Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Brain Sci ; 13(3)2023 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-36979236

RESUMEN

Schizophrenia is regarded as a neurodevelopmental disorder with its course progressing throughout life. However, the aetiology and development of schizophrenia are still under investigation. Several data suggest that the dysfunction of epigenetic mechanisms is known to be involved in the pathomechanism of this mental disorder. The present article revised the epigenetic background of schizophrenia based on the data available in online databases (PubMed, Scopus). This paper focused on the role of epigenetic regulation, such as DNA methylation, histone modifications, and interference of non-coding RNAs, in schizophrenia development. The article also reviewed the available data related to epigenetic regulation that may modify the severity of the disease as a possible target for schizophrenia pharmacotherapy. Moreover, the effects of antipsychotics on epigenetic malfunction in schizophrenia are discussed based on preclinical and clinical results. The obtainable data suggest alterations of epigenetic regulation in schizophrenia. Moreover, they also showed the important role of epigenetic modifications in antipsychotic action. There is a need for more data to establish the role of epigenetic mechanisms in schizophrenia therapy. It would be of special interest to find and develop new targets for schizophrenia therapy because patients with schizophrenia could show little or no response to current pharmacotherapy and have treatment-resistant schizophrenia.

2.
Genes (Basel) ; 14(2)2023 01 18.
Artículo en Inglés | MEDLINE | ID: mdl-36833173

RESUMEN

Schizophrenia pathogenesis remains challenging to define; however, there is strong evidence that the interaction of genetic and environmental factors causes the disorder. This paper focuses on transcriptional abnormalities in the prefrontal cortex (PFC), a key anatomical structure that determines functional outcomes in schizophrenia. This review summarises genetic and epigenetic data from human studies to understand the etiological and clinical heterogeneity of schizophrenia. Gene expression studies using microarray and sequencing technologies reported the aberrant transcription of numerous genes in the PFC in patients with schizophrenia. Altered gene expression in schizophrenia is related to several biological pathways and networks (synaptic function, neurotransmission, signalling, myelination, immune/inflammatory mechanisms, energy production and response to oxidative stress). Studies investigating mechanisms driving these transcriptional abnormalities focused on alternations in transcription factors, gene promoter elements, DNA methylation, posttranslational histone modifications or posttranscriptional regulation of gene expression mediated by non-coding RNAs.


Asunto(s)
Epigénesis Genética , Esquizofrenia , Humanos , Esquizofrenia/genética , Metilación de ADN , Corteza Prefrontal/metabolismo , Expresión Génica
3.
Int J Mol Sci ; 23(20)2022 Oct 14.
Artículo en Inglés | MEDLINE | ID: mdl-36293185

RESUMEN

The expression of the Calcium/Calmodulin-Dependent Protein Kinase I gamma (encoded by the Camk1g gene) depends on the activation of glucocorticoid receptors (GR) and is strongly regulated by stress. Since Camk1g is primarily expressed in neuronal cells of the limbic system in the brain, we hypothesized that it could be involved in signaling mechanisms that underlie the adaptive or maladaptive responses to stress. Here, we find that restraint-induced stress and the GR agonist dexamethasone robustly increase the expression of Camk1g in neurons of the amygdalar nuclei in the mouse brain. To assess the functional role of Camk1g expression, we performed a virally induced knock-down of the transcript. Mice with bilateral amygdala-specific Camk1g knock-down showed increased anxiety-like behaviors in the light-dark box, and an increase in freezing behavior after fear-conditioning, but normal spatial working memory during exploration of a Y-maze. Thus, we confirm that Camk1g is a neuron-specific GR-regulated transcript, and show that it is specifically involved in behaviors related to anxiety, as well as responses conditioned by aversive stimuli.


Asunto(s)
Núcleo Amigdalino Central , Glucocorticoides , Ratones , Animales , Glucocorticoides/farmacología , Núcleo Amigdalino Central/metabolismo , Receptores de Glucocorticoides/genética , Receptores de Glucocorticoides/metabolismo , Calcio , Proteína Quinasa Tipo 1 Dependiente de Calcio Calmodulina/metabolismo , Ansiedad/metabolismo , Dexametasona/farmacología , Conducta Animal
4.
Neuropharmacology ; 198: 108779, 2021 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-34481835

RESUMEN

The basal amygdala (BA) has been implicated in encoding fear and its extinction. The level of serotonin (5-HT) in the BA increases due to arousal and stress related to aversive stimuli. The effects of 5-HT7 receptor (5-HT7R) activation and blockade on the activity of BA neurons have not yet been investigated. In the present study, a transgenic mouse line carrying green fluorescent protein (GFP) reporter gene was used to identify neurons that express the 5-HT7R. GFP immunoreactivity was present mainly in cells that also expressed GAD67 or parvalbumin (PV), the phenotypic markers for GABAergic interneurons. Most cells showing GFP fluorescence demonstrated firing patterns characteristic of BA inhibitory interneurons. Activation of 5-HT7Rs resulted in a depolarization and/or occurrence of spontaneous spiking activity of BA interneurons that was accompanied by an increase in the mean frequency and mean amplitude of spontaneous inhibitory postsynaptic currents (sIPSCs) recorded from BA principal neurons. These effects were blocked by a specific 5-HT7R antagonist, SB269970 and were absent in slices from 5-HT7R knockout mice. Activation of 5-HT7Rs also decreased the mean frequency of spontaneous excitatory postsynaptic currents (sEPSCs) recorded from BA principal neurons, which was blocked by the GABAA receptor antagonist picrotoxin. Neither inhibitory nor excitatory miniature postsynaptic currents (mIPSCs/mEPSCs) were affected by 5-HT7R activation. These results show that in the BA 5-HT7Rs stimulate an activity-dependent enhancement of inhibitory input from local interneurons to BA principal neurons and provide insights about the possible involvement of BA serotonergic receptors in neuronal mechanisms underlying fear memory.


Asunto(s)
Amígdala del Cerebelo/efectos de los fármacos , Neuronas/efectos de los fármacos , Receptores de Serotonina/efectos de los fármacos , Antagonistas de la Serotonina/farmacología , Agonistas de Receptores de Serotonina/farmacología , Sinapsis/efectos de los fármacos , Animales , Fenómenos Electrofisiológicos , Potenciales Postsinápticos Excitadores/efectos de los fármacos , Neuronas GABAérgicas/efectos de los fármacos , Proteínas Fluorescentes Verdes , Interneuronas/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Fenoles/farmacología , Picrotoxina/farmacología , Receptores de GABA-A/efectos de los fármacos , Receptores de Serotonina/genética , Sulfonamidas/farmacología
5.
Int J Mol Sci ; 22(16)2021 Aug 13.
Artículo en Inglés | MEDLINE | ID: mdl-34445411

RESUMEN

BACKGROUND: The present study investigated the role of proteins from the bromodomain and extra-terminal (BET) family in schizophrenia-like abnormalities in a neurodevelopmental model of schizophrenia induced by prenatal methylazoxymethanol (MAM) administration (MAM-E17). METHODS: An inhibitor of BET proteins, JQ1, was administered during adolescence on postnatal days (P) 23-P29, and behavioural responses (sensorimotor gating, recognition memory) and prefrontal cortical (mPFC) function (long-term potentiation (LTP), molecular and proteomic analyses) studies were performed in adult males and females. RESULTS: Deficits in sensorimotor gating and recognition memory were observed only in MAM-treated males. However, adolescent JQ1 treatment affected animals of both sexes in the control but not MAM-treated groups and reduced behavioural responses in both sexes. An electrophysiological study showed LTP impairments only in male MAM-treated animals, and JQ1 did not affect LTP in the mPFC. In contrast, MAM did not affect activity-dependent gene expression, but JQ1 altered gene expression in both sexes. A proteomic study revealed alterations in MAM-treated groups mainly in males, while JQ1 affected both sexes. CONCLUSIONS: MAM-induced schizophrenia-like abnormalities were observed only in males, while adolescent JQ1 treatment affected memory recognition and altered the molecular and proteomic landscape in the mPFC of both sexes. Thus, transient adolescent inhibition of the BET family might prompt permanent alterations in the mPFC.


Asunto(s)
Azepinas/administración & dosificación , Acetato de Metilazoximetanol/análogos & derivados , Corteza Prefrontal/crecimiento & desarrollo , Esquizofrenia/fisiopatología , Triazoles/administración & dosificación , Adolescente , Desarrollo del Adolescente/efectos de los fármacos , Animales , Azepinas/farmacología , Modelos Animales de Enfermedad , Femenino , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Humanos , Potenciación a Largo Plazo/efectos de los fármacos , Masculino , Acetato de Metilazoximetanol/toxicidad , Corteza Prefrontal/efectos de los fármacos , Corteza Prefrontal/metabolismo , Proteómica , Ratas , Reconocimiento en Psicología/efectos de los fármacos , Esquizofrenia/inducido químicamente , Esquizofrenia/metabolismo , Caracteres Sexuales , Triazoles/farmacología
6.
Behav Brain Res ; 406: 113231, 2021 05 21.
Artículo en Inglés | MEDLINE | ID: mdl-33737089

RESUMEN

Adolescent social isolation (SI) might change the trajectory of brain development. In the present study, we investigated the effect of short-term adolescent SI on fear memory, anxiety and protein levels in the adult medial prefrontal cortex of rats prenatally treated with methylazoxymethanol, MAM-E17 model of schizophrenia. The animals were maintained in standard housing (SH) or social isolation (P30-P40, SI) conditions. Behavioural tests (trace or delay fear conditioning, light/dark box) were performed in late adolescence and early adulthood. The results showed that MAM treatment did not alter fear memory, which was investigated with the use of either trace or delay fear conditioning, at any age, and SI decreased the fear response in adult control animals only under trace conditioning. Neither MAM nor SI influenced anxiety-related behaviour measured in the light/dark box. A proteomics study showed that both MAM and SI changed the protein levels related to synapse maturation and cytoskeletal organization, energy transfer and metabolic processes. Prenatal or adolescent environmental factors are able to change the expression of proteins that are correlated with behavioural impairments. Moreover, SI reversed some alterations in proteins induced by MAM. Thus, normally developing brains showed different responses to adolescent SI than those with altering courses of MAM administration.


Asunto(s)
Conducta Animal/fisiología , Condicionamiento Clásico/fisiología , Miedo/fisiología , Corteza Prefrontal , Efectos Tardíos de la Exposición Prenatal , Esquizofrenia , Aislamiento Social , Factores de Edad , Animales , Femenino , Masculino , Acetato de Metilazoximetanol/análogos & derivados , Acetato de Metilazoximetanol/farmacología , Corteza Prefrontal/metabolismo , Corteza Prefrontal/fisiopatología , Embarazo , Efectos Tardíos de la Exposición Prenatal/inducido químicamente , Efectos Tardíos de la Exposición Prenatal/metabolismo , Efectos Tardíos de la Exposición Prenatal/fisiopatología , Proteoma , Ratas Wistar , Esquizofrenia/etiología , Esquizofrenia/metabolismo , Esquizofrenia/fisiopatología , Teratógenos/farmacología
7.
Behav Brain Res ; 402: 113095, 2021 03 26.
Artículo en Inglés | MEDLINE | ID: mdl-33359366

RESUMEN

The amygdala is a key structure involved in both physiological and behavioural effects of fearful and stressful stimuli. The central stress response is controlled by the activity of the hypothalamic-pituitary-adrenal (HPA) axis via glucocorticoid hormones, acting mainly through glucocorticoid receptors (GR), widely expressed among different brain regions, including the central nucleus of the amygdala (CeA). Although to date, neuronal GR was postulated to be involved in the mediating stress effects, increasing evidence points to the vital role of glial GR. Here, we aimed to evaluate the role of astrocytic GR in CeA in various aspects of the stress response. We used a lentiviral vector to disrupt an astrocytic GR in the CeA of Aldh1l1-Cre transgenic mice. Astrocytic GR knockdown mice (GR KD) exhibited an attenuated expression of fear-related memory in the fear conditioning paradigm. Interestingly, the consolidation of non-stressful memory in the novel object recognition test remained unchanged. Moreover, GR KD group presented reduced anxiety, measured in the open field test. However, knockdown of astrocytic GR in the CeA did not affect an acute response to stress in the tail suspension test. Taken together, obtained results suggest that astrocytic GR in the CeA promotes aversive memory consolidation and some aspects of anxiety behaviour.


Asunto(s)
Ansiedad/fisiopatología , Astrocitos/metabolismo , Núcleo Amigdalino Central/fisiología , Condicionamiento Clásico/fisiología , Miedo/fisiología , Consolidación de la Memoria/fisiología , Receptores de Glucocorticoides/metabolismo , Estrés Psicológico/fisiopatología , Animales , Ansiedad/metabolismo , Conducta Animal/fisiología , Núcleo Amigdalino Central/metabolismo , Modelos Animales de Enfermedad , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Estrés Psicológico/metabolismo
8.
Neuropsychopharmacology ; 45(2): 404-415, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31254970

RESUMEN

To date, neurons have been the primary focus of research on the role of glucocorticoids in the regulation of brain function and pathological behaviors, such as addiction. Astrocytes, which are also glucocorticoid-responsive, have been recently implicated in the development of drug abuse, albeit through as yet undefined mechanisms. Here, using a spectrum of tools (whole-transcriptome profiling, viral-mediated RNA interference in vitro and in vivo, behavioral pharmacology and electrophysiology), we demonstrate that astrocytes in the nucleus accumbens (NAc) are an important locus of glucocorticoid receptor (GR)-dependent transcriptional changes that regulate rewarding effects of morphine. Specifically, we show that targeted knockdown of the GR in the NAc astrocytes enhanced conditioned responses to morphine, with a concomitant inhibition of morphine-induced neuronal excitability and plasticity. Interestingly, GR knockdown did not influence sensitivity to cocaine. Further analyses revealed GR-dependent regulation of astroglial metabolism. Notably, GR knockdown inhibited induced by glucocorticoids lactate release in astrocytes. Finally, lactate administration outbalanced conditioned responses to morphine in astroglial GR knockdown mice. These findings demonstrate a role of GR-dependent regulation of astrocytic metabolism in the NAc and a key role of GR-expressing astrocytes in opioid reward processing.


Asunto(s)
Analgésicos Opioides/farmacología , Astrocitos/metabolismo , Condicionamiento Psicológico/fisiología , Ácido Láctico/metabolismo , Morfina/farmacología , Receptores de Glucocorticoides/metabolismo , Animales , Astrocitos/efectos de los fármacos , Células Cultivadas , Condicionamiento Psicológico/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos
9.
Metab Brain Dis ; 34(1): 341-352, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30519836

RESUMEN

Altered parvalbumin (PV) expression is observed in the prefrontal cortex of subjects with schizophrenia. Environmental context, particularly during adolescence, might regulate PV expression. In the present study, we investigated the effect of adolescent social isolation (SI) on PV expression in the medial prefrontal cortex in a neurodevelopmental model (MAM-E17) of schizophrenia. SI exposure occurred from postnatal day 30 to 40, followed by resocialization until late adolescence or early adulthood. PV mRNA and protein levels, as well as the number of PV cells, were analysed at these ages. Moreover, epigenetic regulation of PV expression by histone methylation was examined by measuring the total and PV gene-bound H3K4me3 levels. MAM only decreased levels of the PV mRNA and protein in adulthood. Decreases in total H3K4me3 levels and its level at the PV gene were also observed at this age. In contrast, in late adolescence, SI induced a decrease in the expression of the PV mRNA in the MAM group that was related to the reduction in total and PV gene-bound H3K4me3 levels. However, at this age, SI increased the levels of the PV protein in both the control and MAM groups. In adulthood, SI did not affect PV mRNA or H3K4me3 levels but decreased levels of the PV protein in both groups. Both MAM and SI failed to change the number of PV cells at any age. The results indicate that adolescent SI accelerated epigenetic impairments of PV expression in MAM-E17 rats; however, subsequent resocialization abolished this dysfunction, but failed to prevent alterations in PV protein.


Asunto(s)
Histonas/metabolismo , Parvalbúminas/metabolismo , Corteza Prefrontal/metabolismo , Esquizofrenia/metabolismo , Aislamiento Social , Animales , Modelos Animales de Enfermedad , Epigénesis Genética , Femenino , Masculino , Metilación , Neuronas/metabolismo , Ratas , Ratas Wistar
10.
Glia ; 61(4): 623-35, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23339081

RESUMEN

Chronic opioid use leads to the structural reorganization of neuronal networks, involving genetic reprogramming in neurons and glial cells. Our previous in vivo studies have revealed that a significant fraction of the morphine-induced alterations to the striatal transcriptome included glucocorticoid (GC) receptor (GR)-dependent genes. Additional analyses suggested glial cells to be the locus of these changes. In the current study, we aimed to differentiate the direct transcriptional effects of morphine and a GR agonist on primary striatal neurons and astrocytes. Whole-genome transcriptional profiling revealed that while morphine had no significant effect on gene expression in both cell types, dexamethasone significantly altered the transcriptional profile in astrocytes but not neurons. We obtained a complete dataset of genes undergoing the regulation, which includes genes related to glucose metabolism (Pdk4), circadian activity (Per1) and cell differentiation (Sox2). There was also an overlap between morphine-induced transcripts in striatum and GR-dependent transcripts in cultured astrocytes. We further analyzed the regulation of expression of one gene belonging to both groups, serum and GC regulated kinase 1 (Sgk1). We identified two transcriptional variants of Sgk1 that displayed selective GR-dependent upregulation in cultured astrocytes but not neurons. Moreover, these variants were the only two that were found to be upregulated in vivo by morphine in a GR-dependent fashion. Our data suggest that the morphine-induced, GR-dependent component of transcriptome alterations in the striatum is confined to astrocytes. Identification of this mechanism opens new directions for research on the role of astrocytes in the central effects of opioids.


Asunto(s)
Astrocitos/metabolismo , Marcación de Gen/métodos , Morfina/administración & dosificación , Neuronas/fisiología , Receptores de Glucocorticoides/fisiología , Transducción de Señal/fisiología , Animales , Astrocitos/efectos de los fármacos , Astrocitos/fisiología , Células Cultivadas , Masculino , Ratones , Ratones Endogámicos C57BL , Neuronas/efectos de los fármacos , Transducción de Señal/efectos de los fármacos
11.
Genome Biol ; 11(5): R48, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20459597

RESUMEN

BACKGROUND: Various drugs of abuse activate intracellular pathways in the brain reward system. These pathways regulate the expression of genes that are essential to the development of addiction. To reveal genes common and distinct for different classes of drugs of abuse, we compared the effects of nicotine, ethanol, cocaine, morphine, heroin and methamphetamine on gene expression profiles in the mouse striatum. RESULTS: We applied whole-genome microarray profiling to evaluate detailed time-courses (1, 2, 4 and 8 hours) of transcriptome alterations following acute drug administration in mice. We identified 42 drug-responsive genes that were segregated into two main transcriptional modules. The first module consisted of activity-dependent transcripts (including Fos and Npas4), which are induced by psychostimulants and opioids. The second group of genes (including Fkbp5 and S3-12), which are controlled, in part, by the release of steroid hormones, was strongly activated by ethanol and opioids. Using pharmacological tools, we were able to inhibit the induction of particular modules of drug-related genomic profiles. We selected a subset of genes for validation by in situ hybridization and quantitative PCR. We also showed that knockdown of the drug-responsive genes Sgk1 and Tsc22d3 resulted in alterations to dendritic spines in mice, possibly reflecting an altered potential for plastic changes. CONCLUSIONS: Our study identified modules of drug-induced genes that share functional relationships. These genes may play a critical role in the early stages of addiction.


Asunto(s)
Regulación de la Expresión Génica/efectos de los fármacos , Redes Reguladoras de Genes/efectos de los fármacos , Drogas Ilícitas/farmacología , Neostriado/efectos de los fármacos , Neostriado/metabolismo , Trastornos Relacionados con Sustancias/genética , Transcripción Genética/efectos de los fármacos , Análisis de Varianza , Animales , Conducta Animal/efectos de los fármacos , Sitios de Unión , Células Cultivadas , Estimulantes del Sistema Nervioso Central/farmacología , Cromosomas de los Mamíferos/genética , Análisis por Conglomerados , Espinas Dendríticas/efectos de los fármacos , Espinas Dendríticas/genética , Perfilación de la Expresión Génica , Técnicas de Silenciamiento del Gen , Ratones , Ratones Endogámicos C57BL , Análisis de Secuencia por Matrices de Oligonucleótidos , Reacción en Cadena de la Polimerasa , ARN Mensajero/genética , ARN Mensajero/metabolismo , Reproducibilidad de los Resultados , Recompensa , Factores de Transcripción/metabolismo
12.
Eur J Neurosci ; 30(6): 1101-10, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19735294

RESUMEN

Persistent changes that take place during the development of opioid addiction are thought to be due to reorganization of synaptic connections in relevant brain circuits. This neuronal plasticity requires trafficking of signaling molecules that are controlled by kinesins. In neurons, kinesin light chain 1 (KLC1) acts as the primary regulator of kinesin action. We observed that KLC1 was enriched in sub-cortical regions of the brain in C57Bl/6J mice. KLC1 expression was especially enriched in the striatum, hippocampus and amygdala, which are known to be involved in opioid addiction. Our study revealed that conditioning of C57Bl/6J mice with morphine elevated KLC1 levels in the amygdala, frontal cortex and hippocampus, but not in the striatum. Further study revealed that alterations in KLC1 protein levels in the studied brain regions correlated with the expression of morphine-induced conditioned place preference. In the cortex, hippocampus and amygdala, KLC1 co-localized with calcium/calmodulin-dependent protein kinase II (CaMKII), suggesting that KLC1 was present in the cell bodies and dendrites of pyramidal neurons. Our findings indicate that KLC1, a molecule involved in dendritic and axonal transport in the brain, is affected during chronic morphine treatment and may be involved in the development of opioid addiction.


Asunto(s)
Amígdala del Cerebelo/metabolismo , Cuerpo Estriado/metabolismo , Proteínas Asociadas a Microtúbulos/metabolismo , Morfina/farmacología , Recompensa , Análisis de Varianza , Animales , Western Blotting , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/metabolismo , Condicionamiento Psicológico/efectos de los fármacos , Condicionamiento Psicológico/fisiología , Técnica del Anticuerpo Fluorescente , Hibridación in Situ , Cinesinas , Masculino , Ratones , Ratones Endogámicos C57BL , Morfina/metabolismo , Actividad Motora/efectos de los fármacos , Neuronas/metabolismo
13.
J Neurosci Res ; 82(4): 563-70, 2005 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-16211563

RESUMEN

Activity-regulated cytoskeleton-associated protein (Arc) is an effector immediate early gene product implicated in long-term potentiation and other forms of neuroplasticity. Earlier studies demonstrated Arc induction in discrete brain regions by several psychoactive substances, including drugs of abuse. In the present experiments, the influence of morphine on Arc expression was assessed by quantitative reverse transcription real-time PCR and Western blotting in vivo in the mouse striatum/nucleus accumbens and, in vitro, in the mouse Neuro2A MOR1A cell line, expressing mu-opioid receptor. An acute administration of morphine produced a marked increase in Arc mRNA and protein level in the mouse striatum/nucleus accumbens complex. After prolonged opiate treatment, tolerance to the stimulatory effect of morphine on Arc expression developed. No changes in the striatal Arc mRNA levels were observed during spontaneous or opioid antagonist-precipitated morphine withdrawal. In Neuro2A MOR1A cells, acute, but not prolonged, morphine treatment elevated Arc mRNA level by activation of mu-opioid receptor. This was accompanied by a corresponding increase in Arc protein level. Inhibition experiments revealed that morphine induced Arc expression in Neuro2A MOR1A cells via intracellular signaling pathways involving mitogen-activated protein (MAP) kinases and protein kinase C. These results lend further support to the notion that stimulation of opioid receptors may exert an activating influence on some intracellular pathways and leads to induction of immediate early genes. They also demonstrate that Arc is induced in the brain in vivo after morphine administration and thus may play a role in neuroadaptations produced by the drug.


Asunto(s)
Complejo Relacionado con el SIDA/metabolismo , Cuerpo Estriado/efectos de los fármacos , Expresión Génica/efectos de los fármacos , Morfina/administración & dosificación , Narcóticos/administración & dosificación , Receptores Opioides mu/metabolismo , Complejo Relacionado con el SIDA/genética , Análisis de Varianza , Animales , Western Blotting/métodos , Línea Celular Tumoral , Cuerpo Estriado/metabolismo , Relación Dosis-Respuesta a Droga , Esquema de Medicación , Interacciones Farmacológicas , Encefalina Ala(2)-MeFe(4)-Gli(5)/farmacología , Inhibidores Enzimáticos/farmacología , Flavonoides/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/fisiología , Indoles/farmacología , Masculino , Maleimidas/farmacología , Ratones , Ratones Endogámicos C57BL , Neuroblastoma/metabolismo , ARN Mensajero/biosíntesis , Receptores Opioides mu/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa/métodos , Somatostatina/análogos & derivados , Somatostatina/farmacología , Factores de Tiempo
14.
J Neurosci ; 25(20): 4996-5003, 2005 May 18.
Artículo en Inglés | MEDLINE | ID: mdl-15901780

RESUMEN

Chronic exposure to opiates produces dependence and addiction, which may result from neuroadaptations in the dopaminergic reward pathway and its target brain regions. The neuronal protein alpha-synuclein has been implicated in neuronal plasticity and proposed to serve as a negative regulator of dopamine neurotransmission. Thus, alpha-synuclein could mediate some effects of opiates in the brain. The present study investigated the influence of acute and chronic morphine administration on alpha-synuclein mRNA and protein expression in the brains of mice. Downregulation of alpha-synuclein mRNA was observed in the basolateral amygdala, dorsal striatum, nucleus accumbens, and ventral tegmental area of mice withdrawn from chronic morphine treatment. The changes were the most pronounced after longer periods of withdrawal (48 h). In contrast, levels of alpha-synuclein protein, as assessed by Western blotting, were significantly increased in the amygdala and striatum/accumbens (but not in the mesencephalon) of morphine-withdrawn mice. In both brain regions, levels of alpha-synuclein were elevated for as long as 2 weeks after treatment cessation. Because alpha-synuclein is a presynaptic protein, the detected opposite changes in its mRNA and protein levels are likely to take place in different populations of projection neurons whose somata are in different brain areas. Axonal localization of alpha-synuclein was confirmed by immunofluorescent labeling. An attempt to identify postsynaptic neurons innervated by alpha-synuclein-containing axon terminals revealed their selective apposition to calbindin D28K-negative projection neurons in the basolateral amygdala. The observed changes in alpha-synuclein levels are discussed in connection with their putative role in mediating suppression of dopaminergic neurotransmission during opiate withdrawal.


Asunto(s)
Regulación de la Expresión Génica/efectos de los fármacos , Sistema Límbico/efectos de los fármacos , Morfina/administración & dosificación , Narcóticos/administración & dosificación , alfa-Sinucleína/metabolismo , Análisis de Varianza , Animales , Western Blotting/métodos , Diagnóstico por Imagen/métodos , Esquema de Medicación , Inmunohistoquímica/métodos , Hibridación in Situ/métodos , Masculino , Ratones , Ratones Endogámicos C57BL , ARN Mensajero/genética , ARN Mensajero/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa/métodos , Factores de Tiempo , alfa-Sinucleína/genética
15.
J Neurochem ; 90(4): 874-82, 2004 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-15287893

RESUMEN

Addiction to opiates depend on drug-induced neuroplastic changes and are underlain by alterations of gene expression. Transcription factors Ca2+/cAMP responsive element binding protein (CREB) and activator protein 1 (AP-1) may constitute a direct link between the opioid-regulated signal transduction pathways and modulation of gene expression. Acute treatment of Neuro2a MOR neuroblastoma cells with opioids stimulated CREB activity; prolonged treatment normalized it, while withdrawal from the drug again elicited an increase in phosphorylated CREB levels. Protein kinase C was responsible for the activation of transcription following acute opioid administration whereas the cAMP pathway activated similar mechanisms during withdrawal, making CREB a kind of 'a trigger' reacting to the presence or withdrawal of the opioid signal. Apart from the elevated CREB phosphorylation, CRE binding activity and expression of luciferase reporter gene regulated by CRE elements were increased after single administration and during withdrawal from the prolonged opioid treatment. Along with CREB, AP-1 binding activity and AP-1-directed transcription were stimulated after single administration and during withdrawal from the opioid. These results provide evidence that both single opioid administration and opioid withdrawal activate CREB and CRE-dependent transcriptional mechanisms via distinct intracellular signaling pathways.


Asunto(s)
Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Expresión Génica/efectos de los fármacos , Neuroblastoma/metabolismo , Receptores Opioides mu/metabolismo , Factor de Transcripción AP-1/metabolismo , Analgésicos Opioides/farmacología , Animales , Línea Celular , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/efectos de los fármacos , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , ADN/metabolismo , Encefalina Ala(2)-MeFe(4)-Gli(5)/farmacología , Genes Reporteros/efectos de los fármacos , Ratones , Morfina/farmacología , Fosforilación/efectos de los fármacos , Proteína Quinasa C/metabolismo , Receptores Opioides mu/efectos de los fármacos , Factor de Transcripción AP-1/efectos de los fármacos
16.
Neurosci Lett ; 358(2): 79-82, 2004 Mar 25.
Artículo en Inglés | MEDLINE | ID: mdl-15026153

RESUMEN

Recent reports have demonstrated effectiveness of melanocortin antagonists as potent analgesics, and have suggested that the spinal melanocortin 4 receptor (MC4-R) mediates their effects on pain transmission. These findings prompted us to investigate the changes in MC4-R mRNA level in the spinal cord and dorsal root ganglia (DRG) of neuropathic animals at different time points after sciatic nerve injury by quantitative real-time PCR. The spinal MC4-R mRNA level was not affected by sciatic nerve injury. In contrast, down-regulation of MC4-R mRNA in DRG developed 2 weeks after the injury and was parallel with the attenuated effectiveness of MC4-R ligands in neuropathic animals. The MC4-R adaptation in DRG observed in neuropathic rats indicates their important role in presynaptic modulation of activity of the primary afferents in neuropathic pain.


Asunto(s)
Regulación hacia Abajo/fisiología , Ganglios Espinales/metabolismo , Dolor/metabolismo , Receptor de Melanocortina Tipo 4/biosíntesis , Neuropatía Ciática/metabolismo , Animales , Masculino , Umbral del Dolor/fisiología , ARN Mensajero/biosíntesis , Ratas , Ratas Wistar , Médula Espinal/metabolismo
17.
Brain Res ; 990(1-2): 113-9, 2003 Nov 14.
Artículo en Inglés | MEDLINE | ID: mdl-14568335

RESUMEN

The relationships between the CRF, which enhances the proopiomelanocortin (POMC) biosynthesis, and POMC-derived peptides (opioids and melanocortins) might be a new target for rational treatment of morphine tolerance. In the present study, we investigated the effect of acute and chronic morphine administration on the level of CRF1 and melanocortin 4 receptor (MC4-R) mRNAs in the rat amygdala by quantitative real-time PCR method. Moreover, we investigated the effect of antagonists of melanocortin and CRF receptors, SHU9119 and alpha-helical CRF (alphah-CRF), respectively, administered bilaterally into the central nucleus of the amygdala, on morphine tolerance using tail-flick and paw withdrawal tests. Our study demonstrated that acute morphine administration decreased the level of MC4-R mRNA in the rat amygdala. This decrease was attenuated following chronic morphine administration, and mRNA level of MC4 receptors was gradually increased and, on 9th day of morphine administration, i.e. in the period when morphine tolerance already developed, the level was significantly increased in comparison with control and with the effect after single morphine dose. In contrast, morphine did not affect the CRF receptor. In behavioral study, we demonstrated that SHU9119 and alphah-CRF significantly increased the antinociceptive effect of morphine, when they were injected into the amygdala prior to morphine administration in tolerant rats. We have shown for the first time the contribution of amygdalar melanocortin receptors to morphine tolerance, and we conclude that the altered melanocortin receptor function may play an important role in the development of morphine-induced tolerance. CRF and melanocortin peptides can modulate the phenomena in the same direction, in opposition to opioids. Therefore, antagonists of melanocortin receptors may be regarded as possible therapeutic modulators of morphine tolerance.


Asunto(s)
Amígdala del Cerebelo/metabolismo , Analgésicos Opioides/farmacología , Morfina/farmacología , ARN Mensajero/biosíntesis , Receptor de Melanocortina Tipo 4/biosíntesis , Receptores de Hormona Liberadora de Corticotropina/biosíntesis , Amígdala del Cerebelo/efectos de los fármacos , Animales , Hormona Liberadora de Corticotropina/farmacología , Tolerancia a Medicamentos , Antagonistas de Hormonas/farmacología , Masculino , Hormonas Estimuladoras de los Melanocitos/farmacología , Microinyecciones , Dimensión del Dolor/efectos de los fármacos , Fragmentos de Péptidos/farmacología , Ratas , Ratas Wistar , Receptor de Melanocortina Tipo 4/antagonistas & inhibidores , Receptores de Hormona Liberadora de Corticotropina/antagonistas & inhibidores , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
18.
Neurosci Lett ; 325(2): 107-10, 2002 Jun 07.
Artículo en Inglés | MEDLINE | ID: mdl-12044633

RESUMEN

The development of morphine tolerance and sciatic nerve injury-induced allodynia after functional knockdown of spinal opioid receptors using antisense oligonucleotides targeting beta-arrestin was investigated. Ineffectiveness of morphine in neuropathic pain suggests an implication of the same mechanism in these two processes. The development of morphine tolerance (10 microg intrathecally (i.th.), every 12 h) was significantly inhibited in rats, which received i.th. beta-arrestin antisenses (2 nM). Acute and chronic (6 days) i.th. administration of antisenses antagonized the allodynia in the rat model of neuropathic pain. Our results demonstrated that i.th. administration of beta-arrestin antisenses delayed development of tolerance to morphine and nerve injury-induced cold allodynia, which suggest that both of the investigated phenomena may be mediated by a similar mechanism, e.g. receptor desensitization. Moreover, the antisense oligonucleotides targeting beta-arrestin may constitute a new approach to the therapy of neuropathic pain.


Asunto(s)
Analgésicos Opioides/farmacología , Arrestinas/genética , Hiperestesia/fisiopatología , Morfina/farmacología , Antagonistas de Narcóticos , Oligonucleótidos Antisentido/farmacología , Médula Espinal/metabolismo , Analgésicos Opioides/administración & dosificación , Animales , Frío , Tolerancia a Medicamentos , Hiperestesia/etiología , Inyecciones Espinales , Masculino , Morfina/administración & dosificación , Ratas , Ratas Wistar , Nervio Ciático/lesiones , Heridas y Lesiones/complicaciones , beta-Arrestinas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...