Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 3 de 3
Filter
Add more filters










Database
Language
Publication year range
1.
Transl Psychiatry ; 9(1): 100, 2019 02 21.
Article in English | MEDLINE | ID: mdl-30792491

ABSTRACT

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.


Subject(s)
Brain-Derived Neurotrophic Factor/metabolism , Cytokines/metabolism , Paroxetine/pharmacology , RNA Editing , Receptor, Serotonin, 5-HT2C/metabolism , Stress Disorders, Post-Traumatic/metabolism , Amygdala/metabolism , Animals , Anxiety/genetics , Behavior, Animal/drug effects , Brain-Derived Neurotrophic Factor/genetics , Disease Models, Animal , Fear , Hippocampus/metabolism , Male , Maze Learning , Mice , Mice, Inbred C57BL , Mice, Transgenic , RNA, Messenger/genetics , Receptor, Serotonin, 5-HT2C/genetics , Signal Transduction , Stress Disorders, Post-Traumatic/drug therapy
2.
Br J Pharmacol ; 174(15): 2471-2483, 2017 08.
Article in English | MEDLINE | ID: mdl-28493335

ABSTRACT

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.


Subject(s)
Antidepressive Agents/pharmacology , Citalopram/pharmacology , Receptors, Serotonin, 5-HT3/metabolism , Social Behavior , Stress, Psychological/drug therapy , Animals , Antidepressive Agents/administration & dosage , Citalopram/administration & dosage , Dose-Response Relationship, Drug , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Oxidative Stress/drug effects , Receptors, Serotonin, 5-HT3/deficiency
3.
Sci Rep ; 8: 45907, 2017 04 04.
Article in English | MEDLINE | ID: mdl-28374847

ABSTRACT

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.


Subject(s)
Acetamides/administration & dosage , Cognitive Dysfunction/drug therapy , Memory Disorders/drug therapy , Stress, Psychological/drug therapy , Animals , Antidepressive Agents/administration & dosage , Brain-Derived Neurotrophic Factor/genetics , Cognitive Dysfunction/genetics , Cognitive Dysfunction/pathology , Depressive Disorder/drug therapy , Depressive Disorder/genetics , Depressive Disorder/pathology , Disease Models, Animal , Disks Large Homolog 4 Protein/genetics , Gene Expression Regulation/drug effects , Hippocampus/metabolism , Hippocampus/pathology , Humans , Memory Disorders/genetics , Memory Disorders/pathology , Mice , Microfilament Proteins/genetics , Nerve Tissue Proteins/genetics , Neuronal Plasticity/genetics , Stress, Psychological/genetics , Stress, Psychological/pathology , Synapsins/genetics , Synaptophysin/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...