38 research outputs found

    Abnormal expression of cortical cell cycle regulators underlying anxiety and depressive-like behavior in mice exposed to chronic stress

    Get PDF
    BackgroundThe cell cycle is a critical mechanism for proper cellular growth, development and viability. The p16INK4a and p21Waf1/Cip1 are important regulators of the cell cycle progression in response to internal and external stimuli (e.g., stress). Accumulating evidence indicates that the prefrontal cortex (PFC) is particularly vulnerable to stress, where stress induces, among others, molecular and morphological alterations, reflecting behavioral changes. Here, we investigated if the p16INK4a and p21Waf1/Cip1 expression are associated with behavioral outcomes.MethodsPrefrontal cortex mRNA and protein levels of p16INK4A and p21Waf1/Cip1 of mice (six independent groups of C57BL/6J, eight mice/group, 50% female) exposed from 0 to 35 days of chronic restraint stress (CRS) were quantified by qPCR and Western Blot, respectively. Correlation analyses were used to investigate the associations between cyclin-dependent kinase inhibitors (CKIs) expression and anxiety- and depression-like behaviors.ResultsOur results showed that the PFC activated the cell cycle regulation pathways mediated by both CKIs p16INK4A and p21Waf1/Cip1 in mice exposed to CRS, with overall decreased mRNA expression and increased protein expression. Moreover, correlation analysis revealed that mRNA and protein levels are statistically significant correlated with anxiety and depressive-like behavior showing a greater effect in males than females.ConclusionOur present study extends the existing literature providing evidence that PFC cells respond to chronic stress exposure by overexpressing CKIs. Furthermore, our findings indicated that abnormal expression of p16INK4A and p21Waf1/Cip1 may significantly contribute to non-adaptive behavioral responses

    Decreased SGK1 Expression and Function Contributes to Behavioral Deficits Induced by Traumatic Stress

    No full text
    Exposure to extreme stress can trigger the development of major depressive disorder (MDD) as well as post-traumatic stress disorder (PTSD). The molecular mechanisms underlying the structural and functional alterations within corticolimbic brain regions, including the prefrontal cortex (PFC) and amygdala of individuals subjected to traumatic stress, remain unknown. In this study, we show that serum and glucocorticoid regulated kinase 1 (SGK1) expression is down-regulated in the postmortem PFC of PTSD subjects. Furthermore, we demonstrate that inhibition of SGK1 in the rat medial PFC results in helplessness- and anhedonic-like behaviors in rodent models. These behavioral changes are accompanied by abnormal dendritic spine morphology and synaptic dysfunction. Together, the results are consistent with the possibility that altered SGK1 signaling contributes to the behavioral and morphological phenotypes associated with traumatic stress pathophysiology

    Region- and phase-dependent effects of 5-HT(1A) and 5-HT(2C) receptor activation on adult neurogenesis.

    No full text
    International audienceAdult neurogenesis and serotoninergic transmission are associated to mood disorders and their treatments. The present study focused on the effects of chronic activation of 5-HT(1A) and 5-HT(2C) receptors on newborn cell survival in the dentate gyrus (DG) and olfactory bulb (OB), and examined whether potential neurogenic zones as the prefrontal cortex (PFC) and striatum (ST) are reactive to these treatments. Administration of 8-OH-DPAT, but not RO600,175 increases neurogenesis and survival of late differentiating cells (15-21days) in the DG. Both 8-OH-DPAT and RO600,175 increase neurogenesis in the OB, but only 8-OH-DPAT affected cell survival, inducing a parallel decrease in the number of BrdU cells in the OB and increase in the SVZ, which suggests an impaired migration. In the PFC and ST, 8-OH-DPAT and R0600,175 increase gliogenesis (NG2-labeled cells). This study provides new insights on the serotonergic regulation of critical phases of neurogenesis helpful to understand the neurogenic and gliogenic effects of antidepressant treatments in different brain regions

    Dépression et neuroplasticité : implication des systèmes sérotoninergiques

    No full text
    L'analyse des effets des antidépresseurs sur la neuroplasticité, et plus particulièrement sur la formation de nouveaux neurones dans le cerveau adulte, constitue aujourd'hui un thème de recherche novateur compte tenu de l'implication d'une modification de la connectivité neuronale dans la physiopathologie des troubles de l'humeur. Les modèles animaux de stress chronique ont permis de révéler des altérations structurales au niveau de l'hippocampe notamment, qui sont réversibles avec le temps et les traitements antidépresseurs. Chez des patients atteints de dépression majeure, on observe aussi une diminution du volume hippocampique qui a été associée à des déficits cognitifs. Dans ce contexte, la sérotonine (5-HT) apparaît jouer un rôle clé, par ses effets notables dans la réponse au stress et le traitement de la dépression, comme dans son implication dans les processus de la neuroplasticité et sa faculté de stimuler la neurogenèse secondaire. D'autres antidépresseurs, comme l'agomélatine ayant des propriétés à la fois sérotoninergique et mélatoninergique, entraînent aussi une augmentation de ce processus, et bien que ces observations ne permettent pas pour l'instant de proposer un mécanisme sous-jacent commun à l'ensemble de ces effets, leurs conséquences pourraient avoir une implication importante dans le traitement des troubles de l'humeur

    Data_Sheet_1_Abnormal expression of cortical cell cycle regulators underlying anxiety and depressive-like behavior in mice exposed to chronic stress.PDF

    No full text
    BackgroundThe cell cycle is a critical mechanism for proper cellular growth, development and viability. The p16INK4a and p21Waf1/Cip1 are important regulators of the cell cycle progression in response to internal and external stimuli (e.g., stress). Accumulating evidence indicates that the prefrontal cortex (PFC) is particularly vulnerable to stress, where stress induces, among others, molecular and morphological alterations, reflecting behavioral changes. Here, we investigated if the p16INK4a and p21Waf1/Cip1 expression are associated with behavioral outcomes.MethodsPrefrontal cortex mRNA and protein levels of p16INK4A and p21Waf1/Cip1 of mice (six independent groups of C57BL/6J, eight mice/group, 50% female) exposed from 0 to 35 days of chronic restraint stress (CRS) were quantified by qPCR and Western Blot, respectively. Correlation analyses were used to investigate the associations between cyclin-dependent kinase inhibitors (CKIs) expression and anxiety- and depression-like behaviors.ResultsOur results showed that the PFC activated the cell cycle regulation pathways mediated by both CKIs p16INK4A and p21Waf1/Cip1 in mice exposed to CRS, with overall decreased mRNA expression and increased protein expression. Moreover, correlation analysis revealed that mRNA and protein levels are statistically significant correlated with anxiety and depressive-like behavior showing a greater effect in males than females.ConclusionOur present study extends the existing literature providing evidence that PFC cells respond to chronic stress exposure by overexpressing CKIs. Furthermore, our findings indicated that abnormal expression of p16INK4A and p21Waf1/Cip1 may significantly contribute to non-adaptive behavioral responses.</p

    Chronic agomelatine treatment corrects behavioral, cellular, and biochemical abnormalities induced by prenatal stress in rats.

    No full text
    International audienceRATIONALE AND OBJECTIVES: The rat model of prenatal restraint stress (PRS) replicates factors that are implicated in the etiology of anxious/depressive disorders. We used this model to test the therapeutic efficacy of agomelatine, a novel antidepressant that behaves as a mixed MT1/MT2 melatonin receptor agonist/5-HT(2c) serotonin receptor antagonist. RESULTS: Adult PRS rats showed behavioral, cellular, and biochemical abnormalities that were consistent with an anxious/depressive phenotype. These included an increased immobility in the forced swim test, an anxiety-like behavior in the elevated plus maze, reduced hippocampal levels of phosphorylated cAMP-responsive element binding protein (p-CREB), reduced hippocampal levels of mGlu2/3 and mGlu5 metabotropic glutamate receptors, and reduced neurogenesis in the ventral hippocampus, the specific portion of the hippocampus that encodes memories related to stress and emotions. All of these changes were reversed by a 3- or 6-week treatment with agomelatine (40-50 mg/kg, i.p., once a day). Remarkably, agomelatine had no effect in age-matched control rats, thereby behaving as a "disease-dependent" drug. CONCLUSIONS: These data indicate that agomelatine did not act on individual symptoms but corrected all aspects of the pathological epigenetic programming triggered by PRS. Our findings strongly support the antidepressant activity of agomelatine and suggest that the drug impacts mechanisms that lie at the core of anxious/depressive disorders
    corecore