Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Behav Brain Res ; 373: 112079, 2019 11 05.
Artículo en Inglés | MEDLINE | ID: mdl-31301411

RESUMEN

Previous studies show that chronic stress induces synaptic structural alterations in brain regions involved in emotional processing such as the prefrontal cortex (PFC) and the basolateral amygdala (BLA). Yet, these studies are based mainly in animal exposure to unpredictable stressors or to restraint stress. On the other hand, studies using the chronic social defeat stress (CSDS), a relevant model of depression based on social conflict, are lacking. Here we aim to study the acute (24 h after CSDS) and long-term (one month after CSDS) effects of CSDS on dendritic and synaptic structures in the PFC and BLA of C57BL/6 mice. Specifically, BLA and PFC dendritic spine densities as well as BLA arborisation were analysed. Subsequently, we investigate in these regions the synaptic response to a friendly (interaction with a same strain mouse) or a fearful (interaction with a dominant strain mouse) social stimulus. Spine densities of the apical dendrites from the PFC pyramidal neurons were decreased by CSDS in the long-term (one month after CSDS). In addition, CSDS increased BLA stellate neurons spine density in the short-term (24 h after CSDS) and dendritic arborisation in the long-term. Moreover, long-term CSDS mice exposed to a fearful stimulus experienced a marked social avoidance and showed a significant increase in the expression of the immature form of the brain derived neurotrophic factor (proBDNF) in the amygdala. Taken together these results suggest the existence of persistent neuronal adaptations in the PFC and BLA in socially defeated mice. Specifically, spine density retraction in the PFC and increased BLA dendritic arborisation could represent an adaptive structural change allowing rapid expression of synaptic markers in response to fearful experiences.


Asunto(s)
Estrés Psicológico/fisiopatología , Sinapsis/metabolismo , Transmisión Sináptica/fisiología , Amígdala del Cerebelo/metabolismo , Amígdala del Cerebelo/fisiología , Animales , Complejo Nuclear Basolateral/metabolismo , Conducta Animal , Encéfalo/metabolismo , Dendritas/metabolismo , Espinas Dendríticas/metabolismo , Depresión/metabolismo , Trastorno Depresivo/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Plasticidad Neuronal/fisiología , Neuronas/metabolismo , Corteza Prefrontal/metabolismo , Corteza Prefrontal/fisiología , Células Piramidales/metabolismo , Conducta Social , Estrés Psicológico/metabolismo
2.
Behav Brain Res ; 335: 128-131, 2017 09 29.
Artículo en Inglés | MEDLINE | ID: mdl-28778545

RESUMEN

Some histone deacetylase (HDACs) enzymes have been proposed as epigenetic targets involved in the pathophysiology of depression and antidepressant-like action. Among them, we have recently identified SIRT2, a class III NAD+-dependent HDAC, as being oppositely regulated by stress and antidepressants. Moreover, SIRT2 inhibition has shown antianhedonic-like action in the chronic mild stress model of depression. Here we have extended the study using an alternative model of depression based in a genetic manipulation of glutamate function. Specifically, mice heterozygous for the vesicular glutamate transporter 1 (VGLUT1+/-) were used. Firstly, mRNA expression of the different members of the HDAC superfamily in the prefrontal cortex (PFC) of VGLUT1+/- mice and WT littermates were studied by RT-PCR. Secondly, the effect of repeated treatment with the selective SIRT2 inhibitor 33i and the antidepressant imipramine on anhedonic behaviour of VGLUT1+/- mice was studied by weekly monitoring of sucrose intake. Further, the interaction of 33i towards specific monoaminergic targets such as serotonin or noradrenaline transporters as well as the monoaminooxidase enzyme was studied. The mRNA occurance of the different members of HDAC superfamily was not altered in the PFC of VGLUT1+/- mice. While repeated imipramine showed an anti-anhedonic action in both VGLUT1+/- and WT, the selective SIRT2 inhibitor 33i fully reversed anhedonia of VGLUT1+/-. Further, 33i showed no interaction with the above mentioned monoaminergic molecular targets. These results confirm that SIRT2 inhibition is able to reverse anhedonia in different animal models and highlight the need to further investigate the role of SIRT2 inhibitors as new antidepressant agents.


Asunto(s)
Anhedonia/efectos de los fármacos , Depresión/tratamiento farmacológico , Depresión/metabolismo , Sirtuina 2/antagonistas & inhibidores , Proteína 1 de Transporte Vesicular de Glutamato/genética , Anhedonia/fisiología , Animales , Antidepresivos/farmacología , Depresión/genética , Modelos Animales de Enfermedad , Ácido Glutámico/metabolismo , Histona Desacetilasas/genética , Histona Desacetilasas/metabolismo , Imipramina/farmacología , Masculino , Ratones , Ratones Endogámicos C57BL , Corteza Prefrontal/metabolismo , Corteza Prefrontal/fisiología , ARN Mensajero/genética , ARN Mensajero/metabolismo , Serotonina/metabolismo , Sirtuina 2/metabolismo , Estrés Psicológico/genética , Estrés Psicológico/metabolismo , Proteína 1 de Transporte Vesicular de Glutamato/metabolismo
3.
Eur Neuropsychopharmacol ; 25(11): 2036-48, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26433268

RESUMEN

Changes in histone acetylation could contribute to the pathogenesis of depression and antidepressant therapy. Using the chronic social defeat stress (CSDS) model of depression and different antidepressant treatments we studied the regulation of histone deacetylases (Hdac׳s) and synaptic plasticity markers in the prefrontal cortex (PFC). Further, functional implication of identified Hdac׳s in brain plasticity was explored. Mice were exposed to CSDS (10 days) followed by saline or imipramine (4 weeks). PFC Hdac׳s mRNA abundance was studied and compared to human׳s. Further, protein expression of acetylated histones (AcH3 and AcH4), neuroplasticity markers (CREB and pro-BDNF) and selected Hdac׳s were analyzed. Moreover, other antidepressants (fluoxetine and reboxetine) and selective HDAC inhibitors were studied. CSDS increased Hdac5 and Sirt2 mRNA whereas repeated imipramine did the opposite. Accordingly, stress and imipramine induced opposite changes on AcH3, AcH4 and CREB expression. At protein level, CSDS upregulated nuclear fraction of Hdac5 and repeated imipramine and reboxetine increased its phosphorylated form (p-Hdac5), mainly located in the cytoplasm. Moreover, Sirt2 was downregulated by all monoaminergic antidepressants. Further, repeated treatment with the class IIa Hdac inhibitor MC1568 and the Sirt2 inhibitor 33i for three weeks increased synaptic plasticity in the prefrontal cortex. Our results suggest that Hdac5 and Sirt2 upregulation could constitute stable stress-induced neuronal adaptations. Noteworthy, the SIRT2 upregulation in depressed patients supports the interest of this target for therapeutic intervention. On the other hand, cytoplasmic Hdac5 export and Sirt2 downregulation induced by monoaminergic antidepressants could contribute to the well-known beneficial effects of antidepressants on brain plasticity.


Asunto(s)
Antidepresivos/farmacología , Histona Desacetilasas/metabolismo , Plasticidad Neuronal/fisiología , Sirtuinas/metabolismo , Estrés Psicológico/fisiopatología , Adulto , Animales , Enfermedad Crónica , Trastorno Depresivo/metabolismo , Fluoxetina/farmacología , Inhibidores de Histona Desacetilasas/farmacología , Humanos , Imipramina/farmacología , Masculino , Ratones Endogámicos C57BL , Persona de Mediana Edad , Morfolinas/farmacología , Plasticidad Neuronal/efectos de los fármacos , Corteza Prefrontal/efectos de los fármacos , Corteza Prefrontal/metabolismo , ARN Mensajero/metabolismo , Reboxetina , Estrés Psicológico/tratamiento farmacológico
4.
Pharmacol Biochem Behav ; 135: 227-36, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-26051025

RESUMEN

Many studies suggest that the prefrontal cortex (PFC) is a target limbic region for stress response because a dysfunction here is linked to anhedonia, a decrease in reactivity to rewards, and to anxiety. It is suggested that stress-induced persistent molecular changes in this brain region could bring some light on the mechanisms perpetuating depressive episodes. In order to address this issue, here we have studied the long-term PFC gene expression pattern and behavioral effects induced by a chronic mild stress (CMS) model and antidepressant treatment in mice. CMS was applied to mice for six weeks and imipramine (10mg/kg, i.p.) or saline treatment was administered for five weeks starting from the third week of CMS. Mice were sacrificed one month after CMS and following two weeks after the discontinuation of drug treatment and the PFC was dissected and prepared for gene (mRNA) and protein expression studies. Using the same experimental design, a separate group of mice was tested for anhedonia, recognition memory, social interaction and anxiety. CMS induced a long-term altered gene expression profile in the PFC that was partially reverted by imipramine. Specifically, the circadian rhythm signaling pathway and functions such as gene expression, cell proliferation, survival and apoptosis as well as neurological and psychiatric disorders were affected. Of these, some changes of the circadian rhythm pathway (Hdac5, Per1, and Per2) were validated by RT-PCR and western-blot. Moreover, CMS induced long-lasting anhedonia that was reverted by imipramine treatment. Impaired memory, decreased social interaction and anxiety behavior were also induced by chronic stress. We have identified in the PFC molecular targets oppositely regulated by CMS and imipramine that could be relevant for chronic depression and antidepressant action. Among these, a possible candidate for further investigation could be the circadian rhythm pathway.


Asunto(s)
Antidepresivos Tricíclicos/farmacología , Conducta Animal/efectos de los fármacos , Expresión Génica/efectos de los fármacos , Imipramina/farmacología , Corteza Prefrontal/metabolismo , Estrés Psicológico/genética , Estrés Psicológico/psicología , Anhedonia/efectos de los fármacos , Animales , Ansiedad/psicología , Ritmo Circadiano/efectos de los fármacos , Histona Desacetilasas/biosíntesis , Histona Desacetilasas/genética , Relaciones Interpersonales , Masculino , Memoria/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Corteza Prefrontal/efectos de los fármacos , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Reconocimiento en Psicología/efectos de los fármacos
5.
Eur Neuropsychopharmacol ; 23(7): 697-708, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22743048

RESUMEN

Major depression is a mental disorder often preceded by exposure to chronic stress or stressful life events. Recently, animal models based on social conflict such as chronic social defeat stress (CSDS) are proposed to be more relevant to stress-induced human psychopathology compared to environmental models like the chronic mild stress (CMS). However, while CMS reproduces specifically core depressive symptoms such as anhedonia and helplessness, CSDS studies rely on the analysis of stress-induced social avoidance, addressing different neuropsychiatric disorders. Here, we study comparatively the two models from a behavioural and neurochemical approach and their possible relevance to human depression. Mice (C57BL/6) were exposed to CMS or CSDS for six weeks and ten days. Anhedonia was periodically evaluated. A battery of test applied during the fourth week after the stress procedure included motor activity, memory, anxiety, social interaction and helplessness. Subsequently, we examined glutamate, GABA, 5-HT and dopamine levels in the prefrontal cortex, hippocampus and brainstem. CMS induced a clear depressive-like profile including anhedonia, helplessness and memory impairment. CSDS induced anhedonia, hyperactivity, anxiety and social avoidance, signs also common to anxiety and posttraumatic stress disorders. While both models disrupted the excitatory inhibitory balance in the prefrontal cortex, CMS altered importantly this balance in the brainstem. Moreover, CSDS decreased dopamine in the prefrontal cortex and brainstem. We suggests that while depressive-like behaviours might be associated to altered aminoacid neurotransmission in cortical and brain stem areas, CSDS induced anxiety behaviours might be linked to specific alteration of dopaminergic pathways involved in rewarding processes.


Asunto(s)
Conducta Animal , Depresión/metabolismo , Depresión/psicología , Modelos Animales de Enfermedad , Neurotransmisores/metabolismo , Predominio Social , Estrés Psicológico/metabolismo , Estrés Psicológico/psicología , Animales , Ansiedad/metabolismo , Ansiedad/psicología , Tronco Encefálico/metabolismo , Dopamina/metabolismo , Ácido Glutámico/metabolismo , Hipocampo/metabolismo , Masculino , Ratones , Corteza Prefrontal/metabolismo , Serotonina/metabolismo , Trastornos por Estrés Postraumático/metabolismo , Trastornos por Estrés Postraumático/psicología , Ácido gamma-Aminobutírico/metabolismo
6.
Psychopharmacology (Berl) ; 224(2): 313-25, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22707231

RESUMEN

RATIONALE: Chronic social defeat stress (CSDS) has been proposed as a model of depression. However, most CSDS studies rely only on the analysis of stress-induced social avoidance. Moreover, the predictive validity of the model has been poorly analyzed, let alone its interaction with biological risk factors. OBJECTIVES: Here, we explore the validity of CSDS as a depression model. Further, the effect of decreased vesicular glutamate transporter 1 (VGLUT1), as a potential factor enhancing a depressive-like phenotype, was studied. METHODS: Mice were exposed to CSDS (10 days) followed by saline, venlafaxine, fluoxetine, or tianeptine treatment (30 days). The battery of behaviors included motor activity, memory, anxiety, social interaction, helplessness, and anhedonic-like behavior. Moreover, the behavioral effect of CSDS in VGLUT1 heterozygous (VGLUT1+/-) mice was studied, as well as the regulation of VGLUT1 mRNA. RESULTS: CSDS induced anhedonia, helplessness, hyperactivity, anxiety, social avoidance, and freezing, as well as downregulation of VGLUT1 mRNA in the amygdala. Repeated venlafaxine showed antidepressant-like activity and both venlafaxine and tianeptine behaved as effective anxiolytics. CSDS-induced social avoidance was reverted by tianeptine. Fluoxetine failed to revert most of the behavioral alterations. VGLUT1+/- mice showed an enhanced vulnerability to stress-induced social avoidance. CONCLUSION: We suggest that CSDS is not a pure model of depression. Indeed, it addresses relevant aspects of anxiety-related disorders. Firstly, CSDS-induced anhedonia and social avoidance are not associated in this model. Moreover, CSDS might be affecting brain areas mainly involved in the processing of social behavior, such as the amygdala, where the glutamatergic mechanism could play a key role.


Asunto(s)
Antidepresivos/farmacología , Estrés Psicológico/tratamiento farmacológico , Estrés Psicológico/genética , Animales , Ansiedad/tratamiento farmacológico , Ansiedad/genética , Conducta Animal , Ciclohexanoles/farmacología , Depresión/tratamiento farmacológico , Depresión/genética , Trastorno Depresivo/tratamiento farmacológico , Trastorno Depresivo/genética , Modelos Animales de Enfermedad , Fluoxetina/farmacología , Masculino , Ratones , Ratones Endogámicos C57BL , ARN Mensajero/biosíntesis , Factores de Riesgo , Conducta Social , Tiazepinas/farmacología , Clorhidrato de Venlafaxina , Proteína 1 de Transporte Vesicular de Glutamato
7.
Eur Neuropsychopharmacol ; 21(1): 23-32, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-20937555

RESUMEN

Major depression might originate from both environmental and genetic risk factors. The environmental chronic mild stress (CMS) model mimics some environmental factors contributing to human depression and induces anhedonia and helplessness. Mice heterozygous for the synaptic vesicle protein (SVP) vesicular glutamate transporter 1 (VGLUT1) have been proposed as a genetic model of deficient glutamate function linked to depressive-like behaviour. Here, we aimed to identify, in these two experimental models, gene expression changes in the frontal cortex, common to stress and impaired glutamate function. Both VGLUT1(+/-) and CMS mice showed helpless and anhedonic-like behavior. Microarray studies in VGLUT1(+/-) mice revealed regulation of genes involved in apoptosis, neurogenesis, synaptic transmission, protein metabolic process or learning and memory. In addition, RT-PCR studies confirmed gene expression changes in several glutamate, GABA, dopamine and serotonin neurotransmitter receptors. On the other hand, CMS affected the regulation of 147 transcripts, some of them involved in response to stress and oxidoreductase activity. Interestingly, 52 genes were similarly regulated in both models. Specifically, a dowregulation in genes that promote cell proliferation (Anapc7), cell growth (CsnK1g1), cell survival (Hdac3), and inhibition of apoptosis (Dido1) was observed. Genes linked to cytoskeleton (Hspg2, Invs), psychiatric disorders (Grin1, MapK12) or an antioxidant enzyme (Gpx2) were also downregulated. Moreover, genes that inhibit the MAPK pathways (Dusp14), stimulate oxidative metabolism (Eif4a2) and enhance glutamate transmission (Rab8b) were upregulated. We suggest that these genes could form part of the altered "molecular context" underlying depressive-like behaviour in animal models. The clinical relevance of these findings is discussed.


Asunto(s)
Depresión/genética , Trastorno Depresivo/genética , Lóbulo Frontal/metabolismo , Ácido Glutámico/genética , Estrés Psicológico/genética , Proteína 1 de Transporte Vesicular de Glutamato/genética , Animales , Conducta Animal , Depresión/fisiopatología , Trastorno Depresivo/metabolismo , Modelos Animales de Enfermedad , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Ácido Glutámico/metabolismo , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Análisis por Micromatrices , Trastornos del Humor/genética , Neurotransmisores/genética , Neurotransmisores/metabolismo , Fenotipo , Placer , ARN/análisis , Estrés Psicológico/metabolismo , Sacarosa , Factores de Tiempo , Proteína 1 de Transporte Vesicular de Glutamato/deficiencia , Proteína 1 de Transporte Vesicular de Glutamato/metabolismo
8.
Psychopharmacology (Berl) ; 199(1): 1-14, 2008 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-18470507

RESUMEN

RATIONALE: Many studies support the validity of the chronic mild stress (CMS) model of depression in rodents. However, most of them focus on analysis of reactivity to rewards during the CMS and/or depressive-like behavior shortly after stress. In this study, we investigate acute and long-term effects of CMS and antidepressant treatment on depressive, anxiety-like behavior and learning. MATERIALS AND METHODS: Mice (C57BL/6) were exposed to CMS for 6 weeks and anhedonia was evaluated by weekly monitoring of sucrose intake. Paroxetine (10 mg kg(-1)day(-1) i.p.) or saline were administered the last 3 weeks of CMS and continued for 2 weeks thereafter. Behavioral tests were performed over the last week of CMS (acute effects) and 1 month later (long-term effects). RESULTS: Mice exposed to CMS displayed both acute and long-term decreased sucrose intake, increased immobility in the forced swimming test (FST) and impaired memory in the novel object recognition test. It is interesting to note that a correlation was found between the cognitive deficits and the helpless behavior in the FST induced by CMS. During the CMS procedure, paroxetine treatment reverted partially recognition memory impairment but failed to prevent the increased immobility in the FST. Moreover, it decreased on its own sucrose intake. Importantly, the long-term effects of CMS were partially prevented by chronic paroxetine. CONCLUSIONS: CMS leads to a long-term altered behavioral profile that could be partially reverted by chronic antidepressant treatment. This study brings novel features regarding the long-term effects of CMS and on the predictive validity of this depression animal model.


Asunto(s)
Antidepresivos de Segunda Generación/farmacología , Ansiedad/psicología , Depresión/psicología , Recuerdo Mental/efectos de los fármacos , Paroxetina/farmacología , Reconocimiento en Psicología/efectos de los fármacos , Estrés Psicológico/complicaciones , Animales , Ingestión de Líquidos/efectos de los fármacos , Reacción de Fuga/efectos de los fármacos , Conducta Exploratoria/efectos de los fármacos , Miedo/efectos de los fármacos , Desamparo Adquirido , Inyecciones Intraperitoneales , Cuidados a Largo Plazo , Masculino , Ratones , Motivación , Actividad Motora , Reconocimiento Visual de Modelos/efectos de los fármacos , Sacarosa/administración & dosificación , Gusto/efectos de los fármacos
9.
Eur J Neurosci ; 25(1): 281-90, 2007 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-17241289

RESUMEN

Three isoforms of a vesicular glutamate transporter (VGLUT1-3) have been identified. Of these, VGLUT1 is the major isoform in the cerebral cortex and hippocampus where it is selectively located on synaptic vesicles of excitatory glutamatergic terminals. Variations in VGLUT1 expression levels have a major impact on the efficacy of glutamate synaptic transmission. Given evidence linking alterations in glutamate neurotransmission to various neuropsychiatric disorders, we investigated the possible influence of a down-regulation of VGLUT1 transporter on anxiety, depressive-like behaviour and learning. The behavioural phenotype of VGLUT1-heterozygous mice (C57BL/6) was compared to wild-type (WT) littermates. Moreover, VGLUT1-3 expression, hippocampal excitatory terminal ultrastructure and neurochemical phenotype were analysed. VGLUT1-heterozygous mice displayed normal spontaneous locomotor activity, increased anxiety in the light-dark exploration test and depressive-like behaviour in the forced swimming test: no differences were shown in the elevated plus-maze model of anxiety. In the novel object recognition test, VGLUT1(+/-) mice showed normal short-term but impaired long-term memory. Spatial memory in the Morris water maze was unaffected. Western blot analysis confirmed that VGLUT1 heterozygotes expressed half the amount of transporter compared to WT. In addition, a reduction in the reserve pool of synaptic vesicles of hippocampal excitatory terminals and a 35-45% reduction in GABA in the frontal cortex and the hippocampus were observed in the mutant mice. These observations suggest that a VGLUT1-mediated presynaptic alteration of the glutamatergic synapses, in specific brain regions, leads to a behavioural phenotype resembling certain aspects of psychiatric and cognitive disorders.


Asunto(s)
Ansiedad/metabolismo , Depresión/metabolismo , Trastornos de la Memoria/metabolismo , Proteína 1 de Transporte Vesicular de Glutamato/deficiencia , Animales , Animales Recién Nacidos , Ansiedad/genética , Encéfalo/metabolismo , Encéfalo/ultraestructura , Depresión/genética , Conducta Exploratoria/fisiología , Femenino , Masculino , Aprendizaje por Laberinto/fisiología , Trastornos de la Memoria/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Microscopía Electrónica de Transmisión/métodos , Actividad Motora/genética , Neurotransmisores/metabolismo , Tiempo de Reacción/genética , Reconocimiento en Psicología/fisiología , Natación/fisiología , Sinapsis/genética , Sinapsis/ultraestructura
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...