Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
1.
Pharmaceutics ; 13(12)2021 Dec 08.
Article in English | MEDLINE | ID: mdl-34959395

ABSTRACT

Despite promising initial reports, corticotropin-releasing factor receptor type-1 (CRF-R1) antagonists have mostly failed to display efficacy in clinical trials for anxiety or depression. Rather than broad-spectrum antidepressant/anxiolytic-like drugs, they may represent an 'antistress' solution for single stressful situations or for patients with chronic stress conditions. However, the impact of prolonged CRF-R1 antagonist treatments on the hypothalamic-pituitary-adrenal (HPA) axis under chronic stress conditions remained to be characterized. Hence, our study investigated whether a chronic CRF-R1 antagonist (crinecerfont, formerly known as SSR125543, 20 mg·kg-1·day-1 ip, 5 weeks) would alter HPA axis basal circadian activity and negative feedback sensitivity in mice exposed to either control or chronic stress conditions (unpredictable chronic mild stress, UCMS, 7 weeks), through measures of fecal corticosterone metabolites, plasma corticosterone, and dexamethasone suppression test. Despite preserving HPA axis parameters in control non-stressed mice, the 5-week crinercerfont treatment improved the negative feedback sensitivity in chronically stressed mice, but paradoxically exacerbated their basal corticosterone secretion nearly all along the circadian cycle. The capacity of chronic CRF-R1 antagonists to improve the HPA negative feedback in UCMS argues in favor of a potential therapeutic benefit against stress-related conditions. However, the treatment-related overactivation of HPA circadian activity in UCMS raise questions about possible physiological outcomes with long-standing treatments under ongoing chronic stress.

2.
Brain Behav Immun ; 94: 159-174, 2021 05.
Article in English | MEDLINE | ID: mdl-33609652

ABSTRACT

BACKGROUND: Several lines of evidence suggest that neuroinflammation might be a key neurobiological mechanism of depression. In particular, the P2X7 receptor (P2X7R), an ATP-gated ion channel involved in activation of the pro-inflammatory interleukin IL-1ß, has been shown to be a potential new pharmacological target in depression. The aim of this study was to explore the impact of unpredictable chronic mild stress (UCMS) on behavioural changes, hippocampal neurogenesis, and cellular characterisation of brain immune cells, in P2X7R Knock-Out (KO) mice. METHODS: P2X7R KO and wild-type (WT) mice were subjected to a 6-week UCMS protocol and received a conventional oral antidepressant (15 mg.kg-1 fluoxetine) or water per os. The mice then underwent behavioural tests consisting of the tail suspension test (TST), the elevated plus maze (EPM) test, the open field test, the splash test and the nest building test (week 7). Doublecortin immunostaining (DCX) of brain slices was used to assess neurogenesis in the dentate gyrus. Iba1 and TMEM119 immunostaining was used to characterise brain immune cells, Iba1 as a macrophage marker (including microglial cells) and TMEM119 as a potential specific resident microglial cells marker. RESULTS: After a 6-week UCMS exposure, P2X7R KO mice exhibited less deterioration of their coat state, spent a significantly smaller amount of time immobile in the TST and spent a larger amount of time in the open arms of the EPM. As expected, adult ventral hippocampal neurogenesis was significantly decreased by UCMS in WT mice, while P2X7R KO mice maintained ventral hippocampal neurogenesis at similar levels in both control and UCMS conditions. In stress-related brain regions, P2X7R KO mice also exhibited less recruitment of Iba1+/TMEM119+ and Iba1+/TMEM119- cells in the brain. The ratio between these two staining patterns revealed that brain immune cells were mostly composed of Iba1+/TMEM119+ cells (87 to 99%), and this ratio was affected neither by P2X7R genetic depletion nor by antidepressant treatment. DISCUSSION: Behavioural patterns, neurogenesis levels and density of brain immune cells in P2X7R KO mice after exposure to UCMS significantly differed from control conditions. Brain immune cells were mostly increased in brain regions known to be sensitive to UCMS exposure in WT but not in P2X7R KO mice. Considering Iba1+/TMEM119- staining might characterize peripheral immune cells, the ratio between Iba1+/TMEM119+ cells and IBA1+/TMEM119- cells, suggests that the rate of peripheral immune cells recruitment may not be modified neither by P2X7R gene expression nor by antidepressant treatment.


Subject(s)
Depression , Stress, Psychological , Animals , Antidepressive Agents , Disease Models, Animal , Doublecortin Protein , Hippocampus , Mice , Mice, Knockout , Receptors, Purinergic P2X7/genetics
3.
Eur J Neurosci ; 53(1): 151-171, 2021 01.
Article in English | MEDLINE | ID: mdl-32150310

ABSTRACT

Some recent clinical and preclinical evidence suggests that neuroinflammation is a key factor that interacts with the three neurobiological correlates of major depressive disorder: depletion of brain serotonin, dysregulation of the hypothalamus-pituitary-adrenal (HPA) axis and alteration of the continuous production of adult-generated neurons in the dentate gyrus of the hippocampus. This review discusses the main players in brain immunity as well as how inflammation interacts with the above three mechanisms. It is reported that kynurenine (KYN) pathway alteration in favour of its excitotoxic component and HPA axis dysregulation have the common effect of increasing extracellular glutamate levels and glutamate neurotransmission, which can impact hippocampal neurogenesis. This pathophysiological cascade appears to be triggered or sustained and reinforced by any chronic inflammatory condition involving increased circulating markers of inflammation that are able to cross the blood-brain barrier and activate microglia; it can also be the consequence of primary brain neuroinflammation, such as in neurodegenerative disorders with early manifestations that are frequently depressive symptoms. Further recent data indicate that primary microglial activation may also result from a direct impact of chronic stress on vascular function. The intricated dynamic crosstalk between neuroinflammation and other relevant neurobiological correlates of depression add to evidence that neuroinflammation may be a key therapeutic target for future therapeutic strategies in major depressive disorder.


Subject(s)
Depressive Disorder, Major , Hypothalamo-Hypophyseal System , Depression , Hippocampus , Humans , Neurogenesis , Pituitary-Adrenal System
4.
Front Mol Neurosci ; 10: 248, 2017.
Article in English | MEDLINE | ID: mdl-28848385

ABSTRACT

Major depressive disorder (MDD) is a highly prevalent mental illness whose therapy management remains uncertain, with more than 20% of patients who do not achieve response to antidepressants. Therefore, identification of reliable biomarkers to predict response to treatment will greatly improve MDD patient medical care. Due to the inaccessibility and lack of brain tissues from living MDD patients to study depression, researches using animal models have been useful in improving sensitivity and specificity of identifying biomarkers. In the current study, we used the unpredictable chronic mild stress (UCMS) model and correlated stress-induced depressive-like behavior (n = 8 unstressed vs. 8 stressed mice) as well as the fluoxetine-induced recovery (n = 8 stressed and fluoxetine-treated mice vs. 8 unstressed and fluoxetine-treated mice) with transcriptional signatures obtained by genome-wide microarray profiling from whole blood, dentate gyrus (DG), and the anterior cingulate cortex (ACC). Hierarchical clustering and rank-rank hypergeometric overlap (RRHO) procedures allowed us to identify gene transcripts with variations that correlate with behavioral profiles. As a translational validation, some of those transcripts were assayed by RT-qPCR with blood samples from 10 severe major depressive episode (MDE) patients and 10 healthy controls over the course of 30 weeks and four visits. Repeated-measures ANOVAs revealed candidate trait biomarkers (ARHGEF1, CMAS, IGHMBP2, PABPN1 and TBC1D10C), whereas univariate linear regression analyses uncovered candidates state biomarkers (CENPO, FUS and NUBP1), as well as prediction biomarkers predictive of antidepressant response (CENPO, NUBP1). These data suggest that such a translational approach may offer new leads for clinically valid panels of biomarkers for MDD.

5.
Biol Psychiatry ; 82(5): 370-379, 2017 09 01.
Article in English | MEDLINE | ID: mdl-28359564

ABSTRACT

BACKGROUND: Depression is frequently associated with chronic pain or chronic stress. Among cortical areas, the anterior cingulate cortex (ACC, areas 24a and 24b) appears to be important for mood disorders and constitutes a neuroanatomical substrate for investigating the underlying molecular mechanisms. The current work aimed at identifying ACC molecular factors subserving depression. METHODS: Anxiodepressive-like behaviors in C57BL/6J male mice were induced by neuropathic pain, unpredictable chronic mild stress, and optogenetic ACC stimulation and were evaluated using novelty suppressed feeding, splash, and forced swim tests. ACC molecular changes in chronic pain-induced depression were uncovered through whole-genome expression analysis. Further mechanistic insights were provided by chromatin immunoprecipitation, Western blot, and immunostaining. The causal link between molecular changes and depression was studied using knockout, pharmacological antagonism, and local viral-mediated gene knockdown. RESULTS: Under chronic pain-induced depression, gene expression changes in the ACC highlighted the overexpression of a regulator of the mitogen-activated protein kinase pathway, mitogen-activated protein kinase phosphatase-1 (MKP-1). This upregulation is associated with the presence of transcriptionally active chromatin marks (acetylation) at its proximal promoter region as well as increased cyclic adenosine monophosphate response element-mediated transcriptional activity and phosphorylation of cyclic adenosine monophosphate response element binding protein and activating transcription factor. MKP-1 overexpression is also observed with unpredictable chronic mild stress and repeated ACC optogenetic stimulation and is reversed by fluoxetine. A knockout, an antagonist, or a local silencing of MKP-1 attenuates depressive-like behaviors, pointing to an important role of this phosphatase in depression. CONCLUSIONS: These data point to ACC MKP-1 as a key factor in the pathophysiology of depression and a potential target for treatment development.


Subject(s)
Depressive Disorder/enzymology , Dual Specificity Phosphatase 1/metabolism , Gyrus Cinguli/enzymology , Animals , Antidepressive Agents, Second-Generation/pharmacology , Chronic Pain/enzymology , Depressive Disorder/drug therapy , Disease Models, Animal , Dual Specificity Phosphatase 1/genetics , Epigenesis, Genetic , Fluoxetine/pharmacology , Gene Expression/drug effects , Gyrus Cinguli/drug effects , Male , Mice, Inbred C57BL , Mice, Transgenic , Promoter Regions, Genetic , Proto-Oncogene Proteins c-fos/metabolism , Stress, Psychological/drug therapy , Stress, Psychological/enzymology , Up-Regulation/drug effects
6.
Mol Neurobiol ; 54(7): 5361-5374, 2017 09.
Article in English | MEDLINE | ID: mdl-27590138

ABSTRACT

In amyotrophic lateral sclerosis (ALS), motor neuron degeneration is associated with systemic metabolic impairment. However, the evolution of metabolism alteration is partially unknown and its link with disease progression has never been described. For the first time, we ran a study focused on (1) the evolution of metabolism disturbance during disease progression through omics approaches and (2) the relation between metabolome profile and clinical evolution. SOD1-G93A (mSOD1) transgenic mice (n = 11) and wild-type (WT) littermates (n = 17) were studied during 20 weeks. Metabolomic profile of muscle and cerebral cortex was analysed at week 20, and plasma samples were assessed at four time points over 20 weeks. The relevant metabolic pathways highlighted by metabolomic analysis were explored by a targeted transcriptomic approach in mice. Plasma metabolomics were also performed in 24 ALS patients and 24 gender- and age-matched controls. Metabolomic analysis of muscle and cerebral cortex enabled an excellent discrimination between mSOD1 and WT mice (p < 0.001). These alterations included especially tryptophan, arginine, and proline metabolism pathways (including polyamines) as also revealed by transcriptomic analysis and findings in ALS patients. Multivariate models performed to explain clinical findings in ALS mice, and patients were excellent (p < 0.01) and highlighted three main metabolic pathways: arginine and proline, tryptophan, and branched amino acid metabolism. This work is the first longitudinal study that evaluates metabolism alteration in ALS, including the analysis of different tissues and using a combination of omics methods. We particularly identified arginine and proline metabolism. This pathway is also associated with disease progression and may open new perspectives of therapeutic targets.


Subject(s)
Amyotrophic Lateral Sclerosis/metabolism , Arginine/metabolism , Motor Neurons/metabolism , Proline/metabolism , Animals , Disease Models, Animal , Female , Humans , Longitudinal Studies , Male , Metabolomics/methods , Mice, Transgenic , Superoxide Dismutase/genetics , Tryptophan/metabolism
7.
Neurotherapeutics ; 13(4): 905-917, 2016 10.
Article in English | MEDLINE | ID: mdl-27444617

ABSTRACT

In amyotrophic lateral sclerosis (ALS), motor neuron degeneration occurs simultaneously with systemic metabolic impairment and neuroinflammation. Playing an important role in the regulation of both phenomena, interleukin (IL)-6, a major cytokine of the inflammatory response has been proposed as a target for management of ALS. Although a pilot clinical trial provided promising results in humans, another recent preclinical study showed that knocking out the IL-6 gene in mice carrying ALS did not improve clinical outcome. In this study, we aimed to determine the relevance of the IL-6 pathway blockade in a mouse model of ALS by using a pharmacological antagonist of IL-6, a murine surrogate of tocilizumab, namely MR16-1. We analyzed the immunological and metabolic effects of IL-6 blockade by cytokine measurement, blood cell immunophenotyping, targeted metabolomics, and transcriptomics. A deleterious clinical effect of MR16-1 was revealed, with a speeding up of weight loss (p = 0.0041) and decreasing body weight (p < 0.05). A significant increase in regulatory T-cell count (p = 0.0268) and a decrease in C-X-C ligand-1 concentrations in plasma (p = 0.0479) were observed. Metabolomic and transcriptomic analyses revealed that MR16-1 mainly affected branched-chain amino acid, lipid, arginine, and proline metabolism. IL-6 blockade negatively affected body weight, despite a moderated anti-inflammatory effect. Metabolic effects of IL-6 were mild compared with metabolic disturbances observed in ALS, but a modification of lipid metabolism by therapy was identified. These results indicate that IL-6 blockade did not improve clinical outcome of a mutant superoxide dismutase 1 mouse model of ALS.


Subject(s)
Amyotrophic Lateral Sclerosis/drug therapy , Amyotrophic Lateral Sclerosis/metabolism , Antibodies, Monoclonal, Humanized/pharmacology , Antibodies, Monoclonal, Humanized/therapeutic use , Interleukin-6/metabolism , Lipid Metabolism/drug effects , Amyotrophic Lateral Sclerosis/genetics , Animals , Body Weight/genetics , Cytokines/blood , Disability Evaluation , Disease Models, Animal , Follow-Up Studies , Gene Regulatory Networks , Immunologic Factors/pharmacology , Immunologic Factors/therapeutic use , Interleukin-6/genetics , Locomotion/drug effects , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Superoxide Dismutase/genetics , Superoxide Dismutase/metabolism , T-Lymphocytes, Regulatory/drug effects
8.
Neurobiol Dis ; 82: 332-341, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26188176

ABSTRACT

The serotonin 1A receptor (5-HT1A), a critical regulator of the brain serotonergic tone, is implicated in major depressive disorder (MDD) where it is often found to be dys-regulated. However, the extent to which stress and antidepressant treatment impact 5-HT1A expression in adults remains unclear. To address this issue, we subjected adult male BALB/c mice to unpredictable chronic mild stress (UCMS) to induce a depression-like phenotype that was reversed by chronic treatment with the antidepressant imipramine. In prefrontal cortex (PFC) and midbrain tissue, UCMS increased 5-HT1A RNA and protein levels, changes that are expected to decrease the brain serotonergic activity. The stress-induced increase in 5-HT1A expression was paralleled by a specific increase in DNA methylation of the conserved -681 CpG promoter site, located within a Sp1-like element. We show that the -681 CpG site is recognized and repressed by Sp4, the predominant neuronal Sp1-like factor and that Sp4-induced repression is attenuated by DNA methylation, despite a stress-induced increase in PFC Sp4 levels. These results indicate that adult life stress induces DNA methylation of a conserved promoter site, antagonizing Sp4 repression to increase 5-HT1A expression. Chronic imipramine treatment fully reversed the UCMS-induced increase in methylation of the -681 CpG site in the PFC but not midbrain of stressed animals and also increased 5-HT1A expression in the PFC of control animals. Incomplete reversal by imipramine of stress-induced changes in 5-HT1A methylation and expression indicates a persistence of stress vulnerability, and that sustained reversal of behavioral impairments may require additional pathways.


Subject(s)
Antidepressive Agents, Tricyclic/pharmacology , DNA Methylation/drug effects , Depressive Disorder/drug therapy , Depressive Disorder/metabolism , Receptor, Serotonin, 5-HT1A/genetics , Receptor, Serotonin, 5-HT1A/metabolism , Animals , Chronic Disease , Conserved Sequence , CpG Islands , DNA Methylation/physiology , Depressive Disorder/genetics , Disease Models, Animal , Dorsal Raphe Nucleus/drug effects , Dorsal Raphe Nucleus/metabolism , Imipramine/pharmacology , Male , Mice, Inbred BALB C , Prefrontal Cortex/drug effects , Prefrontal Cortex/metabolism , Promoter Regions, Genetic , RNA, Messenger/metabolism , Stress, Psychological/drug therapy , Stress, Psychological/genetics , Stress, Psychological/metabolism , Transcription, Genetic/drug effects , Transcription, Genetic/physiology
9.
Front Pharmacol ; 4: 146, 2013 Nov 22.
Article in English | MEDLINE | ID: mdl-24319431

ABSTRACT

Depression is one of the most frequent and severe mental disorder. Since the discovery of antidepressant (AD) properties of the imipramine and then after of other tricyclic compounds, several classes of psychotropic drugs have shown be effective in treating major depressive disorder (MDD). However, there is a wide range of variability in response to ADs that might lead to non response or partial response or in increased rate of relapse or recurrence. The mechanisms of response to AD therapy are poorly understood, and few biomarkers are available than can predict response to pharmacotherapy. Here, we will first review markers that can be used to predict response to pharmacotherapy, such as markers of drug metabolism or blood-brain barrier (BBB) function, the activity of specific brain areas or neurotransmitter systems, hormonal dysregulations or plasticity, and related molecular targets. We will describe both clinical and preclinical studies and describe factors that might affect the expression of these markers, including environmental or genetic factors and comorbidities. This information will permit us to suggest practical recommendations and innovative treatment strategies to improve therapeutic outcomes.

10.
CNS Drugs ; 27(6): 411-22, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23657787

ABSTRACT

Depression is a devastating mental disorder with an increasing impact throughout the world, whereas the efficacy of currently available pharmacological treatment is still limited. Growing evidence from preclinical and clinical studies suggests that orexins (neuropeptides that are also known as hypocretins) and their receptors are involved in the physiopathology of depression. Indeed, the orexinergic system regulates functions that are disturbed in depressive states such as sleep, reward system, feeding behavior, the stress response and monoaminergic neurotransmission. Nevertheless, the precise role of orexins in behavioral and neurophysiological impairments observed in depression is still unclear. Both hypoactivity and hyperactivity of orexin signaling pathways have been found to be associated with depression. These discrepancies in the literature prompted the necessity for additional investigations, as the orexinergic system appears to be a promising target to treat the symptoms of depression. This assumption is underlined by recent data suggesting that pharmacological blockade of orexin receptors induces a robust antidepressant-like effect in an animal model of depression. Further preclinical and clinical studies are needed to progress the overall understanding of the orexinergic alterations in depression, which will eventually translate preliminary observations into real therapeutic potential. The aim of this paper is to provide an overview of human and animal research dedicated to the study of the specific involvement of orexins in depression, and to propose a framework in which disturbances of the orexinergic system are regarded as an integral component of the etiology of depression.


Subject(s)
Antidepressive Agents/pharmacology , Depression/etiology , Intracellular Signaling Peptides and Proteins/metabolism , Neuropeptides/metabolism , Orexin Receptor Antagonists , Animals , Antidepressive Agents/administration & dosage , Antidepressive Agents/therapeutic use , Biogenic Monoamines/metabolism , Depression/drug therapy , Depression/metabolism , Depression/psychology , Humans , Orexins , Signal Transduction/drug effects
11.
J Neuroinflammation ; 10: 19, 2013 Feb 01.
Article in English | MEDLINE | ID: mdl-23374751

ABSTRACT

BACKGROUND: Toxoplasmosis is one of the most common parasitic infections in humans. It can establish chronic infection and is characterized by the formation of tissue cysts in the brain. The cysts remain largely quiescent for the life of the host, but can reactivate and cause life-threatening toxoplasmic encephalitis in immunocompromised patients, such as those with AIDS, neoplastic diseases and organ transplants. Toll-like receptor (TLR) adaptor MyD88 activation is required for the innate sensing of Toxoplasma gondii. Mice deficient in MyD88 have defective IL-12 and Th1 effector responses, and are highly susceptible to the acute phase of T. gondii infection. However, the role of this signaling pathway during cerebral infection is poorly understood and requires examination. METHOD: MyD88-deficient mice and control mice were orally infected with T. gondii cysts. Cellular and parasite infiltration in the peripheral organs and in the brain were determined by histology and immunohistochemistry. Cytokine levels were determined by ELISA and chemokine mRNA levels were quantified by real-time PCR (qPCR). RESULTS: Thirteen days after infection, a higher parasite burden was observed but there was no histological change in the liver, heart, lungs and small intestine of MyD88⁻/⁻ and MyD88⁺/⁺ mice. However, MyD88⁻/⁻ mice compared to MyD88⁺/⁺ mice were highly susceptible to cerebral infection, displayed high parasite migration to the brain, severe neuropathological signs of encephalitis and succumbed within 2 weeks of oral infection. Susceptibility was primarily associated with lower expression of Th1 cytokines, especially IL-12, IFN-γ and TNF-α, significant decrease in the expression of CCL3, CCL5, CCL7 and CCL19 chemokines, marked defect of CD8⁺ T cells, and infiltration of CD11b⁺ and F4/80⁺ cells in the brain. CONCLUSION: MyD88 is essential for the protection of mice during the cerebral installation of T. gondii infection. These results establish a role for MyD88 in T cell-mediated control of T. gondii in the central nervous system (CNS).


Subject(s)
Brain/metabolism , Immunity, Cellular/immunology , Myeloid Differentiation Factor 88/deficiency , Toxoplasma , Toxoplasmosis, Animal/metabolism , Toxoplasmosis, Cerebral/metabolism , Animals , Brain/immunology , Brain/parasitology , Female , Mice , Mice, Inbred BALB C , Mice, Knockout , Toxoplasma/immunology , Toxoplasmosis, Animal/immunology , Toxoplasmosis, Animal/prevention & control , Toxoplasmosis, Cerebral/immunology , Toxoplasmosis, Cerebral/prevention & control
12.
Neuropsychopharmacology ; 37(10): 2210-21, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22713907

ABSTRACT

Growing evidence indicates that an increase of orexin (or hypocretin) signaling is involved in the pathophysiology of major depression, but little is known regarding the causal link between the orexinergic system and depressive-like states. Here we blocked orexin receptors in mice subjected to unpredictable chronic mild stress (UCMS) to investigate putative antidepressant-like effects of this treatment, as well as the underlying mechanisms. BALB/c mice were exposed to 9 weeks of UCMS and from the third week onward treated daily with fluoxetine (20 mg/kg per day, per os) or with the dual orexin receptor antagonist almorexant (100 mg/kg per day, per os). The effects of UCMS regimen and pharmacological treatments were assessed by physical measures and behavioral testing. The dexamethasone suppression test was performed to examine the integrity of the negative feedback of the hypothalamic-pituitary-adrenal (HPA) axis, and immunohistochemical markers were used to assess cell proliferation (Ki-67), immature newborn neurons (doublecortin), and mature newborn neurons (5-bromo-2'-deoxyuridine/NeuN) in the dorsal and ventral parts of the hippocampus. Our results show that 7 weeks of fluoxetine or almorexant treatments counteract the UCMS-induced physical and behavioral alterations. Both treatments prevented the HPA axis dysregulation caused by UCMS, but only fluoxetine reversed the UCMS-induced decrease of hippocampal cell proliferation and neurogenesis, while chronic almorexant treatment decreased cell proliferation and neurogenesis specifically in the ventral hippocampus. Taken together, this is the first evidence that pharmacological blockade of the orexinergic system induces a robust antidepressant-like effect and the restoration of stress-related HPA axis defect independently from a neurogenic action.


Subject(s)
Acetamides/therapeutic use , Depression/drug therapy , Isoquinolines/therapeutic use , Receptors, G-Protein-Coupled/antagonists & inhibitors , Receptors, Neuropeptide/antagonists & inhibitors , Animals , Antidepressive Agents/therapeutic use , Behavior, Animal/drug effects , Biomarkers , Bromodeoxyuridine/analysis , Cell Proliferation , Disease Models, Animal , Doublecortin Domain Proteins , Fluoxetine/therapeutic use , Hypothalamo-Hypophyseal System/drug effects , Intracellular Signaling Peptides and Proteins/physiology , Ki-67 Antigen/analysis , Mice , Mice, Inbred BALB C , Microtubule-Associated Proteins/analysis , Neurogenesis/physiology , Neuropeptides/analysis , Neuropeptides/physiology , Orexin Receptors , Orexins , Pituitary-Adrenal System/drug effects
13.
Behav Brain Res ; 231(1): 130-7, 2012 May 16.
Article in English | MEDLINE | ID: mdl-22465167

ABSTRACT

Unipolar depression is one of the leading causes of disability. The pathophysiology of depression is poorly understood. Evidence suggests that inflammation is associated with depression. For instance, pro-inflammatory cytokines are found to be elevated in the peripheral blood of depressed subjects. Cytokine immunotherapy itself is known to induce depressive symptoms. While the epidemiological and biochemical relationship between inflammation and depression is strong, little is known about the possible existence of neuroinflammation in depression. The use of animal models of depression such as the Unpredictable Chronic Mild Stress (UCMS) has already contributed to the elucidation of the pathophysiological mechanisms of depression such as decreased neurogenesis and HPA axis alterations. We used this model to explore the association of depressive-like behavior in mice with changes in peripheral pro-inflammatory cytokines IL-1ß, TNFα and IL-6 level as well as the neuroinflammation by quantifying CD11b expression in brain areas known to be involved in the pathophysiology of depression. These areas include the cerebral cortex, the nucleus accumbens, the bed nucleus of the stria terminalis, the caudate putamen, the amygdala and the hippocampus. The results indicate that microglial activation is significantly increased in the infralimbic, cingulate and medial orbital cortices, nucleus accumbens, caudate putamen, amygdala and hippocampus of the mouse brain as a function of UCMS, while levels of pro-inflammatory cytokines did not differ among the groups. This finding suggests that neuroinflammation occurs in depression and may be implicated in the subject's behavioral response. They also suggest that UCMS could be a potentially reliable model to study depression-induced neuroinflammation.


Subject(s)
Brain/physiopathology , Depression/physiopathology , Disease Models, Animal , Encephalitis/physiopathology , Stress, Physiological/physiology , Stress, Psychological/physiopathology , Animals , Behavior, Animal , Brain/metabolism , Cytokines/metabolism , Depression/metabolism , Encephalitis/metabolism , Male , Mice , Stress, Psychological/metabolism
14.
Neuropharmacology ; 61(1-2): 336-46, 2011.
Article in English | MEDLINE | ID: mdl-21530551

ABSTRACT

Chronic stressful life events are risk factors for depression often accompanied by homeostatic disturbances. Hypothalamic neuropeptides, such as orexins (OXs) and melanin-concentrating hormone (MCH), are involved in regulation of several autonomic functions that are altered in depression. However, little is known about the link between orexinergic or MCH-ergic systems and depression. Using double immunohistochemical labeling for OX- or MCH-containing neurons and Fos protein, we studied the effects of a chronic selective serotonin reuptake inhibitor antidepressant treatment (fluoxetine) on the OX and MCH neuronal activation in mice exposed to unpredictable chronic mild stress (UCMS), a rodent model of depression. Western blot was also performed to assess OX and MCH receptor expression in various brain areas. Finally, almorexant, a dual OX receptor antagonist, was assessed in the tail suspension test. UCMS induced physical and behavioral disturbances in mice reversed by 6-week fluoxetine treatment. Orexinergic neurons were more activated in the dorsomedial and perifornical hypothalamic area (DMH-PFA) of UCMS-subjected mice compared to the lateral hypothalamus (LH), and this increase was reversed by 6-week fluoxetine treatment. UCMS also reduced expression of OX-receptor 2 in the thalamus and hypothalamus, but not in animals chronically treated with fluoxetine. MCH neurons were neither affected by UCMS nor by antidepressant treatment, while UCMS modulated MCH receptor 1 expression in thalamus and hippocampus. Finally, chronic but not acute administration of almorexant, induced antidepressant-like effect in the tail suspension test. These data suggest that OX neurons in the DMH-PFA and MCH-ergic system may contribute to the pathophysiology of depressive disorders.


Subject(s)
Antidepressive Agents/metabolism , Depression/metabolism , Disease Models, Animal , Dorsomedial Hypothalamic Nucleus/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Neurons/metabolism , Neuropeptides/metabolism , Animals , Antidepressive Agents/pharmacology , Antidepressive Agents/therapeutic use , Depression/drug therapy , Dorsomedial Hypothalamic Nucleus/drug effects , Hypothalamus/drug effects , Hypothalamus/metabolism , Male , Mice , Mice, Inbred BALB C , Neurons/drug effects , Orexins , Random Allocation
15.
Med Sci (Paris) ; 25(8-9): 687-91, 2009.
Article in French | MEDLINE | ID: mdl-19765381

ABSTRACT

Schizophrenia is one of the most severe and disabling psychiatric disease that affects about 1 % of the adult worldwide population. Aetiology of schizophrenia is still unknown but genetic and environmental factors are suspected to play a major role in its onset. Recent epidemiologic studies indicate that infectious agents may contribute to some cases of schizophrenia. In particular, several epidemiological, behavioural and neurochemical studies suggested the existence of an association between schizophrenia and past history of primo-infection by the Toxoplasma gondii. However, they are some limitations for this hypothesis among which the lack of correlation between the geographic distribution of both diseases and of direct evidence for the presence of the parasite in schizophrenic patients. Nevertheless the identification of physiopathological mechanisms related to the parasite could provide a better comprehension to the outcome of schizophrenia. Studies on the link between toxoplasmosis and schizophrenia may provide interesting data for the diagnosis and the development of new treatments for this disorder.


Subject(s)
Schizophrenia/complications , Schizophrenia/epidemiology , Toxoplasmosis/complications , Adult , Animals , Humans , Incidence , Pedigree , Schizophrenia/genetics , Schizophrenia/therapy , Toxoplasma , Toxoplasmosis/epidemiology , Treatment Outcome
16.
Behav Brain Res ; 202(2): 245-51, 2009 Sep 14.
Article in English | MEDLINE | ID: mdl-19463708

ABSTRACT

Despite growing evidences of an association between brain-derived neurotrophic factor (BDNF) and antidepressant effects, the neurotrophic hypothesis of depression is challenged by the paucity of direct links between BDNF deficit and depressive-like behaviors. The unpredictable chronic mild stress (UCMS) paradigm might take our understanding a step further by examining whether a decrease in bdnf expression can lead to enhanced vulnerability to stress and prevent antidepressant efficacy in all or specific UCMS-induced alterations. Wild-type bdnf(+/+) and heterozygous bdnf(+/-) mice were exposed to an 8-week UCMS regimen and, from the third week onward, treated with either vehicle or imipramine (20mg/kg/day, ip). Physical, behavioral and biological (plasma corticosterone levels, bdnf expression in the dentate gyrus) measures were further analyzed regarding to the genotype and the treatment. Heterozygous bdnf(+/-) mice displayed hyperactivity and increase of body weight but no enhancement of the sensitivity to stress exposure in all the measures investigated here. In contrast, while imipramine treatment reduced anxiety-like behaviors in the novelty-suppressed feeding test in both genotypes, it decreased aggressiveness in the resident/intruder test and immobility in the tail suspension test in wild-type but not in heterozygous mice. Furthermore, imipramine induced a twofold increase of bdnf expression in the dentate gyrus in both genotypes, while bdnf(+/-) mice displayed roughly half-reduced level for the same treatment. In summary, we demonstrate here that depletion in BDNF dampened the antidepressant effects in several behaviors but failed to increase vulnerability to chronic stress exposure.


Subject(s)
Brain-Derived Neurotrophic Factor/deficiency , Stress, Psychological/physiopathology , Stress, Psychological/psychology , Aggression/drug effects , Animals , Antidepressive Agents, Tricyclic/administration & dosage , Anxiety/drug therapy , Body Weight/drug effects , Body Weight/physiology , Brain-Derived Neurotrophic Factor/genetics , Brain-Derived Neurotrophic Factor/metabolism , Corticosterone/blood , Dentate Gyrus/metabolism , Eating/drug effects , Eating/physiology , Exploratory Behavior/drug effects , Exploratory Behavior/physiology , Genotype , Imipramine/administration & dosage , Mice , Motor Activity/physiology , RNA, Messenger/metabolism , Stress, Psychological/drug therapy , Time Factors
17.
Neuropsychopharmacology ; 34(6): 1363-80, 2009 May.
Article in English | MEDLINE | ID: mdl-18536703

ABSTRACT

Gene microarrays may enable the elucidation of neurobiological changes underlying the pathophysiology and treatment of major depression. However, previous studies of antidepressant treatments were performed in healthy normal rather than 'depressed' animals. Since antidepressants are devoid of mood-changing effects in normal individuals, the clinically relevant rodent transcriptional changes could remain undetected. We investigated antidepressant-related transcriptome changes in a corticolimbic network of mood regulation in the context of the unpredictable chronic mild stress (UCMS), a naturalistic model of depression based on socio-environmental stressors. Mice subjected to a 7-week UCMS displayed a progressive coat state deterioration, reduced weight gain, and increased agonistic and emotion-related behaviors. Chronic administration of an effective (fluoxetine) or putative antidepressant (corticotropin-releasing factor-1 (CRF1) antagonist, SSR125543) reversed all physical and behavioral effects. Changes in gene expression differed among cingulate cortex (CC), amygdala (AMY) and dentate gyrus (DG) and were extensively reversed by both drugs in CC and AMY, and to a lesser extent in DG. Fluoxetine and SSR125543 also induced additional and very similar molecular profiles in UCMS-treated mice, but the effects of the same drug differed considerably between control and UCMS states. These studies established on a large-scale that the molecular impacts of antidepressants are region-specific and state-dependent, revealed common transcriptional changes downstream from different antidepressant treatments and supported CRF1 targeting as an effective therapeutic strategy. Correlations between UCMS, drug treatments, and gene expression suggest distinct AMY neuronal and oligodendrocyte molecular phenotypes as candidate systems for mood regulation and therapeutic interventions.


Subject(s)
Antidepressive Agents/therapeutic use , Brain/drug effects , Brain/physiopathology , Depressive Disorder/drug therapy , Depressive Disorder/physiopathology , Gene Expression Profiling , Affect/drug effects , Affect/physiology , Agonistic Behavior/drug effects , Agonistic Behavior/physiology , Amygdala/drug effects , Amygdala/physiopathology , Animals , Corticotropin-Releasing Hormone/antagonists & inhibitors , Dentate Gyrus/drug effects , Dentate Gyrus/physiopathology , Depressive Disorder/genetics , Disease Models, Animal , Emotions/drug effects , Emotions/physiology , Fluoxetine/therapeutic use , Gene Expression/drug effects , Gene Expression/physiology , Gyrus Cinguli/drug effects , Gyrus Cinguli/physiopathology , Hydrocarbons, Halogenated/therapeutic use , Male , Mice , Mice, Inbred BALB C , Stress, Psychological/drug therapy , Stress, Psychological/physiopathology , Thiazines/therapeutic use , Weight Gain/drug effects , Weight Gain/physiology
18.
Biol Psychiatry ; 64(4): 293-301, 2008 Aug 15.
Article in English | MEDLINE | ID: mdl-18406399

ABSTRACT

BACKGROUND: Depression and anxiety disorders have been linked to dysfunction of the hypothalamo-pituitary-adrenal (HPA) axis and structural changes within the hippocampus. Unpredictable chronic mild stress (UCMS) can recapitulate these effects in a mouse model, and UCMS-induced changes, including downregulation of hippocampal neurogenesis, can be reversed by antidepressant (AD) treatment. We investigated causality between changes in hippocampal neurogenesis and the effects of both chronic stress and chronic ADs. METHODS: Mice were treated with either a sham procedure or focal hippocampal irradiation to disrupt cell proliferation before being confronted with 5 weeks of UCMS. From the third week onward, we administered monoaminergic ADs (imipramine, fluoxetine), the corticotropin-releasing factor 1 (CRF(1)) antagonist SSR125543, or the vasopressin 1b (V(1b)) antagonist SSR149415 daily. The effects of UCMS regimen, AD treatments, and irradiation were assessed by physical measures (coat state, weight), behavioral testing (Splash test, Novelty-Suppressed feeding test, locomotor activity), and hippocampal BrdU labeling. RESULTS: Our results show that elimination of hippocampal neurogenesis has no effect on animals' sensitivity to UCMS in several behavioral assays, suggesting that reduced neurogenesis is not a cause of stress-related behavioral deficits. Second, we present evidence for both neurogenesis-dependent and -independent mechanisms for the reversal of stress-induced behaviors by AD drugs. Specifically, loss of neurogenesis completely blocked the effects of monoaminergic ADs (imipramine, fluoxetine) but did not prevent most effects of the CRF(1) and the V(1b) antagonists. CONCLUSIONS: Hippocampal neurogenesis might thus be used by the monoaminergic ADs to counteract the effects of stress, whereas similar effects could be achieved by directly targeting the HPA axis and related neuropeptides.


Subject(s)
Antidepressive Agents/pharmacology , Cell Proliferation/drug effects , Depression/pathology , Hippocampus/pathology , Neurons/drug effects , Analysis of Variance , Animals , Antidepressive Agents/classification , Antidepressive Agents/therapeutic use , Behavior, Animal/drug effects , Biogenic Monoamines/metabolism , Bromodeoxyuridine/metabolism , Cell Count/methods , Cell Proliferation/radiation effects , Corticotropin-Releasing Hormone/metabolism , Depression/drug therapy , Disease Models, Animal , Exploratory Behavior/drug effects , Exploratory Behavior/radiation effects , Hemibody Irradiation/adverse effects , Hemibody Irradiation/methods , Hydrocarbons, Halogenated/pharmacology , Indoles/pharmacology , Male , Mice , Mice, Inbred BALB C , Motor Activity/drug effects , Neurons/radiation effects , Pyrrolidines/pharmacology , Reaction Time/drug effects , Reaction Time/radiation effects , Thiazines/pharmacology , Time Factors
19.
Physiol Behav ; 93(3): 512-20, 2008 Feb 27.
Article in English | MEDLINE | ID: mdl-18045628

ABSTRACT

Tonic immobility (TI) is an unlearned fear response induced by a brief physical restraint and characterized by a marked autonomic nervous system involvement. This experiment aimed at studying the relative involvement of both autonomic sub-systems, the sympathetic and parasympathetic nervous systems, during TI, by analyzing Heart Rate Variability. Quail selected genetically for long (LTI) or short (STI) TI duration and quail from a control line (CTI) were used. The animals were surgically fitted with a telemetric device to record electrocardiograms before and during a TI test. Heart rate did not differ between lines at rest. The induction of TI, whether effective or not, induced an increase in HR characterized by a shift of the sympathovagal balance towards a higher sympathetic dominance. Parasympathetic activity was lower during effective than during non-effective inductions in CTI quail. During TI, the increase in sympathetic dominance was initially maintained and then declined, while relative parasympathetic activity remained low, especially in CTI and STI lines. The end of tonic immobility was characterized by a rise in overall autonomic activity in all lines and an increase in parasympathetic influence in CTI and STI quail. To conclude, the susceptibility to TI cannot be explained only by autonomic reflex changes. It is probably strongly related to the perception of the test by the quail. During TI, the differences between lines in autonomic responses probably reflect behavioural differences in the fear response.


Subject(s)
Heart Rate/physiology , Immobility Response, Tonic/physiology , Quail/physiology , Adrenergic beta-Antagonists/pharmacology , Analysis of Variance , Animals , Atropine/pharmacology , Behavior, Animal/drug effects , Electrocardiography/methods , Female , Heart Rate/drug effects , Immobility Response, Tonic/drug effects , Parasympatholytics/pharmacology , Propranolol/pharmacology , Restraint, Physical/methods , Telemetry/methods , Time Factors
20.
J Neurosci Methods ; 167(2): 198-206, 2008 Jan 30.
Article in English | MEDLINE | ID: mdl-17889939

ABSTRACT

Gene expression profiling holds great promise for identifying molecular pathologies of central nervous system disorders. However, the analysis of brain tissue poses unique analytical challenges, as typical microarray signals represent averaged transcript levels across neuronal and glial cell populations. Here we have generated ratios of gene transcript levels between gray and adjacent white matter samples to estimate the relative cellular origins of expression. We show that incorporating these ratios into transcriptome analysis (i) provides new analytical perspectives, (ii) increases the potential for biological insight obtained from postmortem transcriptome studies, (iii) expands knowledge about glial and neuronal cellular programs and (iv) facilitates the generation of cell-type specific hypotheses. This approach represents a robust and cost-effective "add-on" to transcriptome analyses of the mammalian brain. As this approach can be applied post hoc, we provide tables of ratios for analysis of existing mouse and human brain datasets.


Subject(s)
Brain/cytology , Brain/metabolism , Gene Expression Profiling/methods , RNA, Messenger/genetics , Transcription, Genetic , Animals , Cluster Analysis , Cohort Studies , Databases, Genetic , Gene Expression Regulation , Humans , Mice , Microarray Analysis/methods , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neuroglia/metabolism , Neurons/metabolism , Postmortem Changes , RNA, Messenger/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...