Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 3 de 3
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Transl Psychiatry ; 9(1): 100, 2019 02 21.
Artículo en Inglés | MEDLINE | ID: mdl-30792491

RESUMEN

Post-traumatic stress disorder (PTSD) is a trauma- and stress-related disorder with dysregulated fear responses and neurobiological impairments, notably at neurotrophic and inflammation levels. Understanding the mechanisms underlying this disease is crucial to develop PTSD models that meet behavioral and neurobiological validity criteria as well as innovative therapeutic approaches. Serotonin 2C receptors (5-HT2CR) are known for their important role in anxiety, and mice having only the fully edited VGV isoform of 5-HT2CR, which thereby overexpressed brain 5-HT2CR, are of special interest to study PTSD predisposition. Innate and conditioned fear-related behaviors were assessed in VGV and wild-type mice. mRNA expression of brain-derived neurotrophic factor (BDNF), tissue-plasminogen activator (tPA), and pro-inflammatory cytokines (IL-6, IL-1ß, and calcineurin) were measured by qRT-PCR. The effect of acute and chronic paroxetine was evaluated on both behavior and gene expression. VGV mice displayed greater fear expression, extensive fear extinction deficits, and fear generalization. Paroxetine restored fear extinction in VGV mice when administered acutely and decreased innate fear and fear generalization when administered chronically. In parallel, Bdnf, tPA, and pro-inflammatory cytokines mRNA levels were dysregulated in VGV mice. Bdnf and tPA mRNA expression was decreased in the hippocampus but increased in the amygdala, and chronic paroxetine normalized Bdnf mRNA levels both in the amygdala and the hippocampus. Amygdalar calcineurin mRNA level in VGV mice was also normalized by chronic paroxetine. VGV-transgenic mice displayed behavioral and neurobiological features that could be accessory to the investigation of PTSD and its treatment. Furthermore, these data point out to the role of 5-HT2CR in neuroplasticity and neuroinflammation.


Asunto(s)
Factor Neurotrófico Derivado del Encéfalo/metabolismo , Citocinas/metabolismo , Paroxetina/farmacología , Edición de ARN , Receptor de Serotonina 5-HT2C/metabolismo , Trastornos por Estrés Postraumático/metabolismo , Amígdala del Cerebelo/metabolismo , Animales , Ansiedad/genética , Conducta Animal/efectos de los fármacos , Factor Neurotrófico Derivado del Encéfalo/genética , Modelos Animales de Enfermedad , Miedo , Hipocampo/metabolismo , Masculino , Aprendizaje por Laberinto , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , ARN Mensajero/genética , Receptor de Serotonina 5-HT2C/genética , Transducción de Señal , Trastornos por Estrés Postraumático/tratamiento farmacológico
2.
Br J Pharmacol ; 174(15): 2471-2483, 2017 08.
Artículo en Inglés | MEDLINE | ID: mdl-28493335

RESUMEN

BACKGROUND AND PURPOSE: It has recently been suggested that 5-HT3 receptor blockade enhances the efficacy of selective 5-HT (serotonin) reuptake inhibitor (SSRI) antidepressants and may reverse stress-induced deficits in rodents. EXPERIMENTAL APPROACH: To further explore this hypothesis, we used mice lacking the 5-HT3 receptor (Htr3a KO) and their wild-type (WT) controls to assess their response in behavioural paradigms relevant to anxiety and depression. Mice were studied under basal, antidepressant treatments and chronic social defeat stress (CSDS) conditions. KEY RESULTS: In basal conditions, Htr3a KO mice displayed anxiolytic- and antidepressant-like behaviours in the elevated plus maze, the social interaction and the forced swim tests (FST), but behaved as WT mice in response to acute citalopram in the FST. However, the effects of fluoxetine were blunted in Htr3a KO mice in these same tests. In an in vitro electrophysiological paradigm, a low-dose citalopram treatment triggered 5-HT1A receptor desensitization only in the dorsal raphe nucleus of Htr3a KO, although a high dose desensitized 5-HT1A autoreceptor function equally in Htr3a KO and WT mice, suggesting that citalopram may become effective at lower doses when 5-HT3 receptors are inactivated. In addition, Htr3a deletion blocked CSDS-induced modification in the cortical expression of two genes involved in oxidative stress, CaMKIIa and SOD1. CONCLUSIONS AND IMPLICATIONS: Taken together, these data show that Htr3a deletion promotes SSRI efficacy and prevents the occurrence of stress-induced deleterious effects, suggesting that the 5-HT3 receptor may represent an interesting target for the treatment of stress-related disorders.


Asunto(s)
Antidepresivos/farmacología , Citalopram/farmacología , Receptores de Serotonina 5-HT3/metabolismo , Conducta Social , Estrés Psicológico/tratamiento farmacológico , Animales , Antidepresivos/administración & dosificación , Citalopram/administración & dosificación , Relación Dosis-Respuesta a Droga , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Estrés Oxidativo/efectos de los fármacos , Receptores de Serotonina 5-HT3/deficiencia
3.
Sci Rep ; 8: 45907, 2017 04 04.
Artículo en Inglés | MEDLINE | ID: mdl-28374847

RESUMEN

Chronic stress is known to induce not only anxiety and depressive-like phenotypes in mice but also cognitive impairments, for which the action of classical antidepressant compounds remains unsatisfactory. In this context, we investigated the effects of chronic social defeat stress (CSDS) on anxiety-, social- and cognitive-related behaviors, as well as hippocampal Bdnf, synaptic plasticity markers (PSD-95, Synaptophysin, Spinophilin, Synapsin I and MAP-2), and epigenetic modifying enzymes (MYST2, HDAC2, HDAC6, MLL3, KDM5B, DNMT3B, GADD45B) gene expression in C57BL/6J mice. CSDS for 10 days provoked long-lasting anxious-like phenotype in the open field and episodic memory deficits in the novel object recognition test. While total Bdnf mRNA level was unchanged, Bdnf exon IV, MAP-2, HDAC2, HDAC6 and MLL3 gene expression was significantly decreased in the CSDS mouse hippocampus. In CSDS mice treated 3 weeks with 50 mg/kg/d agomelatine, an antidepressant with melatonergic receptor agonist and 5-HT2C receptor antagonist properties, the anxious-like phenotype was not reversed, but the treatment successfully prevented the cognitive impairments and hippocampal gene expression modifications. Altogether, these data evidenced that, in mice, agomelatine was effective in alleviating stress-induced altered cognitive functions, possibly through a mechanism involving BDNF signaling, synaptic plasticity and epigenetic remodeling.


Asunto(s)
Acetamidas/administración & dosificación , Disfunción Cognitiva/tratamiento farmacológico , Trastornos de la Memoria/tratamiento farmacológico , Estrés Psicológico/tratamiento farmacológico , Animales , Antidepresivos/administración & dosificación , Factor Neurotrófico Derivado del Encéfalo/genética , Disfunción Cognitiva/genética , Disfunción Cognitiva/patología , Trastorno Depresivo/tratamiento farmacológico , Trastorno Depresivo/genética , Trastorno Depresivo/patología , Modelos Animales de Enfermedad , Homólogo 4 de la Proteína Discs Large/genética , Regulación de la Expresión Génica/efectos de los fármacos , Hipocampo/metabolismo , Hipocampo/patología , Humanos , Trastornos de la Memoria/genética , Trastornos de la Memoria/patología , Ratones , Proteínas de Microfilamentos/genética , Proteínas del Tejido Nervioso/genética , Plasticidad Neuronal/genética , Estrés Psicológico/genética , Estrés Psicológico/patología , Sinapsinas/genética , Sinaptofisina/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...