Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 105
Filtrar
1.
FASEB J ; 38(11): e23718, 2024 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-38847487

RESUMEN

Female carriers of a Duchenne muscular dystrophy (DMD) gene mutation manifest exercise intolerance and metabolic anomalies that may be exacerbated following menopause due to the loss of estrogen, a known regulator of skeletal muscle function and metabolism. Here, we studied the impact of estrogen depletion (via ovariectomy) on exercise tolerance and muscle mitochondrial metabolism in female mdx mice and the potential of estrogen replacement therapy (using estradiol) to protect against functional and metabolic perturbations. We also investigated the effect of estrogen depletion, and replacement, on the skeletal muscle proteome through an untargeted proteomic approach with TMT-labelling. Our study confirms that loss of estrogen in female mdx mice reduces exercise capacity, tricarboxylic acid cycle intermediates, and citrate synthase activity but that these deficits are offset through estrogen replacement therapy. Furthermore, ovariectomy downregulated protein expression of RNA-binding motif factor 20 (Rbm20), a critical regulator of sarcomeric and muscle homeostasis gene splicing, which impacted pathways involving ribosomal and mitochondrial translation. Estrogen replacement modulated Rbm20 protein expression and promoted metabolic processes and the upregulation of proteins involved in mitochondrial dynamics and metabolism. Our data suggest that estrogen mitigates dystrophinopathic features in female mdx mice and that estrogen replacement may be a potential therapy for post-menopausal DMD carriers.


Asunto(s)
Estrógenos , Ratones Endogámicos mdx , Músculo Esquelético , Proteínas de Unión al ARN , Animales , Femenino , Ratones , Estrógenos/metabolismo , Estrógenos/farmacología , Proteínas de Unión al ARN/metabolismo , Proteínas de Unión al ARN/genética , Músculo Esquelético/metabolismo , Músculo Esquelético/efectos de los fármacos , Distrofia Muscular de Duchenne/metabolismo , Distrofia Muscular de Duchenne/genética , Ratones Endogámicos C57BL , Ovariectomía , Mitocondrias/metabolismo , Mitocondrias Musculares/metabolismo , Mitocondrias Musculares/efectos de los fármacos
2.
Neurobiol Dis ; 162: 105559, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34774794

RESUMEN

Skeletal muscle dysfunction may contribute to the progression and severity of amyotrophic lateral sclerosis (ALS). In the present study, we characterized the skeletal muscle pathophysiology in an inducible transgenic mouse model (rNLS8) that develops a TAR-DNA binding protein (TDP-43) proteinopathy and ALS-like neuropathology and disease progression; representative of >90% of all familial and sporadic ALS cases. As we previously observed elevated levels of miR-23a in skeletal muscle of patients with familial and sporadic ALS, we also investigated the effect of miR-23a suppression on skeletal muscle pathophysiology and disease severity in rNLS8 mice. Five weeks after disease onset TDP-43 protein accumulation was observed in tibialis anterior (TA), quadriceps (QUAD) and diaphragm muscle lysates and associated with skeletal muscle atrophy. In the TA muscle TDP-43 was detected in muscle fibres that appeared atrophied and angular in appearance and that also contained ß-amyloid aggregates. These fibres were also positive for neural cell adhesion molecule (NCAM), but not embryonic myosin heavy chain (eMHC), indicating TDP-43/ ß-amyloid localization in denervated muscle fibres. There was an upregulation of genes associated with myogenesis and NMJ degeneration and a decrease in the MURF1 atrophy-related protein in skeletal muscle. Suppression of miR-23a impaired rotarod performance and grip strength and accelerated body weight loss during early stages of disease progression. This was associated with increased AchRα mRNA expression and decreased protein levels of PGC-1α. The TDP-43 proteinopathy-induced impairment of whole body and skeletal muscle functional performance is associated with muscle wasting and elevated myogenic and NMJ stress markers. Suppressing miR-23a in the rNLS8 mouse model of ALS contributes to an early acceleration of disease progression as measured by decline in motor function.


Asunto(s)
Esclerosis Amiotrófica Lateral , Proteínas de Unión al ADN , MicroARNs , Proteinopatías TDP-43 , Esclerosis Amiotrófica Lateral/metabolismo , Animales , Proteínas de Unión al ADN/genética , Modelos Animales de Enfermedad , Humanos , Ratones , Ratones Transgénicos , MicroARNs/genética , Proteinopatías TDP-43/genética
3.
FASEB J ; 35(12): e22034, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34780665

RESUMEN

Mutation to the gene encoding dystrophin can cause Duchenne muscular dystrophy (DMD) and increase the sensitivity to stress in vertebrate species, including the mdx mouse model of DMD. Behavioral stressors can exacerbate some dystrophinopathy phenotypes of mdx skeletal muscle and cause hypotension-induced death. However, we have discovered that a subpopulation of mdx mice present with a wildtype-like response to mild (forced downhill treadmill exercise) and moderate (scruff restraint) behavioral stressors. These "stress-resistant" mdx mice are more physically active, capable of super-activating the hypothalamic-pituitary-adrenal and renin-angiotensin-aldosterone pathways following behavioral stress and they express greater levels of mineralocorticoid and glucocorticoid receptors in striated muscle relative to "stress-sensitive" mdx mice. Stress-resistant mdx mice also presented with a less severe striated muscle histopathology and greater exercise and skeletal muscle oxidative capacity at rest. Most interestingly, female mdx mice were more physically active following behavioral stressors compared to male mdx mice; a response abolished after ovariectomy and rescued with estradiol. We demonstrate that the response to behavioral stress greatly impacts disease severity in mdx mice suggesting the management of stress in patients with DMD be considered as a therapeutic approach to ameliorate disease progression.


Asunto(s)
Conducta Animal , Distrofia Muscular Animal/patología , Distrofia Muscular de Duchenne/patología , Condicionamiento Físico Animal , Estrés Psicológico/complicaciones , Animales , Modelos Animales de Enfermedad , Distrofina/deficiencia , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos mdx , Ratones Noqueados , Distrofia Muscular Animal/etiología , Distrofia Muscular Animal/psicología , Distrofia Muscular de Duchenne/etiología , Distrofia Muscular de Duchenne/psicología , Factores Sexuales
4.
J Strength Cond Res ; 36(8): 2306-2312, 2022 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-32881839

RESUMEN

ABSTRACT: Allsopp, GL, Hoffmann, SM, Feros, SA, Pasco, JA, Russell, AP, and Wright, CR. The effect of normobaric hypoxia on resistance training adaptations in older adults. J Strength Cond Res 36(8): 2306-2312, 2022-The effect of normobaric hypoxia on strength, body composition, and cardiovascular fitness was investigated after a resistance training intervention in older adults. A single-blinded, randomized control trial recruited 20 healthy adults aged 60-75 years for an 8-week resistance training intervention in normoxia ( n = 10) or normobaric hypoxia (14.4% O 2 ; n = 10). Subjects performed 2 sessions per week of upper-body and lower-body exercises at 70% of 1 repetition maximum (1RM). Pretraining and post-training, maximal oxygen uptake (V̇O 2 max), muscular endurance (30 maximal knee flexions/extensions), and 5RM were assessed, with 5RM used to calculate 1RM. Subjects underwent whole-body dual-energy x-ray absorptiometry (DXA) at pretraining and post-training for fat and lean mass quantification. Significance was set at p < 0.05. Subjects in both groups substantially improved their calculated 1RM strength for leg extension, pectoral fly, row, and squat (normoxia; 30, 38, 27, and 29%, hypoxia; 43, 50, 28, and 64%, respectively); however, hypoxia did not augment this response. Hypoxia did not enhance V̇O 2 max or muscular endurance responses after the training intervention, with no improvements seen in either group. Fat mass and lean mass remained unchanged in both groups after the intervention. In summary, 8 weeks of resistance training in hypoxia was well tolerated in healthy older adults and increased upper-body and lower-body strength. However, the magnitude of strength and lean muscle improvements in hypoxia was no greater than normoxia; therefore, there is currently no evidence to support the use of hypoxic resistance training in older adults.


Asunto(s)
Entrenamiento de Fuerza , Adaptación Fisiológica , Composición Corporal/fisiología , Humanos , Hipoxia , Fuerza Muscular/fisiología , Músculo Esquelético/fisiología
5.
Exp Physiol ; 106(7): 1597-1611, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-33963617

RESUMEN

NEW FINDINGS: What is the central question of this study? Striated muscle activator of rho signalling (STARS) is an actin-binding protein that regulates transcriptional pathways controlling muscle function, growth and myogenesis, processes that are impaired in dystrophic muscle: what is the regulation of the STARS pathway in Duchenne muscular dystrophy (DMD)? What is the main finding and its importance? Members of the STARS signalling pathway are reduced in the quadriceps of patients with DMD and in mouse models of muscular dystrophy. Overexpression of STARS in the dystrophic deficient mdx mouse model increased maximal isometric specific force and upregulated members of the actin cytoskeleton and oxidative phosphorylation pathways. Regulating STARS may be a therapeutic approach to enhance muscle health. ABSTRACT: Duchenne muscular dystrophy (DMD) is characterised by impaired cytoskeleton organisation, cytosolic calcium handling, oxidative stress and mitochondrial dysfunction. This results in progressive muscle damage, wasting and weakness and premature death. The striated muscle activator of rho signalling (STARS) is an actin-binding protein that activates the myocardin-related transcription factor-A (MRTFA)/serum response factor (SRF) transcriptional pathway, a pathway regulating cytoskeletal structure and muscle function, growth and repair. We investigated the regulation of the STARS pathway in the quadriceps muscle from patients with DMD and in the tibialis anterior (TA) muscle from the dystrophin-deficient mdx and dko (utrophin and dystrophin null) mice. Protein levels of STARS, SRF and RHOA were reduced in patients with DMD. STARS, SRF and MRTFA mRNA levels were also decreased in DMD muscle, while Stars mRNA levels were decreased in the mdx mice and Srf and Mrtfa mRNAs decreased in the dko mice. Overexpressing human STARS (hSTARS) in the TA muscles of mdx mice increased maximal isometric specific force by 13% (P < 0.05). This was not associated with changes in muscle mass, fibre cross-sectional area, fibre type, centralised nuclei or collagen deposition. Proteomics screening followed by pathway enrichment analysis identified that hSTARS overexpression resulted in 31 upregulated and 22 downregulated proteins belonging to the actin cytoskeleton and oxidative phosphorylation pathways. These pathways are impaired in dystrophic muscle and regulate processes that are vital for muscle function. Increasing the STARS protein in dystrophic muscle improves muscle force production, potentially via synergistic regulation of cytoskeletal structure and energy production.


Asunto(s)
Distrofia Muscular de Duchenne , Fosforilación Oxidativa , Citoesqueleto de Actina/metabolismo , Animales , Modelos Animales de Enfermedad , Distrofina/genética , Distrofina/metabolismo , Humanos , Ratones , Ratones Endogámicos mdx , Proteínas de Microfilamentos , Músculo Esquelético/metabolismo , Distrofia Muscular de Duchenne/genética , Distrofia Muscular de Duchenne/metabolismo
6.
Am J Physiol Cell Physiol ; 319(2): C432-C440, 2020 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-32608991

RESUMEN

microRNAs (miRNAs) are important regulators of cellular homeostasis and exert their effect by directly controlling protein expression. We have previously reported an age-dependent negative association between microRNA-99b (miR-99b-5p) expression and muscle protein synthesis in human muscle in vivo. Here we investigated the role of miR-99b-5p as a potential negative regulator of protein synthesis via inhibition of mammalian target for rapamycin (MTOR) signaling in human primary myocytes. Overexpressing miR-99b-5p in human primary myotubes from young and old subjects significantly decreased protein synthesis with no effect of donor age. A binding interaction between miR-99b-5p and its putative binding site within the MTOR 3'-untranslated region (UTR) was confirmed in C2C12 myoblasts. The observed decline in protein synthesis was, however, not associated with a suppression of the MTOR protein but of its regulatory associated protein of mTOR complex 1 (RPTOR). These results demonstrate that modulating the expression levels of a miRNA can regulate protein synthesis in human muscle cells and provide a potential mechanism for muscle wasting in vivo.


Asunto(s)
MicroARNs/genética , Fibras Musculares Esqueléticas/metabolismo , Biosíntesis de Proteínas/genética , Serina-Treonina Quinasas TOR/genética , Regiones no Traducidas 3'/genética , Animales , Proliferación Celular/genética , Regulación de la Expresión Génica/genética , Humanos , Diana Mecanicista del Complejo 1 de la Rapamicina/genética , Ratones , Mioblastos/metabolismo , Transducción de Señal/genética
7.
Am J Physiol Endocrinol Metab ; 319(6): E1008-E1018, 2020 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-32954829

RESUMEN

Skeletal muscle is sensitive to environmental cues that are first present in utero. Maternal overnutrition is a model of impaired muscle development leading to structural and metabolic dysfunction in adult life. In this study, we investigated the effect of an obesogenic maternal environment on growth and postnatal myogenesis in the offspring. Male C57BL/6J mice born to chow- or high-fat-diet-fed mothers were allocated to four different groups at the end of weaning. For the following 10 wk, half of the pups were maintained on the same diet as their mother and half of the pups were switched to the other diet (chow or high-fat). At 12 wk of age, muscle injury was induced using an intramuscular injection of barium chloride. Seven days later, mice were humanely killed and muscle tissue was harvested. A high-fat maternal diet impaired offspring growth patterns and downregulated satellite cell activation and markers of postnatal myogenesis 7 days after injury without altering the number of newly synthetized fibers over the whole 7-day period. Importantly, a healthy postnatal diet could not reverse any of these effects. In addition, we demonstrated that postnatal myogenesis was associated with a diet-independent upregulation of three miRNAs, mmu-miR-31-5p, mmu-miR-136-5p, and mmu-miR-296-5p. Furthermore, in vitro analysis confirmed the role of these miRNAs in myocyte proliferation. Our findings are the first to demonstrate that maternal overnutrition impairs markers of postnatal myogenesis in the offspring and are particularly relevant to today's society where the incidence of overweight/obesity in women of childbearing age is increasing.


Asunto(s)
Dieta Alta en Grasa , Crecimiento y Desarrollo/fisiología , Desarrollo de Músculos/fisiología , Efectos Tardíos de la Exposición Prenatal , Células Satélite del Músculo Esquelético/fisiología , Animales , Biomarcadores/análisis , Biomarcadores/metabolismo , Dieta Alta en Grasa/efectos adversos , Femenino , Masculino , Fenómenos Fisiologicos Nutricionales Maternos , Ratones , Ratones Endogámicos C57BL , MicroARNs/genética , MicroARNs/metabolismo , Obesidad/etiología , Obesidad/fisiopatología , Hipernutrición/complicaciones , Hipernutrición/fisiopatología , Embarazo , Complicaciones del Embarazo/etiología , Complicaciones del Embarazo/fisiopatología , Efectos Tardíos de la Exposición Prenatal/etiología , Efectos Tardíos de la Exposición Prenatal/fisiopatología
8.
Exp Physiol ; 105(8): 1326-1338, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32468595

RESUMEN

NEW FINDINGS: What is the central question of this study? Do elevated levels of the stress-response protein NDRG2 protect against fasting and chronic disease in mouse skeletal muscle? What is the main finding and its importance? NDRG2 levels increased in the tibialis anterior muscle in response to fasting and the effects of motor neurone disease. No alleviation of the stress-related and proteasomal pathways, mitochondrial dysfunction or muscle mass loss was observed even with the addition of exogenous NDRG2 indicating that the increase in NDRG2 is a normal adaptive response. ABSTRACT: Skeletal muscle mass loss and dysfunction can arise from stress, which leads to enhanced protein degradation and metabolic impairment. The expression of N-myc downstream-regulated gene 2 (NDRG2) is induced in response to different stressors and is protective against the effects of stress in some tissues and cell types. Here, we investigated the endogenous NDRG2 response to the stress of fasting and chronic disease in mice and whether exogenous NDRG2 overexpression through adeno-associated viral (AAV) treatment ameliorated the response of skeletal muscle to these conditions. Endogenous levels of NDRG2 increased in the tibialis anterior muscle in response to 24 h fasting and with the development of the motor neurone disease, amyotrophic lateral sclerosis, in SOD1G93A transgenic mice. Despite AAV-induced overexpression and increased expression with fasting, NDRG2 was unable to protect against the activation of proteasomal and stress pathways in response to fasting. Furthermore, NDRG2 was unable to reduce muscle mass loss, mitochondrial dysfunction and elevated oxidative and endoplasmic reticulum stress levels in SOD1G93A mice. Conversely, elevated NDRG2 levels did not exacerbate these stress responses. Overall, increasing NDRG2 levels might not be a useful therapeutic strategy to alleviate stress-related disease pathologies in skeletal muscle.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/genética , Músculo Esquelético/metabolismo , Estrés Fisiológico , Animales , Enfermedad Crónica , Modelos Animales de Enfermedad , Estrés del Retículo Endoplásmico , Ayuno , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Mitocondrias , Estrés Oxidativo , Transducción de Señal , Superóxido Dismutasa/metabolismo
9.
Hum Mol Genet ; 26(6): 1087-1103, 2017 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-28087734

RESUMEN

Spinal and bulbar muscular atrophy (SBMA) is a neuromuscular disorder caused by polyglutamine expansion in the androgen receptor (AR) and characterized by the loss of lower motor neurons. Here we investigated pathological processes occurring in muscle biopsy specimens derived from SBMA patients and, as controls, age-matched healthy subjects and patients suffering from amyotrophic lateral sclerosis (ALS) and neurogenic atrophy. We detected atrophic fibers in the muscle of SBMA, ALS and neurogenic atrophy patients. In addition, SBMA muscle was characterized by the presence of a large number of hypertrophic fibers, with oxidative fibers having a larger size compared with glycolytic fibers. Polyglutamine-expanded AR expression was decreased in whole muscle, yet enriched in the nucleus, and localized to mitochondria. Ultrastructural analysis revealed myofibrillar disorganization and streaming in zones lacking mitochondria and degenerating mitochondria. Using molecular (mtDNA copy number), biochemical (citrate synthase and respiratory chain enzymes) and morphological (dark blue area in nicotinamide adenine dinucleotide-stained muscle cross-sections) analyses, we found a depletion of the mitochondria associated with enhanced mitophagy. Mass spectrometry analysis revealed an increase of phosphatidylethanolamines and phosphatidylserines in mitochondria isolated from SBMA muscles, as well as a 50% depletion of cardiolipin associated with decreased expression of the cardiolipin synthase gene. These observations suggest a causative link between nuclear polyglutamine-expanded AR accumulation, depletion of mitochondrial mass, increased mitophagy and altered mitochondrial membrane composition in SBMA muscle patients. Given the central role of mitochondria in cell bioenergetics, therapeutic approaches toward improving the mitochondrial network are worth considering to support SBMA patients.


Asunto(s)
Esclerosis Amiotrófica Lateral/genética , Trastornos Musculares Atróficos/genética , Péptidos/genética , Receptores Androgénicos/genética , Adulto , Anciano , Anciano de 80 o más Años , Esclerosis Amiotrófica Lateral/fisiopatología , Andrógenos/metabolismo , Animales , Biopsia , ADN Mitocondrial/genética , Femenino , Humanos , Masculino , Persona de Mediana Edad , Mitofagia/genética , Neuronas Motoras/metabolismo , Neuronas Motoras/patología , Músculo Esquelético/irrigación sanguínea , Músculo Esquelético/metabolismo , Músculo Esquelético/patología , Trastornos Musculares Atróficos/fisiopatología
10.
Muscle Nerve ; 57(5): 838-847, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29236291

RESUMEN

INTRODUCTION: The pathology of amyotrophic lateral sclerosis (ALS) is associated with impaired RNA processing and microRNA (miRNA) dysregulation. Here we investigate the regulation of the members of the miRNA biogenesis pathways and total miRNA levels at different stages of the disease. METHODS: Muscle, brain, and spinal cord tissue were obtained from presymptomatic, symptomatic, and end-stage superoxide dismutase 1 (SOD1)G93A mice. miRNA and transcript levels were measured by quantitative polymerase chain reaction. RESULTS: As the diseases progresses, several genes involved in miRNA biogenesis as well as the miRNA/total RNA (totRNA) ratio increased in the tibialis anterior (TA) muscle but not in the soleus or in neural tissue. DISCUSSION: We propose that a dysregulation in the miRNA/totRNA ratio in the TA muscle from SOD1G93A mice reflects a pathological increase in miRNA biogenesis machinery. Alterations in the miRNA/totRNA ratio influence the levels of reference noncoding RNAs and may therefore potentially compromise the accuracy of commonly used miRNA normalization strategies. Muscle Nerve 57: 838-847, 2018.


Asunto(s)
Esclerosis Amiotrófica Lateral/patología , Regulación de la Expresión Génica/genética , MicroARNs/metabolismo , Músculo Esquelético/metabolismo , ARN/metabolismo , Factores de Edad , Esclerosis Amiotrófica Lateral/genética , Análisis de Varianza , Animales , Estudios de Cohortes , Modelos Animales de Enfermedad , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Neuronas/metabolismo , Superóxido Dismutasa/genética
11.
Eur J Nutr ; 57(1): 363-372, 2018 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-27785566

RESUMEN

PURPOSE: Telomere length is a biomarker of cellular ageing, with longer telomeres associated with longevity and reduced risk of chronic disease in older age. Consumption of a healthy diet may contribute to longevity via its impact on cellular ageing, but studies on diet and telomere length to date have been limited and their findings equivocal. The aim of this study was to examine associations between three indices of diet quality and telomere length in older men and women. METHODS: Adults aged 57-68 years participating in the Wellbeing, Eating and Exercise for a Long Life (WELL) study in Victoria, Australia (n = 679), completed a postal survey including an 111-item food frequency questionnaire in 2012. Diet quality was assessed via three indices: the Dietary Guideline Index, the Recommended Food Score, and the Mediterranean Diet Score. Relative telomere length was measured by quantitative polymerase chain reaction. Associations between diet quality and telomere length were assessed using linear regression adjusted for covariates. RESULTS: After adjustment for age, sex, education, smoking, physical activity, and body mass index (BMI), there were no significant associations between diet quality and relative telomere length. CONCLUSIONS: In a sample of older adults residing in Victoria, Australia, men and women aged 57-68 years with better-quality diets did not have longer telomeres. Further investigation in longitudinal studies will determine whether diet can influence telomere length over time in an ageing population.


Asunto(s)
Envejecimiento/fisiología , Dieta Saludable , Homeostasis del Telómero/fisiología , Anciano , Índice de Masa Corporal , Supervivencia Celular , Estudios Transversales , Dieta , Encuestas sobre Dietas , Ejercicio Físico , Femenino , Humanos , Longevidad/fisiología , Estudios Longitudinales , Masculino , Persona de Mediana Edad , Victoria
12.
Nature ; 484(7394): 394-8, 2012 Apr 04.
Artículo en Inglés | MEDLINE | ID: mdl-22495301

RESUMEN

Duchenne muscular dystrophy (DMD) is a severe and progressive muscle wasting disorder caused by mutations in the dystrophin gene that result in the absence of the membrane-stabilizing protein dystrophin. Dystrophin-deficient muscle fibres are fragile and susceptible to an influx of Ca(2+), which activates inflammatory and muscle degenerative pathways. At present there is no cure for DMD, and existing therapies are ineffective. Here we show that increasing the expression of intramuscular heat shock protein 72 (Hsp72) preserves muscle strength and ameliorates the dystrophic pathology in two mouse models of muscular dystrophy. Treatment with BGP-15 (a pharmacological inducer of Hsp72 currently in clinical trials for diabetes) improved muscle architecture, strength and contractile function in severely affected diaphragm muscles in mdx dystrophic mice. In dko mice, a phenocopy of DMD that results in severe spinal curvature (kyphosis), muscle weakness and premature death, BGP-15 decreased kyphosis, improved the dystrophic pathophysiology in limb and diaphragm muscles and extended lifespan. We found that the sarcoplasmic/endoplasmic reticulum Ca(2+)-ATPase (SERCA, the main protein responsible for the removal of intracellular Ca(2+)) is dysfunctional in severely affected muscles of mdx and dko mice, and that Hsp72 interacts with SERCA to preserve its function under conditions of stress, ultimately contributing to the decreased muscle degeneration seen with Hsp72 upregulation. Treatment with BGP-15 similarly increased SERCA activity in dystrophic skeletal muscles. Our results provide evidence that increasing the expression of Hsp72 in muscle (through the administration of BGP-15) has significant therapeutic potential for DMD and related conditions, either as a self-contained therapy or as an adjuvant with other potential treatments, including gene, cell and pharmacological therapies.


Asunto(s)
Progresión de la Enfermedad , Proteínas del Choque Térmico HSP72/metabolismo , Músculo Esquelético/fisiología , Distrofia Muscular de Duchenne/metabolismo , Distrofia Muscular de Duchenne/fisiopatología , Animales , ATPasas Transportadoras de Calcio/metabolismo , Diafragma/efectos de los fármacos , Diafragma/fisiología , Modelos Animales de Enfermedad , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Proteínas del Choque Térmico HSP72/biosíntesis , Proteínas del Choque Térmico HSP72/genética , Cifosis/tratamiento farmacológico , Longevidad/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos mdx , Ratones Transgénicos , Músculo Esquelético/efectos de los fármacos , Músculo Esquelético/fisiopatología , Distrofia Muscular de Duchenne/genética , Distrofia Muscular de Duchenne/patología , Oximas/farmacología , Piperidinas/farmacología , Ratas
13.
Eur J Appl Physiol ; 118(3): 617-627, 2018 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-29350278

RESUMEN

PURPOSE: We examined the concurrent characteristics of the remote development of strength and cross-sectional area (CSA) of upper body skeletal muscle in response to lower body resistance training performed with an applied blood flow restriction (BFR). METHODS: Males allocated to an experimental BFR group (EXP; n = 12) or a non-BFR control group (CON; n = 12) completed 7-weeks of resistance training comprising three sets of unilateral bicep curls [50% 1-repetition maximum (1-RM)], then four sets of bilateral knee extension and flexion exercises (30% 1-RM). EXP performed leg exercises with an applied BFR (60% limb occlusion pressure). 1-RM strength was measured using bilateral leg exercises and unilateral bicep curls in both trained and untrained arms. Muscle CSA was measured via peripheral quantitative computed tomography in the dominant leg and both arms. RESULTS: 1-RM in the trained arm increased more in EXP (2.5 ± 0.4 kg; mean ± SEM) than the contralateral untrained arm (0.8 ± 0.4 kg), and the trained arm of CON (0.6 ± 0.3 kg, P < 0.05). The increase in knee extension 1-RM was twofold that of CON (P < 0.01). Knee flexion 1-RM, leg CSA, and trained arm CSA increased similarly between groups (P > 0.05), while untrained arm CSA did not change (P > 0.05). CONCLUSION: Lower limb BFR training increased trained arm strength more than the contralateral untrained arm, and the trained arm of controls. However, there was no additional effect on muscle CSA. These findings support evidence for a BFR training-derived remote strength transfer that may be relevant to populations with localised movement disorders.


Asunto(s)
Adaptación Fisiológica , Brazo/fisiología , Pierna/irrigación sanguínea , Fuerza Muscular , Músculo Esquelético/fisiología , Acondicionamiento Físico Humano/métodos , Flujo Sanguíneo Regional , Adulto , Humanos , Aparatos de Compresión Neumática Intermitente , Pierna/fisiología , Masculino , Acondicionamiento Físico Humano/instrumentación
14.
Proc Natl Acad Sci U S A ; 112(49): E6780-9, 2015 Dec 08.
Artículo en Inglés | MEDLINE | ID: mdl-26598680

RESUMEN

Classic physiology studies dating to the 1930s demonstrate that moderate or transient glucocorticoid (GC) exposure improves muscle performance. The ergogenic properties of GCs are further evidenced by their surreptitious use as doping agents by endurance athletes and poorly understood efficacy in Duchenne muscular dystrophy (DMD), a genetic muscle-wasting disease. A defined molecular basis underlying these performance-enhancing properties of GCs in skeletal muscle remains obscure. Here, we demonstrate that ergogenic effects of GCs are mediated by direct induction of the metabolic transcription factor KLF15, defining a downstream pathway distinct from that resulting in GC-related muscle atrophy. Furthermore, we establish that KLF15 deficiency exacerbates dystrophic severity and muscle GC-KLF15 signaling mediates salutary therapeutic effects in the mdx mouse model of DMD. Thus, although glucocorticoid receptor (GR)-mediated transactivation is often associated with muscle atrophy and other adverse effects of pharmacologic GC administration, our data define a distinct GR-induced gene regulatory pathway that contributes to therapeutic effects of GCs in DMD through proergogenic metabolic programming.


Asunto(s)
Glucocorticoides/farmacología , Músculo Esquelético/efectos de los fármacos , Distrofia Muscular de Duchenne/tratamiento farmacológico , Animales , Femenino , Glucocorticoides/uso terapéutico , Humanos , Factores de Transcripción de Tipo Kruppel/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Músculo Esquelético/fisiopatología , Proteínas Nucleares/fisiología , Receptores de Glucocorticoides/fisiología
15.
BMC Genomics ; 18(1): 52, 2017 01 07.
Artículo en Inglés | MEDLINE | ID: mdl-28061746

RESUMEN

BACKGROUND: MiRNAs are essential regulators of skeletal muscle development and homeostasis. To date, the role and regulation of miRNAs in myogenesis have been mostly studied in tissue culture and during embryogenesis. However, little information relating to miRNA regulation during early post-natal skeletal muscle growth in mammals is available. Using a high-throughput miRNA qPCR-based array, followed by stringent statistical and bioinformatics analysis, we describe the expression pattern and putative role of 768 miRNAs in the quadriceps muscle of mice aged 2 days, 2 weeks, 4 weeks and 12 weeks. RESULTS: Forty-six percent of all measured miRNAs were expressed in mouse quadriceps muscle during the first 12 weeks of life. We report unprecedented changes in miRNA expression levels over time. The expression of a majority of miRNAs significantly decreased with post-natal muscle maturation in vivo. MiRNA clustering identified 2 subsets of miRNAs that are potentially involved in cell proliferation and differentiation, mainly via the regulation of non-muscle specific targets. CONCLUSION: Collective miRNA expression in mouse quadriceps muscle is subjected to substantial levels of regulation during the first 12 weeks of age. This study identified a new suite of highly conserved miRNAs that are predicted to influence early muscle development. As such it provides novel knowledge pertaining to post-natal myogenesis and muscle regeneration in mammals.


Asunto(s)
Perfilación de la Expresión Génica , MicroARNs/genética , Desarrollo de Músculos/genética , Músculo Esquelético/crecimiento & desarrollo , Animales , Masculino , Ratones , Ratones Endogámicos C57BL , Factores Reguladores Miogénicos/genética
16.
FASEB J ; 30(2): 674-87, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26481306

RESUMEN

Skeletal muscle mitochondrial content and oxidative capacity are important determinants of muscle function and whole-body health. Mitochondrial content and function are enhanced by endurance exercise and impaired in states or diseases where muscle function is compromised, such as myopathies, muscular dystrophies, neuromuscular diseases, and age-related muscle atrophy. Hence, elucidating the mechanisms that control muscle mitochondrial content and oxidative function can provide new insights into states and diseases that affect muscle health. In past studies, we identified Perm1 (PPARGC1- and ESRR-induced regulator, muscle 1) as a gene induced by endurance exercise in skeletal muscle, and regulating mitochondrial oxidative function in cultured myotubes. The capacity of Perm1 to regulate muscle mitochondrial content and function in vivo is not yet known. In this study, we use adeno-associated viral (AAV) vectors to increase Perm1 expression in skeletal muscles of 4-wk-old mice. Compared to control vector, AAV1-Perm1 leads to significant increases in mitochondrial content and oxidative capacity (by 40-80%). Moreover, AAV1-Perm1-transduced muscles show increased capillary density and resistance to fatigue (by 33 and 31%, respectively), without prominent changes in fiber-type composition. These findings suggest that Perm1 selectively regulates mitochondrial biogenesis and oxidative function, and implicate Perm1 in muscle adaptations that also occur in response to endurance exercise.


Asunto(s)
Regulación de la Expresión Génica/fisiología , Mitocondrias/metabolismo , Fatiga Muscular/fisiología , Proteínas Musculares/metabolismo , Músculo Esquelético/metabolismo , Animales , Dependovirus , Ratones , Ratones Endogámicos C57BL , Proteínas Musculares/genética , Oxidación-Reducción
17.
Mediators Inflamm ; 2017: 7517350, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29362521

RESUMEN

Granulocyte colony-stimulating factor (G-CSF) was originally discovered in the context of hematopoiesis. However, the identification of the G-CSF receptor (G-CSFR) being expressed outside the hematopoietic system has revealed wider roles for G-CSF, particularly in tissue repair and regeneration. Skeletal muscle damage, including that following strenuous exercise, induces an elevation in plasma G-CSF, implicating it as a potential mediator of skeletal muscle repair. This has been supported by preclinical studies and clinical trials investigating G-CSF as a potential therapeutic agent in relevant disease states. This review focuses on the growing literature associated with G-CSF and G-CSFR in skeletal muscle under healthy and disease conditions and highlights the current controversies.


Asunto(s)
Factor Estimulante de Colonias de Granulocitos/farmacología , Músculo Esquelético/efectos de los fármacos , Receptores de Factor Estimulante de Colonias de Granulocito/fisiología , Regeneración/efectos de los fármacos , Animales , Factor Estimulante de Colonias de Granulocitos/sangre , Factor Estimulante de Colonias de Granulocitos/uso terapéutico , Humanos , Músculo Esquelético/fisiología , Enfermedades Musculares/tratamiento farmacológico , Fosfatidilinositol 3-Quinasas/fisiología , Transducción de Señal/efectos de los fármacos
18.
Am J Physiol Regul Integr Comp Physiol ; 310(11): R1297-311, 2016 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-27101297

RESUMEN

We compared the effects of concurrent exercise, incorporating either high-intensity interval training (HIT) or moderate-intensity continuous training (MICT), on mechanistic target of rapamycin complex 1 (mTORC1) signaling and microRNA expression in skeletal muscle, relative to resistance exercise (RE) alone. Eight males (mean ± SD: age, 27 ± 4 yr; V̇o2 peak , 45.7 ± 9 ml·kg(-1)·min(-1)) performed three experimental trials in a randomized order: 1) RE (8 × 5 leg press repetitions at 80% 1-repetition maximum) performed alone and RE preceded by either 2) HIT cycling [10 × 2 min at 120% lactate threshold (LT); HIT + RE] or 3) work-matched MICT cycling (30 min at 80% LT; MICT + RE). Vastus lateralis muscle biopsies were obtained immediately before RE, either without (REST) or with (POST) preceding endurance exercise and +1 h (RE + 1 h) and +3 h (RE + 3 h) after RE. Prior HIT and MICT similarly reduced muscle glycogen content and increased ACC(Ser79) and p70S6K(Thr389) phosphorylation before subsequent RE (i.e., at POST). Compared with MICT, HIT induced greater mTOR(Ser2448) and rps6(Ser235/236) phosphorylation at POST. RE-induced increases in p70S6K and rps6 phosphorylation were not influenced by prior HIT or MICT; however, mTOR phosphorylation was reduced at RE + 1 h for MICT + RE vs. both HIT + RE and RE. Expression of miR-133a, miR-378, and miR-486 was reduced at RE + 1 h for HIT + RE vs. both MICT + RE and RE. Postexercise mTORC1 signaling following RE is therefore not compromised by prior HIT or MICT, and concurrent exercise incorporating HIT, but not MICT, reduces postexercise expression of miRNAs implicated in skeletal muscle adaptation to RE.


Asunto(s)
Entrenamiento de Intervalos de Alta Intensidad/métodos , MicroARNs/metabolismo , Complejos Multiproteicos/metabolismo , Músculo Esquelético/fisiología , Resistencia Física/fisiología , Entrenamiento de Fuerza/métodos , Serina-Treonina Quinasas TOR/metabolismo , Adaptación Fisiológica/fisiología , Adulto , Humanos , Masculino , Diana Mecanicista del Complejo 1 de la Rapamicina , Acondicionamiento Físico Humano/métodos , Transducción de Señal/fisiología
19.
Nat Chem Biol ; 10(2): 113-21, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24362705

RESUMEN

Phenotypic screening is making a comeback in drug discovery as the maturation of chemical proteomics methods has facilitated target identification for bioactive small molecules. A limitation of these approaches is that time-consuming genetic methods or other means are often required to determine the biologically relevant target (or targets) from among multiple protein-compound interactions that are typically detected. Here, we have combined phenotypic screening of a directed small-molecule library with competitive activity-based protein profiling to map and functionally characterize the targets of screening hits. Using this approach, we identify carboxylesterase 3 (Ces3, also known as Ces1d) as a primary molecular target of bioactive compounds that promote lipid storage in adipocytes. We further show that Ces3 activity is markedly elevated during adipocyte differentiation. Treatment of two mouse models of obesity-diabetes with a Ces3 inhibitor ameliorates multiple features of metabolic syndrome, illustrating the power of the described strategy to accelerate the identification and pharmacologic validation of new therapeutic targets.


Asunto(s)
Hidrolasas de Éster Carboxílico/genética , Hidrolasas de Éster Carboxílico/metabolismo , Diabetes Mellitus/genética , Obesidad/genética , Fenotipo , Bibliotecas de Moléculas Pequeñas , Animales , Células Cultivadas , Modelos Animales de Enfermedad , Sistemas de Liberación de Medicamentos , Descubrimiento de Drogas , Ratones , Análisis por Matrices de Proteínas , Proteómica
20.
Biochim Biophys Acta ; 1840(4): 1276-84, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24291686

RESUMEN

BACKGROUND: Maintaining skeletal muscle mitochondrial content and function is important for sustained health throughout the lifespan. Exercise stimulates important key stress signals that control skeletal mitochondrial biogenesis and function. Perturbations in mitochondrial content and function can directly or indirectly impact skeletal muscle function and consequently whole-body health and wellbeing. SCOPE OF REVIEW: This review will describe the exercise-stimulated stress signals and molecular mechanisms positively regulating mitochondrial biogenesis and function. It will then discuss the major myopathies, neuromuscular diseases and conditions such as diabetes and ageing that have dysregulated mitochondrial function. Finally, the impact of exercise and potential pharmacological approaches to improve mitochondrial function in diseased populations will be discussed. MAJOR CONCLUSIONS: Exercise activates key stress signals that positively impact major transcriptional pathways that transcribe genes involved in skeletal muscle mitochondrial biogenesis, fusion and metabolism. The positive impact of exercise is not limited to younger healthy adults but also benefits skeletal muscle from diseased populations and the elderly. Impaired mitochondrial function can directly influence skeletal muscle atrophy and contribute to the risk or severity of disease conditions. Pharmacological manipulation of exercise-induced pathways that increase skeletal muscle mitochondrial biogenesis and function in critically ill patients, where exercise may not be possible, may assist in the treatment of chronic disease. GENERAL SIGNIFICANCE: This review highlights our understanding of how exercise positively impacts skeletal muscle mitochondrial biogenesis and function. Exercise not only improves skeletal muscle mitochondrial health but also enables us to identify molecular mechanisms that may be attractive targets for therapeutic manipulation. This article is part of a Special Issue entitled Frontiers of mitochondrial research.


Asunto(s)
Ejercicio Físico/fisiología , Salud , Mitocondrias Musculares/fisiología , Músculo Esquelético/fisiología , Enfermedades Musculares/etiología , Adulto , Animales , Humanos , MicroARNs/fisiología , Enfermedades Mitocondriales/terapia , Mitofagia/fisiología , Músculo Esquelético/ultraestructura , Enfermedades Musculares/terapia
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda