Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Cells ; 10(8)2021 08 10.
Artículo en Inglés | MEDLINE | ID: mdl-34440812

RESUMEN

ALS is a fatal neurodegenerative disease that is associated with muscle atrophy, motoneuron degeneration and denervation. Different mechanisms have been proposed to explain the pathogenesis of the disease; in this context, microRNAs have been described as biomarkers and potential pathogenetic factors for ALS. MyomiRs are microRNAs produced by skeletal muscle, and they play an important role in tissue homeostasis; moreover, they can be released in blood circulation in pathological conditions, including ALS. However, the functional role of myomiRs in muscle denervation has not yet been fully clarified. In this study, we analyze the levels of two myomiRs, namely miR-206 and miR-133a, in skeletal muscle and blood samples of denervated mice, and we demonstrate that surgical denervation reduces the expression of both miR-206 and miR-133a, while miR-206 but not miR-133a is upregulated during the re-innervation process. Furthermore, we quantify the levels of miR-206 and miR-133a in serum samples of two ALS mouse models, characterized by different disease velocities, and we demonstrate a different modulation of circulating myomiRs during ALS disease, according to the velocity of disease progression. Moreover, taking into account surgical and pathological denervation, we describe a different response to increasing amounts of circulating miR-206, suggesting a hormetic effect of miR-206 in relation to changes in neuromuscular communication.


Asunto(s)
Esclerosis Amiotrófica Lateral/patología , MicroARNs/sangre , Músculo Esquelético/metabolismo , Esclerosis Amiotrófica Lateral/genética , Esclerosis Amiotrófica Lateral/cirugía , Animales , Biomarcadores/metabolismo , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Regulación de la Expresión Génica , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , MicroARNs/metabolismo , Desnervación Muscular , Músculo Esquelético/inervación , Superóxido Dismutasa-1/genética , Superóxido Dismutasa-1/metabolismo
2.
Aging Cell ; 18(3): e12954, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30953403

RESUMEN

The decline in skeletal muscle mass and strength occurring in aging, referred as sarcopenia, is the result of many factors including an imbalance between protein synthesis and degradation, changes in metabolic/hormonal status, and in circulating levels of inflammatory mediators. Thus, factors that increase muscle mass and promote anabolic pathways might be of therapeutic benefit to counteract sarcopenia. Among these, the insulin-like growth factor-1 (IGF-1) has been implicated in many anabolic pathways in skeletal muscle. IGF-1 exists in different isoforms that might exert different role in skeletal muscle. Here we study the effects of two full propeptides IGF-1Ea and IGF-1Eb in skeletal muscle, with the aim to define whether and through which mechanisms their overexpression impacts muscle aging. We report that only IGF-1Ea expression promotes a pronounced hypertrophic phenotype in young mice, which is maintained in aged mice. Nevertheless, examination of aged transgenic mice revealed that the local expression of either IGF-1Ea or IGF-1Eb transgenes was protective against age-related loss of muscle mass and force. At molecular level, both isoforms activate the autophagy/lysosome system, normally altered during aging, and increase PGC1-α expression, modulating mitochondrial function, ROS detoxification, and the basal inflammatory state occurring at old age. Moreover, morphological integrity of neuromuscular junctions was maintained and preserved in both MLC/IGF-1Ea and MLC/IGF-1Eb mice during aging. These data suggest that IGF-1 is a promising therapeutic agent in staving off advancing muscle weakness.


Asunto(s)
Factor I del Crecimiento Similar a la Insulina/metabolismo , Músculo Esquelético/metabolismo , Sarcopenia/metabolismo , Envejecimiento , Animales , Inflamación/metabolismo , Factor I del Crecimiento Similar a la Insulina/genética , Ratones , Ratones Transgénicos , Mitocondrias/metabolismo , Músculo Esquelético/crecimiento & desarrollo , Unión Neuromuscular/genética , Unión Neuromuscular/metabolismo , Unión Neuromuscular/patología , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma/metabolismo , Isoformas de Proteínas/química , Isoformas de Proteínas/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Sarcopenia/patología , Transducción de Señal/genética , Regulación hacia Arriba/genética
3.
Front Physiol ; 9: 831, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30042688

RESUMEN

Amyotrophic lateral sclerosis (ALS) is a severe neurodegenerative disorder, classified into sporadic or familial forms and characterized by motor neurons death, muscle atrophy, weakness, and paralysis. Among the familial cases of ALS, approximately 20% are caused by dominant mutations in the gene coding for superoxide dismutase (SOD1) protein. Of note, mutant SOD1 toxicity is not necessarily limited to the central nervous system. ALS is indeed a multi-systemic and multifactorial disease that affects whole body physiology and induces severe metabolic changes in several tissues, including skeletal muscle. Nevertheless, whether alterations in the plasticity, heterogeneity, and metabolism of muscle fibers are the result of motor neuron degeneration or alternatively occur independently of it remain to be elucidated. To address this issue, we made use of a mouse model (MLC/SOD1G93A) that overexpresses the SOD1 mutant gene selectively in skeletal muscle. We found an alteration in the metabolic properties of skeletal muscle characterized by alteration in fiber type composition and metabolism. Indeed, we observed an alteration of muscle glucose metabolism associated with the induction of Phosphofructokinases and Pyruvate dehydrogenase kinase 4 expression. The upregulation of Pyruvate dehydrogenase kinase 4 led to the inhibition of Pyruvate conversion into Acetyl-CoA. Moreover, we demonstrated that the MLC/SOD1G93A transgene was associated with an increase of lipid catabolism and with the inhibition of fat deposition inside muscle fibers. All together these data demonstrate that muscle expression of the SOD1G93A gene induces metabolic changes, along with a preferential use of lipid energy fuel by muscle fibers. We provided evidences that muscle metabolic alterations occurred before disease symptoms and independently of motor neuron degeneration, indicating that skeletal muscle is likely an important therapeutic target in ALS.

4.
Physiol Rep ; 5(7)2017 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-28364027

RESUMEN

Skeletal muscle myopathy is universal in cirrhotic patients, however, little is known about the main mechanisms involved. The study aims to investigate skeletal muscle morphological, histological, and functional modifications in experimental models of cirrhosis and the principal molecular pathways responsible for skeletal muscle myopathy. Cirrhosis was induced by bile duct ligation (BDL) and carbon tetrachloride (CCl4) administration in mice. Control animals (CTR) underwent bile duct exposure or vehicle administration only. At sacrifice, peripheral muscles were dissected and weighed. Contractile properties of extensor digitorum longus (EDL) were studied in vitro. Muscle samples were used for histological and molecular analysis. Quadriceps muscle histology revealed a significant reduction in cross-sectional area of muscle and muscle fibers in cirrhotic mice with respect to CTR. Kinetic properties of EDL in both BDL and CCl4 were reduced with respect to CTR; BDL mice also showed a reduction in muscle force and a decrease in the resistance to fatigue. Increase in myostatin expression associated with a decrease in AKT-mTOR expressions was observed in BDL mice, together with an increase in LC3 protein levels. Upregulation of the proinflammatory citochines TNF-a and IL6 and an increased expression of NF-kB and MuRF-1 were observed in CCl4 mice. In conclusion, skeletal muscle myopenia was present in experimental models of BDL and CCl4-induced cirrhosis. Moreover, reduction in protein synthesis and activation of protein degradation were the main mechanisms responsible for myopenia in BDL mice, while activation of ubiquitin-pathway through inflammatory cytokines seems to be the main potential mechanism involved in CCl4 mice.


Asunto(s)
Cirrosis Hepática Biliar/complicaciones , Cirrosis Hepática Experimental/complicaciones , Enfermedades Musculares/etiología , Animales , Tetracloruro de Carbono , Modelos Animales de Enfermedad , Interleucina-6/metabolismo , Ligadura , Cirrosis Hepática Biliar/metabolismo , Cirrosis Hepática Biliar/patología , Cirrosis Hepática Experimental/inducido químicamente , Cirrosis Hepática Experimental/metabolismo , Cirrosis Hepática Experimental/patología , Ratones , Contracción Muscular/fisiología , Proteínas Musculares/metabolismo , Músculo Esquelético/patología , Enfermedades Musculares/metabolismo , Enfermedades Musculares/patología , FN-kappa B/metabolismo , Proteínas de Motivos Tripartitos/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo
6.
Int J Cardiol ; 214: 137-47, 2016 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-27060274

RESUMEN

BACKGROUND: Exercise intolerance is one of the main clinical symptoms of heart failure (HF) and is associated with skeletal muscle wasting due to an imbalance between proteolysis and protein synthesis. In this study, we tested whether aerobic exercise training (AET) would counteract skeletal muscle atrophy by activating IGF-I/Akt/mTOR pathway in HF mice. METHODS: Sympathetic hyperactivity induced HF mice were assigned into 8-week moderate intensity AET. Untrained wild type and HF mice were used as control. Soleus cross sectional area was evaluated by histochemistry and motor performance by rotarod. 26S proteasome activity was assessed by fluorimetric assay, and components of IGF-I/Akt/mTOR pathway or myostatin pathway by qRT-PCR or immunoblotting. A different subset of mice was used to evaluate the relative contribution of mTOR inhibition (rapamycin) or activation (leucine) on AET-induced changes in muscle mass regulation. RESULTS: AET prevented exercise intolerance and impaired motor performance in HF mice. These effects were associated with attenuation of soleus atrophy. Rapamycin treatment precluded AET effects on soleus mass in HF mice suggesting the involvement of IGF signaling pathway in this response. In fact, AET increased IGF-I Ea and IGF-I Pan mRNA levels, while it reduced myostatin and Smad2 mRNA levels in HF mice. At protein levels, AET prevented reduced expression levels of IGF-I, pAkt (at basal state), as well as, p4E-BP1 and pP70(S6K) (leucine-stimulated state) in HF mice. Additionally, AET prevented 26S proteasome hyperactivity in HF mice. CONCLUSIONS: Taken together, our data provide evidence for AET-induced activation of IGF-I/Akt/mTOR signaling pathway counteracting HF-induced muscle wasting.


Asunto(s)
Insuficiencia Cardíaca/metabolismo , Músculo Esquelético/metabolismo , Atrofia Muscular/metabolismo , Condicionamiento Físico Animal/fisiología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Animales , Insuficiencia Cardíaca/terapia , Factor I del Crecimiento Similar a la Insulina/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Atrofia Muscular/prevención & control , Condicionamiento Físico Animal/métodos , ARN Mensajero/metabolismo , Transducción de Señal/fisiología
7.
Physiol Rep ; 4(24)2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-28039397

RESUMEN

Age-related sarcopenia is characterized by a progressive loss of muscle mass with decline in specific force, having dramatic consequences on mobility and quality of life in seniors. The etiology of sarcopenia is multifactorial and underlying mechanisms are currently not fully elucidated. Physical exercise is known to have beneficial effects on muscle trophism and force production. Alterations of mitochondrial Ca2+ homeostasis regulated by mitochondrial calcium uniporter (MCU) have been recently shown to affect muscle trophism in vivo in mice. To understand the relevance of MCU-dependent mitochondrial Ca2+ uptake in aging and to investigate the effect of physical exercise on MCU expression and mitochondria dynamics, we analyzed skeletal muscle biopsies from 70-year-old subjects 9 weeks trained with either neuromuscular electrical stimulation (ES) or leg press. Here, we demonstrate that improved muscle function and structure induced by both trainings are linked to increased protein levels of MCU Ultrastructural analyses by electron microscopy showed remodeling of mitochondrial apparatus in ES-trained muscles that is consistent with an adaptation to physical exercise, a response likely mediated by an increased expression of mitochondrial fusion protein OPA1. Altogether these results indicate that the ES-dependent physiological effects on skeletal muscle size and force are associated with changes in mitochondrial-related proteins involved in Ca2+ homeostasis and mitochondrial shape. These original findings in aging human skeletal muscle confirm the data obtained in mice and propose MCU and mitochondria-related proteins as potential pharmacological targets to counteract age-related muscle loss.


Asunto(s)
Envejecimiento , Canales de Calcio/metabolismo , Ejercicio Físico , Mitocondrias/fisiología , Músculo Esquelético/metabolismo , Músculo Esquelético/fisiología , Sarcopenia/metabolismo , Anciano , Atrofia , Estimulación Eléctrica , Femenino , Humanos , Factor I del Crecimiento Similar a la Insulina/metabolismo , Contracción Isométrica , Masculino , Mitocondrias/ultraestructura , Músculo Esquelético/ultraestructura , Sarcopenia/prevención & control , Conducta Sedentaria
8.
Mol Ther ; 23(7): 1189-1200, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25896247

RESUMEN

Insulin-like growth factor 1 (IGF-1) is a potent enhancer of tissue regeneration, and its overexpression in muscle injury leads to hastened resolution of the inflammatory phase. Here, we show that monocytes/macrophages constitute an important initial source of IGF-1 in muscle injury, as conditional deletion of the IGF-1 gene specifically in mouse myeloid cells (ϕIGF-1 CKO) blocked the normal surge of local IGF-1 in damaged muscle and significantly compromised regeneration. In injured muscle, Ly6C+ monocytes/macrophages and CD206+ macrophages expressed equivalent IGF-1 levels, which were transiently upregulated during transition from the inflammation to repair. In injured ϕIGF-1 CKO mouse muscle, accumulation of CD206+ macrophages was impaired, while an increase in Ly6C+ monocytes/macrophages was favored. Transcriptional profiling uncovered inflammatory skewing in ϕIGF-1 CKO macrophages, which failed to fully induce a reparative gene program in vitro or in vivo, revealing a novel autocrine role for IGF-1 in modulating murine macrophage phenotypes. These data establish local macrophage-derived IGF-1 as a key factor in inflammation resolution and macrophage polarization during muscle regeneration.


Asunto(s)
Factor I del Crecimiento Similar a la Insulina/biosíntesis , Músculo Esquelético/crecimiento & desarrollo , Regeneración/genética , Cicatrización de Heridas , Animales , Comunicación Autocrina/genética , Regulación del Desarrollo de la Expresión Génica , Inflamación/genética , Inflamación/patología , Factor I del Crecimiento Similar a la Insulina/genética , Macrófagos/metabolismo , Macrófagos/patología , Ratones , Monocitos/metabolismo , Músculo Esquelético/metabolismo
9.
In Vivo ; 28(5): 993-6, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25189920

RESUMEN

BACKGROUND: After a review of clinical cases of the Unit of Plastic Surgery of the University of Siena, Italy, we found that 22 patients undergoing lipofilling for breast recontruction needed less pain drugs compared to 18 patients which did not undergo lipofilling. In this work, the postoperative pain was analyzed in two groups of patients: a cohort treated with prosthesis and a cohort treated with prosthesis implant together with a lipofilling procedure. PATIENTS AND METHODS: During the immediate postoperative period, a visual analog scale for pain was submitted to every patient every eight hours until they were discharged, then every day for a week, every two days during the second week and once a week in the first three months. The administration of analgesics was also registered. RESULTS: Pain intensity was lower in the group treated with prosthesis and lipofilling. CONCLUSION: Fat transplant is a procedure well-tolerated by patients, resulting in a lower rate of pain.


Asunto(s)
Implantación de Mama/efectos adversos , Dolor Postoperatorio/etiología , Adulto , Analgésicos/uso terapéutico , Femenino , Humanos , Persona de Mediana Edad , Dimensión del Dolor , Dolor Postoperatorio/diagnóstico , Dolor Postoperatorio/tratamiento farmacológico , Resultado del Tratamiento
10.
PLoS One ; 9(9): e107753, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25229238

RESUMEN

Muscle regeneration involves the activation of satellite cells, is regulated at the genetic and epigenetic levels, and is strongly influenced by gene activation and environmental conditions. The aim of this study was to determine whether the overexpression of mIGF-1 can modify functional features of satellite cells during the differentiation process, particularly in relation to modifications of intracellular Ca2+ handling. Satellite cells were isolated from wild-type and MLC/mIGF-1 transgenic mice. The cells were differentiated in vitro, and morphological analyses, intracellular Ca2+ measurements, and ionic current recordings were performed. mIGF-1 overexpression accelerates satellite cell differentiation and promotes myotube hypertrophy. In addition, mIGF-1 overexpression-induced potentiation of myogenesis triggers both quantitative and qualitative changes to the control of intracellular Ca2+ handling. In particular, the differentiated MLC/mIGF-1 transgenic myotubes have reduced velocity and amplitude of intracellular Ca2+ increases after stimulation with caffeine, KCl and acetylcholine. This appears to be due, at least in part, to changes in the physico-chemical state of the sarcolemma (increased membrane lipid oxidation, increased output currents) and to increased expression of dihydropyridine voltage-operated Ca2+ channels. Interestingly, extracellular ATP and GTP evoke intracellular Ca2+ mobilization to greater extents in the MLC/mIGF-1 transgenic satellite cells, compared to the wild-type cells. These data suggest that these MLC/mIGF-1 transgenic satellite cells are more sensitive to trophic stimuli, which can potentiate the effects of mIGF-1 on the myogenic programme.


Asunto(s)
Calcio/metabolismo , Diferenciación Celular , Factor I del Crecimiento Similar a la Insulina/metabolismo , Células Satélite del Músculo Esquelético/metabolismo , Células Satélite del Músculo Esquelético/patología , Animales , Fenómenos Electrofisiológicos , Espacio Extracelular/metabolismo , Homeostasis , Hipertrofia/metabolismo , Factor I del Crecimiento Similar a la Insulina/genética , Fluidez de la Membrana , Ratones , Ratones Transgénicos , Fibras Musculares Esqueléticas/patología , Fibras Musculares Esqueléticas/fisiología , Cadenas Ligeras de Miosina/genética , Estrés Oxidativo
11.
Front Aging Neurosci ; 6: 189, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25104935

RESUMEN

The loss in muscle mass coupled with a decrease in specific force and shift in fiber composition are hallmarks of aging. Training and regular exercise attenuate the signs of sarcopenia. However, pathologic conditions limit the ability to perform physical exercise. We addressed whether electrical stimulation (ES) is an alternative intervention to improve muscle recovery and defined the molecular mechanism associated with improvement in muscle structure and function. We analyzed, at functional, structural, and molecular level, the effects of ES training on healthy seniors with normal life style, without routine sport activity. ES was able to improve muscle torque and functional performances of seniors and increased the size of fast muscle fibers. At molecular level, ES induced up-regulation of IGF-1 and modulation of MuRF-1, a muscle-specific atrophy-related gene. ES also induced up-regulation of relevant markers of differentiating satellite cells and of extracellular matrix remodeling, which might guarantee shape and mechanical forces of trained skeletal muscle as well as maintenance of satellite cell function, reducing fibrosis. Our data provide evidence that ES is a safe method to counteract muscle decline associated with aging.

12.
Biomed Res Int ; 2014: 206026, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24967341

RESUMEN

IL-6 is a multifaceted pleiotropic cytokine, which is produced by a variety of cell types and targets different cells and tissues. In physiological conditions, IL-6 can be locally and transiently produced by skeletal muscle and plays an important role in muscle homeostasis. Circulating IL-6 levels are normally very low or undetectable but are dramatically increased in several pathologic conditions. In this study, we aimed to define the potential molecular mechanisms underlying the effects of IL-6 on myogenic program. We explored the molecular mechanisms through which exogenous IL-6, or the conditioned medium from the murine C-26 adenocarcinoma cells (a cellular model that secretes high levels of IL-6 and induces cancer cachexia in mice), interferes with the myogenic program. Our study revealed that IL-6 induces the activation of the Stat3 signaling and promotes the downmodulation of the p90RSK/eEF2 and mTOR/p70S6K axes, while it does not affect the activation of AKT. We thus identified potential molecular mediators of the inhibitory effects of IL-6 on myogenic program.


Asunto(s)
Diferenciación Celular/fisiología , Factor 2 Eucariótico de Iniciación/metabolismo , Interleucina-6/metabolismo , Desarrollo de Músculos/fisiología , Fibras Musculares Esqueléticas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas Quinasas S6 Ribosómicas 70-kDa/metabolismo , Proteínas Quinasas S6 Ribosómicas 90-kDa/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Animales , Línea Celular Tumoral , Ratones , Fibras Musculares Esqueléticas/citología , Factor de Transcripción STAT3/metabolismo , Transducción de Señal/fisiología
13.
PLoS One ; 8(8): e73709, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23977392

RESUMEN

Human aging is associated with a progressive loss of muscle mass and strength and a concomitant fat accumulation in form of inter-muscular adipose tissue, causing skeletal muscle function decline and immobilization. Fat accumulation can also occur as intra-muscular triglycerides (IMTG) deposition in lipid droplets, which are associated with perilipin proteins, such as Perilipin2 (Plin2). It is not known whether Plin2 expression changes with age and if this has consequences on muscle mass and strength. We studied the expression of Plin2 in the vastus lateralis (VL) muscle of both healthy subjects and patients affected by lower limb mobility limitation of different age. We found that Plin2 expression increases with age, this phenomenon being particularly evident in patients. Moreover, Plin2 expression is inversely correlated with quadriceps strength and VL thickness. To investigate the molecular mechanisms underpinning this phenomenon, we focused on IGF-1/p53 network/signalling pathway, involved in muscle physiology. We found that Plin2 expression strongly correlates with increased p53 activation and reduced IGF-1 expression. To confirm these observations made on humans, we studied mice overexpressing muscle-specific IGF-1, which are protected from sarcopenia. These mice resulted almost negative for the expression of Plin2 and p53 at two years of age. We conclude that fat deposition within skeletal muscle in form of Plin2-coated lipid droplets increases with age and is associated with decreased muscle strength and thickness, likely through an IGF-1- and p53-dependent mechanism. The data also suggest that excessive intramuscular fat accumulation could be the initial trigger for p53 activation and consequent loss of muscle mass and strength.


Asunto(s)
Proteínas de la Membrana/metabolismo , Debilidad Muscular/metabolismo , Debilidad Muscular/patología , Músculo Esquelético/metabolismo , Músculo Esquelético/patología , Sarcopenia/metabolismo , Sarcopenia/patología , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Envejecimiento/metabolismo , Envejecimiento/patología , Animales , Humanos , Factor I del Crecimiento Similar a la Insulina/metabolismo , Ratones , Fuerza Muscular , Debilidad Muscular/complicaciones , Debilidad Muscular/fisiopatología , Músculo Esquelético/fisiopatología , Perilipina-2 , Sarcopenia/complicaciones , Sarcopenia/fisiopatología , Transducción de Señal , Proteína p53 Supresora de Tumor/metabolismo , Adulto Joven
14.
Biogerontology ; 14(3): 273-92, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23666344

RESUMEN

Although adult skeletal muscle is composed of fully differentiated fibers, it retains the capacity to regenerate in response to injury and to modify its contractile and metabolic properties in response to changing demands. The major role in the growth, remodeling and regeneration is played by satellite cells, a quiescent population of myogenic precursor cells that reside between the basal lamina and plasmalemma and that are rapidly activated in response to appropriate stimuli. However, in pathologic conditions or during aging, the complete regenerative program can be precluded by fibrotic tissue formation and resulting in functional impairment of the skeletal muscle. Our study, along with other studies, demonstrated that although the regenerative program can also be impaired by the limited proliferative capacity of satellite cells, this limit is not reached during normal aging, and it is more likely that the restricted muscle repair program in aging is presumably due to missing signals that usually render the damaged muscle a permissive environment for regenerative activity.


Asunto(s)
Envejecimiento/fisiología , Proliferación Celular , Músculo Esquelético/fisiología , Regeneración/fisiología , Células Satélite del Músculo Esquelético/fisiología , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Biopsia , Células Cultivadas , Inhibidor p16 de la Quinasa Dependiente de Ciclina , Humanos , Músculo Esquelético/metabolismo , Músculo Esquelético/patología , Proteínas de Neoplasias/metabolismo , Estrés Oxidativo/fisiología , Sarcopenia/metabolismo , Sarcopenia/patología , Sarcopenia/fisiopatología , Células Satélite del Músculo Esquelético/metabolismo , Células Satélite del Músculo Esquelético/patología , Telómero/ultraestructura , Proteína p53 Supresora de Tumor/metabolismo , Adulto Joven
15.
Sci Rep ; 3: 1420, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23478253

RESUMEN

The object of this study was to develop an in vitro bioengineered three-dimensional vascularized skeletal muscle tissue, named eX-vivo Muscle Engineered Tissue (X-MET). This new tissue contains cells that exhibit the characteristics of differentiated myotubes, with organized contractile machinery, undifferentiated cells, and vascular cells capable of forming "vessel-like" networks. X-MET showed biomechanical properties comparable with that of adult skeletal muscles; thus it more closely mimics the cellular complexity typical of in vivo muscle tissue than myogenic cells cultured in standard monolayer conditions. Transplanted X-MET was able to mimic the activity of the excided EDL muscle, restoring the functionality of the damaged muscle. Our results suggest that X-MET is an ideal in vitro 3D muscle model that can be employed to repair muscle defects in vivo and to perform in vitro studies, limiting the use of live animals.


Asunto(s)
Músculo Esquelético/irrigación sanguínea , Músculo Esquelético/trasplante , Ingeniería de Tejidos/métodos , Animales , Técnicas de Cultivo de Célula , Diferenciación Celular , Tratamiento Basado en Trasplante de Células y Tejidos , Ratones , Ratones Endogámicos C57BL , Fibras Musculares Esqueléticas
16.
Cell Metab ; 12(4): 341-351, 2010 Oct 06.
Artículo en Inglés | MEDLINE | ID: mdl-20727829

RESUMEN

In Duchenne muscular dystrophy (DMD) the absence of dystrophin at the sarcolemma delocalizes and downregulates nitric oxide synthase (nNOS); this alters S-nitrosylation of HDAC2 and its chromatin association. We show that the differential HDAC2 nitrosylation state in Duchenne versus wild-type conditions deregulates the expression of a specific subset of microRNA genes. Several circuitries controlled by the identified microRNAs, such as the one linking miR-1 to the G6PD enzyme and the redox state of cell, or miR-29 to extracellular proteins and the fibrotic process, explain some of the DMD pathogenetic traits. We also show that, at variance with other myomiRs, miR-206 escapes from the dystrophin-nNOS control being produced in activated satellite cells before dystrophin expression; in these cells, it contributes to muscle regeneration through repression of the satellite specific factor, Pax7. We conclude that the pathway activated by dystrophin/nNOS controls several important circuitries increasing the robustness of the muscle differentiation program.


Asunto(s)
Distrofina/metabolismo , MicroARNs/fisiología , Distrofia Muscular Animal/enzimología , Óxido Nítrico Sintasa de Tipo I/metabolismo , Animales , Distrofina/fisiología , Regulación de la Expresión Génica , Histona Desacetilasa 2/metabolismo , Ratones , Ratones Endogámicos mdx , MicroARNs/genética , Músculo Esquelético/citología , Músculo Esquelético/fisiología , Regeneración , Células Satélite del Músculo Esquelético/fisiología
17.
Methods Mol Biol ; 633: 101-11, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20204623

RESUMEN

Muscle tissue culture provides a system for studying the growth and differentiation of muscle cells in a controlled environment. In mature muscle tissue, terminally differentiated myocytes form multinucleate syncytia in which structural and regulatory genes are expressed and the contractile apparatus is assembled. Adult muscle fibres are characterized by the presence of satellite cells. These are a quiescent population of myogenic cells that reside between the basal lamina and the plasmalemma of terminally differentiated muscle fibres and are rapidly activated in response to appropriate stimuli. This chapter describes protocols used in our laboratory for isolating and culturing satellite cells isolated from mouse skeletal muscles. In particular we discuss the technical aspect of satellite cell isolation, the methods necessary to enrich the satellite cell fraction, and the culture conditions which optimize proliferation and myotube formation of mouse satellite cells.


Asunto(s)
Técnicas de Cultivo de Célula/métodos , Separación Celular/métodos , Células Satélite del Músculo Esquelético/citología , Animales , Diferenciación Celular , Proliferación Celular , Disección , Miembro Posterior , Ratones , Músculo Esquelético/citología , Músculo Esquelético/metabolismo , Pronasa/metabolismo , Células Satélite del Músculo Esquelético/metabolismo , Tripsina/metabolismo
18.
Muscle Nerve ; 41(6): 828-35, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20151462

RESUMEN

The stromal cell-derived factor (SDF)-1/CXC receptor 4 (CXCR4) axis has been shown to play a role in skeletal muscle development, but its contribution to postnatal myogenesis and the role of the alternate SDF-1 receptor, CXC receptor 7 (CXCR7), are poorly characterized. Western blot analysis and real-time polymerase chain reaction (PCR) were performed to evaluate in vitro the effect of SDF-1 and CXCR4 and CXCR7 inhibition on myogenic differentiation. Proliferating myoblasts express CXCR4, CXCR7, and SDF-1; during myogenic differentiation, CXCR4 and CXCR7 levels are downregulated, and SDF-1 release is decreased. SDF-1 anticipates myosin heavy chain accumulation and myotube formation in both C2C12 myoblasts and satellite cells. Interestingly, inhibition of CXCR4 and CXCR7 signaling, either by drugs or RNA interfererence, blocks myogenic differentiation. Further, the CXCR4 antagonist, 4F-benzoyl-TN14003, inhibits myoblast cell cycle withdrawal and decreases the retinoblastoma gene (pRb) product accumulation in its hypophosphorylated form. Our experiments demonstrate that SDF-1 regulates myogenic differentiation via both CXCR4 and CXCR7 chemokine receptors.


Asunto(s)
Quimiocina CXCL12/genética , Mioblastos/citología , Receptores CXCR4/genética , Receptores CXCR/genética , Animales , Ciclo Celular/genética , Diferenciación Celular/efectos de los fármacos , Quimiocina CXCL12/farmacología , Citometría de Flujo , Ratones , Mioblastos/efectos de los fármacos , Reacción en Cadena de la Polimerasa , ARN Mensajero/genética , ARN Interferente Pequeño/genética , Receptores CXCR4/antagonistas & inhibidores
19.
Endocr Dev ; 14: 29-37, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19293573

RESUMEN

In the last decade, dramatic progress has been made in elucidating the molecular defects underlying a number of muscle diseases. With the characterization of mutations responsible for muscle dysfunction in several inherited pathologies, and the identification of novel signaling pathways, subtle alterations in which can lead to significant defects in muscle metabolism, the field is poised to devise successful strategies for treatment of this debilitating and often fatal group of human ailments. Yet progress has been slow in therapeutic applications of our newly gained knowledge. The complexity of muscle types, the intimate relationship between structural integrity and mechanical function, and the sensitivity of skeletal muscle to metabolic perturbations have impeded rapid progress in successful clinical intervention. The relatively poor regenerative properties of striated muscle compound the devastating effects of muscle degeneration. Perhaps the most difficult hurdle is the sheer volume of tissue that must be treated to effect a significant improvement in quality of life. Recent studies on the role of insulin-like growth factor-1 in skeletal muscle growth and homeostasis have excited new interest in this important mediator of anabolic pathways and suggest promising new avenues for intervention in catabolic disease. In this review, we will discuss the potential therapeutic role of local insulin-like growth factor 1 in the treatment of muscle wasting associated with muscle diseases.


Asunto(s)
Factor I del Crecimiento Similar a la Insulina/metabolismo , Músculo Esquelético/metabolismo , Enfermedades Musculares , Transducción de Señal/fisiología , Humanos , Enfermedades Musculares/genética , Enfermedades Musculares/metabolismo , Enfermedades Musculares/terapia
20.
Mol Reprod Dev ; 73(10): 1239-46, 2006 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-16850445

RESUMEN

Mature spermatozoa of most animal species can spontaneously take up foreign DNA molecules which can be delivered to embryos upon fertilization. Following this procedure, transgenic animals of various species have been generated. We recently discovered a reverse transcriptase (RT) activity in mouse spermatozoa that can reverse-transcribe exogenous RNA molecules into cDNA copies. These cDNA copies are transferred to embryos at fertilization, mosaic propagated as non-integrated structures in tissues of founder individuals and further transmitted to F1 progeny. Reverse-transcribed sequences behave as functional genes, being correctly expressed in tissues of F0 and F1 animals. To learn more about this mechanism and further characterize the reverse transcription step, we have now incubated spermatozoa with a plasmid harboring a green fluorescent protein (EGFP) retrotransposition cassette interrupted by an intron in the opposite orientation to the EGFP gene. We found that reverse-transcribed spliced EGFP DNA sequences are generated in sperm cells and transmitted to embryos in IVF assays. After implantation in foster mothers, embryos developed into mice that expressed EGFP in the blood vessel endothelia of a variety of organs. The EGFP-encoding cDNA sequences were detected in positive tissues as extrachromosomal mosaic-propagated structures, maintained in low-copy number (<1 copy/genome), and mosaic transmitted from founders to the F1 progeny. These results indicate that an efficient machinery is present in mature spermatozoa, which can transcribe, splice, and reverse-transcribe exogenous DNA molecules. This mechanism is implicated in the genesis and non-Mendelian propagation of new genetic information besides that contained in chromosomes.


Asunto(s)
ADN Complementario/metabolismo , Embrión de Mamíferos/metabolismo , ADN Polimerasa Dirigida por ARN/metabolismo , Espermatozoides/enzimología , Animales , Transporte Biológico , ADN Complementario/análisis , Embrión de Mamíferos/química , Feto/química , Feto/metabolismo , Proteínas Fluorescentes Verdes/análisis , Proteínas Fluorescentes Verdes/genética , Intrones , Masculino , Ratones , Plásmidos/genética , Plásmidos/metabolismo , Empalme del ARN , Espermatozoides/metabolismo , Distribución Tisular , Transcripción Genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...