Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters










Publication year range
1.
Article in Russian | MEDLINE | ID: mdl-32929929

ABSTRACT

OBJECTIVE: To compare the effects of cortexin, cerebrolysin and actovegin on memory impairment, cerebral circulation and morphological changes in the hippocampus of rats with chronic brain ischemia. MATERIAL AND METHODS: The study was conducted using male rats with chronic brain ischemia caused by stenosis of the common carotid arteries by 50%. Animals received cortexin (0,3; 1 or 3 mg/kg), cerebrolysin (0,8; 2,5 or 7,5 ml/kg) and actovegin (5 ml/kg) in two 10-day courses with 10 days of treatment break. The severity of cognitive impairment was evaluated using the Morris water maze, passive and active avoidance tests. Cerebral circulation using laser flowmetry and brain hippocampus structures were studied in the end of treatment. RESULTS: Cognitive impairment in animals with chronic brain ischemia was accompanied by the development of pathological changes in the CA1 and CA4 regions of the hippocampus. Administration of cortexin (1 and 3 mg/kg) and cerebrolysin (2.5 and 7.5 ml/kg) to rats with chronic brain ischemia had almost no effect on cerebral blood flow, but contributed to the improvement in memory formation and retrieval processes in the Morris water maze. The treatment effect was comparable for both drugs and persisted after 10 days of treatment break. Morphological assessment showed a decrease in the severity of pathological changes in the hippocampal regions. CONCLUSION: The course-administration of cortexin and cerebrolysin lead to a decrease in the severity of memory impairment and pathomorphological changes in the hippocampus in rats with chronic brain ischemia.


Subject(s)
Brain Ischemia , Amino Acids , Animals , Cerebrovascular Circulation , Heme/analogs & derivatives , Hippocampus , Intercellular Signaling Peptides and Proteins , Male , Rats , Rats, Wistar
2.
Neurobiol Stress ; 11: 100171, 2019 Nov.
Article in English | MEDLINE | ID: mdl-31193462

ABSTRACT

This perspective highlights research presented as part of the symposium entitled, "Stress and Glucocorticoid Modulation of Feeding and Metabolism" at the 2018 Neurobiology of Stress Workshop held in Banff, AB, Canada. The symposium comprised five researchers at different career stages who each study different aspects of the interaction between the stress response and metabolic control. Their collective results reveal the complexity of this relationship in terms of behavioural and physiological outcomes. Their work emphasizes the need to consider the level of interaction (cellular, tissue, systems) as well as the timing and context in which the interaction is studied. Rather than a comprehensive review on the work presented at the Symposium, here we discuss recurring themes that emerged at the biennial workshop, which address new avenues of research that will drive the field forward.

3.
Mol Psychiatry ; 23(3): 533-543, 2018 03.
Article in English | MEDLINE | ID: mdl-28696432

ABSTRACT

Single-nucleotide polymorphisms (SNPs) in CACNA1C, the α1C subunit of the voltage-gated L-type calcium channel Cav1.2, rank among the most consistent and replicable genetics findings in psychiatry and have been associated with schizophrenia, bipolar disorder and major depression. However, genetic variants of complex diseases often only confer a marginal increase in disease risk, which is additionally influenced by the environment. Here we show that embryonic deletion of Cacna1c in forebrain glutamatergic neurons promotes the manifestation of endophenotypes related to psychiatric disorders including cognitive decline, impaired synaptic plasticity, reduced sociability, hyperactivity and increased anxiety. Additional analyses revealed that depletion of Cacna1c during embryonic development also increases the susceptibility to chronic stress, which suggest that Cav1.2 interacts with the environment to shape disease vulnerability. Remarkably, this was not observed when Cacna1c was deleted in glutamatergic neurons during adulthood, where the later deletion even improved cognitive flexibility, strengthened synaptic plasticity and induced stress resilience. In a parallel gene × environment design in humans, we additionally demonstrate that SNPs in CACNA1C significantly interact with adverse life events to alter the risk to develop symptoms of psychiatric disorders. Overall, our results further validate Cacna1c as a cross-disorder risk gene in mice and humans, and additionally suggest a differential role for Cav1.2 during development and adulthood in shaping cognition, sociability, emotional behavior and stress susceptibility. This may prompt the consideration for pharmacological manipulation of Cav1.2 in neuropsychiatric disorders with developmental and/or stress-related origins.


Subject(s)
Calcium Channels, L-Type/genetics , Calcium Channels, L-Type/physiology , Mental Disorders/genetics , Adult , Black or African American , Animals , Bipolar Disorder/genetics , Calcium Channels/genetics , Depressive Disorder, Major/genetics , Disease Models, Animal , Female , Genetic Predisposition to Disease/genetics , Genetic Variation/genetics , Humans , Male , Mice/embryology , Mice, Transgenic/genetics , Neurons/metabolism , Polymorphism, Single Nucleotide/genetics , Schizophrenia/genetics
4.
Transl Psychiatry ; 7(9): e1228, 2017 09 05.
Article in English | MEDLINE | ID: mdl-28872640

ABSTRACT

Nectin-3, a cell adhesion molecule enriched in hippocampal neurons, has been implicated in stress-related cognitive disorders. Nectin-3 is expressed by granule cells in the dentate gyrus (DG), but it remains unclear whether nectin-3 in DG modulates the structural plasticity of dentate granule cells and hippocampus-dependent memory. In this study, we found that DG nectin-3 expression levels were developmentally regulated and reduced by early postnatal stress exposure in adult mice. Most importantly, knockdown of nectin-3 levels in all DG neuron populations by adeno-associated virus (AAV) mimicked the cognitive effects of early-life stress, and impaired long-term spatial memory and temporal order memory. Moreover, AAV-mediated DG nectin-3 knockdown increased the density of doublecortin-immunoreactive differentiating cells under proliferation and calretinin-immunoreactive immature neurons, but markedly decreased calbindin immunoreactivity, indicating that nectin-3 modulates the differentiation and maturation of adult-born DG granule cells. Using retrovirus to target newly generated DG neurons, we found that selective nectin-3 knockdown in new DG neurons also impaired long-term spatial memory. In addition, suppressing nectin-3 expression in new DG neurons evoked a reduction of dendritic spines, especially thin spines. Our data indicate that nectin-3 expressed in DG neurons may modulate adult neurogenesis, dendritic spine plasticity and the cognitive effects of early-life stress.


Subject(s)
Dendritic Spines/physiology , Dentate Gyrus/physiology , Memory, Long-Term/physiology , Nectins/physiology , Neurogenesis/physiology , Neuronal Plasticity/physiology , Spatial Memory/physiology , Stress, Psychological/physiopathology , Animals , Behavior, Animal/physiology , Dendritic Spines/genetics , Dentate Gyrus/cytology , Disease Models, Animal , Female , Gene Knockdown Techniques , Male , Mice , Mice, Inbred C57BL , Nectins/genetics , Neurogenesis/genetics , Neuronal Plasticity/genetics , Stress, Psychological/genetics
5.
Mol Psychiatry ; 22(3): 466-475, 2017 03.
Article in English | MEDLINE | ID: mdl-27240530

ABSTRACT

Anxiety disorders constitute a major disease and social burden worldwide; however, many questions concerning the underlying molecular mechanisms still remain open. Besides the involvement of the major excitatory (glutamate) and inhibitory (gamma aminobutyric acid (GABA)) neurotransmitter circuits in anxiety disorders, the stress system has been directly implicated in the pathophysiology of these complex mental illnesses. The glucocorticoid receptor (GR) is the major receptor for the stress hormone cortisol (corticosterone in rodents) and is widely expressed in excitatory and inhibitory neurons, as well as in glial cells. However, currently it is unknown which of these cell populations mediate GR actions that eventually regulate fear- and anxiety-related behaviors. In order to address this question, we generated mice lacking the receptor specifically in forebrain glutamatergic or GABAergic neurons by breeding GRflox/flox mice to Nex-Cre or Dlx5/6-Cre mice, respectively. GR deletion specifically in glutamatergic, but not in GABAergic, neurons induced hypothalamic-pituitary-adrenal axis hyperactivity and reduced fear- and anxiety-related behavior. This was paralleled by reduced GR-dependent electrophysiological responses in the basolateral amygdala (BLA). Importantly, viral-mediated GR deletion additionally showed that fear expression, but not anxiety, is regulated by GRs in glutamatergic neurons of the BLA. This suggests that pathological anxiety likely results from altered GR signaling in glutamatergic circuits of several forebrain regions, while modulation of fear-related behavior can largely be ascribed to GR signaling in glutamatergic neurons of the BLA. Collectively, our results reveal a major contribution of GRs in the brain's key excitatory, but not inhibitory, neurotransmitter system in the regulation of fear and anxiety behaviors, which is crucial to our understanding of the molecular mechanisms underlying anxiety disorders.


Subject(s)
Anxiety Disorders/physiopathology , Receptors, Glucocorticoid/metabolism , Receptors, Glutamate/metabolism , Amygdala/metabolism , Animals , Anxiety/physiopathology , Basolateral Nuclear Complex/metabolism , Corticosterone/metabolism , Excitatory Amino Acid Agents/metabolism , Fear/physiology , GABA Agents/metabolism , GABAergic Neurons/metabolism , Glutamic Acid/metabolism , Hypothalamo-Hypophyseal System/metabolism , Mice , Mice, Knockout , Neurons/metabolism , Pituitary-Adrenal System/metabolism , Prosencephalon/metabolism , Synaptic Transmission/physiology , gamma-Aminobutyric Acid/metabolism
6.
Stress ; 19(1): 83-90, 2016.
Article in English | MEDLINE | ID: mdl-26585320

ABSTRACT

Major depression is a multifactorial disease, involving both environmental and genetic risk factors. Recently, SLC6A15 - a neutral amino acid transporter mainly expressed in neurons - was proposed as a new candidate gene for major depression and stress vulnerability. Risk allele carriers for a single nucleotide polymorphism (SNP) in a SLC6A15 regulatory region display altered hippocampal volume, glutamate levels, and hypothalamus-pituitary-adrenal axis activity, all markers associated with major depression. Despite this genetic link between SLC6A15 and depression, its functional role with regard to the development and maintenance of depressive disorder is still unclear. The aim of the current study was therefore to characterize the role of mouse slc6a15 in modulating brain function and behavior, especially in relation to stress as a key risk factor for the development of mood disorders. We investigated the effects of slc6a15 manipulation using two mouse models, a conventional slc6a15 knock-out mouse line (SLC-KO) and a virus-mediated hippocampal slc6a15 overexpression (SLC-OE) model. Mice were tested under basal conditions and following chronic social stress. We found that SLC-KO animals displayed a similar behavioral profile to wild-type littermates (SLC-WT) under basal conditions. Interestingly, following chronic social stress SLC-KO animals showed lower levels of anxiety- and depressive-like behavior compared to stressed WT littermates. In support of these findings, SLC-OE animals displayed increased anxiety-like behavior already under basal condition. We also provide evidence that GluR1 expression in the dentate gyrus, but not GluR2 or NR1, are regulated by slc6a15 expression, and may contribute to the difference in stress responsiveness observed between SLC-KO and SLC-WT animals. Taken together, our data demonstrate that slc6a15 plays a role in modulating emotional behavior, possibly mediated by its impact on glutamatergic neurotransmission.


Subject(s)
Amino Acid Transport Systems, Neutral/genetics , Anxiety/genetics , Behavior, Animal , Depression/genetics , Hippocampus/metabolism , RNA, Messenger/metabolism , Stress, Psychological/genetics , Alleles , Animals , Corticosterone/blood , Dentate Gyrus/metabolism , Disease Models, Animal , Gene Expression Regulation , Gene Knock-In Techniques , Genotype , Male , Mice , Mice, Knockout , Mood Disorders/genetics , Nerve Tissue Proteins/genetics , Polymorphism, Single Nucleotide , Receptors, AMPA/genetics , Receptors, N-Methyl-D-Aspartate/genetics , Risk Factors , Stress, Psychological/metabolism
7.
Mol Psychiatry ; 21(2): 277-89, 2016 Feb.
Article in English | MEDLINE | ID: mdl-25849320

ABSTRACT

Psychotropic medications target glycogen synthase kinase 3ß (GSK3ß), but the functional integration with other factors relevant for drug efficacy is poorly understood. We discovered that the suggested psychiatric risk factor FK506 binding protein 51 (FKBP51) increases phosphorylation of GSK3ß at serine 9 (pGSK3ß(S9)). FKBP51 associates with GSK3ß mainly through its FK1 domain; furthermore, it also changes GSK3ß's heterocomplex assembly by associating with the phosphatase PP2A and the kinase cyclin-dependent kinase 5. FKBP51 acts through GSK3ß on the downstream targets Tau, ß-catenin and T-cell factor/lymphoid enhancing factor (TCF/LEF). Lithium and the antidepressant (AD) paroxetine (PAR) functionally synergize with FKBP51, as revealed by reporter gene and protein association analyses. Deletion of FKBP51 blunted the PAR- or lithium-induced increase in pGSK3ß(S9) in cells and mice and attenuated the behavioral effects of lithium treatment. Clinical improvement in depressive patients was predicted by baseline GSK3ß pathway activity and by pGSK3ß(S9) reactivity to ex vivo treatment of peripheral blood mononuclear lymphocytes with lithium or PAR. In sum, FKBP51-directed GSK3ß activity contributes to the action of psychotropic medications. Components of the FKBP51-GSK3ß pathway may be useful as biomarkers predicting AD response and as targets for the development of novel ADs.


Subject(s)
Glycogen Synthase Kinase 3/metabolism , Tacrolimus Binding Proteins/genetics , Adult , Animals , Antidepressive Agents/pharmacology , Biomarkers/blood , Cell Culture Techniques , Cell Line , Cyclin-Dependent Kinase 5 , Female , Glycogen Synthase Kinase 3 beta , HEK293 Cells , Humans , Leukocytes, Mononuclear/metabolism , Lithium , Male , Mice , Middle Aged , Phosphorylation/drug effects , Psychotropic Drugs/pharmacology , Signal Transduction/drug effects , Tacrolimus Binding Proteins/metabolism , beta Catenin/metabolism
8.
Adv Gerontol ; 29(1): 59-67, 2016.
Article in Russian | MEDLINE | ID: mdl-28423247

ABSTRACT

Animals were subjected to seven days combined stress in a special chamber (6 isolated compartments of equal area) with removable multi-modal stressors (noise, vibration, pulsating bright light) every 5 minutes on the stochastic scheme with restraint and temperature rise in the chamber during 30-minute stressing time sessions. After exposure to combined stress in the ventral hippocampus of old rats (24 months) compared with adult animals (12 months) following changes were revealed: marked dystrophic changes and increased inducible nitric oxide synthase expression in pyramidal neurons of CA3 field, signs of impaired hemodynamic disorders in the microvasculature, perivascular edema, decreased endothelial nitric oxide synthase expression in microvascular endothelial cells, as well as decreased expression of serine racemase in the neuropil of the radial layer of CA1 field.


Subject(s)
Hippocampus , Aging , Animals , Nitric Oxide Synthase , Nitric Oxide Synthase Type II , Nitric Oxide Synthase Type III , Pyramidal Cells , Rats
9.
Neuroscience ; 290: 147-58, 2015 Apr 02.
Article in English | MEDLINE | ID: mdl-25637808

ABSTRACT

Understanding the molecular mechanisms that promote stress resilience might open up new therapeutic avenues to prevent stress-related disorders. We recently characterized a stress and glucocorticoid-regulated gene, down-regulated in renal cell carcinoma - DRR1 (Fam107A). DRR1 is expressed in the mouse brain; it is up-regulated by stress and glucocorticoids and modulates neuronal actin dynamics. In the adult mouse, DRR1 was shown to facilitate specific behaviors which might be protective against some of the deleterious consequences of stress exposure: in the hippocampal CA3 region, DRR1 improved cognitive performance whereas in the septum, it specifically increased social behavior. Therefore DRR1 was suggested as a candidate protein promoting stress-resilience. Fam107B (family with sequence similarity 107, member B) is the unique paralog of DRR1, and both share high sequence similarities, predicted glucocorticoid response elements, heat-shock induction and tumor suppressor properties. So far, the role of Fam107B in the central nervous system was not studied. The aim of the present investigation, therefore, was to analyze whether Fam107B and DRR1 display comparable mRNA expression patterns in the brain and whether both are modulated by stress and glucocorticoids. Spatio-temporal mapping of Fam107B mRNA expression in the embryonic and adult mouse brain, by means of in situ hybridization, showed that Fam107B was expressed during embryogenesis and in the adulthood, with particularly high and specific expression in the forming telencephalon suggestive of an involvement in corticogenesis. In the adult mouse, expression was restricted to neurogenic niches, like the dentate gyrus. In contrast to DRR1, Fam107B mRNA expression failed to be modulated by glucocorticoids and social stress in the adult mouse. In summary, Fam107B and DRR1 show different spatio-temporal expression patterns in the central nervous system, suggesting at least partially different functional roles in the brain, and where the glucocorticoid receptor (GR)-induced regulation appears to be a unique property of DRR1.


Subject(s)
Brain/growth & development , Brain/metabolism , Stress, Psychological/metabolism , Tumor Suppressor Proteins/metabolism , Acute Disease , Amino Acid Sequence , Animals , Brain/drug effects , Chronic Disease , Dexamethasone/pharmacology , Dominance-Subordination , Glucocorticoids/pharmacology , In Situ Hybridization , Male , Mice , Mice, Inbred C57BL , Molecular Sequence Data , RNA, Messenger/metabolism , Receptors, Glucocorticoid/agonists , Receptors, Glucocorticoid/metabolism , Sequence Homology, Amino Acid , Stem Cell Niche/drug effects , Stem Cell Niche/physiology , Time Factors , Tumor Suppressor Proteins/genetics
10.
Eur J Neurosci ; 40(4): 2691-700, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24840018

ABSTRACT

It has been shown previously (Sotnikov et al., ) that mice selectively inbred for high anxiety-related behavior (HAB) vs. low anxiety-related behavior in the elevated plus maze differentially respond to trimethylthiazoline (TMT), a synthetic fox fecal odor. However, less is known about whether environmental factors can rescue these extreme phenotypes. Here, we found that an enriched environment (EE) provided during early adolescence induced anxiolytic effects in HAB (HAB-EE) mice, rescuing their strong avoidance behavior induced by TMT. In a series of experiments, the contribution of maternal, juvenile and adolescent behavior to the anxiolytic effects elicited by EE was investigated. At the molecular level, using c-fos expression mapping, we found that the activity of the medial and basolateral amygdala was significantly reduced in HAB-EE mice after TMT exposure. We further analysed the expression of Crhr1, as its amount in the amygdala has been reported to be important for the regulation of anxiety-related behavior after EE. Indeed, in situ hybridisation indicated significantly decreased Crhr1 expression in the basolateral and central amygdala of HAB-EE mice. To further test the involvement of Crhr1 in TMT-induced avoidance, we exposed conditional glutamatergic-specific Crhr1-knockout mice to the odor. The behavioral response of Crhr1-knockout mice mimicked that of HAB-EE mice, and c-fos expression in the amygdala after TMT exposure was significantly lower compared with controls, thereby further supporting a critical involvement of Crhr1 in environmentally-induced anxiolysis. Altogether, our results indicate that EE can rescue strong avoidance of TMT by HAB mice with Crhr1 expression in the amygdala being critically involved.


Subject(s)
Amygdala/metabolism , Anxiety/metabolism , Environment, Controlled , Proto-Oncogene Proteins c-fos/metabolism , Receptors, Corticotropin-Releasing Hormone/metabolism , Animals , Anxiety/chemically induced , Anxiety/genetics , Brain/metabolism , Genes, Immediate-Early , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Motor Activity , Receptors, Corticotropin-Releasing Hormone/genetics , Thiazoles/toxicity
11.
Neuroscience ; 250: 112-20, 2013 Oct 10.
Article in English | MEDLINE | ID: mdl-23838393

ABSTRACT

Synaptic plasticity, specifically long-term potentiation and long-term depression, is thought to be the underlying cellular mechanism for learning and memory processes in the brain. About two decades ago a new concept was introduced, namely metaplasticity, which comprises changes that modify the properties of synaptic plasticity due to a priming or preconditioning event. While metaplasticity was initially defined and studied predominantly on a synaptic and cellular level, it soon became apparent that the term could also be very useful to describe plasticity changes on a more global level, including environmental stressors as priming events and altered behavior as outcome measures. We consider here whether it is helpful to conceptualize these latter effects as "behavioral metaplasticity", and in which sense this view fits into the original concept of metaplasticity. By integrating the literature on environmental effects on plasticity, especially stress, plus developmental aspects as well as genetic and epigenetic modifications, we shape the framework in which the term "behavioral metaplasticity" should be considered and discuss research directions that can help to unravel the mechanisms involved in both synaptic and behavioral metaplasticity.


Subject(s)
Neuronal Plasticity/physiology , Stress, Psychological/physiopathology , Synapses/physiology , Animals , Behavior/physiology , Behavior, Animal/physiology , Humans , Learning/physiology , Memory/physiology
12.
Neuroscience ; 232: 161-8, 2013 Mar 01.
Article in English | MEDLINE | ID: mdl-23238574

ABSTRACT

The onset of action of antidepressants (ADs) usually takes several weeks, but first molecular responses to these drugs may appear already after acute administration. The Extracellular Signal-regulated Kinase/Mitogen-Activated Protein Kinase (ERK/MAPK) signaling pathway is a target of ADs and an important pathway involved in cellular plasticity. In major depressive disorder (MDD), especially the prefrontal cortex (PFC) and hippocampus (Hip) are most likely affected in depressive patients and recent work revealed a hyperactivated ERK signaling in the rat PFC after chronic stress, a precipitating factor for MDD. Strong evidences support that not only neurons but also astrocytes participate in neuronal activity and may therefore additionally be a substrate of AD action. In this study, we show by Western blot that neither fluoxetine (FLX) nor desipramine (DMI) preferentially affects the activation of one of the two ERK isoforms, ERK1 and ERK2, with respect to the other. Further immunohistochemical analysis in the PFC revealed that basal levels of phospho-activated ERK (pERK) are mostly found in neurons in contrast to very few astrocytes. Both ADs can inhibit neuronal pERK as early as 15 min after drug administration with peculiar regional and layer specificities. Contrarily, at this time point none of the two ADs shows a clear modulation of astrocytic pERK. We propose that this mechanism of action of ADs may be protective against an exacerbated cortical ERK activity that may exert detrimental effects on susceptible neuronal populations. Our findings on acute effects of AD treatment in the adult mouse PFC encourage to examine further how this treatment might influence pERK in animal models of depression to identify early targets of AD action.


Subject(s)
Antidepressive Agents/pharmacology , Astrocytes/drug effects , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Neurons/drug effects , Prefrontal Cortex/drug effects , Animals , Astrocytes/enzymology , Blotting, Western , Corpus Striatum/drug effects , Corpus Striatum/enzymology , Desipramine/pharmacology , Fluorescent Antibody Technique , Fluoxetine/pharmacology , Hippocampus/drug effects , Hippocampus/enzymology , Male , Mice , Microscopy, Confocal , Mitogen-Activated Protein Kinase 1/antagonists & inhibitors , Mitogen-Activated Protein Kinase 3/antagonists & inhibitors , Neurons/enzymology , Prefrontal Cortex/enzymology
13.
Transl Psychiatry ; 2: e177, 2012 Oct 23.
Article in English | MEDLINE | ID: mdl-23092981

ABSTRACT

Despite the overt need for improved treatment modalities in depression, efforts to develop conceptually novel antidepressants have been relatively unsuccessful so far. Here we present a translational approach combining results from hypothesis-free animal experiments with data from a genetic association study in depression. Comparing genes regulated by chronic paroxetine treatment in the mouse hippocampus with genes showing nominally significant association with antidepressant treatment response in two pharmacogenetic studies, the activin pathway was the only one to show this dual pattern of association and therefore selected as a candidate. We examined the regulation of activin A and activin receptor type IA mRNA following antidepressant treatment. We investigated the effects of stereotaxic infusion of activin into the hippocampus and the amygdala in a behavioural model of depression. To analyse whether variants in genes in the activin signalling pathway predict antidepressant treatment response, we performed a human genetic association study. Significant changes in the expression of genes in the activin signalling pathway were observed following 1 and 4 weeks of treatment. Injection of activin A into the hippocampus exerts acute antidepressant-like effects. Polymorphisms in the betaglycan gene, a co-receptor mediating functional antagonism of activin signalling, significantly predict treatment outcome in our system-wide pharmacogenetics study in depression. We provide convergent evidence from mouse and human data that genes in the activin signalling pathway are promising novel candidates involved in the neurobiogical mechanisms underlying antidepressant mechanisms of action. Further, our data suggest this pathway to be a target for more rapid-acting antidepressants in the future.


Subject(s)
Activins , Antidepressive Agents , Brain , Depressive Disorder , Paroxetine/pharmacology , Proteoglycans/genetics , RNA, Messenger/analysis , Receptors, Transforming Growth Factor beta/genetics , Activin Receptors, Type I/genetics , Activin Receptors, Type I/metabolism , Activins/genetics , Activins/metabolism , Activins/pharmacology , Adult , Aged , Amygdala/drug effects , Animals , Antidepressive Agents/pharmacology , Antidepressive Agents/therapeutic use , Behavior, Animal/drug effects , Brain/drug effects , Brain/metabolism , Dentate Gyrus/drug effects , Depressive Disorder/drug therapy , Depressive Disorder/genetics , Female , Genetic Association Studies , Genotype , Humans , Male , Mice , Middle Aged , Pharmacogenetics , Polymorphism, Single Nucleotide , Signal Transduction
14.
Horm Behav ; 57(4-5): 415-20, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20100488

ABSTRACT

Major depression is one of the most prevalent stress-related psychiatric diseases. Next to environmental influences such as chronic social stress, gender is among the strongest risk factors for major depression, with women having a twice as high risk to develop the disease compared to men. While there is abundant literature on the effects of chronic social stress in male rodents, there is a serious lack of information on gender-specific effects. Especially in mice, which due to the wide availability of transgenic lines offer a unique opportunity to study gene x environment interactions, there is no existing model of chronic social stress that is applicable to both sexes. We here describe the effects of chronic social stress based on the disruption of the social network in a group-housed situation in female mice, a model that was recently described and validated for male mice. In this model, the group composition of the mice is changed twice per week for a period of 7 weeks, covering the adolescent and early adulthood period. We observed that housing in an unpredictable social environment resulted in chronic stress in female mice. The observed effects, which included increased adrenal weight, decreased thymus weight, increased corticosterone levels, and increased anxiety-like behavior, were very similar to the described effects of this paradigm in male mice. In addition, we observed a distinct expression of stress system-related genes in female mice following chronic stress exposure. Our results validate this model as a suitable approach to study chronic social stress in female mice and open up the opportunity to use this model with transgenic or knockout mouse lines.


Subject(s)
Social Environment , Stress, Psychological/psychology , Adrenocorticotropic Hormone/blood , Animals , Anxiety/psychology , Arginine Vasopressin/biosynthesis , Arginine Vasopressin/genetics , Body Weight/physiology , Corticosterone/blood , Corticotropin-Releasing Hormone/biosynthesis , Corticotropin-Releasing Hormone/genetics , Disease Models, Animal , Feeding Behavior/physiology , Female , Gene Expression , Hierarchy, Social , In Situ Hybridization , Male , Mice , Motor Activity/physiology , Organ Size/physiology , Receptors, Gonadotropin/biosynthesis , Receptors, Gonadotropin/genetics , Sex Characteristics
15.
Int J Dev Neurosci ; 28(2): 217-24, 2010 Apr.
Article in English | MEDLINE | ID: mdl-19897026

ABSTRACT

One of the striking characteristics of the developing neuroendocrine system of rats and mice is the stress hypo-responsive period (SHRP), i.e. low basal corticosterone secretion and the inability to increase corticosterone in response to mild stressors during the first 2 weeks of life. However, immediately after 24 h of deprivation from maternal care the response of the hypothalamic-pituitary-adrenal (HPA) axis to mild stressors is enhanced. This study examines in CD1 mouse pups the recovery pattern of markers of HPA axis (re)activity from maternal deprivation (once for 24 h from postnatal days (pnds) 3 to 4). As expected, deprivation induced a profound corticosterone response to novelty immediately after deprivation. In contrast, 1 day after reunion with the mother (pnd 5), this effect was abolished, lasting for at least 3 days. Basal corticosterone remained even below control levels. Corticotropin-releasing hormone (CRH) mRNA expression in the hypothalamic paraventricular nucleus (PVN) was suppressed for 2 days, exceeded control levels at pnds 7 and 8, and subsequently followed the gradual decline observed in controls until pnd 12. Delayed and rather short-lasting changes were found for adrenocorticotropic hormone (low at pnd 5), and glucocorticoid receptor mRNA expression (decreased in the PVN at pnd 4, and in the hippocampal CA1 area at pnd 5). Hippocampal mineralocorticoid receptor mRNA expression was unaffected. From pnds 9 to 13, both deprived and control pups gradually emerged from the SHRP in a similar temporal pattern. In conclusion, maternal deprivation at pnd 3 augments hypo-responsiveness of corticosterone secretion to mild stress for several days, but does not affect the duration of the SHRP. Whether CRH and glucocorticoid receptor changes are cause or consequence remains to be established.


Subject(s)
Anxiety, Separation/physiopathology , Corticosterone/metabolism , Hypothalamo-Hypophyseal System/physiopathology , Mothers , Pituitary-Adrenal System/physiopathology , Animals , Animals, Newborn , Mice , Pituitary-Adrenal System/drug effects
16.
Psychoneuroendocrinology ; 35(5): 635-43, 2010 Jun.
Article in English | MEDLINE | ID: mdl-19857931

ABSTRACT

Chronic stress is a key risk factor for a variety of diseases, including depression. There is a large degree of individual variation in the ability to recover successfully from a chronic stress exposure, but the determinants of this individual stress susceptibility are still poorly understood. We recently developed a novel mouse paradigm for chronic social stress during adolescence, which closely mimics the human condition of chronic social stress in respect to construct, face and predictive validity. By applying this chronic stress model to a large number of animals we aimed at identifying individuals that are either resilient or vulnerable to the persistent effects of chronic social stress exposure. Animals showing markedly elevated basal corticosterone levels 5 weeks following the end of the stress paradigm were considered "vulnerable", whereas individuals recovering quickly and being indistinguishable from controls were classified as "resilient". Stress vulnerability was associated with an increased level of corticotropin-releasing hormone in the paraventricular nucleus, decreased hippocampal mineralocorticoid receptor expression as well as increased anxiety- and depression-like behavior compared to resilient and control animals. In summary, we show that by using a large cohort of animals it is possible to select individuals that are vulnerable or resilient to the lasting effects of chronic social stress. The vulnerable phenotype mimics many aspects of stress-related human affective disorders and this may be used as a novel approach to study depression in an animal model, ultimately contributing to a better understanding and treatment of stress-related disorders.


Subject(s)
Brain/metabolism , Corticosterone/blood , Depression/metabolism , Disease Models, Animal , Disease Susceptibility/metabolism , Social Behavior , Stress, Psychological/metabolism , Stress, Psychological/psychology , Animals , Anxiety/metabolism , Behavior, Animal , Body Weight , Corticotropin-Releasing Hormone/metabolism , Male , Mice , Mice, Inbred Strains , Organ Size , Phenotype , Receptors, Mineralocorticoid/metabolism , Time Factors
17.
J Neuroendocrinol ; 21(5): 473-80, 2009 May.
Article in English | MEDLINE | ID: mdl-19302188

ABSTRACT

The importance of restraining stress-induced activation of the hypothalamic-pituitary-adrenocortical (HPA) system within tolerable limits requires efficient mechanisms for feedback inhibition. Recently, central corticotrophin-releasing hormone (CRH) receptor type 1 (CRHR1) has been shown to mediate HPA system feedback inhibition. To date, most of the data regarding stress-associated expression changes of CRHR1 and CRHR2 mRNA and their ligand CRH have been generated in rats. Taken considerable species differences into consideration, and with the growing importance of transgenic mice, a systematic analysis of the time course of expression changes of CRH and its two receptors in the mouse brain is needed to provide more insight into the regulation of the HPA system, both under physiological and pathophysiological conditions in this species. We analysed in detail the time course of expression changes of CRH, CRHR1 and CRHR2 mRNA after of restraint stress in mice in stress-relevant brain regions (paraventricular nucleus, hippocampus, neocortex). We could show a rapid, strong and long-lasting decrease in cortical and hippocampal CRHR1 mRNA expression after stress, whereas CRHR2 mRNA increased in the same neuroanatomical areas. In situ hybridisation analyses could be further confirmed at the protein level by CRH receptor autoradiography with changes in CRH binding that persisted even 7 days after a single episode of restraint stress. Our observation that stress has opposing effects on CRHR1 and CRHR2 neuronal systems supports the idea that regulation of the relative contribution of the two CRH receptors to brain CRH pathways may be essential in coordinating physiological responses to stress. We further hypothesise that the sustained alteration of CRH receptor expression and binding after a single episode of stress could mediate the long-term effects of stress on neuroendocrine function and emotional regulation.


Subject(s)
Brain/metabolism , Receptors, Corticotropin-Releasing Hormone/metabolism , Restraint, Physical , Stress, Psychological , Animals , Autoradiography , Brain/anatomy & histology , Corticotropin-Releasing Hormone/genetics , Corticotropin-Releasing Hormone/metabolism , Humans , Hypothalamo-Hypophyseal System/physiology , Hypothalamo-Hypophyseal System/physiopathology , In Situ Hybridization , Male , Mice , Mice, Inbred C57BL , Pituitary-Adrenal System/physiology , Pituitary-Adrenal System/physiopathology , Rats , Receptors, Corticotropin-Releasing Hormone/genetics
18.
Stress ; 12(1): 89-94, 2009 Jan.
Article in English | MEDLINE | ID: mdl-18951248

ABSTRACT

Obesity and visceral fat accumulation are key features of the metabolic syndrome that represents one of the main health problems in western societies due to its neurovascular and cardiovascular complications. Epidemiological studies have identified chronic stress exposure as an important risk factor for the development of obesity and metabolic syndrome, but also psychiatric diseases, especially affective disorders. However, it is still unclear if chronic stress has merely transient or potentially lasting effects on body composition. Here, we investigated the effects of chronic social stress during the adolescent period on body fat composition in mice one year after the cessation of the stressor. We found that stress exposure during the adolescent period decreases subcutaneous fat content, without change in visceral fat, and consequently increases the visceral fat/subcutaneous fat ratio in adulthood. Further, we demonstrated that treatment with a selective serotonin reuptake inhibitor (paroxetine) during stress exposure prevented later effects on body fat distribution. These results from a recently validated chronic stress paradigm in mice provide evidence that stressful experiences during adolescence can alter body fat distribution in adulthood, thereby possibly contributing to an increased risk for metabolic diseases. Antidepressant treatment disrupted this effect underlining the link between the stress hormone system, metabolic homeostasis and affective disorders.


Subject(s)
Abdominal Fat/pathology , Antidepressive Agents/therapeutic use , Paroxetine/therapeutic use , Social Behavior , Stress, Psychological/pathology , Abdominal Fat/drug effects , Animals , Behavior, Animal/drug effects , Blood Glucose/drug effects , Corticosterone/blood , Eating/drug effects , Male , Mice , Models, Animal , Motor Activity/drug effects , Stress, Psychological/drug therapy , Subcutaneous Fat/drug effects , Subcutaneous Fat/pathology
19.
Psychoneuroendocrinology ; 32(5): 417-29, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17449187

ABSTRACT

Chronic stress is widely regarded as a key risk factor for a variety of diseases. A large number of paradigms have been used to induce chronic stress in rodents. However, many of these paradigms do not consider the etiology of human stress-associated disorders, where the stressors involved are mostly of social nature and the effects of the stress exposure persist even if the stressor is discontinued. In addition, many chronic stress paradigms are problematic with regard to stress adaptation, continuity, duration and applicability. Here we describe and validate a novel chronic social stress paradigm in male mice during adolescence. We demonstrate persistent effects of chronic social stress after 1 week of rest, including altered adrenal sensitivity, decreased expression of corticosteroid receptors in the hippocampus and increased anxiety. In addition, pharmacological treatments with the antidepressant paroxetine (SSRI) or with the corticotropin-releasing hormone receptor 1 antagonist DMP696 were able to prevent aversive long-term consequences of chronic social stress. In conclusion, this novel chronic stress paradigm results in persistent alterations of hypothalamus-pituitary-adrenal axis function and behavior, which are reversible by pharmacological treatment. Moreover, this paradigm allows to investigate the interaction of genetic susceptibility and environmental risk factors.


Subject(s)
Corticosterone/blood , Exploratory Behavior/physiology , Receptors, Steroid/metabolism , Social Behavior , Stress, Psychological/physiopathology , Adaptation, Physiological , Adaptation, Psychological , Adrenal Glands/metabolism , Adrenocorticotropic Hormone/blood , Age Factors , Analysis of Variance , Animals , Antidepressive Agents, Second-Generation/therapeutic use , Chronic Disease , Disease Models, Animal , Hierarchy, Social , Hippocampus/metabolism , Hypothalamo-Hypophyseal System/metabolism , Male , Mice , Paroxetine/therapeutic use , Pituitary-Adrenal System/metabolism , Stress, Psychological/drug therapy , Stress, Psychological/psychology
20.
J Neurosci Methods ; 162(1-2): 180-6, 2007 May 15.
Article in English | MEDLINE | ID: mdl-17320190

ABSTRACT

Many behavioural tests in rodents are based on the premise that basal locomotor activity of the animals is similar between the tested groups. The measurement of basal home cage activity is therefore an essential parameter, that should be included in all studies which employ tests of anxiety or cognition. Currently available systems for the assessment of home cage locomotion are often complex and expensive. Here we describe and validate a novel, simple and cost-efficient apparatus for the assessment of basic home cage locomotor activity in rodents. Circadian dark-light activity patterns can be reliably obtained with the home cage activity counter. Furthermore, changes in locomotion induced by novelty or pharmacological treatment were reliably and sensitively detected by the apparatus. Thus, the here presented home cage activity counter can be used for the measurement of basal home cage locomotor activity.


Subject(s)
Motor Activity/physiology , Acclimatization/drug effects , Animals , Dextroamphetamine/pharmacology , Dizocilpine Maleate/pharmacology , Environment , Equipment Design , Housing, Animal , Male , Mice , Motor Activity/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...