Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 5 de 5
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Int J Biochem Cell Biol ; 83: 56-64, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-28013147

RESUMEN

Hexim-1 is an inhibitor of RNA polymerase II transcription elongation. Decreased Hexim-1 expression in animal models of chronic diseases such as left ventricular hypertrophy, obesity and cancer triggered significant changes in adaptation and remodeling. The main aim of this study was to evaluate the role of Hexim1 in lipid metabolism focused in the progression of atherosclerosis and steatosis. We used the C57BL6 apolipoprotein E (ApoE null) crossed bred to C57BL6Hexim1 heterozygous mice to obtain ApoE null - Hexim1 heterozygous mice (ApoE-HT). Both ApoE null backgrounds were fed high fat diet for twelve weeks. Then, we evaluated lipid metabolism, atherosclerotic plaque formation and liver steatosis. In order to understand changes in the transcriptome of both backgrounds during the progression of steatosis, we performed Affymetrix mouse 430 2.0 microarray. After 12 weeks of HFD, ApoE null and ApoE-HT showed similar increase of cholesterol and triglycerides in plasma. Plaque composition was altered in ApoE-HT. Additionally, liver triglycerides and steatosis were decreased in ApoE-HT mice. Affymetrix analysis revealed that decreased steatosis might be due to impaired inducible SOCS3 expression in ApoE-HT mice. In conclusion, decreased Hexim-1 expression does not alter cholesterol metabolism in ApoE null background after HFD. However, it promotes stable atherosclerotic plaque and decreased steatosis by promoting the anti-inflammatory TGFß pathway and blocking the expression of the inducible and pro-inflammatory expression of SOCS3 respectively.


Asunto(s)
Apolipoproteínas E/deficiencia , Dieta Aterogénica/efectos adversos , Placa Aterosclerótica/genética , Placa Aterosclerótica/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Animales , Aorta Torácica/metabolismo , Apolipoproteínas E/genética , Colesterol/metabolismo , Dieta Alta en Grasa/efectos adversos , Progresión de la Enfermedad , Hígado Graso/etiología , Hígado Graso/genética , Hígado Graso/metabolismo , Heterocigoto , Metabolismo de los Lípidos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , MicroARNs/genética , MicroARNs/metabolismo , Placa Aterosclerótica/patología , Proteínas de Unión al ARN , Proteína 3 Supresora de la Señalización de Citocinas/genética , Proteína 3 Supresora de la Señalización de Citocinas/metabolismo
2.
Mol Endocrinol ; 30(3): 314-24, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26859361

RESUMEN

Leptin triggers signaling events with significant transcriptional responses that are essential to metabolic processes affecting obesity and glucose disposal. We asked whether hexamethylene bis-acetamide inducible-1 (Hexim1), an inhibitor of RNA II polymerase-dependent transcription elongation, regulates leptin-Janus kinase 2 signaling axis in the hypothalamus. We subjected C57BL6 Hexim1 heterozygous (HT) mice to high-fat diet and when compared with wild type, HT mice were resistant to high-fat diet-induced weight gain and remain insulin sensitive. HT mice exhibited increased leptin-pY(705)Stat3 signaling in the hypothalamus, with normal adipocyte size, increased type I oxidative muscle fiber density, and enhanced glucose transporter 4 expression. We also observed that normal Hexim1 protein level is required to facilitate the expression of CCAAT/enhancer-binding proteins (C/EBPs) required for adipogenesis and inducible suppressor of cytokine signaling 3 (SOCS) expression. Further support on the role of Hexim1 regulating C/EBPs during adipocyte differentiation was shown when HT 3T3L1 fibroblasts failed to undergo adipogenesis. Hexim1 selectively modulates leptin-mediated signal transduction pathways in the hypothalamus, the expression of C/EBPs and peroxisome proliferator-activated receptor-γ (PPAR γ) in skeletal muscle and adipose tissue during the adaptation to metabolic stress. We postulate that Hexim1 might be a novel factor involved in maintaining whole-body energy balance.


Asunto(s)
Glucosa/metabolismo , Leptina/metabolismo , Obesidad/metabolismo , Factores de Transcripción/metabolismo , Células 3T3 , Adipogénesis/efectos de los fármacos , Animales , Núcleo Arqueado del Hipotálamo/metabolismo , Dieta Alta en Grasa , Transportador de Glucosa de Tipo 4/metabolismo , Células HEK293 , Haploinsuficiencia , Heterocigoto , Humanos , Janus Quinasa 2/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Músculo Esquelético/metabolismo , Fosfotirosina/metabolismo , Proteínas de Unión al ARN , Factor de Transcripción STAT3/metabolismo , Transducción de Señal , Aumento de Peso
3.
J Clin Invest ; 122(11): 3873-87, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23023707

RESUMEN

The native capacity of adult skeletal muscles to regenerate is vital to the recovery from physical injuries and dystrophic diseases. Currently, the development of therapeutic interventions has been hindered by the complex regulatory network underlying the process of muscle regeneration. Using a mouse model of skeletal muscle regeneration after injury, we identified hexamethylene bisacetamide inducible 1 (HEXIM1, also referred to as CLP-1), the inhibitory component of the positive transcription elongation factor b (P-TEFb) complex, as a pivotal regulator of skeletal muscle regeneration. Hexim1-haplodeficient muscles exhibited greater mass and preserved function compared with those of WT muscles after injury, as a result of enhanced expansion of satellite cells. Transplanted Hexim1-haplodeficient satellite cells expanded and improved muscle regeneration more effectively than WT satellite cells. Conversely, HEXIM1 overexpression restrained satellite cell proliferation and impeded muscle regeneration. Mechanistically, dissociation of HEXIM1 from P-TEFb and subsequent activation of P-TEFb are required for satellite cell proliferation and the prevention of early myogenic differentiation. These findings suggest a crucial role for the HEXIM1/P-TEFb pathway in the regulation of satellite cell­mediated muscle regeneration and identify HEXIM1 as a potential therapeutic target for degenerative muscular diseases.


Asunto(s)
Diferenciación Celular/fisiología , Proteínas Musculares/metabolismo , Músculo Esquelético/lesiones , Músculo Esquelético/metabolismo , Regeneración/fisiología , Células Satélite del Músculo Esquelético/metabolismo , Factores de Transcripción/metabolismo , Animales , Proliferación Celular , Ratones , Ratones Mutantes , Proteínas Musculares/genética , Factor B de Elongación Transcripcional Positiva/metabolismo , Proteínas de Unión al ARN , Factores de Transcripción/genética
4.
J Biol Chem ; 287(16): 13084-93, 2012 Apr 13.
Artículo en Inglés | MEDLINE | ID: mdl-22308025

RESUMEN

It is well known that the renin-angiotensin system contributes to left ventricular hypertrophy and fibrosis, a major determinant of myocardial stiffness. TGF-ß1 and renin-angiotensin system signaling alters the fibroblast phenotype by promoting its differentiation into morphologically distinct pathological myofibroblasts, which potentiates collagen synthesis and fibrosis and causes enhanced extracellular matrix deposition. However, the atrial natriuretic peptide, which is induced during left ventricular hypertrophy, plays an anti-fibrogenic and anti-hypertrophic role by blocking, among others, the TGF-ß-induced nuclear localization of Smads. It is not clear how the hypertrophic and fibrotic responses are transcriptionally regulated. CLP-1, the mouse homolog of human hexamethylene bis-acetamide inducible-1 (HEXIM-1), regulates the pTEFb activity via direct association with pTEFb causing inhibition of the Cdk9-mediated serine 2 phosphorylation in the carboxyl-terminal domain of RNA polymerase II. It was recently reported that the serine kinase activity of Cdk9 not only targets RNA polymerase II but also the conserved serine residues of the polylinker region in Smad3, suggesting that CLP-1-mediated changes in pTEFb activity may trigger Cdk9-dependent Smad3 signaling that can modulate collagen expression and fibrosis. In this study, we evaluated the role of CLP-1 in vivo in induction of left ventricular hypertrophy in angiotensinogen-overexpressing transgenic mice harboring CLP-1 heterozygosity. We observed that introduction of CLP-1 haplodeficiency in the transgenic α-myosin heavy chain-angiotensinogen mice causes prominent changes in hypertrophic and fibrotic responses accompanied by augmentation of Smad3/Stat3 signaling. Together, our findings underscore the critical role of CLP-1 in remodeling of the genetic response during hypertrophy and fibrosis.


Asunto(s)
Angiotensina II/metabolismo , Cardiomegalia/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Factor de Crecimiento Transformador beta1/metabolismo , Remodelación Ventricular/genética , Angiotensinógeno/genética , Animales , Cardiomegalia/genética , Cardiomegalia/patología , Matriz Extracelular/metabolismo , Matriz Extracelular/patología , Fibroblastos/metabolismo , Fibroblastos/patología , Fibrosis/metabolismo , Fibrosis/patología , Heterocigoto , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Cadenas Pesadas de Miosina/genética , Proteínas de Unión al ARN , Factor de Transcripción STAT3/metabolismo , Transducción de Señal/fisiología , Proteína smad3/metabolismo , Transcripción Genética/fisiología
5.
Dev Dyn ; 230(4): 767-72, 2004 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-15254910

RESUMEN

Transcripts encoding trkC and full-length (catalytic) TrkC receptors were detected in the outflow tract of the chicken heart during early development (stage 17; embryonic day [E] 2.5) before the start of septation. Expression of trkC mRNA persisted through early septation (stage 25, E4.5-E5) but was no longer evident by the end of septation (stage 34, E8). Neurotrophin-3 (NT-3) mRNA was also shown to be present in the outflow tract throughout cardiac development. Quail-chick chimeras were used to confirm that cardiac neural crest cells were not present in the outflow tract at stage 17 (E2.5). Our results show that NT-3 interacts with cells in the outflow tract that are not of neural crest origin. This finding indicates that, in addition to effects on neural crest cells, NT-3 may be important for cardiac development due to its interaction with cells in the outflow tract such as those arising from the secondary heart field.


Asunto(s)
Corazón/embriología , Miocardio/metabolismo , Neurotrofina 3/biosíntesis , Neurotrofina 3/fisiología , Receptor trkC/biosíntesis , Receptor trkC/fisiología , Animales , Catálisis , Pollos , Quimera , Immunoblotting , Inmunohistoquímica , Hibridación in Situ , Cresta Neural/embriología , Codorniz , ARN Mensajero/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factores de Tiempo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA