Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Genes (Basel) ; 11(10)2020 10 20.
Artículo en Inglés | MEDLINE | ID: mdl-33092227

RESUMEN

miR-23a, a member of the miR-23a/24-2/27a cluster, has been demonstrated to play pivotal roles in many cellular activities. However, the mechanisms of how bta-miR-23a controls the myogenic differentiation (MD) of PDGFRα- bovine progenitor cells (bPCs) remain poorly understood. In the present work, bta-miR-23a expression was increased during the MD of PDGFRα- bPCs. Moreover, bta-miR-23a overexpression significantly promoted the MD of PDGFRα- bPCs. Luciferase reporter assays showed that the 3'-UTR region of MDFIC (MyoD family inhibitor domain containing) could be a promising target of bta-miR-23a, which resulted in its post-transcriptional down-regulation. Additionally, the knockdown of MDFIC by siRNA facilitated the MD of PDGFRα- bPCs, while the overexpression of MDFIC inhibited the activating effect of bta-miR-23a during MD. Of note, MDFIC might function through the interaction between MyoG transcription factor and MEF2C promoter. This study reveals that bta-miR-23a can promote the MD of PDGFRα- bPCs through post-transcriptional downregulation of MDFIC.


Asunto(s)
Diferenciación Celular , Regulación del Desarrollo de la Expresión Génica , MicroARNs/genética , Desarrollo de Músculos , Músculo Esquelético/citología , Factores Reguladores Miogénicos/antagonistas & inhibidores , Células Madre/citología , Animales , Bovinos , Músculo Esquelético/metabolismo , Células Madre/metabolismo
2.
Int J Cardiol ; 167(3): 698-703, 2013 Aug 10.
Artículo en Inglés | MEDLINE | ID: mdl-22464481

RESUMEN

BACKGROUND: Although skeletal muscle atrophy and changes in myosin heavy chain (MyHC) isoforms have often been observed during heart failure, their pathophysiological mechanisms are not completely defined. In this study we tested the hypothesis that skeletal muscle phenotype changes are related to myogenic regulatory factors and myostatin/follistatin expression in spontaneously hypertensive rats (SHR) with heart failure. METHODS: After developing tachypnea, SHR were subjected to transthoracic echocardiogram. Pathological evidence of heart failure was assessed during euthanasia. Age-matched Wistar-Kyoto (WKY) rats were used as controls. Soleus muscle morphometry was analyzed in histological sections, and MyHC isoforms evaluated by electrophoresis. Protein levels were assessed by Western blotting. STATISTICAL ANALYSIS: Student'st test and Pearson correlation. RESULTS: All SHR presented right ventricular hypertrophy and seven had pleuropericardial effusion. Echocardiographic evaluation showed dilation in the left chambers and left ventricular hypertrophy with systolic and diastolic dysfunction in SHR. Soleus weight and fiber cross sectional areas were lower (WKY 3615 ± 412; SHR 2035 ± 224 µm(2); P<0.001), and collagen fractional volume was higher in SHR. The relative amount of type I MyHC isoform was increased in SHR. Myogenin, myostatin, and follistatin expression was lower and MRF4 levels higher in SHR. Myogenin and follistatin expression positively correlated with fiber cross sectional areas and MRF4 levels positively correlated with I MyHC isoform. CONCLUSION: Reduced myogenin and follistatin expression seems to participate in muscle atrophy while increased MRF4 protein levels can modulate myosin heavy chain isoform shift in skeletal muscle of spontaneously hypertensive rats with heart failure.


Asunto(s)
Insuficiencia Cardíaca/patología , Músculo Esquelético/patología , Enfermedades Musculares/metabolismo , Factores Reguladores Miogénicos/antagonistas & inhibidores , Factores Reguladores Miogénicos/biosíntesis , Animales , Proteínas Relacionadas con la Folistatina/antagonistas & inhibidores , Proteínas Relacionadas con la Folistatina/biosíntesis , Insuficiencia Cardíaca/genética , Insuficiencia Cardíaca/metabolismo , Masculino , Músculo Esquelético/metabolismo , Atrofia Muscular/genética , Atrofia Muscular/metabolismo , Atrofia Muscular/patología , Enfermedades Musculares/genética , Enfermedades Musculares/patología , Cadenas Pesadas de Miosina/biosíntesis , Cadenas Pesadas de Miosina/genética , Ratas , Ratas Endogámicas SHR , Ratas Endogámicas WKY , Especificidad de la Especie
3.
J Biol Chem ; 287(48): 40360-70, 2012 Nov 23.
Artículo en Inglés | MEDLINE | ID: mdl-23055528

RESUMEN

BACKGROUND: Notch3 is expressed in myogenic precursors, but its function is not well known. RESULTS: Notch3 represses the activity of Mef2c and is in turn inhibited by the microRNAs-1 and -206. CONCLUSION: Notch3 serves as a regulator for preventing premature myogenic differentiation. SIGNIFICANCE: Understanding how precocious differentiation is prevented is critical for designing therapy for skeletal muscle regeneration. The Notch signaling pathway is a well known regulator of skeletal muscle stem cells known as satellite cells. Loss of Notch1 signaling leads to spontaneous myogenic differentiation. Notch1, normally expressed in satellite cells, is targeted for proteasomal degradation by Numb during differentiation. A homolog of Notch1, Notch3, is also expressed in these cells but is not inhibited by Numb. We find that Notch3 is paradoxically up-regulated during the early stages of differentiation by an enhancer that requires both MyoD and activated Notch1. Notch3 itself strongly inhibits the myogenic transcription factor Mef2c, most likely by increasing the p38 phosphatase Mkp1, which inhibits the Mef2c activator p38 MAP kinase. Active Notch3 decreases differentiation. Mef2c, however, induces microRNAs miR-1 and miR-206, which directly down-regulate Notch3 and allow differentiation to proceed. Thus, the myogenic differentiation-induced microRNAs miR-1 and -206 are important for differentiation at least partly because they turn off Notch3. We suggest that the transient expression of Notch3 early in differentiation generates a temporal lag between myoblast activation by MyoD and terminal differentiation into myotubes directed by Mef2c.


Asunto(s)
Diferenciación Celular , Fosfatasa 1 de Especificidad Dual/metabolismo , MicroARNs/metabolismo , Mioblastos/citología , Factores Reguladores Miogénicos/metabolismo , Receptores Notch/metabolismo , Animales , Línea Celular , Regulación hacia Abajo , Fosfatasa 1 de Especificidad Dual/genética , Factores de Transcripción MEF2 , Ratones , MicroARNs/genética , Proteína MioD/genética , Proteína MioD/metabolismo , Mioblastos/metabolismo , Factores Reguladores Miogénicos/antagonistas & inhibidores , Factores Reguladores Miogénicos/genética , Receptor Notch3 , Receptores Notch/antagonistas & inhibidores , Receptores Notch/genética , Transducción de Señal
4.
Biochem Biophys Res Commun ; 422(3): 522-6, 2012 Jun 08.
Artículo en Inglés | MEDLINE | ID: mdl-22627135

RESUMEN

CDA-II (cell differentiation agent II), isolated from healthy human urine, is a DNA methyltransferase inhibitor. Previous studies indicated that CDA-II played important roles in the regulation of cell growth and certain differentiation processes. However, it has not been determined whether CDA-II affects skeletal myogenesis. In this study, we investigated effects of CDA-II treatment on skeletal muscle progenitor cell differentiation, migration and proliferation. We found that CDA-II blocked differentiation of murine myoblasts C2C12 in a dose-dependent manner. CDA-II repressed expression of muscle transcription factors, such as Myogenin and Mef2c, and structural proteins, such as myosin heavy chain (Myh3), light chain (Mylpf) and MCK. Moreover, CDA-II inhibited C1C12 cell migration and proliferation. Thus, our data provide the first evidence that CDA-II inhibits growth and differentiation of muscle progenitor cells, suggesting that the use of CDA-II might affect skeletal muscle functions.


Asunto(s)
Diferenciación Celular/efectos de los fármacos , Metilasas de Modificación del ADN/antagonistas & inhibidores , Desarrollo de Músculos/efectos de los fármacos , Mioblastos Esqueléticos/efectos de los fármacos , Mioblastos/efectos de los fármacos , Péptidos/farmacología , Fenilacetatos/farmacología , Animales , Línea Celular , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Expresión Génica/efectos de los fármacos , Factores de Transcripción MEF2 , Ratones , Desarrollo de Músculos/genética , Mioblastos/citología , Mioblastos/inmunología , Mioblastos Esqueléticos/citología , Mioblastos Esqueléticos/enzimología , Factores Reguladores Miogénicos/antagonistas & inhibidores , Miogenina/antagonistas & inhibidores , Cadenas Pesadas de Miosina/antagonistas & inhibidores
5.
Nucleic Acids Res ; 40(12): 5378-88, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22396528

RESUMEN

Enzymes that modify the epigenetic status of cells provide attractive targets for therapy in various diseases. The therapeutic development of epigenetic modulators, however, has been largely limited to direct targeting of catalytic active site conserved across multiple members of an enzyme family, which complicates mechanistic studies and drug development. Class IIa histone deacetylases (HDACs) are a group of epigenetic enzymes that depends on interaction with Myocyte Enhancer Factor-2 (MEF2) for their recruitment to specific genomic loci. Targeting this interaction presents an alternative approach to inhibiting this class of HDACs. We have used structural and functional approaches to identify and characterize a group of small molecules that indirectly target class IIa HDACs by blocking their interaction with MEF2 on DNA.Weused X-ray crystallography and (19)F NMRto show that these compounds directly bind to MEF2. We have also shown that the small molecules blocked the recruitment of class IIa HDACs to MEF2-targeted genes to enhance the expression of those targets. These compounds can be used as tools to study MEF2 and class IIa HDACs in vivo and as leads for drug development.


Asunto(s)
Anilidas/química , Anilidas/farmacología , Inhibidores de Histona Desacetilasas/química , Inhibidores de Histona Desacetilasas/farmacología , Factores Reguladores Miogénicos/antagonistas & inhibidores , Animales , Sitios de Unión , Línea Celular , ADN/química , Células HeLa , Histona Desacetilasas/análisis , Histona Desacetilasas/química , Histona Desacetilasas/metabolismo , Humanos , Factores de Transcripción MEF2 , Modelos Moleculares , Factores Reguladores Miogénicos/química
6.
Fish Physiol Biochem ; 38(2): 585-93, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21773810

RESUMEN

Histone deacetylase 4 (HDAC4) is an important modifier enzyme for chromatin remodeling and plays an essential role in regulating gene expression. Spatio-temporal expression spectrum revealed that zebrafish hdac4 mRNA, ubiquitously distributed in various tissues, were significantly higher at 36 hpf (hours post-fertilization) and 6 dpf (days post-fertilization) than other periods. Trichostatin A (TSA) inhibited the development of zebrafish embryos and transcription of hdac4 and mef2a (myocyte enhancer factor-2A). Moreover, five vectors containing different promoter regions of hdac4 were constructed in order to analyze promoter activity. The vector containing the region from -125 to +160 exhibited maximum luciferase activity that was approximately 30.3-fold and 58.9-fold higher than the control in two kinds of cells, respectively. By comparing the luciferase activities between the region from -302 to +30 and -698 to +30, it was suggested that the region between -698 and -302 might contain mild negative regulatory elements.


Asunto(s)
Proteínas de Peces/metabolismo , Histona Desacetilasas/metabolismo , Factores Reguladores Miogénicos/metabolismo , Regiones Promotoras Genéticas , Proteínas de Pez Cebra/metabolismo , Pez Cebra/metabolismo , Animales , Secuencia de Bases , Cricetinae , Desarrollo Embrionario/efectos de los fármacos , Proteínas de Peces/antagonistas & inhibidores , Inhibidores de Histona Desacetilasas/farmacología , Ácidos Hidroxámicos/farmacología , Factores de Transcripción MEF2 , Ratones , Datos de Secuencia Molecular , Factores Reguladores Miogénicos/antagonistas & inhibidores , Células 3T3 NIH , Pez Cebra/genética
7.
J Cell Biol ; 195(3): 403-15, 2011 Oct 31.
Artículo en Inglés | MEDLINE | ID: mdl-22042619

RESUMEN

Histone deacetylase 4 (HDAC4) regulates numerous gene expression programs through its signal-dependent repression of myocyte enhancer factor 2 (MEF2) and serum response factor (SRF) transcription factors. In cardiomyocytes, calcium/calmodulin-dependent protein kinase II (CaMKII) signaling promotes hypertrophy and pathological remodeling, at least in part by phosphorylating HDAC4, with consequent stimulation of MEF2 activity. In this paper, we describe a novel mechanism whereby protein kinase A (PKA) overcomes CaMKII-mediated activation of MEF2 by regulated proteolysis of HDAC4. PKA induces the generation of an N-terminal HDAC4 cleavage product (HDAC4-NT). HDAC4-NT selectively inhibits activity of MEF2 but not SRF, thereby antagonizing the prohypertrophic actions of CaMKII signaling without affecting cardiomyocyte survival. Thus, HDAC4 functions as a molecular nexus for the antagonistic actions of the CaMKII and PKA pathways. These findings have implications for understanding the molecular basis of cardioprotection and other cellular processes in which CaMKII and PKA exert opposing effects.


Asunto(s)
Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Histona Desacetilasas/metabolismo , Factores Reguladores Miogénicos/antagonistas & inhibidores , Animales , Células COS , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/genética , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/metabolismo , Núcleo Celular/metabolismo , Chlorocebus aethiops , Técnica del Anticuerpo Fluorescente , Regulación de la Expresión Génica , Factores de Transcripción MEF2 , Ratones , Miocitos Cardíacos/metabolismo , Factores Reguladores Miogénicos/metabolismo , Proteolisis , Ratas , Ratas Sprague-Dawley , Transducción de Señal
8.
Int J Cancer ; 129(11): 2662-73, 2011 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-21213211

RESUMEN

Tumor-associated factors are related to increased accumulation of CD11b(+) Gr1(+) myeloid-derived suppressor cells (MDSCs). However, the exact mechanism of how genetic factors control the expansion of MDSCs in tumor-bearing hosts remains elusive. Herein, we found that tumor-associated MDSCs and their subsets, mononuclear MDSCs and polymorphonuclear MDSCs, have decreased expression of miR-223 when compared to CD11b(+) Gr1(+) cells from the spleen of disease-free mice. With the differentiation of CD11b(+) Gr1(+) MDSCs from bone marrow cells (BMCs) upon exposure to tumor-associated factors, the expression of both pri-miR-223 and mature miR-223 was downregulated, indicating that the expression of miR-223 could be regulated by tumor-associated factors. Interestingly, miR-223 remarkably inhibits differentiation of BMCs into CD11b(+) Gr1(+) MDSCs in the presence of tumor-associated factors by targeting myocyte enhancer factor 2C (MEF2C). Using reconstituted s.c. tumor models, miR-223 also suppresses accumulation of CD11b(+) Gr1(+) MDSCs, whereas its targeting molecule MEF2C increases the number of MDSCs. Tumor growth is slower in mice infused by miR223-engineered BMCs than in mice infused with control transfected BMCs. As miR-223 and its target molecule MEF2C are highly conserved between mice and humans, the modulation of miR-223 in tumor-induced CD11b(+) Gr1(+) MDSCs may exert an important role in controlling the increased accumulation of CD11b(+) Gr1(+) MDSCs in patients with tumor.


Asunto(s)
Células de la Médula Ósea/inmunología , Antígeno CD11b/metabolismo , Diferenciación Celular/inmunología , MicroARNs/fisiología , Células Mieloides/inmunología , Receptores de Quimiocina/metabolismo , Animales , Western Blotting , Células Cultivadas , Ensayo de Inmunoadsorción Enzimática , Femenino , Citometría de Flujo , Técnicas para Inmunoenzimas , Factores de Transcripción MEF2 , Macrófagos/citología , Macrófagos/inmunología , Macrófagos/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Monocitos/citología , Monocitos/inmunología , Monocitos/metabolismo , Células Mieloides/metabolismo , Células Mieloides/patología , Factores Reguladores Miogénicos/antagonistas & inhibidores , Factores Reguladores Miogénicos/genética , Factores Reguladores Miogénicos/metabolismo , ARN Mensajero/genética , ARN Interferente Pequeño/genética , Especies Reactivas de Oxígeno/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
9.
Neurobiol Dis ; 41(1): 71-82, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-20816781

RESUMEN

α-Synuclein is an abundant neuronal protein that has been linked to both normal synaptic function and neurodegenerative disease, in particular, Parkinson's disease (PD). Evidence from both in vitro and in vivo studies indicate that increased wild type or mutant α-synuclein can cause PD, but the molecular mechanisms that underlie α-synuclein-mediated neurotoxicity remain poorly understood. We reported here that myocyte enhancer factor 2D (MEF2D), a nuclear transcription factor known to promote neuronal survival, is down regulated in response α-synuclein accumulation and aggregation. Our data demonstrated that levels of cytoplasmic and nuclear MEF2D were significantly decreased in PD nigral neurons when compared to the nigra of age-matched controls and Alzheimer's disease (AD) cases. This decrease was significantly greater in the nigral neurons with α-synuclein inclusions. Viral vector-mediated overexpression of human α-synuclein in rats resulted in significantly decreased MEF2D in nigral neurons similar to what was seen in PD. The decline of MEF2D-immunoreactivity was associated with a reduction in TH-immunoreactivity. These results indicate that the neuronal survival factor MEF2D is decreased in human and experimental PD, and this decrease is specifically associated with α-synuclein accumulation and aggregation.


Asunto(s)
Regulación hacia Abajo/genética , Proteínas de Dominio MADS/antagonistas & inhibidores , Factores Reguladores Miogénicos/antagonistas & inhibidores , Neuronas/metabolismo , Enfermedad de Parkinson/metabolismo , Sustancia Negra/metabolismo , alfa-Sinucleína/fisiología , Anciano , Anciano de 80 o más Años , Animales , Modelos Animales de Enfermedad , Femenino , Humanos , Cuerpos de Inclusión/metabolismo , Cuerpos de Inclusión/patología , Proteínas de Dominio MADS/metabolismo , Factores de Transcripción MEF2 , Masculino , Persona de Mediana Edad , Factores Reguladores Miogénicos/metabolismo , Neuronas/patología , Enfermedad de Parkinson/patología , Ratas , Ratas Sprague-Dawley , Ratas Transgénicas , Sustancia Negra/patología , alfa-Sinucleína/genética
10.
Am J Physiol Heart Circ Physiol ; 298(5): H1499-509, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-20207814

RESUMEN

Endoplasmic reticulum (ER) stress (ERS) is involved in various cardiovascular diseases. Our previous study verified that ERS took part in the development of cardiac hypertrophy; however, its mechanism is still unclear. This study aimed to investigate the roles of the calcineurin (CaN) signal pathway in hypertrophy induced by the ERS inductor thapsigargin (TG) in neonatal cardiomyocytes from Sprague-Dawley rats. Investigation of ER chaperone expression, ER staining, and calreticulin immunofluorescence were used to detect the ERS response. mRNA expression of atrial natriuretic peptide and brain natriuretic peptide, total protein synthesis rate, and cell surface area were used to evaluate cardiac hypertrophy induced by TG. TG induced a significant ERS response along with hypertrophy in a dose- and time-dependent manner in cardiomyocytes, which was verified by treatment with tunicamycin, another ERS inducer. Furthermore, TG induced a significant elevation of the intracellular Ca(2+) level, CaN activation, and myocyte enhancer factor 2c (MEF2c) expression in a dose- and time-dependent manner in cardiomyocytes. Cyclosporine A, a CaN inhibitor, markedly suppressed MEF2c nuclear translocation and inhibited TG-induced hypertrophy. These results demonstrate that ERS induces cardiac hypertrophy and that the CaN-MEF2c pathway is involved in ERS-induced hypertrophy in cardiomyocytes.


Asunto(s)
Animales Recién Nacidos/fisiología , Calcineurina/fisiología , Cardiomegalia/patología , Retículo Endoplásmico/patología , Miocitos Cardíacos/fisiología , Factores Reguladores Miogénicos/fisiología , Transducción de Señal/fisiología , Animales , Apoptosis/efectos de los fármacos , Western Blotting , Calcio/metabolismo , Tamaño de la Célula/efectos de los fármacos , Células Cultivadas , Ciclosporina/farmacología , Relación Dosis-Respuesta a Droga , Inhibidores Enzimáticos/farmacología , Técnica del Anticuerpo Fluorescente , L-Lactato Deshidrogenasa/metabolismo , Factores de Transcripción MEF2 , Factores Reguladores Miogénicos/antagonistas & inhibidores , ARN/biosíntesis , ARN/aislamiento & purificación , Ratas , Ratas Sprague-Dawley , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , ATPasas Transportadoras de Calcio del Retículo Sarcoplásmico/metabolismo , Transducción de Señal/efectos de los fármacos , Tapsigargina/farmacología , Tunicamicina/farmacología
11.
Exp Mol Med ; 42(3): 205-15, 2010 Mar 31.
Artículo en Inglés | MEDLINE | ID: mdl-20164678

RESUMEN

Chronic and heavy alcohol consumption is one of the causes of heart diseases. However, the effects of ethanol on insulin sensitivity in myocardium has been unclear. To investigate the effects of ethanol on the expression of AMP-activated protein kinase (AMPK), myocyte enhancer factor 2 (MEF2) and glucose transporter 4 (GLUT4), all of which are involved in the regulation of insulin sensitivity, in the myocardium, we performed three parts of experiments in vivo and in vitro. I: Rats were injected with 5-amino-4-imidazolecarboxamide ribonucleotide (AICAR, 0.8 mg.kg(-l)) for 2 h. II: Rats received different dose (0.5, 2.5 or 5 g.kg(-l).d(-l)) of ethanol for 22-week. III: Primary neonatal rat cardiomyocytes were isolated and treated with or without 100 mM ethanol or 1 mM AICAR for 4 h. The cardiac protein and mRNA expression of AMPKalpha subunits, MEF2 and GLUT4 were observed by western-blotting and RT-PCR, respectively. Serum TNFalpha levels were assessed by ELISA. The results showed chronic ethanol exposure induced insulin resistance. Ethanol decreased the mRNA levels of AMPKalpha1 and alpha2, the protein levels of total- and phospho-AMPKalpha in cardiomyocytes. Similarly, ethanol showed inhibitory effects on both the mRNA and protein levels of MEF2A and 2D, and GLUT4 in a dose-response-like fashion. Correlation analysis implied an association between phospho-AMPKalpha and MEF2A or MEF2D, and between the levels of MEF2 protein and GLUT4 transcription. In addition, ethanol elevated serum TNFalpha level. Taken together, chronic ethanol exposure decreases the expression of AMPKalpha and MEF2, and is associated with GLUT4 decline in rat myocardium.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Etanol/administración & dosificación , Etanol/farmacología , Conducta Alimentaria/efectos de los fármacos , Transportador de Glucosa de Tipo 4/metabolismo , Miocardio/enzimología , Factores Reguladores Miogénicos/metabolismo , Proteínas Quinasas Activadas por AMP/genética , Aminoimidazol Carboxamida/análogos & derivados , Aminoimidazol Carboxamida/farmacología , Animales , Activación Enzimática/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , Transportador de Glucosa de Tipo 4/genética , Insulina/farmacología , Resistencia a la Insulina , Factores de Transcripción MEF2 , Masculino , Factores Reguladores Miogénicos/antagonistas & inhibidores , Factores Reguladores Miogénicos/genética , Isoformas de Proteínas/antagonistas & inhibidores , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ratas , Ratas Wistar , Ribonucleótidos/farmacología , Factores de Tiempo , Factor de Necrosis Tumoral alfa/sangre
12.
FASEB J ; 24(6): 1780-8, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20086047

RESUMEN

Krüppel-like factor 5 (KLF5) is a zinc-finger-type transcription factor that mediates the tissue remodeling in cardiovascular diseases, such as atherosclerosis, restenosis, and cardiac hypertrophy. Our previous studies have shown that KLF5 is induced by angiotensin II (AII), although the precise molecular mechanism is not yet known. Here we analyzed regulatory single nucleotide polymorphisms (SNPs) within the KLF5 locus to identify clinically relevant signaling pathways linking AII and KLF5. One SNP was located at -1282 bp and was associated with an increased risk of hypertension: subjects with the A/A and A/G genotypes at -1282 were at significantly higher risk for hypertension than those with the G/G genotype. Interestingly, a reporter construct corresponding to the -1282G genotype showed much weaker responses to AII than a construct corresponding to -1282A. Electrophoretic mobility shift, chromatin immunoprecipitation, and reporter assays collectively showed that the -1282 SNP is located within a functional myocyte enhancer factor 2 (MEF2) binding site, and that the -1282G genotype disrupts the site and reduces the AII responsiveness of the promoter. Moreover, MEF2 activation via reactive oxygen species and p38 mitogen-activated protein kinase induced KLF5 expression in response to AII, and KLF5 and MEF2 were coexpressed in coronary atherosclerotic plaques. These results suggest that a novel signaling and transcription network involving MEF2A and KLF5 plays an important role in the pathogenesis of cardiovascular diseases such as hypertension.


Asunto(s)
Angiotensina II/farmacología , Aterosclerosis/genética , Hipertensión/genética , Factores de Transcripción de Tipo Kruppel/genética , Proteínas de Dominio MADS/metabolismo , Factores Reguladores Miogénicos/metabolismo , Polimorfismo de Nucleótido Simple/genética , Adulto , Anciano , Anciano de 80 o más Años , Animales , Aorta/efectos de los fármacos , Aorta/metabolismo , Aterosclerosis/metabolismo , Aterosclerosis/patología , Estudios de Casos y Controles , Células Cultivadas , Inmunoprecipitación de Cromatina , Ensayo de Cambio de Movilidad Electroforética , Ensayo de Inmunoadsorción Enzimática , Femenino , Humanos , Hipertensión/metabolismo , Hipertensión/patología , Immunoblotting , Técnicas para Inmunoenzimas , Inmunoprecipitación , Proteínas de Dominio MADS/antagonistas & inhibidores , Proteínas de Dominio MADS/genética , Factores de Transcripción MEF2 , Masculino , Persona de Mediana Edad , Factores Reguladores Miogénicos/antagonistas & inhibidores , Factores Reguladores Miogénicos/genética , Regiones Promotoras Genéticas , ARN Mensajero/genética , ARN Mensajero/metabolismo , ARN Interferente Pequeño/farmacología , Ratas , Especies Reactivas de Oxígeno/metabolismo , Elementos de Respuesta , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
13.
Eur J Heart Fail ; 12(1): 4-12, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-20023039

RESUMEN

AIMS: The transcription factor MEF2 is a downstream target for several hypertrophic signalling pathways in the heart, suggesting that MEF2 may act as a valuable therapeutic target in the treatment of heart failure. METHODS AND RESULTS: In this study, we investigated the potential benefits of overall MEF2 inhibition in a mouse model of chronic pressure overloading, by subjecting transgenic mice expressing a dominant negative form of MEF2 (DN-MEF2 Tg) in the heart, to transverse aortic constriction (TAC). Histological analysis revealed no major differences in cardiac remodelling between DN-MEF2 Tg and control mice after TAC. Surprisingly, echocardiographic analysis revealed that DN-MEF2 Tg mice had a decrease in cardiac function compared with control animals. Analysis of the mitochondrial respiratory chain showed that DN-MEF2 Tg mice displayed lower expression of NADH dehydrogenase subunit 6 (ND6), part of mitochondrial Complex I. The reduced expression of ND6 in DN-MEF2 Tg mice after pressure overload correlated with an increase in cell death secondary to overproduction of reactive oxygen species (ROS). CONCLUSION: Our data suggest that MEF2 transcriptional activity is required for mitochondrial function and its inhibition predisposes the heart to impaired mitochondrial function, overproduction of ROS, enhanced cell death, and cardiac dysfunction, following pressure overload.


Asunto(s)
Cardiomegalia/etiología , Insuficiencia Cardíaca/etiología , Mitocondrias Cardíacas/fisiología , Factores Reguladores Miogénicos/antagonistas & inhibidores , 8-Hidroxi-2'-Desoxicoguanosina , Animales , Apoptosis/genética , Cardiomegalia/metabolismo , Cardiomegalia/patología , Desoxiguanosina/análogos & derivados , Desoxiguanosina/genética , Modelos Animales de Enfermedad , Ecocardiografía , Expresión Génica , Insuficiencia Cardíaca/metabolismo , Insuficiencia Cardíaca/patología , Integrasas/genética , Factores de Transcripción MEF2 , Ratones , Ratones Transgénicos , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , Factores Reguladores Miogénicos/genética , NADH Deshidrogenasa/deficiencia , Transcripción Genética , Resultado del Tratamiento , Remodelación Ventricular/genética
15.
Biochem Biophys Res Commun ; 377(1): 120-5, 2008 Dec 05.
Artículo en Inglés | MEDLINE | ID: mdl-18834865

RESUMEN

We have identified two novel MEK5 inhibitors, BIX02188 and BIX02189, which inhibited catalytic function of purified, MEK5 enzyme. The MEK5 inhibitors blocked phosphorylation of ERK5, without affecting phosphorylation of ERK1/2 in sorbitol-stimulated HeLa cells. The compounds also inhibited transcriptional activation of MEF2C, a downstream substrate of the MEK5/ERK5 signaling cascade, in a cellular trans-reporter assay system. These inhibitors offer novel pharmacological tools to better characterize the role of the MEK5/ERK5 pathway in various biological systems.


Asunto(s)
Compuestos de Anilina/farmacología , Indoles/farmacología , MAP Quinasa Quinasa 5/antagonistas & inhibidores , Proteína Quinasa 7 Activada por Mitógenos/antagonistas & inhibidores , Inhibidores de Proteínas Quinasas/farmacología , Compuestos de Anilina/aislamiento & purificación , Células HeLa , Humanos , Indoles/aislamiento & purificación , Proteínas de Dominio MADS/antagonistas & inhibidores , Proteínas de Dominio MADS/genética , MAP Quinasa Quinasa 5/metabolismo , Factores de Transcripción MEF2 , Proteína Quinasa 7 Activada por Mitógenos/metabolismo , Factores Reguladores Miogénicos/antagonistas & inhibidores , Factores Reguladores Miogénicos/genética , Fosforilación/efectos de los fármacos , Inhibidores de Proteínas Quinasas/aislamiento & purificación , Sorbitol/farmacología , Activación Transcripcional/efectos de los fármacos
16.
Stem Cells Dev ; 17(4): 751-60, 2008 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-18484897

RESUMEN

We previously reported that treatment of icariin could significantly induce cardiomyocyte differentiation of murine embryonic stem (ES) cells in vitro. In the present study, the exact activity initiated by icariin was further confirmed and the underlying molecular mechanism was investigated. We found that cardiomyocyte differentiation was efficiently stimulated only if icariin was administrated between days 5 and 8 in differentiation course, which indicated with elevated percentage of embryoid bodies (EB) and with beating areas and up- regulated expression of alpha-actinin and troponin T. Exposure of icariin triggered intracellular reactive oxygen species (ROS) generation of EBs in 3 h, which was abolished in the presence of either NADPH oxidase inhibitor DPI or antioxidant Trolox. Meanwhile, expression of NOX4, a membrane combined enzyme responsible for ROS generation, was promoted by icariin in a dose-dependent manner. Although p38MAPK (mitogen-activated protein kinase), extracellular signal-regulated kinase (ERK), and c-Jun N-terminal protein kinase (JNK) were spontaneously activated in early differentiation, only the phosphorylation of p38MAPK was enhanced and prolonged when icariin was present, whereas both ERK and JNK showed no response to icariin treatment. Moreover, the inducible effect of icariin was blunted by SB203580, a specific inhibitor of p38MAPK. On the contrary, neither UO126 nor SP600125, the specific inhibitor of ERK and JNK, could abolish icariin-stimulated differentiation. Nuclear location of MEF2C, which played a critical role in cardiomyocyte differentiation and could be activated by p38MAPK, was stimulated after icariin exposure. Taken together, these results suggest that ROS generation and the subsequent activation of p38MAPK are essential for the inducible function of icariin on cardiomyocyte differentiation of murine embryonic stem cells in vitro.


Asunto(s)
Diferenciación Celular/efectos de los fármacos , Células Madre Embrionarias/enzimología , Flavonoides/farmacología , Miocitos Cardíacos/enzimología , Especies Reactivas de Oxígeno/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Transporte Activo de Núcleo Celular/efectos de los fármacos , Transporte Activo de Núcleo Celular/fisiología , Animales , Antioxidantes/farmacología , Diferenciación Celular/fisiología , Línea Celular , Núcleo Celular/enzimología , Cromanos/farmacología , Relación Dosis-Respuesta a Droga , Células Madre Embrionarias/citología , Activación Enzimática/efectos de los fármacos , Activación Enzimática/fisiología , Inhibidores Enzimáticos/farmacología , Quinasas MAP Reguladas por Señal Extracelular/antagonistas & inhibidores , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Proteínas Quinasas JNK Activadas por Mitógenos/antagonistas & inhibidores , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Factores de Transcripción MEF2 , Ratones , Miocitos Cardíacos/citología , Factores Reguladores Miogénicos/antagonistas & inhibidores , Factores Reguladores Miogénicos/metabolismo , NADPH Oxidasa 4 , NADPH Oxidasas/antagonistas & inhibidores , NADPH Oxidasas/metabolismo , Fosforilación/efectos de los fármacos , Factores de Tiempo , Proteínas Quinasas p38 Activadas por Mitógenos/antagonistas & inhibidores
17.
Artículo en Chino | MEDLINE | ID: mdl-17419218

RESUMEN

OBJECTIVE: To review researches of the role of inhibitor of differentiation 2(Id2) in skeletal muscle regeneration. METHODS: The latest original literature concerning Id2 and its role in skeletal muscle regeneration was extensively reviewed. RESULTS: Id2 could form heterodimers by combining with E protein to prevent myogenic regulatory factors (MRFs) forming heterodimers by combining with E protein, to inhibit the transcription activity of MRFs and differentiation of skeletal muscle cell. CONCLUSION: Id2 plays an important role in skeletal muscle regeneration.


Asunto(s)
Proteína 2 Inhibidora de la Diferenciación/fisiología , Músculo Esquelético/fisiología , Factores Reguladores Miogénicos/antagonistas & inhibidores , Regeneración , Animales , Diferenciación Celular , Proliferación Celular , Humanos , Ratones , Músculo Esquelético/citología , Músculo Esquelético/lesiones , Mioblastos/citología , Mioblastos/fisiología
18.
Br J Pharmacol ; 145(8): 1103-11, 2005 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-15912127

RESUMEN

Experimental data implicate calpain activation in the pathways involved in neuronal apoptosis. Indeed, calpain inhibitors confer neuroprotection in response to various neurotoxic stimuli. However, the pathways involved in calpain activation-induced apoptosis are not well known. We demonstrate that apoptosis (40%) induced by serum/potassium (S/K) withdrawal on cerebellar granule cells (CGNs) is inhibited by selective calpain inhibitors PD150606 (up to 15%) and PD151746 (up to 29%), but not PD145305 in CGNs. zVAD-fmk, a broad spectrum inhibitor of caspases, attenuates apoptosis (up to 20%) mediated by S/K deprivation and protects against cell death, as measured by MTT ([3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium]) assay. PD150606 and PD151746 prevented apoptosis mediated by S/K withdrawal through inhibition of calpain. Furthermore, PD151746 was able to inhibit caspase-3 activity. After S/K withdrawal, we observed an increase in cdk5/p25 formation and MEF2 phosphorylation that was prevented by 40 microM PD150606 and PD151746. This indicates that calpain inhibition may be an upstream molecular target that prevents neuronal apoptosis in vitro. Taken together, these data suggest an apoptotic route in S/K withdrawal in CGNs mediated by calpain activation, cdk5/p25 formation and MEF2 inhibition. Calpain inhibitors may attenuate S/K withdrawal-induced apoptosis and may provide a potential therapeutic target for drug treatment in a neurodegenerative process.


Asunto(s)
Apoptosis/efectos de los fármacos , Calpaína/antagonistas & inhibidores , Cerebelo/citología , Factores Reguladores Miogénicos/antagonistas & inhibidores , Neuronas/efectos de los fármacos , Animales , Animales Recién Nacidos , Técnicas de Cultivo de Célula , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Medios de Cultivo , Inhibidores Enzimáticos/farmacología , Factores de Transcripción MEF2 , Neuronas/metabolismo , Neuronas/patología , Ratas , Ratas Sprague-Dawley
19.
Cell ; 117(7): 927-39, 2004 Jun 25.
Artículo en Inglés | MEDLINE | ID: mdl-15210113

RESUMEN

Metastasis is a multistep process during which cancer cells disseminate from the site of primary tumors and establish secondary tumors in distant organs. In a search for key regulators of metastasis in a murine breast tumor model, we have found that the transcription factor Twist, a master regulator of embryonic morphogenesis, plays an essential role in metastasis. Suppression of Twist expression in highly metastatic mammary carcinoma cells specifically inhibits their ability to metastasize from the mammary gland to the lung. Ectopic expression of Twist results in loss of E-cadherin-mediated cell-cell adhesion, activation of mesenchymal markers, and induction of cell motility, suggesting that Twist contributes to metastasis by promoting an epithelial-mesenchymal transition (EMT). In human breast cancers, high level of Twist expression is correlated with invasive lobular carcinoma, a highly infiltrating tumor type associated with loss of E-cadherin expression. These results establish a mechanistic link between Twist, EMT, and tumor metastasis.


Asunto(s)
Neoplasias de la Mama/patología , Carcinoma Lobular/patología , Neoplasias Mamarias Experimentales/patología , Factores Reguladores Miogénicos/antagonistas & inhibidores , Proteínas Nucleares/antagonistas & inhibidores , Proteínas Nucleares/metabolismo , Factores de Transcripción/metabolismo , Animales , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Cadherinas/metabolismo , Carcinoma Lobular/genética , Carcinoma Lobular/metabolismo , Línea Celular , Línea Celular Tumoral , Movimiento Celular , Células Epiteliales/citología , Células Epiteliales/metabolismo , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Luciferasas/metabolismo , Neoplasias Pulmonares/secundario , Neoplasias Mamarias Experimentales/genética , Neoplasias Mamarias Experimentales/metabolismo , Mesodermo , Ratones , Ratones Endogámicos BALB C , Morfogénesis , Invasividad Neoplásica , Metástasis de la Neoplasia , Trasplante de Neoplasias , Tamaño de los Órganos , ARN Mensajero/metabolismo , ARN Interferente Pequeño/metabolismo , Proteínas Represoras , Proteína 1 Relacionada con Twist
20.
EMBO J ; 23(2): 365-75, 2004 Jan 28.
Artículo en Inglés | MEDLINE | ID: mdl-14739931

RESUMEN

Skeletal myogenesis is associated with the activation of four muscle regulatory factors (MRFs): Myf5, MyoD, Myogenin and MRF4. Here we report that p38 mitogen-activated protein kinase represses the transcriptional activity of MRF4 (involved in late stages of myogenesis), resulting in downregulation of specific muscle genes. MRF4 is phosphorylated in vitro and in vivo by p38 on two serines (Ser31 and Ser42) located in the N-terminal transactivation domain, resulting in reduced MRF4-mediated transcriptional activity. In contrast, nonphosphorylatable MRF4 mutants display increased transcriptional activity and are able to advance both myoblast fusion and differentiation. We also show that expression of desmin and alpha-actin, but not muscle creatin kinase, decreased at late stages of muscle differentiation, correlating with the induction of MRF4 and p38 activation. Accordingly, inhibition of p38 during late myogenesis results in the upregulation of both desmin and alpha-actin. We propose that repression of MRF4 activity by p38 phosphorylation may represent a new mechanism for the silencing of specific muscle genes at the terminal stages of muscle differentiation.


Asunto(s)
Silenciador del Gen , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Mioblastos Esqueléticos/metabolismo , Factores Reguladores Miogénicos/química , Factores Reguladores Miogénicos/metabolismo , Actinas/biosíntesis , Secuencia de Aminoácidos , Animales , Proteínas Quinasas Dependientes de Calcio-Calmodulina/metabolismo , Diferenciación Celular , Línea Celular , Desmina/biosíntesis , Regulación hacia Abajo , MAP Quinasa Quinasa 6 , Datos de Secuencia Molecular , Músculo Esquelético/citología , Músculo Esquelético/enzimología , Músculo Esquelético/metabolismo , Mutación , Proteína MioD/metabolismo , Mioblastos Esqueléticos/citología , Mioblastos Esqueléticos/enzimología , Factores Reguladores Miogénicos/antagonistas & inhibidores , Fosforilación , Estructura Terciaria de Proteína , Serina/metabolismo , Activación Transcripcional , Proteínas Quinasas p38 Activadas por Mitógenos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...