Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Eur J Immunol ; 42(5): 1327-36, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22311635

RESUMEN

Transitional and naïve mature peripheral B cells respond very differently to B-cell receptor (BCR) cross-linking. While transitional B cells undergo apoptosis upon BCR engagement, mature B cells survive and proliferate. This differential response correlates with the capacity of mature, but not transitional B cells to transcribe genes that promote cell survival and proliferation, including those encoding c-Myc and the Bcl-2 family members Bcl-xL and A1. We recently demonstrated that transitional B cells fail to assemble transcriptional machinery at the promoter region of these target genes despite equivalent cytoplasmic signaling and nuclear translocation of key transcription factors including NF-κB and nuclear factor of activated T cells (NFAT). The transcription factor myocyte enhancer factor-2C (MEF2C) is regulated by both calcineurin and mitogen-activated protein kinase signaling pathways, and is essential for proliferation and survival downstream of BCR engagement in mature B cells. In this work, we demonstrate that transitional B cells have intrinsically low levels of MEF2C protein and DNA-binding activity, and that this developmental difference in MEF2C expression is functionally significant. Forced expression of MEF2C in transitional B cells promoted cell survival, proliferation, and upregulation of pro-survival genes. Thus, low MEF2C expression limits transitional B-cell responsiveness to BCR engagement before these cells reach maturity.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica , Factores Reguladores Miogénicos/genética , Células Precursoras de Linfocitos B/metabolismo , Receptores de Antígenos de Linfocitos B/metabolismo , Animales , Proteínas Reguladoras de la Apoptosis/genética , Proteína 11 Similar a Bcl2 , Proliferación Celular , Supervivencia Celular , Factores de Transcripción MEF2 , Proteínas de la Membrana/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Factores Reguladores Miogénicos/análisis , Proteínas Proto-Oncogénicas/genética , Regulación hacia Arriba
2.
J Endod ; 37(12): 1647-52, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22099898

RESUMEN

INTRODUCTION: Tissue engineering and regenerative medicine using stem cell biology has been a promising field for treatment of local and systemic intractable diseases. Recently, stem cells from human exfoliated deciduous teeth (SHED) have been identified as a novel population of stem cells. This study focused on the characterization of SHED as compared with bone marrow-derived mesenchymal stem cells (BMMSCs). METHODS: We investigated potential characteristics of SHED by using DNA microarray, real-time reverse transcriptase polymerase chain reaction, and immunofluorescence analysis. RESULTS: Multiple gene expression profiles indicated that the expression of 2753 genes in SHED had changed by ≥2.0-fold as compared with that in BMMSCs. One of the most significant pathways that accelerated in SHED was that of bone morphogenetic protein (BMP) receptor signaling, which contains several cascades such as PKA, JNK, and ASK1. When the BMP signaling pathway was stimulated by BMP-2, the expression of BMP-2, BMP-4, Runx2, and DSPP was up-regulated significantly in SHED than that in BMMSCs. Furthermore, the BMP-4 protein was expressed much higher in SHED but not in BMMSCs, as confirmed by immunofluorescence. CONCLUSIONS: By using the gene expression profiles, this study indicates that SHED is involved in the BMP signaling pathway and suggests that BMP-4 might play a crucial role in this. These results might be useful for effective cell-based tissue regeneration, including that of bone, pulp, and dentin, by applying the characteristics of SHED.


Asunto(s)
Células de la Médula Ósea/fisiología , Células Madre Mesenquimatosas/fisiología , Células Madre/fisiología , Diente Primario/citología , Proteína Morfogenética Ósea 2/análisis , Proteína Morfogenética Ósea 4/análisis , Receptores de Proteínas Morfogenéticas Óseas/análisis , Calcificación Fisiológica/fisiología , Proteína Quinasa Tipo 4 Dependiente de Calcio Calmodulina/análisis , Linaje de la Célula , Subunidad alfa 1 del Factor de Unión al Sitio Principal/análisis , Subunidad RIIbeta de la Proteína Quinasa Dependiente de AMP Cíclico/análisis , Proteínas de la Matriz Extracelular/análisis , Técnica del Anticuerpo Fluorescente , Perfilación de la Expresión Génica , Humanos , Proteínas de Dominio MADS/análisis , MAP Quinasa Quinasa 4/análisis , MAP Quinasa Quinasa 6/análisis , MAP Quinasa Quinasa Quinasa 5/análisis , Sistema de Señalización de MAP Quinasas/fisiología , Factores de Transcripción MEF2 , Factores Reguladores Miogénicos/análisis , Análisis de Secuencia por Matrices de Oligonucleótidos , Receptores Activados del Proliferador del Peroxisoma/análisis , Fosfoproteínas/análisis , Proteínas Quinasas/análisis , Proteína Proto-Oncogénica c-ets-2/análisis , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Sialoglicoproteínas/análisis , Transducción de Señal/fisiología , Proteína Elk-4 del Dominio ets/análisis
3.
J Cell Biochem ; 108(6): 1302-17, 2009 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-19830700

RESUMEN

It is known that the MyoD family members (MyoD, Myf5, myogenin, and MRF4) play a pivotal role in the complex mechanism of skeletal muscle cell differentiation. However, fragmentary information on transcription factor-specific regulation is available and data on their post-transcriptional and post-translational behavior are still missing. In this work, we combined mRNA and protein expression analysis with their subcellular localization. Each myogenic regulator factor (MRF) revealed a specific mRNA trend and a protein quantitative analysis not overlapping, suggesting the presence of post-transcriptional mechanisms. In addition, each MRF showed a specific behavior in situ, characterized by a differentiation stage-dependent localization suggestive of a post-translational regulation also. Consistently with their transcriptional activity, immunogold electron microscopy data revealed MRFs distribution in interchromatin domains. Our results showed a MyoD and Myf5 contrasting expression profile in proliferating myoblasts, as well as myogenin and MRF4 opposite distribution in the terminally differentiated myotubes. Interestingly, MRFs expression and subcellular localization analysis during C2C12 cell differentiation stages showed two main MRFs regulation mechanisms: (i) the protein half-life regulation to modulate the differentiation stage-dependent transcriptional activity and (ii) the cytoplasmic retention, as a translocation process, to inhibit the transcriptional activity. Therefore, our results exhibit that MRFs nucleo-cytoplasmic trafficking is involved in muscle differentiation and suggest that, besides the MRFs expression level, also MRFs subcellular localization, related to their functional activity, plays a key role as a regulatory step in transcriptional control mechanisms.


Asunto(s)
Diferenciación Celular , Músculo Esquelético/citología , Mioblastos/metabolismo , Factores Reguladores Miogénicos/análisis , Factores Reguladores Miogénicos/metabolismo , Animales , Técnica del Anticuerpo Fluorescente , Ratones , Músculo Esquelético/metabolismo
4.
Orthod Craniofac Res ; 12(3): 168-77, 2009 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-19627518

RESUMEN

Our goal was to discover genes differentially expressed in the perichondrium (PC) of the mandibular condylar cartilage (MCC) that might enhance regenerative medicine or orthopaedic therapies directed at the tissues of the temporomandibular joint. We used targeted gene arrays (osteogenesis, stem cell) to identify genes preferentially expressed in the PC and the cartilaginous (C) portions of the MCC in 2-day-old mice. Genes with higher expression in the PC sample related to growth factor ligand-receptor interactions [FGF-13 (6.4x), FGF-18 (4x), NCAM (2x); PGDF receptors, transforming growth factor (TGF)-beta and IGF-1], the Notch isoforms (especially Notch 3 and 4) and their ligands or structural proteins/proteoglycans [collagen XIV (21x), collagen XVIII (4x), decorin (2.5x)]. Genes with higher expression in the C sample consisted mostly of known cartilage-specific genes [aggrecan (11x), procollagens X (33x), XI (14x), IX (4.5x), Sox 9 (4.4x) and Indian hedgehog (6.7x)]. However, the functional or structural roles of several genes that were expressed at higher levels in the PC sample are unclear [myogenic factor (Myf) 9 (9x), tooth-related genes such as tuftelin (2.5x) and dentin sialophosphoprotein (1.6x), VEGF-B (2x) and its receptors (3-4x) and sclerostin (1.7x)]. FGF, Notch and TGF-beta signalling may be important regulators of MCC proliferation and differentiation; the relatively high expression of genes such as Myf6 and VEGF-B and its receptors suggests a degree of unsuspected plasticity in PC cells.


Asunto(s)
Cartílago Articular/metabolismo , Expresión Génica/genética , Cóndilo Mandibular/metabolismo , Proteínas Adaptadoras Transductoras de Señales , Agrecanos/análisis , Animales , Animales Recién Nacidos , Proteínas Morfogenéticas Óseas/análisis , Colágeno/análisis , Colágeno Tipo IX/análisis , Colágeno Tipo X/análisis , Colágeno Tipo XI/análisis , Decorina , Proteínas del Esmalte Dental/análisis , Proteínas de la Matriz Extracelular/análisis , Factores de Crecimiento de Fibroblastos/análisis , Marcadores Genéticos , Glicoproteínas , Proteínas Hedgehog/análisis , Factor I del Crecimiento Similar a la Insulina/análisis , Péptidos y Proteínas de Señalización Intercelular , Ratones , Factores Reguladores Miogénicos/análisis , Moléculas de Adhesión de Célula Nerviosa/análisis , Fosfoproteínas/análisis , Procolágeno/análisis , Precursores de Proteínas/análisis , Proteoglicanos/análisis , Proteínas Proto-Oncogénicas/análisis , Receptor Notch3 , Receptor Notch4 , Receptores Notch/análisis , Receptores del Factor de Crecimiento Derivado de Plaquetas/análisis , Receptores de Factores de Crecimiento Endotelial Vascular/análisis , Factor de Transcripción SOX9/análisis , Sialoglicoproteínas , Factor de Crecimiento Transformador beta/análisis , Factor de Crecimiento Transformador beta/antagonistas & inhibidores , Factor B de Crecimiento Endotelial Vascular/análisis
5.
Anim Sci J ; 80(6): 678-85, 2009 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-20163658

RESUMEN

To clarify muscle type-specific effect of myostatin on myogenic regulatory factors (MRFs), we examined mRNA expression of MRFs in five skeletal muscles of normal (NM) and myostatin-deficient double-muscled (DM) adult Japanese Shorthorn cattle by quantitative reverse-transcribed PCR. Among the four MRFs, namely, Myf5, MyoD, myogenin, and MRF4, MyoD expression was different among the muscles of the DM cattle (P < 0.01) but not of the NM cattle. Meanwhile, MyoD expression was significantly elevated only in masseter (MS) muscle in the DM cattle due to the myostatin deficiency (P < 0.05). Myf5 and MRF4 expression in semitendinosus (ST) was higher in the DM than in the NM cattle (P < 0.05). According to analysis of myosin heavy chain (MyHC) isoform expression, more MyHC-2x and -2a and less -slow isoforms were expressed in the longissimus and ST muscles compared to the MS muscle in both cattle (P < 0.05), but no significant difference in MyHC expression was observed between the NM and DM cattle. Taken together, myostatin has influences on Myf5 and MRF4 expression in faster-type muscles and on MyoD expression in slower-type muscles, suggesting a possible muscle type-specific effect of myostatin in skeletal muscle growth and maintenance.


Asunto(s)
Bovinos/fisiología , Músculo Esquelético/química , Factores Reguladores Miogénicos/análisis , Miostatina/deficiencia , Animales , Músculo Esquelético/anatomía & histología , Factores Reguladores Miogénicos/genética , Miostatina/fisiología
6.
FEBS Lett ; 582(7): 1025-31, 2008 Apr 02.
Artículo en Inglés | MEDLINE | ID: mdl-18314011

RESUMEN

Cardiac differentiation involves cross-regulation of several transcription factors, such as Mef2C, regulated by p38alpha MAP kinase. We analysed the role of p38alpha in cardiac differentiation. Either the absence or inhibition of p38alpha impairs MEF2C nuclear localization in cardiomyocytes, colocalising with vimentin at the perinuclear region. As a consequence, expression of the Mef2C targets, ANF and myocardin, is drastically downregulated. In contrast, Mlc2v and crt are mainly unaltered, probably by the strong Mef2B upregulation, conpensating for the impaired Mef2C transactivity. In addition, p38alpha deficiency leads to a decrease in the phosphorylated Mlc2v fraction and alpha-actinin accumulation causing sarcomere disorganisation. We propose a critical role for p38alpha in early stages of cardiac differentiation by modulation of Mef2C localisation and sarcomeric assembly.


Asunto(s)
Corazón/embriología , Proteína Quinasa 14 Activada por Mitógenos/fisiología , Miocitos Cardíacos/enzimología , Animales , Diferenciación Celular , Núcleo Celular/química , Células Cultivadas , Factores de Transcripción MEF2 , Ratones , Ratones Noqueados , Proteína Quinasa 14 Activada por Mitógenos/genética , Miocitos Cardíacos/citología , Miocitos Cardíacos/metabolismo , Factores Reguladores Miogénicos/análisis , Sarcómeros/ultraestructura , Factores de Transcripción/metabolismo
7.
Ultrastruct Pathol ; 31(3): 209-19, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-17614000

RESUMEN

Elastofibromas have been described as ill-defined tumors, composed of fibroblastic stromal cells and a dense collagenous stroma. A total of 5 elastofibromas from 4 Japanese patients were examined by ultrastructural and immunohistochemical methods. The proliferating fibroblastic stromal cells in the lesion showed Cx43-type gap junctions, isolated cilia, prominent nuclear fibrous laminae, and primitive cellular junctions with incomplete laminae. The active proliferating fibroblastic cells showed positive staining for vimentin, CD34, factor XIIIa, prominin 2 (CD133), and MEF 2. Conspicuous cell-to-matrix interactions were observed with abnormally unique elastins, collagens (type I, III, and IV), laminin, fibronectin, and amorphous extracellular matrix (GAGs; glycosaminoglycans). As for the origin of elastofibromas, the tumors in the present study were suggested to arise from subscapular or periosteal connective tissue, but further revealed some similarities to other tissues, such as human skin dermal tissue, as exemplified by the presence of an abundance of type I and III collagen, CD34/factor XIIIa-expressing stromal fibroblast-like cells, amorphous extracellular matrix, and a unique abnormal elastin. The elastofibromas might have arisen from stromal stem cell candidate populations of stromal fibroblastic cells (CD34(+), MEF2(+), prominin 2(CD133)(+), and factor XIIIa(+)).


Asunto(s)
Biomarcadores/análisis , Tejido Elástico/ultraestructura , Fibroma/ultraestructura , Neoplasias de los Tejidos Blandos/ultraestructura , Células del Estroma/ultraestructura , Anciano , Antígenos CD34/análisis , Proliferación Celular , Conexina 43/análisis , Tejido Elástico/química , Factor XIIIa/análisis , Femenino , Fibroblastos/química , Fibroblastos/ultraestructura , Fibroma/química , Fibroma/cirugía , Uniones Comunicantes/química , Uniones Comunicantes/ultraestructura , Humanos , Inmunohistoquímica , Factores de Transcripción MEF2 , Masculino , Glicoproteínas de Membrana/análisis , Microscopía Electrónica de Transmisión/métodos , Persona de Mediana Edad , Factores Reguladores Miogénicos/análisis , Neoplasias de los Tejidos Blandos/química , Neoplasias de los Tejidos Blandos/cirugía , Células del Estroma/química
8.
Cardiovasc Res ; 74(2): 290-303, 2007 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-17367767

RESUMEN

OBJECTIVE: Cardiac contractility is regulated tightly as an extrinsic and intrinsic homeostatic mechanism to the heart. The molecular basis of the intrinsic system is largely unknown. Here, we test the hypothesis that bone morphogenetic protein-2 (BMP-2) mediates embryonic cardiac contractility upstream of myocyte-specific enhancer factor 2A (MEF2A). METHODS: The BMP-2 and MEF2A expression pattern was analyzed by RT-PCR, Western blotting, whole-mount in situ hybridization, and an in vivo transgenic approach. The cardiac phenotype of BMP-2 and MEF2A knock-down zebrafish embryos was analysed. Cardiac contractions were recorded with a video camera. Myofibrillar organization was observed with transmission electron microscopy. Gene expression profiles were performed by quantitative real-time PCR analysis. RESULTS: We demonstrate that BMP-2 and MEF2A are co-expressed in embryonic and neonatal cardiac myocytes. Furthermore, we provide evidence that BMP-2 is required for cardiac contractility in vitro and in vivo and that MEF2A expression can be activated by BMP-2 signaling in neonatal cardiomyocytes. BMP-2 is involved in the assembly of the cardiac contractile apparatus. Finally, we find that exogenous MEF2A is sufficient to rescue ventricular contractility defects in the absence of BMP-2 function. CONCLUSIONS: In all, these observations indicate that BMP-2 and MEF2A are key components of a pathway that controls the cardiac ventricular contractility and suggest that the BMP2-MEF2A pathway can offer new opportunities for the treatment of heart failure.


Asunto(s)
Proteínas Morfogenéticas Óseas/metabolismo , Proteínas de Dominio MADS/metabolismo , Contracción Miocárdica/fisiología , Miocitos Cardíacos/fisiología , Factores Reguladores Miogénicos/metabolismo , Transducción de Señal/fisiología , Factor de Crecimiento Transformador beta/metabolismo , Animales , Animales Recién Nacidos , Secuencia de Bases , Western Blotting , Proteína Morfogenética Ósea 2 , Proteínas Morfogenéticas Óseas/análisis , Proteínas Morfogenéticas Óseas/genética , Proteínas Portadoras/farmacología , Células Cultivadas , Regulación de la Expresión Génica/efectos de los fármacos , Ingeniería Genética , Corazón/embriología , Humanos , Proteínas de Dominio MADS/análisis , Proteínas de Dominio MADS/genética , Factores de Transcripción MEF2 , Datos de Secuencia Molecular , Factores Reguladores Miogénicos/análisis , Factores Reguladores Miogénicos/genética , Organismos Modificados Genéticamente , Interferencia de ARN , ARN Interferente Pequeño/administración & dosificación , Ratas , Ratas Sprague-Dawley , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factor de Crecimiento Transformador beta/análisis , Factor de Crecimiento Transformador beta/genética , Pez Cebra/embriología , Pez Cebra/metabolismo , Proteínas de Pez Cebra/genética
9.
Am J Physiol Endocrinol Metab ; 292(2): E413-20, 2007 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-16985263

RESUMEN

In vitro binding assays have indicated that the exercise-induced increase in muscle GLUT4 is preceded by increased binding of myocyte enhancer factor 2A (MEF2A) to its cis-element on the Glut4 promoter. Because in vivo binding conditions are often not adequately recreated in vitro, we measured the amount of MEF2A that was bound to the Glut4 promoter in rat triceps after an acute swimming exercise in vivo, using chromatin immunoprecipitation (ChIP) assays. Bound MEF2A was undetectable in nonexercised controls or at 24 h postexercise but was significantly elevated approximately 6 h postexercise. Interestingly, the increase in bound MEF2A was preceded by an increase in autonomous activity of calcium/calmodulin-dependent protein kinase (CaMK) II in the same muscle. To determine if CaMK signaling mediates MEF2A/DNA associations in vivo, we performed ChIP assays on C(2)C(12) myotubes expressing constitutively active (CA) or dominant negative (DN) CaMK IV proteins. We found that approximately 75% more MEF2A was bound to the Glut4 promoter in CA compared with DN CaMK IV-expressing cells. GLUT4 protein increased approximately 70% 24 h after exercise but was unchanged by overexpression of CA CaMK IV in myotubes. These results confirm that exercise increases the binding of MEF2A to the Glut4 promoter in vivo and provides evidence that CaMK signaling is involved in this interaction.


Asunto(s)
Proteínas Quinasas Dependientes de Calcio-Calmodulina/fisiología , Transportador de Glucosa de Tipo 4/genética , Músculo Esquelético/metabolismo , Factores Reguladores Miogénicos/metabolismo , Condicionamiento Físico Animal/fisiología , Animales , Proteína Quinasa Tipo 4 Dependiente de Calcio Calmodulina , Proteínas Quinasas Dependientes de Calcio-Calmodulina/genética , Proteínas Quinasas Dependientes de Calcio-Calmodulina/metabolismo , Células Cultivadas , Proteínas de Unión al ADN/metabolismo , Humanos , Factores de Transcripción MEF2 , Masculino , Fibras Musculares Esqueléticas/química , Músculo Esquelético/enzimología , Factores Reguladores Miogénicos/análisis , Regiones Promotoras Genéticas , Ratas , Ratas Wistar
10.
Dev Dyn ; 235(11): 3132-43, 2006 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-16960856

RESUMEN

Present knowledge indicates that fibre recruitment (hyperplasia) in developing teleost fish occurs in three distinct phases. However, the origin and relationship of the myogenic precursors activated during the different phases remains unclear. Here, we address this issue using molecular techniques on embryos and larvae of pearlfish, a large cyprinid species. Results provide comprehensive molecular characterisation of cell recruitment over the three phases of myogenesis, identifying muscle types as they arise. Specifically, we show that the myogenic cells arising during 2nd phase myogenesis are clearly different from the myogenic cells arising during the 3rd phase and that the dermomyotome is a major source of myogenic cells driving 2nd phase hyperplasia. These findings are discussed in relation to their implications for the generality of vertebrate developmental patterns.


Asunto(s)
Cyprinidae/crecimiento & desarrollo , Desarrollo de Músculos , Mioblastos Esqueléticos/fisiología , Animales , Proliferación Celular , Cyprinidae/genética , Cyprinidae/metabolismo , Expresión Génica , Desarrollo de Músculos/genética , Proteína MioD/análisis , Proteína MioD/genética , Proteína MioD/metabolismo , Mioblastos Esqueléticos/química , Mioblastos Esqueléticos/metabolismo , Factores Reguladores Miogénicos/análisis , Factores Reguladores Miogénicos/genética , Factores Reguladores Miogénicos/metabolismo , Cadenas Pesadas de Miosina/análisis , Cadenas Pesadas de Miosina/genética , Cadenas Pesadas de Miosina/metabolismo , Factor de Transcripción PAX7/análisis , Factor de Transcripción PAX7/metabolismo
11.
Int J Exp Pathol ; 87(3): 219-25, 2006 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-16709230

RESUMEN

Heart failure (HF) is characterized by a skeletal muscle myopathy with increased expression of fast myosin heavy chains (MHCs). The skeletal muscle-specific molecular regulatory mechanisms controlling MHC expression during HF have not been described. Myogenic regulatory factors (MRFs), a family of transcriptional factors that control the expression of several skeletal muscle-specific genes, may be related to these alterations. This investigation was undertaken in order to examine potential relationships between MRF mRNA expression and MHC protein isoforms in Wistar rat skeletal muscle with monocrotaline-induced HF. We studied soleus (Sol) and extensor digitorum longus (EDL) muscles from both HF and control Wistar rats. MyoD, myogenin and MRF4 contents were determined using reverse transcription-polymerase chain reaction while MHC isoforms were separated using polyacrylamide gel electrophoresis. Despite no change in MHC composition of Wistar rat skeletal muscles with HF, the mRNA relative expression of MyoD in Sol and EDL muscles and that of MRF4 in Sol muscle were significantly reduced, whereas myogenin was not changed in both muscles. This down-regulation in the mRNA relative expression of MRF4 in Sol was associated with atrophy in response to HF while these alterations were not present in EDL muscle. Taken together, our results show a potential role for MRFs in skeletal muscle myopathy during HF.


Asunto(s)
Insuficiencia Cardíaca/metabolismo , Músculo Esquelético/metabolismo , Proteína MioD/metabolismo , Factores Reguladores Miogénicos/metabolismo , Animales , Insuficiencia Cardíaca/patología , Modelos Animales , Músculo Esquelético/química , Músculo Esquelético/patología , Atrofia Muscular/metabolismo , Proteína MioD/análisis , Factores Reguladores Miogénicos/análisis , Factores Reguladores Miogénicos/genética , Miogenina/análisis , Miogenina/genética , Cadenas Pesadas de Miosina/análisis , Cadenas Pesadas de Miosina/metabolismo , ARN Mensajero/análisis , Ratas , Ratas Wistar , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
12.
Circ Res ; 98(4): 540-8, 2006 Mar 03.
Artículo en Inglés | MEDLINE | ID: mdl-16439687

RESUMEN

Hypertrophic cardiomyopathy (HCM) is the most common form of sudden death in young competitive athletes. However, exercise has also been shown to be beneficial in the setting of other cardiac diseases. We examined the ability of voluntary exercise to prevent or reverse the phenotypes of a murine model of HCM harboring a mutant myosin heavy chain (MyHC). No differences in voluntary cage wheel performance between nontransgenic (NTG) and HCM male mice were seen. Exercise prevented fibrosis, myocyte disarray, and induction of "hypertrophic" markers including NFAT activity when initiated before established HCM pathology. If initiated in older HCM animals with documented disease, exercise reversed myocyte disarray (but not fibrosis) and "hypertrophic" marker induction. In addition, exercise returned the increased levels of phosphorylated GSK-3beta to those of NTG and decreased levels of phosphorylated CREB in HCM mice to normal levels. Exercise in HCM mice also favorably impacted components of the apoptotic signaling pathway, including Bcl-2 (an inhibitor of apoptosis) and procaspase-9 (an effector of apoptosis) expression, and caspase-3 activity. Remarkably, there were no differences in mortality between exercised NTG and HCM mice. Thus, not only was exercise not harmful but also it was able to prevent and even reverse established cardiac disease phenotypes in this HCM model.


Asunto(s)
Cardiomiopatía Hipertrófica/prevención & control , Condicionamiento Físico Animal , Animales , Apoptosis , Cardiomiopatía Hipertrófica/patología , Cardiomiopatía Hipertrófica/terapia , Caspasa 3 , Caspasas/metabolismo , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Fibrosis , Glucógeno Sintasa Quinasa 3/metabolismo , Glucógeno Sintasa Quinasa 3 beta , Factores de Transcripción MEF2 , Masculino , Ratones , Ratones Endogámicos C57BL , Miocardio/patología , Factores Reguladores Miogénicos/análisis , Cadenas Pesadas de Miosina/genética , Factores de Transcripción NFATC/análisis , Fosforilación , ARN Mensajero/análisis , Transducción de Señal
13.
Proc Natl Acad Sci U S A ; 102(45): 16362-7, 2005 Nov 08.
Artículo en Inglés | MEDLINE | ID: mdl-16260749

RESUMEN

The primary function of the HIV-1 regulatory protein Tat, activation of transcription from the viral LTR, is highly regulated by complex interactions between Tat and a number of host cell proteins. Tat nuclear import, a process mediated by importin beta, is a prerequisite for its activity. Here, we report and characterize the interaction of the human inhibitor of MyoD family domain-containing protein (I-mfa), HIC, with Tat at a biochemical and a functional level. This interaction was shown to occur in vivo and in vitro and to involve the nuclear localization signal and the transactivation responsive element-binding domains of Tat and the I-mfa domain of HIC. Coexpression of HIC and Tat resulted in the down-regulation of transactivation of the HIV-1 LTR, and colocalization studies revealed the cytoplasmic sequestration of Tat by HIC. Functionally this sequestration appears to be the underlying mechanism of LTR transcriptional repression by HIC and represents a unique mechanism for the control of Tat activity and regulation of HIV-1 replication.


Asunto(s)
Citoplasma/metabolismo , Productos del Gen tat/metabolismo , VIH-1/fisiología , Factores Reguladores Miogénicos/metabolismo , Animales , Secuencia de Bases , Células COS , Chlorocebus aethiops , Regulación hacia Abajo , Productos del Gen tat/análisis , Duplicado del Terminal Largo de VIH , VIH-1/genética , Datos de Secuencia Molecular , Factores Reguladores Miogénicos/análisis , Señales de Localización Nuclear , Activación Transcripcional , Productos del Gen tat del Virus de la Inmunodeficiencia Humana
14.
Am J Physiol Endocrinol Metab ; 289(6): E1071-6, 2005 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-16105857

RESUMEN

As the primary glucose transporter in skeletal muscle, GLUT4 is an important factor in the regulation of blood glucose. We previously reported that stimulation of AMP-activated protein kinase (AMPK) with 5-aminoimidazole-4-carboxamide-1-beta-d-ribofuranoside (AICAR) increased GLUT4 expression in muscle. GLUT4 enhancer factor (GEF) and myocyte enhancer factor 2 (MEF2) have been shown to be important for normal GLUT4 expression because deletion or truncation of the consensus sequences on the promoter causes depressed GLUT4 mRNA expression. This led to the current study to investigate possible roles for GEF and MEF2 in mediating the activation of GLUT4 gene transcription in response to AMPK. Here we show that, although AMPK does not appear to phosphorylate MEF2A, AMPK directly phosphorylates the GEF protein in vitro. MEF2 and GEF are activated in response to AMPK as we observed translocation of both to the nucleus after AICAR treatment. Nuclear MEF2 protein content was increased after 2 h, and GEF protein was increased in the nucleus 1 and 2 h post-AICAR treatment. Last, GEF and MEF2 increase in binding to the GLUT4 promoter within 2 h after AICAR treatment. Thus we conclude that GEF and MEF2 mediate the AMPK-induced increase in transcription of skeletal muscle GLUT4. AMPK can phosphorylate GEF and in response to AICAR, GEF, and MEF2 translocate to the nucleus and have increased binding to the GLUT4 promoter.


Asunto(s)
Proteínas de Unión al ADN/genética , Regulación de la Expresión Génica , Transportador de Glucosa de Tipo 4/genética , Complejos Multienzimáticos/metabolismo , Factores Reguladores Miogénicos/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Factores de Transcripción/genética , Proteínas Quinasas Activadas por AMP , Aminoimidazol Carboxamida/análogos & derivados , Aminoimidazol Carboxamida/farmacología , Animales , Transporte Biológico/efectos de los fármacos , Núcleo Celular/química , Núcleo Celular/metabolismo , ADN/metabolismo , Proteínas de Unión al ADN/análisis , Proteínas de Unión al ADN/fisiología , Activación Enzimática/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/fisiología , Humanos , Factores de Transcripción MEF2 , Músculo Esquelético/química , Músculo Esquelético/ultraestructura , Factores Reguladores Miogénicos/análisis , Factores Reguladores Miogénicos/fisiología , Fosforilación , Regiones Promotoras Genéticas/genética , ARN Mensajero/análisis , Ratas , Proteínas Recombinantes/metabolismo , Ribonucleótidos/farmacología , Factores de Transcripción/análisis , Factores de Transcripción/fisiología
15.
Am J Surg Pathol ; 29(8): 1106-13, 2005 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16006807

RESUMEN

The spindle cell variant of rhabdomyosarcoma (RMS) is uncommon and is most often encountered in the paratesticular region of children in whom it has a good prognosis. Only isolated cases in adulthood have been described. Sixteen cases of spindle cell RMS occurring in adults were retrieved from our files. Eleven patients were male and 5 were female. Patient age ranged from 18 to 79 years (median, 32 years). Tumor size varied from 1.5 to 35 cm (median, 6 cm). The head and neck region, including the oral cavity, parotid gland, nasopharynx, and nasal cavity, was the commonest affected area, accounting for >50% of the cases, followed by retroperitoneum, thigh, leg, subscapular area, hand, vulva, and paratesticular region (1 case each). Follow-up was available in 12 cases, ranging from 1 to 102 months (median, 16.5 months). Treatment modalities included surgery, chemotherapy, and radiation. Two patients died of uncontrolled local disease 13 and 27 months after diagnosis; 4 were alive without disease at 12, 17, 24, and 102 months, including 1 patient with metastasis to 10 of 50 pelvic lymph nodes at presentation; 3 are alive with localized disease at 16, 17, and 19 months; and 1 was followed for 6 months and showed persistent local disease. One patient is alive at 10 months after diagnosis with evidence of metastatic disease to bone, lungs, and breast. All the tumors showed long fascicles of spindle cells with elongated, vesicular nuclei and pale indistinct cytoplasm. Scattered spindled or polygonal rhabdomyoblasts with abundant brightly eosinophilic cytoplasm were present in all cases. In 3 cases, focal areas showed pseudovascular, sclerosing features. There were no round cell or pleomorphic areas. Positive immunohistochemical results were as follows: desmin (15 of 15 cases), myf-4 (12 of 12), fast myosin (7 of 9), myoglobin (2 of 3), HHF-35 (9 of 9), and SMA (11 of 14). One tumor was focally positive for keratins and EMA. All tumors were negative for caldesmon, S-100 protein, and GFAP. Spindle cell RMS is a rare neoplasm in adults and appears to have distinct clinicopathologic features when compared with cases occurring in the pediatric population. Specifically, it appears to be most common in the head and neck region, and although only limited follow-up is available so far, these lesions appear to have a more aggressive clinical course in adults.


Asunto(s)
Rabdomiosarcoma Embrionario/patología , Sarcoma/patología , Actinas/análisis , Adolescente , Adulto , Anciano , Desmina/análisis , Femenino , Proteína Ácida Fibrilar de la Glía/análisis , Humanos , Inmunohistoquímica , Queratinas/análisis , Masculino , Persona de Mediana Edad , Mucina-1 , Factores Reguladores Miogénicos/análisis , Mioglobina/análisis , Miosinas/análisis , Rabdomiosarcoma Embrionario/química , Rabdomiosarcoma Embrionario/terapia , Proteínas S100/análisis , Sarcoma/química , Sarcoma/terapia
16.
Proc Natl Acad Sci U S A ; 102(25): 8966-71, 2005 Jun 21.
Artículo en Inglés | MEDLINE | ID: mdl-15951423

RESUMEN

The purpose of this study was to determine whether the heart in large mammals contains cardiac progenitor cells that regulate organ homeostasis and regenerate dead myocardium after infarction. We report that the dog heart possesses a cardiac stem cell pool characterized by undifferentiated cells that are self-renewing, clonogenic, and multipotent. These clonogenic cells and early committed progeny possess a hepatocyte growth factor (HGF)-c-Met and an insulin-like growth factor 1 (IGF-1)-IGF-1 receptor system that can be activated to induce their migration, proliferation, and survival. Therefore, myocardial infarction was induced in chronically instrumented dogs implanted with sonomicrometric crystals in the region of the left ventricular wall supplied by the occluded left anterior descending coronary artery. After infarction, HGF and IGF-1 were injected intramyocardially to stimulate resident cardiac progenitor cells. This intervention led to the formation of myocytes and coronary vessels within the infarct. Newly generated myocytes expressed nuclear and cytoplasmic proteins specific of cardiomyocytes: MEF2C was detected in the nucleus, whereas alpha-sarcomeric actin, cardiac myosin heavy chain, troponin I, and alpha-actinin were identified in the cytoplasm. Connexin 43 and N-cadherin were also present. Myocardial reconstitution resulted in a marked recovery of contractile performance of the infarcted heart. In conclusion, the activation of resident primitive cells in the damaged dog heart can promote a significant restoration of dead tissue, which is paralleled by a progressive improvement in cardiac function. These results suggest that strategies capable of activating the growth reserve of the myocardium may be important in cardiac repair after ischemic injury.


Asunto(s)
Corazón/fisiología , Infarto del Miocardio/fisiopatología , Miocardio/citología , Regeneración , Células Madre/citología , Animales , Diferenciación Celular , División Celular/efectos de los fármacos , Movimiento Celular , Supervivencia Celular , Perros , Electrocardiografía , Corazón/fisiopatología , Factor de Crecimiento de Hepatocito/farmacología , Factor I del Crecimiento Similar a la Insulina/farmacología , Factores de Transcripción MEF2 , Músculo Liso Vascular/citología , Músculo Liso Vascular/patología , Contracción Miocárdica/efectos de los fármacos , Contracción Miocárdica/fisiología , Infarto del Miocardio/patología , Miocardio/patología , Factores Reguladores Miogénicos/análisis , Proteínas Proto-Oncogénicas c-met/metabolismo , Proteínas Proto-Oncogénicas c-met/fisiología , Receptor IGF Tipo 1/fisiología , Regeneración/efectos de los fármacos , Células Madre/efectos de los fármacos
17.
Mol Cell Biol ; 25(6): 2273-87, 2005 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-15743823

RESUMEN

The myocyte enhancer factor-2 (MEF2) family of transcription factors plays an important role in regulating cellular programs like muscle differentiation, neuronal survival, and T-cell apoptosis. Multisite phosphorylation is known to control the transcriptional activity of MEF2 proteins, but it is unclear whether other modifications are involved. Here, we report that human MEF2D, as well as MEF2C, is modified by SUMO2 and SUMO3 at a motif highly conserved among MEF2 proteins from diverse organisms. This motif is located within the C-terminal transcriptional activation domain, and its sumoylation inhibits transcription. As a transcriptional corepressor of MEF2, histone deacetylase 4 (HDAC4) potentiates sumoylation. This potentiation is dependent on the N-terminal region but not the C-terminal deacetylase domain of HDAC4 and is inhibited by the sumoylation of HDAC4 itself. Moreover, HDAC5, HDAC7, and an HDAC9 isoform also stimulate sumoylation of MEF2. Opposing the action of class IIa deacetylases, the SUMO protease SENP3 reverses the sumoylation to augment the transcriptional and myogenic activities of MEF2. Similarly, the calcium/calmodulin-dependent kinases [corrected] and extracellular signal-regulated kinase 5 signaling pathways negatively regulate the sumoylation. These results thus identify sumoylation as a novel regulatory mechanism for MEF2 and suggest that this modification interplays with phosphorylation to promote intramolecular signaling for coordinated regulation in vivo.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Endopeptidasas/fisiología , Histona Desacetilasas/fisiología , Factores Reguladores Miogénicos/metabolismo , Procesamiento Proteico-Postraduccional , Proteínas Modificadoras Pequeñas Relacionadas con Ubiquitina/fisiología , Factores de Transcripción/metabolismo , Secuencias de Aminoácidos , Secuencia de Aminoácidos , Proteína Quinasa Tipo 1 Dependiente de Calcio Calmodulina , Proteínas Quinasas Dependientes de Calcio-Calmodulina/fisiología , Línea Celular , Núcleo Celular/química , Secuencia Conservada , Cisteína Endopeptidasas , Proteínas de Unión al ADN/análisis , Proteínas de Unión al ADN/genética , Histona Desacetilasas/análisis , Histona Desacetilasas/genética , Humanos , Isoenzimas/fisiología , Proteínas de Dominio MADS , Factores de Transcripción MEF2 , Proteína Quinasa 7 Activada por Mitógenos/fisiología , Datos de Secuencia Molecular , Mutación/genética , Factores Reguladores Miogénicos/análisis , Factores Reguladores Miogénicos/genética , Estructura Terciaria de Proteína , Proteínas Modificadoras Pequeñas Relacionadas con Ubiquitina/análisis , Proteínas Modificadoras Pequeñas Relacionadas con Ubiquitina/metabolismo , Factores de Transcripción/análisis , Factores de Transcripción/genética , Transcripción Genética , Regulación hacia Arriba
18.
Differentiation ; 72(7): 327-40, 2004 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-15554944

RESUMEN

Myogenesis is regulated by the MyoD class of myogenic regulatory factors (MRFs). These basic helix-loop-helix transcription factors dimerize with E proteins to bind conserved E-box sequences in the promoter regions of muscle-specific genes. Perhaps due to their expression in a wide array of tissues, the specific interactions of E proteins with different MRFs have been largely ignored. Likewise, the expression of E proteins in muscle tissue remains mostly uncharacterized. We investigated the expression of the E proteins HEB, E12, and E47 in rat L6 myoblasts, which express only embryonic and fast (2X) myosin heavy chains (MyHCs) in vitro, C2C12 myosatellite cells, and a number of muscle tissues, to determine whether myosin heavy chain diversity is mirrored by diversity in E protein or MRF expression. Although L6 and C2C12 myotubes demonstrate strong expression of embryonic and 2X (fast) MyHCs, immunofluorescence demonstrated the additional expression of type 1 (slow), 2A, and 2B MyHCs in the C2C12 cell line. Immunofluorescence and western blot analyses show that HEB was expressed in differentiating L6 myoblasts, C2C12 cells, and neonatal rat primary myotubes. In contrast, E12 and E47 expression was not detected in either cell line or in any adult muscle tissue examined. These data strongly implicate HEB in the development of skeletal muscle. However, because HEB is expressed in L6 myoblasts, C2C12 myosatellite cells, and neonatal hindlimb muscles, it is unlikely to be involved in a fiber type-specific manner, and may have a more general role in differentiation of myotubes.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Desarrollo de Músculos/fisiología , Proteínas Musculares/metabolismo , Músculo Esquelético/crecimiento & desarrollo , Músculo Esquelético/metabolismo , Factores Reguladores Miogénicos/metabolismo , Factores de Transcripción/metabolismo , Animales , Anticuerpos/inmunología , Línea Celular , Núcleo Celular/química , Núcleo Celular/inmunología , Núcleo Celular/metabolismo , Proteínas de Unión al ADN/análisis , Fibras Musculares Esqueléticas/química , Fibras Musculares Esqueléticas/inmunología , Fibras Musculares Esqueléticas/metabolismo , Proteínas Musculares/análisis , Músculo Esquelético/citología , Mioblastos/química , Mioblastos/inmunología , Mioblastos/metabolismo , Factores Reguladores Miogénicos/análisis , Cadenas Pesadas de Miosina/análisis , Cadenas Pesadas de Miosina/metabolismo , Ratas , Factores de Transcripción TCF , Proteína 1 Similar al Factor de Transcripción 7 , Factores de Transcripción/análisis
19.
Acta Physiol Scand ; 180(4): 395-403, 2004 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-15030381

RESUMEN

AIM: We hypothesized that myogenic differentiation of HSMC would yield a more insulin responsive phenotype. METHODS: We assessed expression of several proteins involved in insulin action or myogenesis during differentiation of primary human skeletal muscle cultures (HSMC). RESULTS: Differentiation increased creatine kinase activity and expression of desmin and myocyte enhancer factor (MEF)2C. No change in expression was observed for big mitogen-activated protein kinase (BMK1/ERK5), MEF2A, insulin receptor (IR), hexokinase II, and IR substrates 1 and 2, while expression of glycogen synthase, extracellular signal-regulated kinase 1 and 2 (ERK1/2 MAP kinase) and the insulin responsive aminopeptidase increased after differentiation. In contrast to protein kinase B (PKB)a, expression of (PKB)b increased, with differentiation. Both basal and insulin-stimulated PI 3-kinase activity increased with differentiation. Insulin-mediated phosphorylation of PKB and ERK1/2 MAP kinase increased after differentiation. CONCLUSION: Components of the insulin-signalling machinery are expressed in myoblast and myotube HSMC; however, insulin responsiveness to PKB and ERK MAP kinase phosphorylation increases with differentiation.


Asunto(s)
Diferenciación Celular/fisiología , Proteínas Quinasas Activadas por Mitógenos/análisis , Músculo Esquelético/fisiología , Proteínas Serina-Treonina Quinasas , Proteínas Proto-Oncogénicas/análisis , Biomarcadores/análisis , Western Blotting/métodos , Células Cultivadas , Creatina Quinasa/metabolismo , Proteínas de Unión al ADN/análisis , Desmina/análisis , Femenino , Hexoquinasa/análisis , Humanos , Insulina/metabolismo , Proteínas de Dominio MADS , Factores de Transcripción MEF2 , Masculino , Persona de Mediana Edad , Músculo Esquelético/metabolismo , Factores Reguladores Miogénicos/análisis , Fosfatidilinositol 3-Quinasas/metabolismo , Fosforilación , Proteínas Proto-Oncogénicas c-akt , Receptor de Insulina/análisis , Factores de Transcripción/análisis
20.
Med Sci Sports Exerc ; 35(6): 923-9, 2003 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-12783039

RESUMEN

PURPOSE: This study examined 12 wk of creatine (Cr) supplementation and heavy resistance training on skeletal muscle creatine kinase (M-CK) mRNA expression and the mRNA and protein expression of the myogenic regulatory factors Myo-D, myogenin, MFR-4, and Myf5. METHODS: Twenty-two untrained males were randomly assigned to either a control (CON), placebo (PLC), or Cr (CRT) group in a double-blind fashion. Muscle biopsies were obtained before and after training. PLC and CRT trained thrice weekly using 3 sets of 6-8 repetitions at 85-90% 1-RM on the leg press, knee extension, and knee curl exercises. CRT ingested 6 g.d-1 of Cr for 12 wk while PLC consumed the equal amount of placebo. RESULTS: After training, M-CK mRNA expression, as well as myogenin and MRF-4 mRNA and protein expression, were found to be significantly greater for CRT compared with PLC and CON, whereas PLC was also significantly different from CON (P < 0.05). For Myo-D mRNA and protein, both CRT and PLC were significantly different from CON (P < 0.05), but CRT and PLC were not different from one another. No significant differences were located for Myf5 mRNA or protein (P > 0.05). M-CK mRNA was correlated with myogenin (r = 0.916) and MRF-4 (r = 0.883) protein (P < 0.05). CONCLUSION: When combined with heavy resistance training, Cr supplementation increases M-CK mRNA expression, likely due to concomitant increases in the expression of myogenin and MRF-4. Therefore, increases in myogenin and MRF-4 mRNA and protein may play a role in increasing myosin heavy chain expression, already shown to occur with Cr supplementation.


Asunto(s)
Creatina Quinasa/biosíntesis , Creatina/administración & dosificación , Creatina/farmacología , Regulación de la Expresión Génica , Músculo Esquelético/fisiología , Factores Reguladores Miogénicos/biosíntesis , Levantamiento de Peso , Administración Oral , Adolescente , Adulto , Creatina Quinasa/análisis , Método Doble Ciego , Humanos , Masculino , Músculo Esquelético/efectos de los fármacos , Factores Reguladores Miogénicos/análisis , Placebos , ARN Mensajero/biosíntesis
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...