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.
Proc Natl Acad Sci U S A ; 113(37): E5425-33, 2016 09 13.
Artículo en Inglés | MEDLINE | ID: mdl-27582469

RESUMEN

Testicular tumors, the most common cancer in young men, arise from abnormalities in germ cells during fetal development. Unconventional inheritance for testicular germ cell tumor (TGCT) risk both in humans and mice implicates epigenetic mechanisms. Apolipoprotein B mRNA-editing enzyme complex 1 (APOBEC1) cytidine deaminase and Deadend-1, which are involved in C-to-U RNA editing and microRNA-dependent mRNA silencing, respectively, are potent epigenetic modifiers of TGCT susceptibility in the genetically predisposed 129/Sv inbred mouse strain. Here, we show that partial loss of either APOBEC1 complementation factor (A1CF), the RNA-binding cofactor of APOBEC1 in RNA editing, or Argonaute 2 (AGO2), a key factor in the biogenesis of certain noncoding RNAs, modulates risk for TGCTs and testicular abnormalities in both parent-of-origin and conventional genetic manners. In addition, non-Mendelian inheritance was found among progeny of A1cf and Ago2 mutant intercrosses but not in backcrosses and without fetal loss. Together these findings suggest nonrandom union of gametes rather than meiotic drive or preferential lethality. Finally, this survey also suggested that A1CF contributes to long-term reproductive performance. These results directly implicate the RNA-binding proteins A1CF and AGO2 in the epigenetic control of germ-cell fate, urogenital development, and gamete functions.


Asunto(s)
Desaminasas APOBEC-1/genética , Proteínas Argonautas/genética , Neoplasias de Células Germinales y Embrionarias/genética , Proteínas de Unión al ARN/genética , Neoplasias Testiculares/genética , Desaminasas APOBEC-1/metabolismo , Animales , Proteínas Argonautas/metabolismo , Modelos Animales de Enfermedad , Epigénesis Genética/genética , Predisposición Genética a la Enfermedad , Células Germinativas/metabolismo , Células Germinativas/patología , Humanos , Masculino , Meiosis/genética , Ratones , MicroARNs/genética , Neoplasias de Células Germinales y Embrionarias/patología , Edición de ARN/genética , Proteínas de Unión al ARN/metabolismo , Neoplasias Testiculares/patología
2.
Neuropharmacology ; 73: 31-40, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23688924

RESUMEN

Repeated cocaine exposure induces epigenetic factors such as DNA methyl-binding proteins, indicating that resulting changes in gene expression are mediated by alterations in brain DNA methylation. While the activity of protein phosphatase type-1 (PP1) is involved in cocaine effects and in brain plasticity, the expression of the PP1Cß catalytic subunit gene was identified here as modulated by cocaine. Its expression was induced together with that of PP1Cγ in the brain of Methyl-CpG Binding Protein-2 (Mecp2) mutant mice, whereas PP1Cα expression was not affected, illustrating a different regulation of PP1C isoforms. Repeated cocaine administration was found to increase DNA methylation at the PP1Cß gene together with its binding to Mecp2 in rat caudate putamen, establishing a link between two genes involved in cocaine-related effects and in learning and memory processes. Cocaine also increased DNMT3 expression, resulting in PP1Cß repression that did not occur in the presence of DNMT inhibitor. Cocaine-induced PP1Cß repression was observed in several brain structures, as evaluated by RT-qPCR, immunohistochemistry and Western blot, but did not occur after a single cocaine injection. Our data demonstrate that PP1Cß is a direct MeCP2-target gene in vivo. They suggest that its repression may participate to behavioral adaptations triggered by the drug.


Asunto(s)
Núcleo Caudado/efectos de los fármacos , Cocaína/farmacología , Metilación de ADN/efectos de los fármacos , Proteínas de Unión al ADN/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , Proteína Fosfatasa 1/biosíntesis , Putamen/metabolismo , Animales , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Núcleo Caudado/metabolismo , ADN (Citosina-5-)-Metiltransferasas/biosíntesis , ADN Metiltransferasa 3A , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Regulación de la Expresión Génica/genética , Masculino , Mutación , Subunidades de Proteína/biosíntesis , Putamen/efectos de los fármacos , Ratas
3.
J Psychopharmacol ; 25(2): 222-9, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19939859

RESUMEN

Injection of the histone deacetylases inhibitor trichostatin A to rats has been shown to decrease the reinforcing properties of cocaine. In the present study, we investigated alterations in gene expression patterns in the anterior cingulate cortex, caudate-putamen and nucleus accumbens of rats self-administering cocaine and treated with trichostatin A. As recent studies highlighted the importance of chromatin remodelling in the regulation of gene transcription in neurons, we studied the expression of Mecp2 and of several histone deacetylases. Cocaine self-administration was accompanied by an increased synthesis of Mecp2, HDAC2 and HDAC11 and by a decreased nuclear localization of HDAC5 and of the phospho-form of HDAC5, suggesting a nuclear export of this protein in response to the drug. The latter mechanism was further addressed by the demonstration of an enhanced expression of MEF2C transcription factor. Among the genes we examined, treatment with trichostatin A before each cocaine self-administration session was found to mostly affect Mecp2 and HDAC11 expression. A correlation was found between the modification of Mecp2 and MEF2C gene expression and the reinforcing property of cocaine. The two factors known to regulate gene transcription are likely to play a role in the neurobiological mechanism underlying a decrease in the reinforcing properties of cocaine.


Asunto(s)
Ensamble y Desensamble de Cromatina/efectos de los fármacos , Cocaína/farmacología , Inhibidores de Histona Desacetilasas/farmacología , Ácidos Hidroxámicos/farmacología , Animales , Núcleo Celular/metabolismo , Cocaína/administración & dosificación , Condicionamiento Operante/efectos de los fármacos , Cuerpo Estriado/efectos de los fármacos , Cuerpo Estriado/enzimología , Regulación de la Expresión Génica/efectos de los fármacos , Giro del Cíngulo/efectos de los fármacos , Giro del Cíngulo/enzimología , Histona Desacetilasas/biosíntesis , Factores de Transcripción MEF2 , Masculino , Proteína 2 de Unión a Metil-CpG/biosíntesis , Factores Reguladores Miogénicos/biosíntesis , Núcleo Accumbens/efectos de los fármacos , Núcleo Accumbens/enzimología , Ratas , Ratas Wistar , Autoadministración , Factores de Transcripción/biosíntesis
4.
Neurobiol Dis ; 38(3): 414-24, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20211261

RESUMEN

Rett syndrome and its "early-onset seizure" variant are severe neurodevelopmental disorders associated with mutations within the MECP2 and the CDKL5 genes. Antidepressants and drugs of abuse induce the expression of the epigenetic factor MeCP2, thereby influencing chromatin remodeling. We show that increased MeCP2 levels resulted in the repression of Cdkl5 in rat brain structures in response to cocaine, as well as in cells exposed to serotonin, or overexpressing MeCP2. In contrast, Cdkl5 was induced by siRNA-mediated knockdown of Mecp2 and by DNA-methyltransferase inhibitors, demonstrating its regulation by MeCP2 and by DNA methylation. Cdkl5 gene methylation and its methylation-dependent binding to MeCP2 were increased in the striatum of cocaine-treated rats. Our data demonstrate that Cdkl5 is a MeCP2-repressed target gene providing a link between genes the mutation of which generates overlapping symptoms. They highlight DNA methylation changes as a potential mechanism participating in the long-term plasticity triggered by pharmacological agents.


Asunto(s)
Encéfalo/metabolismo , Proteína 2 de Unión a Metil-CpG/genética , Proteína 2 de Unión a Metil-CpG/metabolismo , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Animales , Encéfalo/efectos de los fármacos , Cocaína/farmacología , Cuerpo Estriado/efectos de los fármacos , Cuerpo Estriado/metabolismo , Metilación de ADN , ADN-Citosina Metilasas/antagonistas & inhibidores , ADN-Citosina Metilasas/metabolismo , Inhibidores de Captación de Dopamina/farmacología , Técnicas de Silenciamiento del Gen , Masculino , Células PC12 , ARN Interferente Pequeño , Ratas , Ratas Wistar , Análisis de Secuencia de ADN , Serotonina/metabolismo
5.
Mol Pharmacol ; 70(2): 487-92, 2006 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16670375

RESUMEN

Once bound to methylated CpG sites, methyl-CpG-binding protein 2 (MeCP2) is thought to silence transcription of downstream genes by recruiting a histone deacetylase (HDAC). Mutations within the MeCP2 gene have been found to cause Rett syndrome, a disorder of arrested neuronal development. Using immunohistochemistry, we found that Mecp2, as well as the methyl-CpG-binding protein MBD1, were significantly induced in normal adult rat brain after repeated injections of fluoxetine or cocaine for 10 days (one injection per day). Mecp2 was not induced by repeated injections of 1-(2-bis(4-fluorphenyl)-methoxy)-ethyl)-4-(3-phenyl-propyl)piperazine (GBR-12909) or nortriptyline. Together, the data indicate that the serotonergic system is predominantly involved. Using real-time reverse transcription-polymerase chain reaction experiments, MBD1 mRNA and both Mecp2_e1 and Mecp2_e2 transcripts were found to be induced by fluoxetine. Induction of the methylbinding proteins was accompanied with enhanced HDAC2 labeling intensity and mRNA synthesis in response to fluoxetine. In tandem, acetylated forms of histone H3 were found to be decreased. The effect was characterized in three serotonin projection areas, the caudate-putamen, the frontal cortex, and the dentate gyrus subregion of hippocampus. Our data highlight GABAergic neurons as major target cells expressing Mecp2 in response to the serotonin-elevating agents and suggest that serotonin signaling enhances gene silencing in postmitotic neurons.


Asunto(s)
Encéfalo/efectos de los fármacos , Cocaína/farmacología , Proteínas de Unión al ADN/biosíntesis , Epigénesis Genética/efectos de los fármacos , Fluoxetina/farmacología , Proteína 2 de Unión a Metil-CpG/biosíntesis , Animales , Encéfalo/metabolismo , Factor Neurotrófico Derivado del Encéfalo/genética , Proteínas de Unión al ADN/análisis , Proteínas de Unión al ADN/genética , Inmunohistoquímica , Masculino , Proteína 2 de Unión a Metil-CpG/análisis , Proteína 2 de Unión a Metil-CpG/genética , ARN Mensajero/análisis , Ratas , Ratas Wistar , Serotonina/metabolismo , Ácido gamma-Aminobutírico/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA