Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Transl Psychiatry ; 7(6): e1146, 2017 06 06.
Article in English | MEDLINE | ID: mdl-28585931

ABSTRACT

Depression is a prevalent psychiatric disorder with an increasing impact in global public health. However, a large proportion of patients treated with currently available antidepressant drugs fail to achieve remission. Recently, antipsychotic drugs have received approval for the treatment of antidepressant-resistant forms of major depression. The modulation of adult neuroplasticity, namely hippocampal neurogenesis and neuronal remodeling, has been considered to have a key role in the therapeutic effects of antidepressants. However, the impact of antipsychotic drugs on these neuroplastic mechanisms remains largely unexplored. In this study, an unpredictable chronic mild stress protocol was used to induce a depressive-like phenotype in rats. In the last 3 weeks of stress exposure, animals were treated with two different antipsychotics: haloperidol (a classical antipsychotic) and clozapine (an atypical antipsychotic). We demonstrated that clozapine improved both measures of depressive-like behavior (behavior despair and anhedonia), whereas haloperidol aggravated learned helplessness in the forced-swimming test and behavior flexibility in a cognitive task. Importantly, an upregulation of adult neurogenesis and neuronal survival was observed in animals treated with clozapine, whereas haloperidol promoted a downregulation of these processes. Furthermore, clozapine was able to re-establish the stress-induced impairments in neuronal structure and gene expression in the hippocampus and prefrontal cortex. These results demonstrate the modulation of adult neuroplasticity by antipsychotics in an animal model of depression, revealing that the atypical antipsychotic drug clozapine reverts the behavioral effects of chronic stress by improving adult neurogenesis, cell survival and neuronal reorganization.


Subject(s)
Affect/drug effects , Antipsychotic Agents/pharmacology , Behavior, Animal/drug effects , Clozapine/pharmacology , Haloperidol/pharmacology , Neuronal Plasticity/drug effects , Animals , Antidepressive Agents/pharmacology , Antidepressive Agents/therapeutic use , Antipsychotic Agents/therapeutic use , Cell Survival , Clozapine/therapeutic use , Depression/drug therapy , Disease Models, Animal , Haloperidol/therapeutic use , Hippocampus/drug effects , Male , Neurogenesis/drug effects , Prefrontal Cortex/drug effects , Rats , Rats, Wistar , Swimming
2.
Mol Psychiatry ; 22(8): 1110-1118, 2017 08.
Article in English | MEDLINE | ID: mdl-28555078

ABSTRACT

Stress, a well-known sculptor of brain plasticity, is shown to suppress hippocampal neurogenesis in the adult brain; yet, the underlying cellular mechanisms are poorly investigated. Previous studies have shown that chronic stress triggers hyperphosphorylation and accumulation of the cytoskeletal protein Tau, a process that may impair the cytoskeleton-regulating role(s) of this protein with impact on neuronal function. Here, we analyzed the role of Tau on stress-driven suppression of neurogenesis in the adult dentate gyrus (DG) using animals lacking Tau (Tau-knockout; Tau-KO) and wild-type (WT) littermates. Unlike WTs, Tau-KO animals exposed to chronic stress did not exhibit reduction in DG proliferating cells, neuroblasts and newborn neurons; however, newborn astrocytes were similarly decreased in both Tau-KO and WT mice. In addition, chronic stress reduced phosphoinositide 3-kinase (PI3K)/mammalian target of rapamycin (mTOR)/glycogen synthase kinase-3ß (GSK3ß)/ß-catenin signaling, known to regulate cell survival and proliferation, in the DG of WT, but not Tau-KO, animals. These data establish Tau as a critical regulator of the cellular cascades underlying stress deficits on hippocampal neurogenesis in the adult brain.


Subject(s)
Neurogenesis/physiology , tau Proteins/metabolism , Animals , Astrocytes/metabolism , Cell Proliferation , Cell Survival , Dentate Gyrus/metabolism , Disease Models, Animal , Glycogen Synthase Kinase 3/metabolism , Hippocampus/metabolism , Male , Mice , Neuronal Plasticity/physiology , Neurons/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation , Signal Transduction , Stress, Physiological , beta Catenin/metabolism
3.
Transl Psychiatry ; 7(3): e1058, 2017 03 14.
Article in English | MEDLINE | ID: mdl-28291258

ABSTRACT

Depression is a highly prevalent and recurrent neuropsychiatric disorder associated with alterations in emotional and cognitive domains. Neuroplastic phenomena are increasingly considered central to the etiopathogenesis of and recovery from depression. Nevertheless, a high number of remitted patients experience recurrent episodes of depression, remaining unclear how previous episodes impact on behavior and neuroplasticity and/or whether modulation of neuroplasticity is important to prevent recurrent depression. Through re-exposure to an unpredictable chronic mild stress protocol in rats, we observed the re-appearance of emotional and cognitive deficits. Furthermore, treatment with the antidepressants fluoxetine and imipramine was effective to promote sustained reversion of a depressive-like phenotype; however, their differential impact on adult hippocampal neuroplasticity triggered a distinct response to stress re-exposure: while imipramine re-established hippocampal neurogenesis and neuronal dendritic arborization contributing to resilience to recurrent depressive-like behavior, stress re-exposure in fluoxetine-treated animals resulted in an overproduction of adult-born neurons along with neuronal atrophy of granule neurons, accounting for an increased susceptibility to recurrent behavioral changes typical of depression. Strikingly, cell proliferation arrest compromised the behavior resilience induced by imipramine and buffered the susceptibility to recurrent behavioral changes promoted by fluoxetine. This study shows that previous exposure to a depressive-like episode impacts on the behavioral and neuroanatomical changes triggered by subsequent re-exposure to similar experimental conditions and reveals that the proper control of adult hippocampal neuroplasticity triggered by antidepressants is essential to counteract recurrent depressive-like episodes.


Subject(s)
Antidepressive Agents/pharmacology , Behavior, Animal/drug effects , Depressive Disorder , Fluoxetine/pharmacology , Hippocampus/drug effects , Imipramine/pharmacology , Neuronal Plasticity/drug effects , Stress, Psychological , Animals , Disease Models, Animal , Disease Susceptibility , Hippocampus/metabolism , Male , Neuronal Plasticity/genetics , Rats , Rats, Wistar , Recurrence
4.
Transl Psychiatry ; 3: e266, 2013 Jun 04.
Article in English | MEDLINE | ID: mdl-23736119

ABSTRACT

There is accumulating evidence that the nucleus accumbens (NAc) has an important role in the pathophysiology of depression. As the NAc is a key component in the neural circuitry of reward, it has been hypothesized that anhedonia, a core symptom of depression, might be related to dysfunction of this brain region. Neuronal morphology and expression of plasticity-related molecules were examined in the NAc of rats displaying anhedonic behavior (measured in the sucrose-consumption test) in response to chronic mild stress. To demonstrate the relevance of our measurements to depression, we tested whether the observed changes were sensitive to reversal with antidepressants (imipramine and fluoxetine). Data show that animals displaying anhedonic behavior display an hypertrophy of medium spiny neurons in the NAc and, in parallel, have increased expression of the genes encoding for brain-derived neurotrophic factor, neural cell adhesion molecule and synaptic protein synapsin 1. Importantly, the reversal of stress-induced anhedonia by antidepressants is linked to a restoration of gene-expression patterns and dendritic morphology in the NAc. Using an animal model of depression, we show that stress induces anhedonic behavior that is associated with specific changes in the neuronal morphology and in the gene-expression profile of the NAc that are effectively reversed after treatment with antidepressants.


Subject(s)
Anhedonia/physiology , Nucleus Accumbens/pathology , Stress, Psychological/complications , Anhedonia/drug effects , Animals , Antidepressive Agents/pharmacology , Behavior, Animal , Fluoxetine/pharmacology , Hypertrophy , Imipramine/pharmacology , Male , Nucleus Accumbens/cytology , Nucleus Accumbens/metabolism , Rats , Rats, Wistar , Real-Time Polymerase Chain Reaction , Stress, Psychological/pathology
5.
Mol Psychiatry ; 18(7): 748-50, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23711984

ABSTRACT

Brain neuroplasticity is increasingly considered to be an important component of both the pathology and treatment of depressive spectrum disorders. Recent studies shed light on the relevance of hippocampal cell genesis and cortico-limbic dendritic plasticity for the development and remission from depressive-like behavior. However, the neurobiological significance of neuroplastic phenomena in this context is still controversial. Here we summarize recent developments in this topic and propose an integrative interpretation of data gathered so far.


Subject(s)
Dendrites/physiology , Depression/physiopathology , Neurogenesis/physiology , Neuronal Plasticity/physiology , Humans , Models, Neurological , Remission Induction
6.
Front Psychiatry ; 4: 21, 2013.
Article in English | MEDLINE | ID: mdl-23565098

ABSTRACT

Obsessive-compulsive disorder (OCD) is achronic psychiatric disorder characterized by recurrent intrusive thoughts and/or repetitive compulsory behaviors. This psychiatric disorder is known to be stress responsive, as symptoms increase during periods of stress but also because stressful events may precede the onset of OCD. However, only a few and inconsistent reports have been published about the stress perception and the stress-response in these patients. Herein, we have characterized the correlations of OCD symptoms with basal serum cortisol levels and scores in a stress perceived questionnaire (PSS-10). The present data reveals that cortisol levels and the stress scores in the PSS-10 were significantly higher in OCD patients that in controls. Moreover, stress levels self-reported by patients using the PSS-10 correlated positively with OCD severity in the Yale-Brown Obsessive-Compulsive Scale (Y-BOCS). Interestingly, PSS-10 scores correlated with the obsessive component, but not with the compulsive component, of Y-BOCS. These results confirm that stress is relevant in the context of OCD, particularly for the obsessive symptomatology.

7.
Transl Psychiatry ; 3: e210, 2013 Jan 15.
Article in English | MEDLINE | ID: mdl-23321807

ABSTRACT

Impairment of hippocampal neurogenesis has been associated with the expression of depressive-like symptoms and some studies have suggested neurogenesis as a critical factor in the normalization of behavior by antidepressant (AD) drugs. This study provides robust evidence that ongoing neurogenesis is essential for the maintenance of behavioral homeostasis and that its pharmacological arrest precipitates symptoms commonly found in depressed patients. Further, the incorporation of newly born neurons and astrocytes into the preexisting hippocampal neurocircuitry is shown to be necessary for the spontaneous recovery from the adverse effects of stress and for long-term benefits of AD treatments.


Subject(s)
Antidepressive Agents/pharmacology , Depression/drug therapy , Fluoxetine/pharmacology , Hippocampus/drug effects , Imipramine/pharmacology , Neurogenesis/drug effects , Neurons/drug effects , Analysis of Variance , Animals , Antidepressive Agents/metabolism , Astrocytes/drug effects , Astrocytes/pathology , Behavior, Animal/drug effects , Cell Proliferation/drug effects , Conditioning, Psychological , Depression/pathology , Fluoxetine/metabolism , Hippocampus/pathology , Imipramine/metabolism , Male , Methylazoxymethanol Acetate/analogs & derivatives , Methylazoxymethanol Acetate/pharmacology , Neurons/pathology , Rats , Rats, Wistar , Stress, Psychological/metabolism , Stress, Psychological/pathology
8.
Mol Psychiatry ; 14(8): 764-73, 739, 2009 Aug.
Article in English | MEDLINE | ID: mdl-18982002

ABSTRACT

The mechanisms underlying the initiation/onset of, and the recovery from, depression are still largely unknown; views that neurogenesis in the hippocampus may be important for the pathogenesis and amelioration of depressive symptoms have gained currency over the years although the original evidence has been challenged. In this study, an unpredictable chronic mild stress protocol was used to induce a depressive-like phenotype in rats. In the last 2 weeks of stress exposure, animals were treated with the antidepressants fluoxetine, imipramine, CP 156,526 or SSR 1494515, alone or combined with methylazoxymethanol, a cytostatic agent used to arrest neurogenesis. We found that antidepressants retain their therapeutic efficacy in reducing both measured indices of depression-like behavior (learned helplessness and anhedonia), even when neurogenesis is blocked. Instead, our experiments suggest re-establishment of neuronal plasticity (dendritic remodeling and synaptic contacts) in the hippocampus and prefrontal cortex, rather than neurogenesis, as the basis for the restoration of behavioral homeostasis by antidepressants.


Subject(s)
Affect/drug effects , Antidepressive Agents/pharmacology , Depression/drug therapy , Hippocampus/drug effects , Neuronal Plasticity/drug effects , Affect/physiology , Analysis of Variance , Animals , Antidiuretic Hormone Receptor Antagonists , Behavior, Animal/drug effects , Behavior, Animal/physiology , Cytostatic Agents/pharmacology , Depression/etiology , Drug Combinations , Fluoxetine/pharmacology , Hippocampus/cytology , Hippocampus/physiology , Imipramine/pharmacology , Indoles/pharmacology , Male , Methylazoxymethanol Acetate/analogs & derivatives , Methylazoxymethanol Acetate/pharmacology , Mitosis/drug effects , Neurogenesis/drug effects , Neurogenesis/physiology , Neuronal Plasticity/physiology , Prefrontal Cortex/cytology , Prefrontal Cortex/drug effects , Prefrontal Cortex/physiology , Pyrrolidines/pharmacology , Rats , Rats, Wistar , Receptors, Corticotropin-Releasing Hormone/antagonists & inhibitors , Stress, Psychological/complications , Stress, Psychological/physiopathology
9.
Behav Brain Res ; 162(1): 135-42, 2005 Jul 01.
Article in English | MEDLINE | ID: mdl-15922074

ABSTRACT

Several variables, including age, are known to influence anxiety. Previous exposure to the elevated-plus maze (EPM) is known to modify emotional behaviour as retesting in the EPM at a standard age of 3 months increases open-arm avoidance and attenuates the effects of anxiolytic drugs. This study analysed whether similar results are obtained when older animals are subjected to these experimental paradigms. Overall, increasing age was associated with more signs of anxiety. Additionally, we observed a paradoxical behaviour pattern in aged-subjects that were re-exposed to the EPM, with mid-aged and old rats failing to display open arm avoidance (OAA) in the second trial; this qualitative shift in emotional behaviour was not associated with decreased locomotion. An examination of how age influences responsiveness to anxiolytic drugs, with or without previous maze experience, was also conducted. Midazolam (0.5 and 1 mg/kg) proved anxiolytic in maize-naive young animals; in marked contrast, in older animals midazolam at 1 mg/kg resulted in sedation but not anxiolyis. One trial tolerance to midazolam was evident in animals of both ages that were subjected to a second EPM trial; the latter phenomenon was apparently accentuated in older animals as they do not show open arm avoidance upon re-exposure to the EPM. These data suggest that the age-associated 'resistance' to anxiolytic drugs might be related to a qualitative shift in emotional behaviour.


Subject(s)
Aging/physiology , Anxiety/physiopathology , Behavior, Animal/physiology , Emotions/physiology , Maze Learning/physiology , Analysis of Variance , Animals , Anti-Anxiety Agents/pharmacology , Anxiety/drug therapy , Avoidance Learning/drug effects , Avoidance Learning/physiology , Behavior, Animal/drug effects , Drug Tolerance , Escape Reaction/drug effects , Escape Reaction/physiology , Exploratory Behavior/drug effects , Male , Maze Learning/drug effects , Midazolam/pharmacology , Rats , Rats, Wistar , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...