Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 76
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Am J Physiol Endocrinol Metab ; 320(1): E150-E159, 2021 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-33284091

RESUMEN

Sepsis remains a leading cause of mortality in critically ill patients. Muscle wasting is a major complication of sepsis and negatively affects clinical outcomes. Despite intense investigation for many years, the molecular mechanisms underlying sepsis-related muscle wasting are not fully understood. In addition, a potential role of muscle wasting in disease development of sepsis has not been studied. Myostatin is a myokine that downregulates skeletal muscle mass. We studied the effects of myostatin deficiency on muscle wasting and other clinically relevant outcomes, including mortality and bacterial clearance, in mice. Myostatin deficiency prevented muscle atrophy along with inhibition of increases in muscle-specific RING finger protein 1 (MuRF-1) and atrogin-1 expression and phosphorylation of signal transducer and activator of transcription protein 3 (STAT3; major players of muscle wasting) in septic mice. Moreover, myostatin deficiency improved survival and bacterial clearance of septic mice. Sepsis-induced liver dysfunction, acute kidney injury, and neutrophil infiltration into the liver and kidney were consistently mitigated by myostatin deficiency, as indicated by plasma concentrations of aspartate aminotransferase (AST), alanine aminotransferase (ALT), and neutrophil gelatinase-associated lipocalin (NGAL) and myeloperoxidase activity in the organs. Myostatin deficiency also inhibited sepsis-induced increases in plasma high-mobility group protein B1 (HMGB1) and macrophage inhibitory cytokine (MIC)-1/growth differentiation factor (GDF)-15 concentrations. These results indicate that myostatin plays an important role not only in muscle wasting but also in other clinically relevant outcomes in septic mice. Furthermore, our data raise the possibility that muscle wasting may not be simply a complication, but myostatin-mediated muscle cachexia and related changes in muscle may actually drive the development of sepsis as well.NEW & NOTEWORTHY Muscle wasting is a major complication of sepsis, but its role in the disease development is not known. Myostatin deficiency improved bacterial clearance and survival and mitigated damage in the liver and kidney in septic mice, which paralleled prevention of muscle wasting. These results raise the possibility that muscle wasting may not simply be a complication of sepsis, but myostatin-mediated cachexic changes may have a role in impaired bacterial clearance and mortality in septic mice.


Asunto(s)
Atrofia Muscular/genética , Miostatina/deficiencia , Miostatina/genética , Sepsis/genética , Lesión Renal Aguda/genética , Animales , Caquexia/genética , Caquexia/prevención & control , Lipocalina 2/sangre , Hepatopatías/etiología , Hepatopatías/genética , Pruebas de Función Hepática , Masculino , Ratones , Proteínas Musculares/biosíntesis , Proteínas Musculares/genética , Atrofia Muscular/prevención & control , Infiltración Neutrófila/genética , Fosforilación , Factor de Transcripción STAT3/biosíntesis , Factor de Transcripción STAT3/genética , Sepsis/microbiología , Sepsis/mortalidad , Análisis de Supervivencia , Proteínas de Motivos Tripartitos/biosíntesis , Proteínas de Motivos Tripartitos/genética , Ubiquitina-Proteína Ligasas/biosíntesis , Ubiquitina-Proteína Ligasas/genética
2.
Transgenic Res ; 29(2): 253-261, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-32078127

RESUMEN

The size of skeletal muscle mass plays a significant role in glucose uptake in healthy and diabetic human subjects. Previously, we have generated myostatin-deficient (MSTN-/-) transgenic pigs via animal cloning technology. MSTN-/- pigs had dramatic phenotype with individual muscle mass increase by 100% over their wild-type controls, which provides a unique large animal model to investigate how enhanced skeletal muscles are beneficial to glucose update in diabetes. We employed intravenous administration of stretozotocin (STZ) to male MSTN-/- and wild-type pigs (100 mg/kg body weight). One month later, blood glucose and insulin concentrations and pancreas histology were examined, STZ-induced diabetes occurred in both MSTN transgenic and wild-type pigs. Histology of pancreas, analysis of pAKT and Glut4 transporter proteins by Western blotting, and real-time qPCR for MSTN gene expression were used in the study. The STZ-treated pigs had increased levels of fasting plasma glucose and insulin levels in comparison with animals receiving sodium citrate buffer, their pancreas also had reduced beta cells and slight increases in lymphocyte. There are significant lower concentrations of fasting plasma glucose and insulin in MSTN-/- pigs than that of wild-type pigs after STZ administration. Detections of pAKT and Glut4 transporter proteins by Western blotting in muscle tissue indicates significant elevations of both proteins in MSTN-/- pigs compared with the wild-type pigs. The results from this pig model suggest that enhanced skeletal muscle by manipulation of myostatin function can improve glucose uptake even in the status of diabetes.


Asunto(s)
Glucemia/análisis , Diabetes Mellitus Experimental/prevención & control , Regulación de la Expresión Génica , Insulina/metabolismo , Desarrollo de Músculos , Músculo Esquelético/citología , Miostatina/deficiencia , Animales , Animales Modificados Genéticamente , Diabetes Mellitus Experimental/etiología , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Experimental/patología , Femenino , Masculino , Miostatina/genética , Fenotipo , Porcinos
3.
Mol Biol Rep ; 47(12): 9531-9540, 2020 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-33225386

RESUMEN

The myostatin (MSTN) gene is of interest in the livestock industry because mutations in this gene are closely related to growth performance and muscle differentiation. Thus, in this study, we established MSTN knockout (KO) quail myoblasts (QM7) and investigated the regulatory pathway of the myogenic differentiation process. We used clustered regularly interspaced short palindromic repeats (CRISPR)/Cas9 to generate MSTN KO QM7 cells and subsequently isolated a single cell-derived MSTN KO QM7 subline with 10- and 16-nucleotide deletions that induced translational frameshift mutations. The differentiation capacity and proliferation rate of MSTN KO QM7 cells were enhanced. We conducted next-generation-sequencing (NGS) analysis to compare the global gene expression profiles of wild-type (WT) QM7 and MSTN KO QM7 cells. Intriguingly, NGS expression profiles showed different expression patterns of p21 and p53 in MSTN KO QM7 cells. Moreover, we identified downregulated expression patterns of leukemia inhibitory factor and DNA Damage Inducible Transcript 4, which are genes in the p53 signaling pathway. Using quantitative RT-PCR (qRT-PCR) analysis and western blotting, we concluded that p53-related genes promote the cell cycle by upregulating p21 and enhancing muscle differentiation in MSTN KO QM7 cells. These results could be applied to improve economic traits in commercial poultry by regulating MSTN-related networks.


Asunto(s)
Desarrollo de Músculos/genética , Mioblastos/metabolismo , Miostatina/genética , Codorniz/genética , Proteína p53 Supresora de Tumor/genética , Animales , Sistemas CRISPR-Cas , Diferenciación Celular , Línea Celular , Proliferación Celular , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/genética , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Mutación del Sistema de Lectura , Edición Génica , Regulación de la Expresión Génica , Secuenciación de Nucleótidos de Alto Rendimiento , Factor Inhibidor de Leucemia/genética , Factor Inhibidor de Leucemia/metabolismo , Músculo Esquelético/citología , Músculo Esquelético/metabolismo , Mioblastos/citología , Miostatina/deficiencia , Codorniz/metabolismo , Transducción de Señal , Proteína p53 Supresora de Tumor/metabolismo
4.
J Anat ; 235(4): 825-835, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31198988

RESUMEN

Obesity is a worldwide nutritional disorder affecting body performance, including skeletal muscle. Inhibition of myostatin not only increases the muscle mass but also it reduces body fat accumulation. We examined the effect of high-fat diet on the phenotypic properties of forelimb muscles from myostatin null mice. Male wild-type and myostatin null mice were fed on either a normal diet or a high-fat diet (45% fat) for 10 weeks. Musculus triceps brachii Caput longum; M. triceps brachii Caput laterale; M. triceps brachii Caput mediale; M. extensor carpi ulnaris and M. flexor carpi ulnaris were processed for fiber type composition using immunohistochemistry and morphometric analysis. Although the muscle mass revealed no change under a high-fat diet, there were morphometric alterations in the absence of myostatin. We show that high-fat diet reduces the cross-sectional area of the fast (IIB and IIX) fibers in M. triceps brachii Caput longum and M. triceps brachii Caput laterale of both genotypes. In contrast, increases of fast fiber areas were observed in both M. extensor carpi ulnaris of wild-type and M. flexor carpi ulnaris of myostatin null mice. Meanwhile, a high-fat diet increased the area of the fast IIA fibers in wild-type mice; myostatin null mice display a muscle-dependent alteration in the area of the same fiber type. The combined high-fat diet and myostatin deletion shows no effect on the area of slow type I fibers. Although a high-fat diet causes a reduction in the area of the peripheral IIB fibers in both genotypes, only myostatin null mice show an increase in the area of the central IIB fibers. We provide evidence that a high-fat diet induces a muscle-dependent fast to slow myofiber shift in the absence of myostatin. The data suggest that the morphological alterations of muscle fibers under a combined high-fat diet and myostatin deletion reflect a functional adaptation of the muscle to utilize the high energy intake.


Asunto(s)
Dieta Alta en Grasa/efectos adversos , Fibras Musculares Esqueléticas/patología , Miostatina/deficiencia , Animales , Miembro Anterior , Hipertrofia/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Músculo Esquelético/patología
5.
Biosci Biotechnol Biochem ; 83(11): 2090-2096, 2019 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-31334687

RESUMEN

Myostatin (Mstn) is an important growth/differentiation factor, and knockdown of Mstn reduces fat content. Here, we knocked down Mstn expression in C2C12 myoblasts and then induced adipogenic trans-differentiation in the cells. The effects of Mstn knockdown on lipid droplet contents and H3K27me3 marker expression on adipocyte-specific genes were detected. The results showed that Mstn knockdown reduced the formation of lipid droplets, downregulated the expression of adipocyte-specific genes, and increased H3K27me3 marker expression on adipocyte-specific genes. Chromatin immunoprecipitation analysis showed that the SMAD2/SMAD3 complex could combine with the Jumonji D3 (Jmjd3) promoter and that Mstn regulated Jmjd3 expression through this process. Jmjd3 overexpression removed the H3K27me3 marker and increased the expression of adipocyte-specific genes. Overall, our results showed that Mstn regulated Jmjd3 expression through SMAD2/SMAD3, thus affecting the H3K27me3 marker on adipocyte-specific genes and the trans-differentiation from myocytes to adipocytes.


Asunto(s)
Adipocitos/citología , Transdiferenciación Celular/genética , Técnicas de Silenciamiento del Gen , Histona Demetilasas con Dominio de Jumonji/metabolismo , Células Musculares/citología , Miostatina/genética , Proteínas Smad Reguladas por Receptores/metabolismo , Adipocitos/metabolismo , Animales , Línea Celular , Regulación hacia Abajo/genética , Histonas/química , Histonas/metabolismo , Gotas Lipídicas/metabolismo , Lisina/metabolismo , Metilación , Ratones , Células Musculares/metabolismo , Miostatina/deficiencia , Proteína Smad2/metabolismo , Proteína smad3/metabolismo
6.
Proc Natl Acad Sci U S A ; 113(47): 13522-13527, 2016 11 22.
Artículo en Inglés | MEDLINE | ID: mdl-27821779

RESUMEN

During fetal development, the uterine environment can have effects on offspring bone architecture and integrity that persist into adulthood; however, the biochemical and molecular mechanisms remain unknown. Myostatin is a negative regulator of muscle mass. Parental myostatin deficiency (Mstntm1Sjl/+) increases muscle mass in wild-type offspring, suggesting an intrauterine programming effect. Here, we hypothesized that Mstntm1Sjl/+ dams would also confer increased bone strength. In wild-type offspring, maternal myostatin deficiency altered fetal growth and calvarial collagen content of newborn mice and conferred a lasting impact on bone geometry and biomechanical integrity of offspring at 4 mo of age, the age of peak bone mass. Second, we sought to apply maternal myostatin deficiency to a mouse model with osteogenesis imperfecta (Col1a2oim), a heritable connective tissue disorder caused by abnormalities in the structure and/or synthesis of type I collagen. Femora of male Col1a2oim/+ offspring from natural mating of Mstntm1Sjl/+ dams to Col1a2oim/+sires had a 15% increase in torsional ultimate strength, a 29% increase in tensile strength, and a 24% increase in energy to failure compared with age, sex, and genotype-matched offspring from natural mating of Col1a2oim/+ dams to Col1a2oim/+ sires. Finally, increased bone biomechanical strength of Col1a2oim/+ offspring that had been transferred into Mstntm1Sjl/+ dams as blastocysts demonstrated that the effects of maternal myostatin deficiency were conferred by the postimplantation environment. Thus, targeting the gestational environment, and specifically prenatal myostatin pathways, provides a potential therapeutic window and an approach for treating osteogenesis imperfecta.


Asunto(s)
Fémur/fisiopatología , Miostatina/metabolismo , Osteogénesis Imperfecta/fisiopatología , Animales , Biomarcadores/sangre , Fenómenos Biomecánicos , Peso Corporal , Colágeno/metabolismo , Modelos Animales de Enfermedad , Implantación del Embrión , Femenino , Fémur/patología , Masculino , Ratones Endogámicos C57BL , Contracción Muscular , Miostatina/deficiencia , Osteoblastos/metabolismo , Osteogénesis Imperfecta/sangre , Osteogénesis Imperfecta/embriología , Tibia/patología , Tibia/fisiopatología
7.
Int J Mol Sci ; 20(3)2019 Feb 02.
Artículo en Inglés | MEDLINE | ID: mdl-30717351

RESUMEN

Myostatin (MSTN) is a member of the TGF-ß superfamily that negatively regulates skeletal muscle growth and differentiation. However, the mechanism by which complete MSTN deletion limits excessive proliferation of muscle cells remains unclear. In this study, we knocked out MSTN in mouse myoblast lines using a Clustered Regularly Interspaced Short Palindromic Repeats (CRISPR/Cas9) system and sequenced the mRNA and miRNA transcriptomes. The results show that complete loss of MSTN upregulates seven miRNAs targeting an interaction network composed of 28 downregulated genes, including TGFB1, FOS and RB1. These genes are closely associated with tumorigenesis and cell proliferation. Our study suggests that complete loss of MSTN may limit excessive cell proliferation via activation of miRNAs. These data will contribute to the treatment of rhabdomyosarcoma (RMS).


Asunto(s)
MicroARNs/genética , Células Musculares/metabolismo , Miostatina/deficiencia , Animales , Línea Celular , Biología Computacional/métodos , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Técnicas de Inactivación de Genes , Orden Génico , Redes Reguladoras de Genes , Vectores Genéticos/genética , Ratones , Anotación de Secuencia Molecular , Interferencia de ARN , ARN Mensajero/genética , Reproducibilidad de los Resultados
8.
Circ Res ; 118(7): 1125-41; discussion 1142, 2016 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-27034275

RESUMEN

Growth differentiation factor 11 (GDF11) and myostatin (or GDF8) are closely related members of the transforming growth factor ß superfamily and are often perceived to serve similar or overlapping roles. Yet, despite commonalities in protein sequence, receptor utilization and signaling, accumulating evidence suggests that these 2 ligands can have distinct functions in many situations. GDF11 is essential for mammalian development and has been suggested to regulate aging of multiple tissues, whereas myostatin is a well-described negative regulator of postnatal skeletal and cardiac muscle mass and modulates metabolic processes. In this review, we discuss the biochemical regulation of GDF11 and myostatin and their functions in the heart, skeletal muscle, and brain. We also highlight recent clinical findings with respect to a potential role for GDF11 and/or myostatin in humans with heart disease. Finally, we address key outstanding questions related to GDF11 and myostatin dynamics and signaling during development, growth, and aging.


Asunto(s)
Proteínas Morfogenéticas Óseas/fisiología , Factores de Diferenciación de Crecimiento/fisiología , Miostatina/fisiología , Adulto , Envejecimiento/fisiología , Secuencia de Aminoácidos , Animales , Proteínas Morfogenéticas Óseas/química , Proteínas Morfogenéticas Óseas/deficiencia , Encéfalo/crecimiento & desarrollo , Encéfalo/fisiología , Dimerización , Femenino , Folistatina/metabolismo , Proteínas Relacionadas con la Folistatina/metabolismo , Factores de Diferenciación de Crecimiento/química , Factores de Diferenciación de Crecimiento/deficiencia , Factores de Diferenciación de Crecimiento/uso terapéutico , Corazón/fisiología , Cardiopatías/metabolismo , Humanos , Masculino , Ratones , Modelos Moleculares , Datos de Secuencia Molecular , Músculos/fisiología , Miocardio/metabolismo , Miostatina/química , Miostatina/deficiencia , Especificidad de Órganos , Conformación Proteica , Estructura Terciaria de Proteína , Ratas , Alineación de Secuencia , Homología de Secuencia de Aminoácido , Transducción de Señal , Relación Estructura-Actividad
9.
Heart Lung Circ ; 27(6): 693-701, 2018 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-28690022

RESUMEN

BACKGROUND: Myostatin inhibits the development of skeletal muscle and regulates the proliferation of skeletal muscle fibroblasts. However, the role of myostatin in regulating cardiac muscle or myofibroblasts, specifically in acute myocardial infarction (MI), is less clear. This study sought to determine whether absence of myostatin altered left ventricular function post-MI. METHODS: Myostatin-null mice (Mstn-/-) and wild-type (WT) mice underwent ligation of the left anterior descending artery to induce MI. Left ventricular function was measured at baseline, days 1 and 28 post-MI. Immunohistochemistry and immunofluorescence were obtained at day 28 for cellular proliferation, collagen deposition, and myofibroblastic activity. RESULTS: Whilst left ventricular function at baseline and size of infarct were similar, significant differences in favour of Mstn-/- compared to WT mice post-MI include a greater recovery of ejection fraction (61.8±1.1% vs 57.1±2.3%, p<0.01), less collagen deposition (41.9±2.8% vs 54.7±3.4%, p<0.05), and lower mortality (0 vs. 20%, p<0.05). There was no difference in the number of BrdU positive cells, percentage of apoptotic cardiomyocytes, or size of cardiomyocytes post-MI between WT and Mstn-/- mice. CONCLUSIONS: Absence of myostatin potentially protects the function of the heart post-MI with improved survival, possibly by limiting extent of fibrosis.


Asunto(s)
Ventrículos Cardíacos/fisiopatología , Infarto del Miocardio/fisiopatología , Miocardio/patología , Miocitos Cardíacos/metabolismo , Miostatina/deficiencia , Función Ventricular Izquierda/fisiología , Remodelación Ventricular , Animales , Apoptosis , Vasos Coronarios/metabolismo , Vasos Coronarios/patología , Vasos Coronarios/fisiopatología , Modelos Animales de Enfermedad , Ecocardiografía , Fibroblastos/metabolismo , Fibroblastos/patología , Ventrículos Cardíacos/metabolismo , Ventrículos Cardíacos/patología , Inmunohistoquímica , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Infarto del Miocardio/metabolismo , Infarto del Miocardio/patología , Miocardio/metabolismo , Miocitos Cardíacos/patología , Miostatina/metabolismo
10.
Biochim Biophys Acta Mol Cell Biol Lipids ; 1862(10 Pt A): 1044-1055, 2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-28676454

RESUMEN

Myostatin (Mstn) deficiency leads to skeletal muscle overgrowth and Mstn inhibition is considered as a promising treatment for muscle-wasting disorders. Mstn gene deletion in mice also causes metabolic changes with decreased mitochondria content, disturbance in mitochondrial respiratory function and increased muscle fatigability. However the impact of MSTN deficiency on these metabolic changes is not fully elucidated. Here, we hypothesized that lack of MSTN will alter skeletal muscle membrane lipid composition in relation with pronounced alterations in muscle function and metabolism. Indeed, phospholipids and in particular cardiolipin mostly present in the inner mitochondrial membrane, play a crucial role in mitochondria function and oxidative phosphorylation process. We observed that Mstn KO muscle had reduced fat membrane transporter levels (FAT/CD36, FABP3, FATP1 and FATP4) associated with decreased lipid oxidative pathway (citrate synthase and ß-HAD activities) and impaired lipogenesis (decreased triglyceride and free fatty acid content), indicating a role of mstn in muscle lipid metabolism. We further analyzed phospholipid classes and fatty acid composition by chromatographic methods in muscle and mitochondrial membranes. Mstn KO mice showed increased levels of saturated and polyunsaturated fatty acids at the expense of monounsaturated fatty acids. We also demonstrated, in this phenotype, a reduction in cardiolipin proportion in mitochondrial membrane versus the proportion of others phospholipids, in relation with a decrease in the expression of phosphatidylglycerolphosphate synthase and cardiolipin synthase, enzymes involved in cardiolipin synthesis. These data illustrate the importance of lipids as a link by which MSTN deficiency can impact mitochondrial bioenergetics in skeletal muscle.


Asunto(s)
Ácidos Grasos/metabolismo , Mitocondrias Musculares/metabolismo , Músculo Esquelético/metabolismo , Miostatina/deficiencia , 3-Hidroxiacil-CoA Deshidrogenasas/genética , 3-Hidroxiacil-CoA Deshidrogenasas/metabolismo , Animales , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Citrato (si)-Sintasa/genética , Citrato (si)-Sintasa/metabolismo , Ácidos Grasos/genética , Masculino , Ratones , Ratones Noqueados , Mitocondrias Musculares/genética , Mitocondrias Musculares/patología , Músculo Esquelético/patología , Oxidación-Reducción
11.
Muscle Nerve ; 55(2): 254-261, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-27312354

RESUMEN

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


Asunto(s)
Contracción Muscular/genética , Fuerza Muscular/genética , Músculo Esquelético/fisiología , Enfermedades Musculares/patología , Miostatina/deficiencia , Animales , Animales Recién Nacidos , Modelos Animales de Enfermedad , Femenino , Masculino , Ratones , Ratones Noqueados , Enfermedades Musculares/genética , Miostatina/genética , Factores Sexuales
12.
Muscle Nerve ; 56(6): E100-E107, 2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-28073155

RESUMEN

INTRODUCTION: Deletion of myostatin in mice (MSTN-/- ) alters structural properties of peripheral axons. However, properties like axon diameter and myelin thickness were analyzed in mixed nerves, so it is unclear whether loss of myostatin affects motor, sensory, or both types of axons. METHODS: Using the MSTN-/- mouse model, we analyzed the effects of increasing the number of muscle fibers on axon diameter, myelin thickness, and internode length in motor and sensory axons. RESULTS: Axon diameter and myelin thickness were increased in motor axons of MSTN-/- mice without affecting internode length or axon number. The number of sensory axons was increased without affecting their structural properties. DISCUSSION: These results suggest that motor and sensory axons establish structural properties by independent mechanisms. Moreover, in motor axons, instructive cues from the neuromuscular junction may play a role in co-regulating axon diameter and myelin thickness, whereas internode length is established independently. Muscle Nerve 56: E100-E107, 2017.


Asunto(s)
Axones/metabolismo , Neuronas Motoras/metabolismo , Miostatina/deficiencia , Conducción Nerviosa/fisiología , Células Receptoras Sensoriales/metabolismo , Animales , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neuronas Motoras/citología , Células Receptoras Sensoriales/citología
13.
Proteomics ; 16(14): 2019-27, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-27214824

RESUMEN

Pharmacologic blockade of the myostatin (Mstn)/activin receptor pathway is being pursued as a potential therapy for several muscle wasting disorders. The functional benefits of blocking this pathway are under investigation, in particular given the findings that greater muscle hypertrophy results from Mstn deficiency arising from genetic ablation compared to post-developmental Mstn blockade. Using high-resolution MS coupled with SILAC mouse technology, we quantitated the relative proteomic changes in gastrocnemius muscle from Mstn knockout (Mstn(-/-) ) and mice treated for 2-weeks with REGN1033, an anti-Mstn antibody. Relative to wild-type animals, Mstn(-/-) mice had a two-fold greater muscle mass and a >1.5-fold change in expression of 12.0% of 1137 quantified muscle proteins. In contrast, mice treated with REGN1033 had minimal changes in muscle proteome (0.7% of 1510 proteins >1.5-fold change, similar to biological difference 0.5% of 1310) even though the treatment induced significant 20% muscle mass increase. Functional annotation of the altered proteins in Mstn(-/-) mice corroborates the mutiple physiological changes including slow-to-fast fiber type switch. Thus, the proteome-wide protein expression differs between Mstn(-/-) mice and mice subjected to specific Mstn blockade post-developmentally, providing molecular-level insights to inform mechanistic hypotheses to explain the observed functional differences.


Asunto(s)
Hipertrofia/genética , Músculo Esquelético/metabolismo , Enfermedades Musculares/genética , Miostatina/genética , Proteoma/genética , Animales , Anticuerpos Monoclonales/farmacología , Modelos Animales de Enfermedad , Femenino , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Ontología de Genes , Humanos , Hipertrofia/metabolismo , Hipertrofia/patología , Marcaje Isotópico , Masculino , Ratones , Ratones Noqueados , Anotación de Secuencia Molecular , Fibras Musculares de Contracción Rápida/efectos de los fármacos , Fibras Musculares de Contracción Rápida/metabolismo , Fibras Musculares de Contracción Rápida/patología , Fibras Musculares de Contracción Lenta/efectos de los fármacos , Fibras Musculares de Contracción Lenta/metabolismo , Fibras Musculares de Contracción Lenta/patología , Músculo Esquelético/efectos de los fármacos , Músculo Esquelético/patología , Enfermedades Musculares/metabolismo , Enfermedades Musculares/patología , Miostatina/antagonistas & inhibidores , Miostatina/deficiencia , Tamaño de los Órganos , Proteoma/metabolismo
14.
Osteoporos Int ; 27(1): 161-70, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26179666

RESUMEN

UNLABELLED: Mice with osteogenesis imperfecta (+/oim), a disorder of bone fragility, were bred to mice with muscle over growth to test whether increasing muscle mass genetically would improve bone quality and strength. The results demonstrate that femora from mice carrying both mutations have greater mechanical integrity than their +/oim littermates. INTRODUCTION: Osteogenesis imperfecta is a heritable connective tissue disorder due primarily to mutations in the type I collagen genes resulting in skeletal deformity and fragility. Currently, there is no cure, and therapeutic strategies encompass the use of antiresorptive pharmaceuticals and surgical bracing, with limited success and significant potential for adverse effects. Bone, a mechanosensing organ, can respond to high mechanical loads by increasing new bone formation and altering bone geometry to withstand increased forces. Skeletal muscle is a major source of physiological loading on bone, and bone strength is proportional to muscle mass. METHODS: To test the hypothesis that congenic increases in muscle mass in the osteogenesis imperfecta murine model mouse (oim) will improve their compromised bone quality and strength, heterozygous (+/oim) mice were bred to mice deficient in myostatin (+/mstn), a negative regulator of muscle growth. The resulting adult offspring were evaluated for hindlimb muscle mass, and bone microarchitecture, physiochemistry, and biomechanical integrity. RESULTS: +/oim mice deficient in myostatin (+/mstn +/oim) were generated and demonstrated that myostatin deficiency increased body weight, muscle mass, and biomechanical strength in +/mstn +/oim mice as compared to +/oim mice. Additionally, myostatin deficiency altered the physiochemical properties of the +/oim bone but did not alter bone remodeling. CONCLUSIONS: Myostatin deficiency partially improved the reduced femoral bone biomechanical strength of adult +/oim mice by increasing muscle mass with concomitant improvements in bone microarchitecture and physiochemical properties.


Asunto(s)
Fémur/fisiopatología , Terapia Genética/métodos , Músculo Esquelético/patología , Miostatina/deficiencia , Osteogénesis Imperfecta/terapia , Animales , Biomarcadores/sangre , Fenómenos Biomecánicos , Peso Corporal/fisiología , Densidad Ósea/fisiología , Remodelación Ósea/fisiología , Colágeno/análisis , Modelos Animales de Enfermedad , Femenino , Fémur/química , Fémur/patología , Masculino , Ratones Endogámicos C57BL , Ratones Mutantes , Mutación , Miostatina/genética , Miostatina/fisiología , Tamaño de los Órganos/fisiología , Osteogénesis Imperfecta/genética , Osteogénesis Imperfecta/patología , Osteogénesis Imperfecta/fisiopatología , Fenotipo , Tibia/patología , Soporte de Peso/fisiología
15.
Circ Res ; 115(2): 296-310, 2014 Jul 07.
Artículo en Inglés | MEDLINE | ID: mdl-24807786

RESUMEN

RATIONALE: Myostatin is a major negative regulator of skeletal muscle mass and initiates multiple metabolic changes, including enhanced insulin sensitivity. However, the function of myostatin in the heart is barely understood, although it is upregulated in the myocardium under several pathological conditions. OBJECTIVE: Here, we aimed to decipher the role of myostatin and myostatin-dependent signaling pathways for cardiac function and cardiac metabolism in adult mice. To avoid potential counterregulatory mechanisms occurring in constitutive and germ-line-based myostatin mutants, we generated a mouse model that allows myostatin inactivation in adult cardiomyocytes. METHODS AND RESULTS: Cardiac MRI revealed that genetic inactivation of myostatin signaling in the adult murine heart caused cardiac hypertrophy and heart failure, partially recapitulating effects of the age-dependent decline of the myostatin paralog growth and differentiation factor 11. We found that myostatin represses AMP-activated kinase activation in the heart via transforming growth factor-ß-activated kinase 1, thereby preventing a metabolic switch toward glycolysis and glycogen accumulation. Furthermore, myostatin stimulated expression of regulator of G-protein signaling 2, a GTPase-activating protein that restricts Gaq and Gas signaling and thereby protects against cardiac failure. Inhibition of AMP-activated kinase in vivo rescued cardiac hypertrophy and prevented enhanced glycolytic flow and glycogen accumulation after inactivation of myostatin in cardiomyocytes. CONCLUSIONS: Our results uncover an important role of myostatin in the heart for maintaining cardiac energy homeostasis and preventing cardiac hypertrophy.


Asunto(s)
Cardiomiopatía Hipertrófica Familiar/genética , Metabolismo Energético/fisiología , Insuficiencia Cardíaca/prevención & control , Miocardio/metabolismo , Miostatina/fisiología , Proteínas Quinasas Activadas por AMP/metabolismo , Animales , Cardiomiopatía Hipertrófica Familiar/complicaciones , Linaje de la Célula , Regulación de la Expresión Génica/fisiología , Glucógeno/metabolismo , Glucólisis/fisiología , Insuficiencia Cardíaca/etiología , Homeostasis/fisiología , Quinasas Quinasa Quinasa PAM/fisiología , Ratones , Ratones Endogámicos C57BL , Miocitos Cardíacos/metabolismo , Miostatina/deficiencia , Proteínas RGS/fisiología , Proteínas Recombinantes de Fusión , Transducción de Señal/fisiología
16.
FASEB J ; 28(4): 1711-23, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24378873

RESUMEN

In models of cancer cachexia, inhibiting type IIB activin receptors (ActRIIBs) reverse muscle wasting and prolongs survival, even with continued tumor growth. ActRIIB mediates signaling of numerous TGF-ß proteins; of these, we demonstrate that activins are the most potent negative regulators of muscle mass. To determine whether activin signaling in the absence of tumor-derived factors induces cachexia, we used recombinant serotype 6 adeno-associated virus (rAAV6) vectors to increase circulating activin A levels in C57BL/6 mice. While mice injected with control vector gained ~10% of their starting body mass (3.8±0.4 g) over 10 wk, mice injected with increasing doses of rAAV6:activin A exhibited weight loss in a dose-dependent manner, to a maximum of -12.4% (-4.2±1.1 g). These reductions in body mass in rAAV6:activin-injected mice correlated inversely with elevated serum activin A levels (7- to 24-fold). Mechanistically, we show that activin A reduces muscle mass and function by stimulating the ActRIIB pathway, leading to deleterious consequences, including increased transcription of atrophy-related ubiquitin ligases, decreased Akt/mTOR-mediated protein synthesis, and a profibrotic response. Critically, we demonstrate that the muscle wasting and fibrosis that ensues in response to excessive activin levels is fully reversible. These findings highlight the therapeutic potential of targeting activins in cachexia.


Asunto(s)
Activinas/genética , Caquexia/genética , Expresión Génica , Atrofia Muscular/genética , Receptores de Activinas Tipo II/genética , Receptores de Activinas Tipo II/metabolismo , Activinas/sangre , Activinas/metabolismo , Animales , Western Blotting , Caquexia/metabolismo , Línea Celular Tumoral , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/genética , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Dependovirus/genética , Vectores Genéticos/genética , Humanos , Células MCF-7 , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Musculares/genética , Proteínas Musculares/metabolismo , Músculo Esquelético/metabolismo , Músculo Esquelético/patología , Atrofia Muscular/metabolismo , Miostatina/deficiencia , Miostatina/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Proteínas Ligasas SKP Cullina F-box/genética , Proteínas Ligasas SKP Cullina F-box/metabolismo , Transducción de Señal/genética
17.
J Cell Biochem ; 115(11): 1908-17, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24909401

RESUMEN

Conversion of skin fibroblasts into myoblasts by transducing the cells with myogenic master regulator MyoD has been in practice for more than two decades. The purpose of such conversion is due to scarcity of muscle biopsies during muscle wasting, hence conversion of fibroblasts to myogenic lineage from various genetic backgrounds offers a great alternative for cell therapies. Here, we have investigated if eliminating Myostatin, a potent negative regulator of myogenesis, could improve the myogenic conversion of fibroblasts. In the present study, we have isolated primary muscle fibroblasts from the skeletal muscles of wild-type (WT) and myostatin null (Mstn(-/-)) mice and transduced the muscle fibroblasts with MyoD using adenoviral, lentiviral transduction, and electroporation methods. In contrast to what we predicted, it is only in WT muscle fibroblasts we detected significant ectopic expression of MyoD, and myogenic conversion. Muscle fibroblasts from Mstn(-/-) genotype failed to take up as much MyoD using the three methods and, therefore, failed to form myotubes. The aforesaid condition of greater MyoD uptake by WT muscle fibroblasts was attributed to the presence of adenoviral receptors, which facilitated adenoviral transduction. However, in Mstn(-/-) fibroblasts we detected negligible levels of adenovirus receptors. Moreover, we also detected significantly higher levels of MyoD antagonists, c-Fos, c-Jun, and cyclin D1 in Mstn(-/-) muscle fibroblasts. Taken together, our results demonstrate that lack of myostatin reduces myogenic potential of muscle fibroblasts by inhibiting MyoD function.


Asunto(s)
Fibroblastos/citología , Desarrollo de Músculos , Músculo Esquelético/citología , Proteína MioD/genética , Miostatina/deficiencia , Animales , Diferenciación Celular , Células Cultivadas , Ciclina D1/metabolismo , Terapia Genética , Ratones , Ratones Transgénicos , Proteína MioD/metabolismo , Proteínas Proto-Oncogénicas c-fos/metabolismo , Proteínas Proto-Oncogénicas c-jun/metabolismo , Transducción Genética
18.
Am J Physiol Regul Integr Comp Physiol ; 307(4): R444-54, 2014 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-24965795

RESUMEN

Myostatin (Mstn) participates in the regulation of skeletal muscle size and has emerged as a regulator of muscle metabolism. Here, we hypothesized that lack of myostatin profoundly depresses oxidative phosphorylation-dependent muscle function. Toward this end, we explored Mstn(-/-) mice as a model for the constitutive absence of myostatin and AAV-mediated overexpression of myostatin propeptide as a model of myostatin blockade in adult wild-type mice. We show that muscles from Mstn(-/-) mice, although larger and stronger, fatigue extremely rapidly. Myostatin deficiency shifts muscle from aerobic toward anaerobic energy metabolism, as evidenced by decreased mitochondrial respiration, reduced expression of PPAR transcriptional regulators, increased enolase activity, and exercise-induced lactic acidosis. As a consequence, constitutively reduced myostatin signaling diminishes exercise capacity, while the hypermuscular state of Mstn(-/-) mice increases oxygen consumption and the energy cost of running. We wondered whether these results are the mere consequence of the congenital fiber-type switch toward a glycolytic phenotype of constitutive Mstn(-/-) mice. Hence, we overexpressed myostatin propeptide in adult mice, which did not affect fiber-type distribution, while nonetheless causing increased muscle fatigability, diminished exercise capacity, and decreased Pparb/d and Pgc1a expression. In conclusion, our results suggest that myostatin endows skeletal muscle with high oxidative capacity and low fatigability, thus regulating the delicate balance between muscle mass, muscle force, energy metabolism, and endurance capacity.


Asunto(s)
Metabolismo Energético , Contracción Muscular , Músculo Esquelético/metabolismo , Miostatina/metabolismo , Resistencia Física , Animales , Genotipo , Glucólisis , Ácido Láctico/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mitocondrias Musculares/metabolismo , Fatiga Muscular , Miostatina/deficiencia , Miostatina/genética , Consumo de Oxígeno , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma , Receptores Activados del Proliferador del Peroxisoma/genética , Receptores Activados del Proliferador del Peroxisoma/metabolismo , Fenotipo , Fosfopiruvato Hidratasa/metabolismo , Carrera , Transducción de Señal , Factores de Tiempo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
19.
Yi Chuan ; 36(7): 685-90, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25076033

RESUMEN

Myostatin (MSTN) can negatively regulate the growth and development of skeletal muscle, and mutations of bovine MSTN gene can cause a "double-muscle" feature. To knock out MSTN gene in bovine fetal fibroblast by transcription activator-like effector nucleases (TALENs) and obtain MSTN knockout cell lines, we constructed one pair of MSTN-TALEN vector and transfected into bovine fetal fibroblast cells by PEI and electroporation. Sequencing results demonstrated that TALEN was available for MSTN knockout. T7 endonuclease 1 (T7E1) was used for the detection of mutation efficiency. The results indicated that knockout efficiency of electroporation transfection was 20.4%, and 10 MSTN(+/-) and MSTN(-/-) cell colonies were obtained via limiting dilution method. The deletion number of nucleotides ranged from 1 to 20, and some of them were frameshift mutation, which could provide the possibility in production of MSTN knockout cattle in the future.


Asunto(s)
Bovinos/genética , Fibroblastos/metabolismo , Técnicas de Inactivación de Genes/métodos , Miostatina/genética , Animales , Secuencia de Bases , Bovinos/embriología , Bovinos/metabolismo , Línea Celular , Desoxirribonucleasas/metabolismo , Electroporación , Datos de Secuencia Molecular , Mutación , Miostatina/deficiencia
20.
Am J Pathol ; 180(2): 575-82, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22155110

RESUMEN

The relationship between liver and body mass is exemplified by the precision with which the liver:body mass ratio is restored after partial hepatic resection. Nevertheless, the compartments, against which liver mass is so exquisitely regulated, currently remain undefined. In the studies reported here, we investigated the role of skeletal muscle mass in the regulation of liver:body mass ratio and liver regeneration via the analysis of myostatin-null mice, in which skeletal muscle is hypertrophied. The results showed that liver mass is comparable and liver:body mass significantly diminished in the null animals compared to age-, sex-, and strain-matched controls. In association with these findings, basal hepatic Akt signaling is decreased, and the expression of the target genes of the constitutive androstane receptor and the integrin-linked kinase are dysregulated in the myostatin-null mice. In addition, the baseline expression levels of the regulators of the G1-S phase cell cycle progression in liver are suppressed in the null mice. The initiation of liver regeneration is not impaired in the null animals, although it progresses toward the lower liver:body mass set point. The data show that skeletal muscle is not the body component against which liver mass is positively regulated, and thus they demonstrate a previously unrecognized systemic compartmental specificity for the regulation of liver:body mass ratio.


Asunto(s)
Composición Corporal/fisiología , Regeneración Hepática/fisiología , Hígado/fisiología , Músculo Esquelético/fisiología , Animales , Ciclinas/metabolismo , Femenino , Hepatectomía , Hígado/anatomía & histología , Sistema de Señalización de MAP Quinasas , Masculino , Ratones , Ratones Endogámicos , Miostatina/deficiencia , Tamaño de los Órganos/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA