Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Mol Neurobiol ; 59(3): 1896-1911, 2022 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-35032317

RESUMEN

Cocaine addiction is a complex pathology inducing long-term neuroplastic changes that, in turn, contribute to maladaptive behaviors. This behavioral dysregulation is associated with transcriptional reprogramming in brain reward circuitry, although the mechanisms underlying this modulation remain poorly understood. The endogenous cannabinoid system may play a role in this process in that cannabinoid mechanisms modulate drug reward and contribute to cocaine-induced neural adaptations. In this study, we investigated whether cocaine self-administration induces long-term adaptations, including transcriptional modifications and associated epigenetic processes. We first examined endocannabinoid gene expression in reward-related brain regions of the rat following self-administered (0.33 mg/kg intravenous, FR1, 10 days) cocaine injections. Interestingly, we found increased Cnr1 expression in several structures, including prefrontal cortex, nucleus accumbens, dorsal striatum, hippocampus, habenula, amygdala, lateral hypothalamus, ventral tegmental area, and rostromedial tegmental nucleus, with most pronounced effects in the hippocampus. Endocannabinoid levels, measured by mass spectrometry, were also altered in this structure. Chromatin immunoprecipitation followed by qPCR in the hippocampus revealed that two activating histone marks, H3K4Me3 and H3K27Ac, were enriched at specific endocannabinoid genes following cocaine intake. Targeting CB1 receptors using chromosome conformation capture, we highlighted spatial chromatin re-organization in the hippocampus, as well as in the nucleus accumbens, suggesting that destabilization of the chromatin may contribute to neuronal responses to cocaine. Overall, our results highlight a key role for the hippocampus in cocaine-induced plasticity and broaden the understanding of neuronal alterations associated with endocannabinoid signaling. The latter suggests that epigenetic modifications contribute to maladaptive behaviors associated with chronic drug use.


Asunto(s)
Cannabinoides , Cocaína , Animales , Cannabinoides/farmacología , Cocaína/farmacología , Hipocampo/metabolismo , Masculino , Núcleo Accumbens/metabolismo , Ratas , Receptores de Cannabinoides/metabolismo , Autoadministración
2.
Genes (Basel) ; 12(8)2021 07 31.
Artículo en Inglés | MEDLINE | ID: mdl-34440369

RESUMEN

The circadian system interacts with the mesocorticolimbic reward system to modulate reward and memory in a time-of-day dependent manner. The circadian discrimination of reward, however, remains difficult to address between natural reinforcers and drugs of abuse. Circadian rhythms control cocaine sensitization and conversely cocaine causes long-term alteration in circadian periodicity in part through the serotonergic neurotransmission. Since neural circuits activated by cocaine and natural reinforcers do not completely overlap, we compared the effect of cocaine with that of sucrose, a strong reinforcer in rodents, by using passive chronic administration. The expression of fifteen genes playing a major role in DNA methylation (Dnmts, Tets), circadian rhythms (Clock, Bmal1, Per1/2, Cry1/2, Rev-Erbß, Dbp1), appetite, and satiety (Orexin, Npy) was analyzed in dopamine projection areas like the prefrontal cortex, the caudate putamen, and the hypothalamus interconnected with the reward system. The corresponding proteins of two genes (Orexin, Per2) were examined by IHC. For many factors controlling biological and cognitive functions, striking opposite responses were found between the two reinforcers, notably for genes controlling DNA methylation/demethylation processes and in global DNA methylation involved in chromatin remodeling. The data are consistent with a repression of critical core-clock genes by cocaine, suggesting that, consequently, both agents differentially modulate day/night cycles. Whether observed cocaine and sucrose-induced changes in DNA methylation in a time dependent manner are long lasting or contribute to the establishment of addiction requires further neuroepigenetic investigation. Understanding the mechanisms dissociating drugs of abuse from natural reinforcers remains a prerequisite for the design of selective therapeutic tools for compulsive behaviors.


Asunto(s)
Encéfalo/efectos de los fármacos , Proteínas CLOCK/genética , Ensamble y Desensamble de Cromatina , Cocaína/farmacología , Metilación de ADN , Sacarosa/farmacología , Animales , Peso Corporal , Encéfalo/metabolismo , Ritmo Circadiano/genética , Masculino , Ratas , Ratas Wistar , Sacarosa/administración & dosificación
3.
Genes (Basel) ; 12(8)2021 08 19.
Artículo en Inglés | MEDLINE | ID: mdl-34440437

RESUMEN

Based on studies describing an increased prevalence of addictive behaviours in several rare sleep disorders and shift workers, a relationship between circadian rhythms and addiction has been hinted for more than a decade. Although circadian rhythm alterations and molecular mechanisms associated with neuropsychiatric conditions are an area of active investigation, success is limited so far, and further investigations are required. Thus, even though compelling evidence connects the circadian clock to addictive behaviour and vice-versa, yet the functional mechanism behind this interaction remains largely unknown. At the molecular level, multiple mechanisms have been proposed to link the circadian timing system to addiction. The molecular mechanism of the circadian clock consists of a transcriptional/translational feedback system, with several regulatory loops, that are also intricately regulated at the epigenetic level. Interestingly, the epigenetic landscape shows profound changes in the addictive brain, with significant alterations in histone modification, DNA methylation, and small regulatory RNAs. The combination of these two observations raises the possibility that epigenetic regulation is a common plot linking the circadian clocks with addiction, though very little evidence has been reported to date. This review provides an elaborate overview of the circadian system and its involvement in addiction, and we hypothesise a possible connection at the epigenetic level that could further link them. Therefore, we think this review may further improve our understanding of the etiology or/and pathology of psychiatric disorders related to drug addiction.


Asunto(s)
Relojes Circadianos/genética , Ritmo Circadiano/genética , Epigénesis Genética/genética , Trastornos Relacionados con Sustancias/genética , Metilación de ADN/genética , Histonas/genética , Humanos , Procesamiento Proteico-Postraduccional/genética , Trastornos Relacionados con Sustancias/metabolismo , Trastornos Relacionados con Sustancias/patología
4.
Mol Neurobiol ; 58(6): 2590-2607, 2021 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-33475949

RESUMEN

In zebrafish, nicotine is known to regulate sensitivity to psychostimulants via epigenetic mechanisms. Little however is known about the regulation of addictive-like behavior by DNA methylation processes. To evaluate the influence of DNA methylation on nicotine-induced conditioned place preference (CPP), zebrafish were exposed to methyl supplementation through oral L-methionine (Met) administration. Met was found to reduce dramatically nicotine-induced CPP as well as behaviors associated with drug reward. The reduction was associated with the upregulation of DNA methyltransferases (DNMT1 and 3) as well as with the downregulation of methyl-cytosine dioxygenase-1 (TET1) and of nicotinic receptor subunits. Met also increased the expression of histone methyltransferases in nicotine-induced CPP groups. It reversed the nicotine-induced reduction in the methylation at α7 and NMDAR1 gene promoters. Treatment with the DNMT inhibitor 5-aza-2'-deoxycytidine (AZA) was found to reverse the effects of Met in structures of the reward pathway. Interestingly, Met did not modify the amount of the phospho-form of CREB (pCREB), a key factor establishing nicotine conditioning, whereas AZA increased pCREB levels. Our data suggest that nicotine-seeking behavior is partially dependent on DNA methylation occurring probably at specific gene loci, such as α7 and NMDAR1 receptor gene promoters. Overall, they suggest that Met should be considered as a potential therapeutic drug to treat nicotine addiction.


Asunto(s)
Conducta Animal/fisiología , Conducta de Elección , Suplementos Dietéticos , Metionina/farmacología , Nicotina/farmacología , Pez Cebra/fisiología , 5-Metilcitosina/metabolismo , Animales , Azacitidina/farmacología , Conducta Animal/efectos de los fármacos , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Condicionamiento Clásico , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Metilación de ADN/efectos de los fármacos , Metilación de ADN/genética , Epigénesis Genética/efectos de los fármacos , Fosforilación/efectos de los fármacos , Subunidades de Proteína/genética , Subunidades de Proteína/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptores de N-Metil-D-Aspartato/genética , Receptores de N-Metil-D-Aspartato/metabolismo , Receptores Nicotínicos/genética , Receptores Nicotínicos/metabolismo , Recompensa , Pez Cebra/genética , Proteínas de Pez Cebra/genética , Proteínas de Pez Cebra/metabolismo
5.
Neurosci Lett ; 764: 135603, 2021 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-33387661

RESUMEN

Cocaine addiction is a serious health issue in Western countries. Despite the regular increase in cocaine consumption across the population, there is no specific treatment for cocaine addiction. Critical roles for glutamate neurotransmission in the rewarding effects of psychostimulants as well as relapse have been suggested and accumulating evidence indicates that targeting mGlu group III receptors could represent a promising strategy to develop therapeutic compounds to treat addiction. In this context, the aim of our study was to examine the effect of LSP2-9166, a mGlu4/mGlu7 receptor orthosteric agonist, on the motivation for cocaine intake. We used an intravenous self-administration paradigm in male Wistar rats as a reliable model of voluntary drug intake. We first evaluated the direct impact of cocaine on Grm4 and Grm7 gene expression. Voluntary cocaine intake under a fixed ratio schedule of injections induced an increase of both mGlu4 and mGlu7 receptor transcripts in nucleus accumbens and hippocampus. We then evaluated the ability of LSP2-9166 to affect cocaine self-administration under a progressive ratio schedule of reinforcement. We found that this compound inhibits the motivation to obtain the drug, although it induced a hypolocomotor effect which could biais motivation index. Our findings demonstrate that mGlu group III receptors represent new targets for decreasing motivation to self-administer cocaine.


Asunto(s)
Aminobutiratos/farmacología , Trastornos Relacionados con Cocaína/tratamiento farmacológico , Motivación/efectos de los fármacos , Receptores de Glutamato Metabotrópico/agonistas , Administración Intravenosa , Aminobutiratos/uso terapéutico , Animales , Cocaína/administración & dosificación , Cocaína/efectos adversos , Trastornos Relacionados con Cocaína/psicología , Modelos Animales de Enfermedad , Ácido Glutámico/metabolismo , Humanos , Masculino , Ratas , Ratas Wistar , Receptores de Glutamato Metabotrópico/metabolismo , Refuerzo en Psicología , Autoadministración , Transmisión Sináptica/efectos de los fármacos
6.
Mol Neurobiol ; 56(8): 5315-5331, 2019 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-30603957

RESUMEN

Inhibitors of DNA methylation and orexin type-1 receptor antagonists modulate the neurobiological effects driving drugs of abuse and natural reinforcers by activating common brain structures of the mesolimbic reward system. In this study, we applied a self-administration paradigm to assess the involvement of factors regulating DNA methylation processes and satiety or appetite signals. These factors include Dnmts and Tets, miR-212/132, orexins, and orx-R1 genes. The study focused on dopamine projection areas such as the prefrontal cortex (PFCx) and caudate putamen (CPu) and in the hypothalamus (HP) that is interconnected with the reward system. Striking changes were observed in response to both reinforcers, but differed depending on contingent and non-contingent delivery. Expression also differed in the PFCx and the CPu. Cocaine and food induced opposite effects on Dnmt3a expression in both brain structures, whereas they repressed both miRs to a different extent, without affecting their primary transcript in the CPu. Unexpectedly, orexin mRNAs were found in the CPu, suggesting a transport from their transcription site in the HP. The orexin receptor1 gene was found to be induced by cocaine in the PFCx, consistent with a regulation by DNA methylation. Global levels of 5-methylcytosines in the PFCx were not significantly altered by cocaine, suggesting that it is rather their distribution that contributes to long-lasting behaviors. Together, our data demonstrate that DNA methylation regulating factors are differentially altered by cocaine and food. At the molecular level, they support the idea that neural circuits activated by both reinforcers do not completely overlap.


Asunto(s)
Encéfalo/metabolismo , Cocaína/administración & dosificación , Metilación de ADN/genética , Alimentos , Orexinas/metabolismo , Autoadministración , Animales , Condicionamiento Operante , ADN (Citosina-5-)-Metiltransferasas/genética , ADN (Citosina-5-)-Metiltransferasas/metabolismo , ADN Metiltransferasa 3A , Conducta Alimentaria , Regulación de la Expresión Génica , Hipotálamo/metabolismo , Masculino , MicroARNs/genética , MicroARNs/metabolismo , Receptores de Orexina/genética , Receptores de Orexina/metabolismo , Péptidos/metabolismo , Corteza Prefrontal/metabolismo , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/metabolismo , Putamen/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ratas Wistar , ADN Metiltransferasa 3B
7.
Med Sci (Paris) ; 31(4): 439-46, 2015 Apr.
Artículo en Francés | MEDLINE | ID: mdl-25958763

RESUMEN

Chronic drug exposure alters gene expression in the brain, which is believed to underlie compulsive drug seeking and drug taking behavior. Recent evidence shows that drug-induced long-term neuroadaptations in the brain are mediated in part by epigenetic mechanisms. By remodeling chromatin, this type of regulation contributes to drug-induced synaptic plasticity that translates into behavioral modifications. How drug-induced alterations in DNA methylation regulate gene expression is reviewed here, with a focus on MeCP2, a protein binding methylated DNA. The importance of histone modifications, especially acetylation is also discussed, with an emphasis on the effects of inhibitors of histone deacetylases on drug-induced behavioral changes. The precise identification of the epigenetic mechanisms that are under the control of drugs of abuse may help to uncover novel targets for the treatment of drug seeking and relapse.


Asunto(s)
Epigénesis Genética , Histona Acetiltransferasas/metabolismo , Histonas/metabolismo , Proteína 2 de Unión a Metil-CpG/fisiología , Trastornos Relacionados con Sustancias/genética , Acetilación , Metilación de ADN , Humanos , Neuronas/metabolismo , Trastornos Relacionados con Sustancias/metabolismo
8.
Addict Biol ; 20(1): 120-31, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23980619

RESUMEN

We previously showed that maternal deprivation predisposes male rats to anxiety, accompanied with an increase in their opiate consumption. In the present report, we searched for brain epigenetic mechanisms that possibly underlie this increase. For that, we examined the expression of the methyl-CpG-binding protein MeCP2 and of the histone deacetylases HDAC2 and HDAC3, as well as the acetylation status of histone H3 and H4 in mesolimbic structures of adult maternally deprived rats, using immunohistochemistry and Western blot analysis. A long-lasting increase in MeCP2 expression was found throughout the striatum of deprived rats. Enhanced HDAC2 expression and increased nuclear HDAC activity in the nucleus accumbens of deprived rats were associated with lower acetylation levels of histone H3 and H4. Treatment for 3 weeks with the HDAC inhibitor sodium valproate abolished HDAC activation together with the decrease in the acetylation levels of histone H4, and was accompanied with normalized oral morphine consumption. The data indicate that epigenetic mechanisms induced by early adverse environment memorize life experience to trigger greater opiate vulnerability during adult life. They suggest that sodium valproate may lessen vulnerability to opiate intake, particularly in subgroups of individuals subjected to adverse postnatal environments.


Asunto(s)
Histona Desacetilasa 2/genética , Histona Desacetilasas/genética , Privación Materna , Proteína 2 de Unión a Metil-CpG/genética , Trastornos Relacionados con Opioides/genética , Acetilación , Analgésicos Opioides , Animales , Conducta Animal , Epigénesis Genética , Interacción Gen-Ambiente , Inhibidores de Histona Desacetilasas/farmacología , Histonas/metabolismo , Morfina , Núcleo Accumbens/metabolismo , Ratas , Ácido Valproico/farmacología
9.
Int J Neuropsychopharmacol ; 17(12): 2031-44, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24936739

RESUMEN

Cocaine exposure induces changes in the expression of numerous genes, in part through epigenetic modifications. We have initially shown that cocaine increases the expression of the chromatin remodeling protein methyl-CpG binding protein 2 (MeCP2) and characterized the protein phosphatase-1Cß (PP1Cß) gene, as repressed by passive i.p. cocaine injections through a Mecp2-mediated mechanism involving de novo DNA methylation. Both proteins being involved in learning and memory processes, we investigated whether voluntary cocaine administration would similarly affect their expression using an operant self-administration paradigm. Passive and voluntary i.v. cocaine intake was found to induce Mecp2 and to repress PP1Cß in the prefrontal cortex and the caudate putamen. This observation is consistent with the role of Mecp2 acting as a transcriptional repressor of PP1Cß and shows that passive intake was sufficient to alter their expression. Surprisingly, striking differences were observed under the same conditions in food-restricted rats tested for food pellet delivery. In the prefrontal cortex and throughout the striatum, both proteins were induced by food operant conditioning, but remained unaffected by passive food delivery. Although cocaine and food activate a common reward circuit, changes observed in the expression of other genes such as reelin and GAD67 provide new insights into molecular mechanisms differentiating neuroadaptations triggered by each reinforcer. The identification of hitherto unknown genes differentially regulated by drugs of abuse and a natural reinforcer should improve our understanding of how two rewarding stimuli differ in their ability to drive behavior.


Asunto(s)
Cocaína/administración & dosificación , Inhibidores de Captación de Dopamina/administración & dosificación , Ingestión de Alimentos/fisiología , Proteína 2 de Unión a Metil-CpG/metabolismo , Proteína Fosfatasa 1/metabolismo , Animales , Encéfalo/efectos de los fármacos , Encéfalo/fisiología , Moléculas de Adhesión Celular Neuronal/metabolismo , Condicionamiento Operante/efectos de los fármacos , Condicionamiento Operante/fisiología , Proteínas de la Matriz Extracelular/metabolismo , Privación de Alimentos/fisiología , Expresión Génica , Glutamato Descarboxilasa/metabolismo , Masculino , Memoria/efectos de los fármacos , Memoria/fisiología , Proteínas del Tejido Nervioso/metabolismo , Distribución Aleatoria , Ratas Wistar , Proteína Reelina , Recompensa , Autoadministración , Serina Endopeptidasas/metabolismo , Volición
10.
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
11.
Int J Neuropsychopharmacol ; 16(7): 1587-97, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23375146

RESUMEN

The C-type natriuretic peptide (CNP) exerts its action via stimulation of the cyclic GMP (cGMP) signalling pathway, which includes the activation of cGMP-dependent protein kinases. The pathway can also be activated by inhibitors of phosphodiesterases (PDE) that hydrolyse cGMP. The present report shows that activation of the cGMP pathway by CNP, by bromo-cGMP, a cell-permeant cGMP analogue, or by the PDE inhibitor zaprinast dose dependently reduces intravenous cocaine self-administration by rats. The effect was found when the compounds were injected in situ into the prefrontal cortex, but not when they were injected into the nucleus accumbens. A decrease in the number of cocaine infusions performed by rats was obtained under the fixed ratio-1 schedule of reinforcement as well as under a progressive ratio schedule, which evaluates the motivation of the animals for the drug. Decrease in cocaine self-administration was accompanied with reduced expression of the epigenetic markers methyl-CpG-binding protein 2 (MeCP2) and histone deacetylase 2 (HDAC2) in dopaminergic projection areas. An increase in the acetylation level of histone H3, but not of histone H4, was also noticed. Since MeCP2 and HDAC2 are known to modulate dynamic functions in the adult brain, such as synaptic plasticity, our results showing that activation of the cGMP signal transduction pathway decreased both cocaine intake and expression of the epigenetic markers strongly suggest that the MeCP2/HDAC2 complex is involved in the analysis of the reinforcing properties of cocaine in the prefrontal cortex.


Asunto(s)
Encéfalo/efectos de los fármacos , Cocaína/administración & dosificación , Inhibidores de Captación de Dopamina/administración & dosificación , Histona Desacetilasa 2/metabolismo , Proteína 2 de Unión a Metil-CpG/metabolismo , Péptido Natriurético Tipo-C/farmacología , Inhibidores de Fosfodiesterasa/farmacología , Análisis de Varianza , Animales , Encéfalo/enzimología , Condicionamiento Operante/efectos de los fármacos , GMP Cíclico/análogos & derivados , GMP Cíclico/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Masculino , Proteínas Serina-Treonina Quinasas/metabolismo , Purinonas/farmacología , Ratas , Ratas Wistar , Esquema de Refuerzo , Autoadministración
12.
Brain Behav ; 2(6): 732-40, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23170236

RESUMEN

Nitric oxide (NO) and the C-type natriuretic peptide (CNP) exert their action via stimulation of the cyclic GMP (cGMP)-signaling pathway, which includes the activation of cGMP-dependent protein kinases (PKG). The present report shows that the activation of PKG by local application of 8-bromo-cGMP in the caudate-putamen reduced the expression of the epigenetic markers, methyl-CpG-binding protein 2 (MeCP2) and histone deacetylase 2 (HDAC2), in dopaminergic projection areas of cocaine-treated rats. An effect of lesser amplitude was observed when rats were not injected with cocaine. We also studied the effect of PKG overexpression by injecting a plasmid vector containing the human PKG-Iα cDNA in either the caudate-putamen or the ventral tegmental area. Injection in the caudate-putamen reduced the epigenetic parameters with higher amplitude than the cGMP analog. The effect was abolished by the injection of a selective PKG inhibitor, confirming that it was due to PKG-dependent phosphorylation. As MeCP2 and HDAC2 modulate dynamic functions in the adult brain such as memory formation and synaptic plasticity, the downregulation of expression by PKG suggests that the cGMP pathway affects cognitive processes through a mechanism that comprises the MeCP2/HDAC2 complex and the subsequent control of gene silencing.

13.
Synapse ; 66(1): 61-70, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21954104

RESUMEN

Distinct forms of MEF2 transcription factor act as positive or negative regulators of dendritic spine formation, with MEF2C playing a key regulator role in synapse plasticity. We report here that acute cocaine treatment of rats induced the expression of MEF2C in the striatum through a recently discovered transduction pathway. Repeated injections were found to induce MEF2C to a lesser extent. The mechanism by which MEF2C was induced involves the subsequent activation of the salt-inducible kinase SIK1 and the phosphorylation of HDAC5, a member of the class IIa of HDACs. Cocaine activated SIK1 by phosphorylation on Thr-182 residue, which was accompanied by the nuclear import of the kinase. In the nuclear compartment, SIK1 then phosphorylated HDAC5 causing the shuttling of its phospho-form from the nucleus to the cytoplasm of striatal cells. Activation of SIK1 by cocaine was further validated by the phosphorylation of TORC1/3, which was followed by the shuttling of TORC proteins from the nucleus to the cytoplasm. Activation of MEF2C was assessed by measuring the expression of the MEF2C gene itself, since the gene is known to be under the control of its own product. Since MEF2C plays a key role in memory/learning processes, activation of this pathway by cocaine is probably involved in plasticity mechanisms whereby the drug establishes its long-term effects such as drug dependence.


Asunto(s)
Cocaína/farmacología , Cuerpo Estriado/efectos de los fármacos , Inhibidores de Captación de Dopamina/farmacología , Histona Desacetilasas/metabolismo , Factores Reguladores Miogénicos/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Animales , Cuerpo Estriado/metabolismo , Activación Enzimática/efectos de los fármacos , Immunoblotting , Inmunohistoquímica , Factores de Transcripción MEF2 , Masculino , Fosforilación , Transporte de Proteínas/efectos de los fármacos , Ratas , Ratas Wistar
14.
Curr Neuropharmacol ; 9(1): 21-5, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21886555

RESUMEN

Drug addiction is a chronic brain disease characterized by a persistent risk of relapse, even after a long period of abstinence. A current hypothesis states that relapse results from lasting neuroadaptations that are induced in response to repeated drug administration. The adaptations require gene expression, some of which being under the control of stable epigenetic regulations. We have previously demonstrated that pretreatment with histone deacetylase (HDAC) inhibitors reduces the cocaine reinforcing properties as well as the motivation of rats for cocaine. We show here that the same HDAC inhibitors, trichostatin A and phenylbutyrate, significantly reduced the cocaine-seeking behavior induced by the combination of a cocaine injection together with the exposure to a light cue previously associated with cocaine taking. Reinstatement of drug-seeking behavior was carried out after a 3-week withdrawal period, which came after ten daily sessions of cocaine intravenous self-administration. Our results suggest that pharmacological treatment aimed at modulating epigenetic regulation, and particularly treatment that would inhibit HDAC activity, could reduce the risk of relapse, a major drawback in the treatment of drug addiction.

15.
Front Psychiatry ; 1: 153, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21423460

RESUMEN

Recent evidence suggests that epigenetic mechanisms play a role in psychiatric diseases. In this study, we considered rats with neonatal ventral hippocampal lesions (NVHL) that are currently used for modeling neurodevelopmental aspects of schizophrenia. Contribution of epigenetic regulation to the effects of the lesion was investigated, using a histone deacetylase (HDAC) inhibitor. Lesioned or sham-operated rats were treated with the general HDAC inhibitor phenylbutyrate, which was injected daily from the day after surgery until adulthood. Changes in the volume of the lesion were monitored by magnetic resonance imaging (MRI). Anxiety was analyzed in the Plus Maze Test. Hypersensitivity of the dopaminergic system was evaluated by measuring the locomotor response to apomorphine. An associative conditioning test rewarded with food was used to evaluate learning abilities. The volume of the lesions expanded long after surgery, independently of the treatment, as assessed by MRI. Removal of the ventral hippocampus reduced anxiety, and this remained unchanged when animals were treated with phenylbutyrate. In contrast, NVHL rats' hypersensitivity to apomorphine and deterioration of the associative learning were reduced by the treatment. Global HDAC activity, which was increased in the prefrontal cortex of lesioned non-treated rats, was found to be reversed by HDAC inhibition. The study provides evidence that chromatin remodeling may be useful for limiting behavioral consequences due to lesioning of the ventral hippocampus at an early age. This represents a novel approach for treating disorders resulting from insults occurring during brain development.

16.
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
17.
J Neurochem ; 116(4): 636-45, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21166804

RESUMEN

Epigenetic mechanisms have recently been shown to be involved in the long-term effects of drugs of abuse. A well described epigenetic mechanism modulating transcriptional activity consists in the binding to DNA of methyl-CpG binding proteins, such as MeCP2, recruiting histone deacetylases (HDACs). Nicotine causes long-term changes in the brain, but little is known concerning the mechanisms involved in nicotine-preference. Using a nicotine-conditioned place preference protocol, we demonstrate here that the histone deacetylase inhibitor phenylbutyrate was able to dramatically reduce the preference for nicotine, without altering the aversive properties of the drug. We measured immunohistochemically the acetylation of lysine-9 of histone H3, and the expression of phosphorylated cAMP-response element-binding protein, HDAC2 and methyl-CpG-binding protein 2 in the striatum and prefrontal cortex of rats displaying nicotine-preference or aversion and treated with phenylbutyrate. We show that, at the dose administered, the inhibitor was effective in inhibiting HDAC activity. The data suggest that phosphorylated cAMP-response element-binding protein participates in the establishment of conditioned place preference, but not in the reduction of nicotine-preference in response to phenylbutyrate. Moreover, striatal expression of HDAC2 in response to phenylbutyrate mirrored the behavioral effects of the inhibitor, suggesting that HDAC2 is involved in promoting synaptic plasticity underlying the preference for nicotine.


Asunto(s)
Reacción de Prevención/fisiología , Condicionamiento Psicológico/fisiología , Histona Desacetilasa 2/antagonistas & inhibidores , Histona Desacetilasa 2/biosíntesis , Inhibidores de Histona Desacetilasas/farmacología , Histona Desacetilasas/metabolismo , Nicotina/farmacología , Animales , Reacción de Prevención/efectos de los fármacos , Encéfalo/efectos de los fármacos , Encéfalo/enzimología , Conducta de Elección/efectos de los fármacos , Conducta de Elección/fisiología , Condicionamiento Psicológico/efectos de los fármacos , Cuerpo Estriado/enzimología , Histona Desacetilasas/biosíntesis , Masculino , Fenilbutiratos/farmacología , Ratas , Ratas Sprague-Dawley
18.
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
19.
J Neurochem ; 113(1): 236-47, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-20132486

RESUMEN

Injection of the histone deacetylase inhibitor trichostatin A (TsA) to rats has been shown to decrease their motivation to self-administer cocaine. In the present study, we investigated alterations in gene expression patterns in the anterior cingulate cortex and nucleus accumbens of rats self-administering cocaine and treated with TsA. Using oligonucleotide microarrays, we identified 722 probe sets in the cortex and 136 probe sets in the nucleus accumbens that were differentially expressed between vehicle and TsA-treated rats that self-administered cocaine. Microarray data were validated by real-time PCR for seven genes. Using immunohistochemistry, we further investigated the expression of Lis1 and reelin genes, because (i) they were similarly regulated by TsA at the mRNA level; (ii) they belong to the same signal transduction pathway; (iii) mutations within both genes cause lissencephaly. Cocaine self-injection was sufficient to activate the two genes at both the mRNA and protein levels. TsA treatment was found to up-regulate both Lis1 and reelin protein expression in the cortex and to down-regulate it in the nucleus accumbens of rats self-administering cocaine. The data suggest that the two proteins contribute to establish neurobiological mechanisms underlying brain plasticity whereby TsA lowers the motivation for cocaine.


Asunto(s)
Moléculas de Adhesión Celular Neuronal/metabolismo , Lisencefalias Clásicas y Heterotopias Subcorticales en Banda/metabolismo , Cocaína/administración & dosificación , Inhibidores de Captación de Dopamina/administración & dosificación , Proteínas de la Matriz Extracelular/metabolismo , Regulación de la Expresión Génica/fisiología , Histona Desacetilasas/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Serina Endopeptidasas/metabolismo , Animales , Conducta Animal/efectos de los fármacos , Encéfalo/anatomía & histología , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Moléculas de Adhesión Celular Neuronal/genética , Lisencefalias Clásicas y Heterotopias Subcorticales en Banda/genética , Biología Computacional/métodos , Condicionamiento Operante/efectos de los fármacos , Proteínas de la Matriz Extracelular/genética , Perfilación de la Expresión Génica/métodos , Regulación de la Expresión Génica/efectos de los fármacos , Inhibidores de Histona Desacetilasas/farmacología , Ácidos Hidroxámicos/farmacología , Masculino , Microdiálisis/métodos , Proteínas del Tejido Nervioso/genética , Análisis de Secuencia por Matrices de Oligonucleótidos/métodos , Ratas , Ratas Wistar , Proteína Reelina , Autoadministración/métodos , Serina Endopeptidasas/genética , Factores de Tiempo
20.
Ann N Y Acad Sci ; 1139: 27-33, 2008 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-18991845

RESUMEN

Alcohol administration is known to alter several brain functions and behaviors in humans and in laboratory animals. One of the targets of ethanol is the mesocorticolimbic dopaminergic reward pathway. We used the "alcohol deprivation effect" test as a rat model of alcohol craving and relapse. The effect is characterized by increased alcohol intake and preference after several weeks of voluntary alcohol consumption followed by a withdrawal phase. The alcohol deprivation effect was found to be considerably reduced by the injection in dopaminergic brain structures of the neuropeptide CNP. This peptide is the most abundant natriuretic peptide in the brain, and signals via an intracellular rise in cyclic GMP. The effect of CNP was observed whether the peptide was injected in situ into the ventral tegmental area or into the prefrontal cortex. It was partially reversed by the injection in the same structures of KT5823, a selective inhibitor of the cGMP-dependent protein kinase. The results indicate that changes of cyclic GMP levels in dopaminergic rat brain areas participate in the neurobiological mechanisms underlying alcohol craving after withdrawal and/or alcohol dependence.


Asunto(s)
Consumo de Bebidas Alcohólicas , Encéfalo/efectos de los fármacos , Dopamina/metabolismo , Péptido Natriurético Tipo-C/farmacología , Neuropéptidos/farmacología , Corteza Prefrontal/efectos de los fármacos , Área Tegmental Ventral/efectos de los fármacos , Trastornos Inducidos por Alcohol/metabolismo , Animales , Encéfalo/metabolismo , Carbazoles/metabolismo , GMP Cíclico/metabolismo , Proteínas Quinasas Dependientes de GMP Cíclico/antagonistas & inhibidores , Proteínas Quinasas Dependientes de GMP Cíclico/metabolismo , Etanol/metabolismo , Humanos , Masculino , Corteza Prefrontal/metabolismo , Ratas , Ratas Wistar , Síndrome de Abstinencia a Sustancias/metabolismo , Área Tegmental Ventral/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...