically in skeletal muscle have an increased number of slow myofibers, elevated mitochondrial activity, and exhibit down-regulation of the fast myofiber gene program, resulting in enhanced muscular endurance. In addition, microarray profiling of Sox6 knockout muscle revealed extensive muscle fiber-type remodeling, and identified numerous genes that display distinctive fiber-type enrichment. Sox6 directly represses the transcription of slow myofiber-enriched genes by binding to conserved cis-regulatory elements. These results identify Sox6 as a robust regulator of muscle contractile phenotype and metabolism, and elucidate a mechanism by which functionally related muscle fiber-type specific gene isoforms are collectively controlled.


Asunto(s)
Regulación de la Expresión Génica , Fibras Musculares de Contracción Rápida/fisiología , Fibras Musculares de Contracción Lenta/fisiología , Músculo Esquelético/fisiología , Factores de Transcripción SOXD/metabolismo , Animales , Calcio/metabolismo , Perfilación de la Expresión Génica , Ratones , Ratones Noqueados , Análisis por Micromatrices , Contracción Muscular/fisiología , Fibras Musculares de Contracción Rápida/citología , Fibras Musculares de Contracción Lenta/citología , Músculo Esquelético/citología , Factores de Transcripción SOXD/genética
15.
Function (Oxf) ; 5(1): zqad066, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38111538

RESUMEN

Alzheimer's disease (AD) develops along a continuum that spans years prior to diagnosis. Decreased muscle function and mitochondrial respiration occur years earlier in those that develop AD; however, it is unknown what causes these peripheral phenotypes in a disease of the brain. Exercise promotes muscle, mitochondria, and cognitive health and is proposed to be a potential therapeutic for AD, but no study has investigated how skeletal muscle adapts to exercise training in an AD-like context. Utilizing 5xFAD mice, an AD model that develops ad-like pathology and cognitive impairments around 6 mo of age, we examined in vivo neuromuscular function and exercise adapations (mitochondrial respiration and RNA sequencing) before the manifestation of overt cognitive impairment. We found 5xFAD mice develop neuromuscular dysfunction beginning as early as 4 mo of age, characterized by impaired nerve-stimulated muscle torque production and compound nerve action potential of the sciatic nerve. Furthermore, skeletal muscle in 5xFAD mice had altered, sex-dependent, adaptive responses (mitochondrial respiration and gene expression) to exercise training in the absence of overt cognitive impairment. Changes in peripheral systems, specifically neural communication to skeletal muscle, may be harbingers for AD and have implications for lifestyle interventions, like exercise, in AD.


Asunto(s)
Enfermedad de Alzheimer , Disfunción Cognitiva , Ratones , Animales , Enfermedad de Alzheimer/genética , Ratones Transgénicos , Encéfalo/metabolismo , Disfunción Cognitiva/etiología , Mitocondrias/metabolismo
16.
J Muscle Res Cell Motil ; 34(5-6): 317-32, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24162313

RESUMEN

The contractile performance of mammalian fast twitch skeletal muscle is history dependent. The effect of previous or ongoing contractile activity to potentiate force, i.e. increase isometric twitch force, is a fundamental property of fast skeletal muscle. The precise manifestation of force potentiation is dependent upon a variety of factors with two general types being identified; staircase potentiation referring to the progressive increase in isometric twitch force observed during low frequency stimulation while posttetanic potentiation refers to the step-like increase in isometric twitch force observed following a brief higher frequency (i.e. tetanic) stimulation. Classic studies established that the magnitude and duration of potentiation depends on a number of factors including muscle fiber type, species, temperature, sarcomere length and stimulation paradigm. In addition to isometric twitch force, more recent work has shown that potentiation also influences dynamic (i.e. concentric and/or isotonic) force, work and power at a range of stimulus frequencies in situ or in vitro, an effect that may translate to enhanced physiological function in vivo. Early studies performed on both intact and permeabilized models established that the primary mechanism for this modulation of performance was phosphorylation of myosin, a modification that increased the Ca(2+) sensitivity of contraction. More recent work from a variety of muscle models indicates, however, the presence of a secondary mechanism for potentiation that may involve altered Ca(2+) handling. The primary purpose of this review is to highlight these recent findings relative to the physiological utility of force potentiation in vivo.


Asunto(s)
Contracción Isométrica/fisiología , Músculo Esquelético/fisiología , Cadenas Ligeras de Miosina/fisiología , Animales , Humanos , Modelos Animales , Músculo Esquelético/metabolismo , Cadenas Ligeras de Miosina/metabolismo , Fosforilación
17.
Mamm Genome ; 23(1-2): 85-108, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22218699

RESUMEN

Duchenne muscular dystrophy (DMD) is an X-linked recessive disorder in which the loss of dystrophin causes progressive degeneration of skeletal and cardiac muscle. Potential therapies that carry substantial risk, such as gene- and cell-based approaches, must first be tested in animal models, notably the mdx mouse and several dystrophin-deficient breeds of dogs, including golden retriever muscular dystrophy (GRMD). Affected dogs have a more severe phenotype, in keeping with that of DMD, so may better predict disease pathogenesis and treatment efficacy. Various phenotypic tests have been developed to characterize disease progression in the GRMD model. These biomarkers range from measures of strength and joint contractures to magnetic resonance imaging. Some of these tests are routinely used in clinical veterinary practice, while others require specialized equipment and expertise. By comparing serial measurements from treated and untreated groups, one can document improvement or delayed progression of disease. Potential treatments for DMD may be broadly categorized as molecular, cellular, or pharmacologic. The GRMD model has increasingly been used to assess efficacy of a range of these therapies. A number of these studies have provided largely general proof-of-concept for the treatment under study. Others have demonstrated efficacy using the biomarkers discussed. Importantly, just as symptoms in DMD vary among patients, GRMD dogs display remarkable phenotypic variation. Though confounding statistical analysis in preclinical trials, this variation offers insight regarding the role that modifier genes play in disease pathogenesis. By correlating functional and mRNA profiling results, gene targets for therapy development can be identified.


Asunto(s)
Modelos Animales de Enfermedad , Músculo Esquelético/patología , Músculo Esquelético/fisiopatología , Distrofia Muscular de Duchenne/fisiopatología , Distrofia Muscular de Duchenne/terapia , Animales , Biomarcadores , Perros , Distrofina/deficiencia , Distrofina/genética , Articulaciones/patología , Distrofia Muscular Animal/genética , Distrofia Muscular Animal/fisiopatología , Distrofia Muscular Animal/terapia , Distrofia Muscular de Duchenne/genética , Distrofia Muscular de Duchenne/patología
18.
Muscle Nerve ; 46(4): 588-91, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22987702

RESUMEN

INTRODUCTION: We established a colony of dogs that harbor an X-linked MTM1 missense mutation.Muscle from affected male dogs exhibits reduction and altered localization of the MTM1 gene product, myotubularin, and provides a model analogous to X-linked myotubular myopathy (XLMTM). METHODS: We studied hindlimb muscle function in age-matched canine XLMTM genotypes between ages 9 and 18 weeks. RESULTS: By the end of the study, affected dogs produce only ∼15% of the torque generated by normals or carriers (0.023 ± 0.005 vs. 0.152 ± 0.007 and 0.154 ± 0.003 N-m/kg body mass, respectively, P < 0.05) and are too weak to stand unassisted. At this age, XLMTM dogs also demonstrate an abnormally low twitch:tetanus ratio, a right-shifted torque-frequency relationship and an increase in torque during repetitive stimulation (P < 0.05). CONCLUSIONS: We hypothesize that muscle weakness results from impaired excitation-contraction (E-C) coupling. Interventions that improve E-C coupling might be translated from the XLMTM dog model to patients.


Asunto(s)
Músculo Esquelético/fisiopatología , Miopatías Estructurales Congénitas/genética , Miopatías Estructurales Congénitas/fisiopatología , Cromosoma X/genética , Animales , Modelos Animales de Enfermedad , Perros , Masculino , Mutación Missense , Proteínas Tirosina Fosfatasas no Receptoras/genética
19.
Nutrients ; 14(4)2022 Feb 18.
Artículo en Inglés | MEDLINE | ID: mdl-35215509

RESUMEN

Prader-Willi Syndrome (PWS) is a human genetic condition that affects up to 1 in 10,000 live births. Affected infants present with hypotonia and developmental delay. Hyperphagia and increasing body weight follow unless drastic calorie restriction is initiated. Recently, our laboratory showed that one of the genes in the deleted locus causative for PWS, Snord116, maintains increased expression of hypothalamic Nhlh2, a basic helix-loop-helix transcription factor. We have previously also shown that obese mice with a deletion of Nhlh2 respond to a conjugated linoleic acid (CLA) diet with weight and fat loss. In this study, we investigated whether mice with a paternal deletion of Snord116 (Snord116m+/p-) would respond similarly. We found that while Snord116m+/p- mice and mice with a deletion of both Snord116 alleles were not significantly obese on a high-fat diet, they did lose body weight and fat on a high-fat/CLA diet, suggesting that the genotype did not interfere with CLA actions. There were no changes in food intake or metabolic rate, and only moderate differences in exercise performance. RNA-seq and microbiome analyses identified hypothalamic mRNAs, and differentially populated gut bacteria, that support future mechanistic analyses. CLA may be useful as a food additive to reduce obesity in humans with PWS.


Asunto(s)
Ácidos Linoleicos Conjugados , Síndrome de Prader-Willi , Animales , Dieta Alta en Grasa/efectos adversos , Ácidos Linoleicos Conjugados/farmacología , Ratones , Obesidad/metabolismo , Síndrome de Prader-Willi/genética , Síndrome de Prader-Willi/metabolismo , ARN Nucleolar Pequeño/genética
20.
Dev Biol ; 337(2): 396-404, 2010 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-19914232

RESUMEN

The mechanisms that regulate skeletal muscle differentiation, fiber type diversity and muscle regeneration are incompletely defined. Forkhead transcription factors are critical regulators of cellular fate determination, proliferation, and differentiation. We identified a forkhead/winged helix transcription factor, Foxj3, which was expressed in embryonic and adult skeletal muscle. To define the functional role of Foxj3, we examined Foxj3 mutant mice. Foxj3 mutant mice are viable but have significantly fewer Type I slow-twitch myofibers and have impaired skeletal muscle contractile function compared to their wild type controls. In response to a severe injury, Foxj3 mutant mice have impaired muscle regeneration. Foxj3 mutant myogenic progenitor cells have perturbed cell cycle kinetics and decreased expression of Mef2c. Examination of the skeletal muscle 5' upstream enhancer of the Mef2c gene revealed an evolutionary conserved forkhead binding site (FBS). Transcriptional assays in C2C12 myoblasts revealed that Foxj3 transcriptionally activates the Mef2c gene in a dose dependent fashion and binds to the conserved FBS. Together, these studies support the hypothesis that Foxj3 is an important regulator of myofiber identity and muscle regeneration through the transcriptional activation of the Mef2c gene.


Asunto(s)
Envejecimiento/metabolismo , Proteínas de Unión al ADN/metabolismo , Fibras Musculares Esqueléticas/metabolismo , Factores Reguladores Miogénicos/genética , Factores de Transcripción/metabolismo , Activación Transcripcional/genética , Animales , Secuencia de Bases , Ciclo Celular , Proliferación Celular , Proteínas de Unión al ADN/genética , Factores de Transcripción Forkhead , Regulación del Desarrollo de la Expresión Génica , Técnicas de Silenciamiento del Gen , Factores de Transcripción MEF2 , Ratones , Ratones Mutantes , Datos de Secuencia Molecular , Contracción Muscular/fisiología , Desarrollo de Músculos/genética , Fibras Musculares Esqueléticas/patología , Fibras Musculares Esqueléticas/ultraestructura , Mioblastos/citología , Mioblastos/metabolismo , Mioblastos/ultraestructura , Factores Reguladores Miogénicos/metabolismo , ARN Interferente Pequeño/metabolismo , Regeneración , Células Madre/citología , Células Madre/metabolismo , Células Madre/ultraestructura , Análisis de Supervivencia , Factores de Transcripción/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA