Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 380
Filter
1.
J Steroid Biochem Mol Biol ; 238: 106461, 2024 04.
Article in English | MEDLINE | ID: mdl-38219844

ABSTRACT

There is growing evidence indicating that mineralocorticoid receptor (MR) expression influences a wide variety of functions in metabolic and immune response. The present study explored if antagonism of the MR reduces neuroinflammation in the spinal cord of mice with experimental autoimmune encephalomyelitis (EAE). Eplerenone (EPLE) (100 mg/kg dissolved in 30% 2-hydroxypropyl-ß-cyclodextrin) was administered intraperitoneally (i.p.) daily from EAE induction (day 0) until sacrificed on day 17 post-induction. The MR blocker (a) significantly decreased the inflammatory parameters TLR4, MYD88, IL-1ß, and iNOS mRNAs; (b) attenuated HMGB1, NLRP3, TGF-ß mRNAs, microglia, and aquaporin4 immunoreaction without modifying GFAP. Serum IL-1ß was also decreased in the EAE+EPLE group. Moreover, EPLE treatment prevented demyelination and improved clinical signs of EAE mice. Interestingly, MR was decreased and GR remained unchanged in EAE mice while EPLE treatment restored MR expression, suggesting that a dysbalanced MR/GR was associated with the development of neuroinflammation. Our results indicated that MR blockage with EPLE attenuated inflammation-related spinal cord pathology in the EAE mouse model of Multiple Sclerosis, supporting a novel therapeutic approach for immune-related diseases.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental , Mice , Animals , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Encephalomyelitis, Autoimmune, Experimental/metabolism , Eplerenone/pharmacology , Eplerenone/therapeutic use , Mineralocorticoid Receptor Antagonists/pharmacology , Mineralocorticoid Receptor Antagonists/therapeutic use , Neuroinflammatory Diseases , Spinal Cord/pathology , Mice, Inbred C57BL
2.
Front Neuroendocrinol ; 49: 124-145, 2018 04.
Article in English | MEDLINE | ID: mdl-29428549

ABSTRACT

Bruce McEwen's discovery of receptors for corticosterone in the rat hippocampus introduced higher brain circuits in the neuroendocrinology of stress. Subsequently, these receptors were identified as mineralocorticoid receptors (MRs) that are involved in appraisal processes, choice of coping style, encoding and retrieval. The MR-mediated actions on cognition are complemented by slower actions via glucocorticoid receptors (GRs) on contextualization, rationalization and memory storage of the experience. These sequential phases in cognitive performance depend on synaptic metaplasticity that is regulated by coordinate MR- and GR activation. The receptor activation includes recruitment of coregulators and transcription factors as determinants of context-dependent specificity in steroid action; they can be modulated by genetic variation and (early) experience. Interestingly, inflammatory responses to damage seem to be governed by a similarly balanced MR:GR-mediated action as the initiating, terminating and priming mechanisms involved in stress-adaptation. We conclude with five questions challenging the MR:GR balance hypothesis.


Subject(s)
Amygdala/metabolism , Cognitive Dysfunction/metabolism , Hippocampus/metabolism , Inflammation/metabolism , Neuronal Plasticity/physiology , Receptors, Glucocorticoid/metabolism , Receptors, Mineralocorticoid/metabolism , Stress, Psychological/metabolism , Animals , Cognitive Dysfunction/etiology , Cognitive Dysfunction/physiopathology , Stress, Psychological/complications , Stress, Psychological/physiopathology
3.
J Neuroendocrinol ; 29(7)2017 07.
Article in English | MEDLINE | ID: mdl-28523794

ABSTRACT

Damage observed in the hippocampus of the adult spontaneously hypertensive rat (SHR) resembles the neuropathology of mineralocorticoid-induced hypertension, supporting a similar endocrine dysfunction in both entities. In the present study, we tested the hypothesis that increased expression of the hippocampal mineralocorticoid receptor (MR) in SHR animals is associated with a prevalent expression of pro-inflammatory over anti-inflammatory factors. Accordingly, in the hippocampus, we measured mRNA expression and immunoreactivity of the MR and glucocorticoid receptor (GR) using a quantitative polymerase chain reaction and histochemistry. We also measured serum-glucocorticoid-activated kinase 1 (Sgk1 mRNA), the number and phenotype of Iba1+ microglia, as well as mRNA expression levels of the pro-inflammatory factors cyclo-oxygenase 2 (Cox2), Nlrp3 inflammasome and tumour necrosis factor α (Tnfα). Expression of anti-inflammatory transforming growth factor (Tgf)ß mRNA and the NADPH-diaphorase activity of nitric oxide synthase (NOS) were also determined. The results showed that, in the hippocampus of SHR rats, expression of MR and the number of immunoreactive MR/GR co-expressing cells were increased compared to Wistar-Kyoto control animals. Expression of Sgk1, Cox2, Nlrp3 and the number of ramified glia cells positive for Iba1+ were also increased, whereas Tgfß mRNA expression and the NADPH-diaphorase activity of NOS were decreased. We propose that, in the SHR hippocampus, increased MR expression causes a bias towards a pro-inflammatory phenotype characteristic for hypertensive encephalopathy.


Subject(s)
Hippocampus/metabolism , Inflammation/metabolism , Neurons/metabolism , Receptors, Mineralocorticoid/metabolism , Animals , Cyclooxygenase 2/genetics , Cyclooxygenase 2/metabolism , Immediate-Early Proteins/genetics , Immediate-Early Proteins/metabolism , Male , Microglia/metabolism , Nitric Oxide Synthase/genetics , Nitric Oxide Synthase/metabolism , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Rats , Rats, Inbred SHR , Rats, Inbred WKY , Receptors, Glucocorticoid/genetics , Receptors, Glucocorticoid/metabolism , Receptors, Mineralocorticoid/genetics , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/metabolism
4.
Neural Plast ; 2016: 6503162, 2016.
Article in English | MEDLINE | ID: mdl-27034848

ABSTRACT

In the forced swim test (FST) rodents progressively show increased episodes of immobility if immersed in a beaker with water from where escape is not possible. In this test, a compound qualifies as a potential antidepressant if it prevents or delays the transition to this passive (energy conserving) behavioural style. In the past decade however the switch from active to passive "coping" was used increasingly to describe the phenotype of an animal that has been exposed to a stressful history and/or genetic modification. A PubMed analysis revealed that in a rapidly increasing number of papers (currently more than 2,000) stress-related immobility in the FST is labeled as a depression-like phenotype. In this contribution we will examine the different phases of information processing during coping with the forced swim stressor. For this purpose we focus on the action of corticosterone that is mediated by the closely related mineralocorticoid receptors (MR) and glucocorticoid receptors (GR) in the limbic brain. The evidence available suggests a model in which we propose that the limbic MR-mediated response selection operates in complementary fashion with dopaminergic accumbens/prefrontal executive functions to regulate the transition between active and passive coping styles. Upon rescue from the beaker the preferred, mostly passive, coping style is stored in the memory via a GR-dependent action in the hippocampal dentate gyrus. It is concluded that the rodent's behavioural response to a forced swim stressor does not reflect depression. Rather the forced swim experience provides a unique paradigm to investigate the mechanistic underpinning of stress coping and adaptation.


Subject(s)
Adaptation, Psychological/physiology , Brain/physiopathology , Receptors, Glucocorticoid/physiology , Receptors, Mineralocorticoid/physiology , Stress, Psychological/physiopathology , Stress, Psychological/psychology , Animals , Behavior, Animal , Brain/metabolism , Glucocorticoids/physiology , Limbic System/metabolism , Limbic System/physiopathology , Mice , Rats , Receptors, Glucocorticoid/metabolism , Receptors, Mineralocorticoid/metabolism , Stress, Psychological/metabolism , Swimming
5.
Mol Psychiatry ; 21(12): 1733-1739, 2016 12.
Article in English | MEDLINE | ID: mdl-26976039

ABSTRACT

Maladaptive glucocorticoid effects contribute to stress-related psychopathology. The glucocorticoid receptor (GR) that mediates many of these effects uses multiple signaling pathways. We have tested the hypothesis that manipulation of downstream factors ('coregulators') can abrogate potentially maladaptive GR-mediated effects on fear-motivated behavior that are linked to corticotropin releasing hormone (CRH). For this purpose the expression ratio of two splice variants of steroid receptor coactivator-1 (SRC-1) was altered via antisense-mediated 'exon-skipping' in the central amygdala of the mouse brain. We observed that a change in splicing towards the repressive isoform SRC-1a strongly reduced glucocorticoid-induced responsiveness of Crh mRNA expression and increased methylation of the Crh promoter. The transcriptional GR target gene Fkbp5 remained responsive to glucocorticoids, indicating gene specificity of the effect. The shift of the SRC-1 splice variants altered glucocorticoid-dependent exploratory behavior and attenuated consolidation of contextual fear memory. In conclusion, our findings demonstrate that manipulation of GR signaling pathways related to the Crh gene can selectively diminish potentially maladaptive effects of glucocorticoids.


Subject(s)
Corticotropin-Releasing Hormone/metabolism , Nuclear Receptor Coactivator 1/metabolism , Alternative Splicing , Amygdala , Animals , Corticosterone/metabolism , Fear , Gene Expression Regulation/drug effects , Glucocorticoids/metabolism , Mice , Nuclear Receptor Coactivator 1/genetics , Promoter Regions, Genetic/drug effects , Protein Isoforms/genetics , RNA Isoforms , RNA, Messenger/metabolism , Receptors, Glucocorticoid/metabolism , Receptors, Steroid , Tacrolimus Binding Proteins/metabolism
6.
J Neuroendocrinol ; 28(8)2016 08.
Article in English | MEDLINE | ID: mdl-26970338

ABSTRACT

Cortisol and corticosterone act on the appraisal process, which comprises the selection of an appropriate coping style and the encoding of the experience for storage in the memory. This action exerted by the stress hormones is mediated by mineralocorticoid receptors (MRs), which are expressed abundantly in the limbic circuitry, particularly in the hippocampus. Limbic MR is down-regulated by chronic stress and during depression but induced by antidepressants. Increased MR activity inhibits hypothalamic-pituitary-adrenal axis activity, promotes slow wave sleep, reduces anxiety and switches circuit connectivity to support coping. Cortisol and emotion-cognition are affected by MR gene haplotypes based on rs5522 and rs2070951. Haplotype 1 (GA) moderates the effects of (early) life stressors, reproductive cycle and oral contraceptives. MR haplotype 2 (CA) is a gain of function variant that protects females against depression by association with an optimistic, resilient phenotype. Activation of MR therefore may offer a target for alleviating depression and cognitive dysfunction. Accordingly, the MR agonist fludrocortisone was found to enhance the efficacy of antidepressants and to improve memory and executive functions in young depressed patients. In conclusion, CORT coordinates via MR the networks underlying how an individual copes with stress, and this action is complemented by the widely distributed lower affinity glucocorticoid receptor (GR) involved in the subsequent management of stress adaptation. In this MR:GR regulation, the MR is an important target for promoting resilience.


Subject(s)
Brain/physiopathology , Corticosterone/physiology , Depression/physiopathology , Receptors, Mineralocorticoid/physiology , Stress, Psychological/physiopathology , Adaptation, Psychological , Animals , Antidepressive Agents/therapeutic use , Brain/metabolism , Corticosterone/metabolism , Depression/metabolism , Fludrocortisone/therapeutic use , Humans , Polymorphism, Single Nucleotide , Receptors, Glucocorticoid/metabolism , Receptors, Glucocorticoid/physiology , Receptors, Mineralocorticoid/agonists , Receptors, Mineralocorticoid/genetics , Receptors, Mineralocorticoid/metabolism , Stress, Psychological/metabolism
7.
PLoS One ; 10(11): e0141564, 2015.
Article in English | MEDLINE | ID: mdl-26529101

ABSTRACT

Emerging data suggest that Electro-Convulsive Treatment (ECT) may reduce depressive symptoms by increasing the expression of Brain-Derived Neurotrophic Factor (BDNF). Yet, conflicting findings have been reported. For this reason we performed a systematic review and meta-analysis of the preclinical and clinical literature on the association between ECT treatment (ECS in animals) and changes in BDNF concentrations and their effect on behavior. In addition, regional brain expression of BDNF in mouse and human brains were compared using Allen Brain Atlas. ECS, over sham, increased BDNF mRNA and protein in animal brain (effect size [Hedge's g]: 0.38-0.54; 258 effect-size estimates, N = 4,284) but not in serum (g = 0.06, 95% CI = -0.05-0.17). In humans, plasma but not serum BDNF increased following ECT (g = 0.72 vs. g = 0.14; 23 effect sizes, n = 281). The gradient of the BDNF increment in animal brains corresponded to the gradient of the BDNF gene expression according to the Allen brain atlas. Effect-size estimates were larger following more ECT sessions in animals (r = 0.37, P < .0001) and in humans (r = 0.55; P = 0.05). There were some indications that the increase in BDNF expression was associated with behavioral changes in rodents, but not in humans. We conclude that ECS in rodents and ECT in humans increase BDNF concentrations but this is not consistently associated with changes in behavior.


Subject(s)
Behavior, Animal , Brain-Derived Neurotrophic Factor/biosynthesis , Depression , Gene Expression Regulation , RNA, Messenger/biosynthesis , Animals , Depression/metabolism , Depression/physiopathology , Depression/therapy , Electric Stimulation Therapy , Humans , Mice
8.
Neuroscience ; 286: 412-22, 2015 Feb 12.
Article in English | MEDLINE | ID: mdl-25497375

ABSTRACT

BACKGROUND: Oral contraceptives (OCs) affect mood in some women and may have more subtle effects on emotional information processing in many more users. Female carriers of mineralocorticoid receptor (MR) haplotype 2 have been shown to be more optimistic and less vulnerable to depression. AIM: To investigate the effects of oral contraceptives on emotional information processing and a possible moderating effect of MR haplotype. METHODS: Cross-sectional study in 85 healthy premenopausal women of West-European descent. RESULTS: We found significant main effects of oral contraceptives on facial expression recognition, emotional memory and decision-making. Furthermore, carriers of MR haplotype 1 or 3 were sensitive to the impact of OCs on the recognition of sad and fearful faces and on emotional memory, whereas MR haplotype 2 carriers were not. LIMITATIONS: Different compounds of OCs were included. No hormonal measures were taken. Most naturally cycling participants were assessed in the luteal phase of their menstrual cycle. CONCLUSIONS: Carriers of MR haplotype 2 may be less sensitive to depressogenic side-effects of OCs.


Subject(s)
Contraceptives, Oral, Hormonal/adverse effects , Decision Making/drug effects , Emotions/drug effects , Mental Recall/drug effects , Receptors, Mineralocorticoid/genetics , Recognition, Psychology/drug effects , Adult , Cross-Sectional Studies , Decision Making/physiology , Emotions/physiology , Facial Expression , Female , Haplotypes , Humans , Mental Recall/physiology , Recognition, Psychology/physiology , Young Adult
9.
J Chem Neuroanat ; 54: 42-9, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24161414

ABSTRACT

This contribution dedicated to Wylie Vale is focused on the action of the glucocorticoid hormone aimed to counterbalance the stress response orchestrated by the corticotrophin releasing factor (CRF) and urocortin (Ucn) family of peptides. It appears that the release and action of these stress hormones themselves are subjected to intrinsic self-regulatory feedback loops that operate as checks and balances in stress adaptation. One of these feedback loops is operated by the mineralocorticoid (MR) and glucocorticoid receptors (GR) that mediate in complementary fashion the action of endogenous cortisol/corticosterone in brain circuits underlying the onset and termination of the stress response. By affecting appraisal processes MR has an important role in coordinating emotional expression and cognitive flexibility with the onset of the stress response, while GR's role is prominent in the management of behavioral and physiological adaptations during the recovery phase. Genetic variation in interaction with environmental input and experience-related factors can modulate this balance between susceptibility and recovery governed by a balanced MR:GR signaling. Thanks to the Wylie Vale School of scientists a parallel balanced regulation between the CRF/CRF-1 and Ucn/CRF-2 receptor systems is being uncovered, leading inexorably to the question: how do the CRF/Ucn and glucocorticoid systems interact in multiple brain sites to maintain homeostasis and health?


Subject(s)
Corticotropin-Releasing Hormone/metabolism , Feedback, Physiological/physiology , Glucocorticoids/metabolism , Neurosecretory Systems/physiology , Stress, Physiological/physiology , Urocortins/metabolism , Adrenal Glands/metabolism , Animals , Brain/metabolism , Humans
10.
Eur J Pharmacol ; 719(1-3): 53-62, 2013 Nov 05.
Article in English | MEDLINE | ID: mdl-23876452

ABSTRACT

This contribution is focused on the action of the naturally occurring corticosteroids, cortisol and corticosterone, which are secreted from the adrenals in hourly pulses and after stress with the goal to maintain resilience and health. To achieve this goal the action of the corticosteroids displays an impressive diversity, because it is cell-specific and context-dependent in coordinating the individual's response to changing environments. These diverse actions of corticosterone are mediated by mineralocorticoid- and glucocorticoid-receptors that operate as a binary system in concert with neurotransmitter and neuropeptide signals to activate and inhibit stress reactions, respectively. Classically MR and GR are gene transcription factors, but recently these receptors appear to mediate also rapid non-genomic actions on excitatory neurotransmission suggesting that they integrate functions over time. Hence the balance of receptor-mediated actions is crucial for homeostasis. This balanced function of mineralo- and glucocorticoid-receptors can be altered epigenetically by a history of traumatic (early) life events and the experience of repeated stressors as well as by predisposing genetic variants in signaling pathways of these receptors. One of these variants, mineralocorticoid receptor haplotype 2, is associated with dispositional optimism in appraisal of environmental challenges. Imbalance in receptor-mediated corticosterone actions was found to leave a genomic signature highlighting the role of master switches such as cAMP response element-binding protein and mammalian target of rapamycin to compromise health, and to promote vulnerability to disease. Diabetic encephalopathy is a pathology of imbalanced corticosterone action, which can be corrected in its pre-stage by a brief treatment with the antiglucocorticoid mifepristone.


Subject(s)
Brain/metabolism , Receptors, Steroid/metabolism , Animals , Brain/physiology , Cognition , Corticosterone/metabolism , Diabetes Mellitus/metabolism , Humans , Stress, Psychological/metabolism
11.
Psychoneuroendocrinology ; 38(9): 1858-73, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23838101

ABSTRACT

Stressful experiences during early-life can modulate the genetic programming of specific brain circuits underlying emotional and cognitive aspects of behavioral adaptation to stressful experiences later in life. Although this programming effect exerted by experience-related factors is an important determinant of mental health, its outcome depends on cognitive inputs and hence the valence an individual assigns to a given environmental context. From this perspective we will highlight, with studies in rodents, non-human primates and humans, the three-hit concept of vulnerability and resilience to stress-related mental disorders, which is based on gene-environment interactions during critical phases of perinatal and juvenile brain development. The three-hit (i.e., hit-1: genetic predisposition, hit-2: early-life environment, and hit-3: later-life environment) concept accommodates the cumulative stress hypothesis stating that in a given context vulnerability is enhanced when failure to cope with adversity accumulates. Alternatively, the concept also points to the individual's predictive adaptive capacity, which underlies the stress inoculation and match/mismatch hypotheses. The latter hypotheses propose that the experience of relatively mild early-life adversity prepares for the future and promotes resilience to similar challenges in later-life; when a mismatch occurs between early and later-life experience, coping is compromised and vulnerability is enhanced. The three-hit concept is fundamental for understanding how individuals can either be prepared for coping with life to come and remain resilient or are unable to do so and succumb to a stress-related mental disorder, under seemingly identical circumstances.


Subject(s)
Adaptation, Psychological , Models, Neurological , Models, Psychological , Resilience, Psychological , Stress, Psychological/psychology , Vulnerable Populations , Animals , Avoidance Learning , Disease Models, Animal , Epigenesis, Genetic , Female , Gene-Environment Interaction , Genetic Predisposition to Disease , Glucocorticoids/physiology , Humans , Hypothalamo-Hypophyseal System/physiopathology , Maternal Behavior , Maternal Deprivation , Neuronal Plasticity , Pituitary-Adrenal System/physiopathology , Pregnancy , Prenatal Exposure Delayed Effects , Primates , Rats , Stress Disorders, Post-Traumatic/physiopathology , Stress Disorders, Post-Traumatic/psychology , Stress, Psychological/physiopathology
12.
Psychoneuroendocrinology ; 38(5): 648-58, 2013 May.
Article in English | MEDLINE | ID: mdl-22980941

ABSTRACT

An imbalance between central glucocorticoid (GR) and mineralocorticoid (MR) receptors is proposed to underlie the HPA axis dysregulation that associates with susceptibility to psychopathology (anxiety, PTSD). To test this 'balance hypothesis' we examined whether the impact of MR levels upon HPA-axis control and behaviour depended on the relative levels of GR and vice versa. Avoiding antenatal maternal 'programming' effects by using littermates, we generated mice with forebrain MR over-expression (MR(hi)) and/or simultaneous global GR under-expression (GR(lo)). We found a significant interaction between MR and GR in control of the HPA-axis under stressed but not basal conditions. With reduced GR levels, HPA-axis activity in response to restraint stress was enhanced, likely due to impaired negative feedback. However, high MR in concert with reduced GR minimised this HPA-axis overshoot in response to stress. MR:GR balance also played a role in determining strategies of spatial memory during a watermaze probe trial: when coupled with GR under-expression, MR(hi) show enhanced perseveration, suggesting enhanced spatial recall or reduced exploratory flexibility. Other alterations in cognitive functions were specific to a single receptor without interaction, with both MR(hi) and GR(lo) manipulations independently impairing reversal learning in spatial and fear memory tasks. Thus, MR and GR interact in specific domains of neuroendocrine and cognitive control, but for other limbic-associated behaviours each receptor mediates its own repertoire of responses. Since modulation of HPA-axis and behavioural dysfunction associated with high levels of MR, selective ligands or transcriptional regulators may afford novel therapeutic approaches to affective psychopathologies.


Subject(s)
Behavior, Animal/physiology , Hypothalamo-Hypophyseal System/metabolism , Pituitary-Adrenal System/metabolism , Receptors, Glucocorticoid/genetics , Receptors, Mineralocorticoid/genetics , Animals , Cognition/physiology , Gene Expression Regulation/physiology , Hypothalamo-Hypophyseal System/physiology , Hypothalamo-Hypophyseal System/physiopathology , Male , Memory/physiology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Pituitary-Adrenal System/physiopathology , Receptors, Glucocorticoid/metabolism , Receptors, Mineralocorticoid/metabolism , Restraint, Physical/physiology
13.
Mol Psychiatry ; 18(9): 993-1005, 2013 Sep.
Article in English | MEDLINE | ID: mdl-22925833

ABSTRACT

Glucocorticoids (GCs) secreted after stress reduce adult hippocampal neurogenesis, a process that has been implicated in cognitive aspects of psychopathology, amongst others. Yet, the exact role of the GC receptor (GR), a key mediator of GC action, in regulating adult neurogenesis is largely unknown. Here, we show that GR knockdown, selectively in newborn cells of the hippocampal neurogenic niche, accelerates their neuronal differentiation and migration. Strikingly, GR knockdown induced ectopic positioning of a subset of the new granule cells, altered their dendritic complexity and increased their number of mature dendritic spines and mossy fiber boutons. Consistent with the increase in synaptic contacts, cells with GR knockdown exhibit increased basal excitability parallel to impaired contextual freezing during fear conditioning. Together, our data demonstrate a key role for the GR in newborn hippocampal cells in mediating their synaptic connectivity and structural as well as functional integration into mature hippocampal circuits involved in fear memory consolidation.


Subject(s)
Hippocampus/cytology , Motivation/genetics , Neurogenesis/genetics , Neurons/physiology , Receptors, Glucocorticoid/deficiency , Animals , Cell Movement/genetics , Conditioning, Classical/physiology , Corticosterone/metabolism , Dendrites/metabolism , Dendrites/ultrastructure , Dendritic Spines/metabolism , Dendritic Spines/ultrastructure , Fear , Genetic Vectors/physiology , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , In Vitro Techniques , Memory Disorders/genetics , Mice , Mice, Inbred BALB C , Mice, Knockout , Nerve Tissue Proteins/metabolism , Neurons/ultrastructure , Presynaptic Terminals/metabolism , RNA, Small Interfering/metabolism , Radioimmunoassay
14.
Behav Brain Res ; 241: 92-5, 2013 Mar 15.
Article in English | MEDLINE | ID: mdl-23219839

ABSTRACT

Stress induces a switch in learning strategies of male C57BL/6J mice from predominantly spatial to more stimulus-response learning. To study generalization of these findings over sex, we investigated female C57BL/6J mice at three phases of the estrous cycle under non stress and acute (10 min) restraint stress conditions. On a circular hole board (CHB) task, about half of the naive female mice used spatial and stimulus-response strategies to solve the task. Under stress, female mice favored spatial over stimulus-response strategies, with 100% of female mice in the estrus phase. Performance expressed as latency to solve the task is only improved in stressed female mice in the estrus phase. We conclude that the use of learning strategies is influenced by sex and this difference between sexes is aggravated by acute stress.


Subject(s)
Estrous Cycle/physiology , Maze Learning/physiology , Spatial Behavior/physiology , Stress, Psychological/physiopathology , Animals , Female , Memory/physiology , Mice , Mice, Inbred C57BL , Reaction Time/physiology
15.
Eur J Neurosci ; 36(8): 3096-102, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22831399

ABSTRACT

Previous studies showed that the mineralocorticoid receptor (MR) is needed for behavioral flexibility in a fear conditioning paradigm. Female mice with forebrain-specific deletion of the MR gene (MR(CaMKCre) ) were unable to show extinction of contextual fear, and could not discriminate between cue and context fear unlike control mice. In the present study, male and female (MR(CaMKCre) ) mice and control littermates were used to study sex-specific fear conditioning, memory performance and extinction. The fear conditioning paradigm assessed both context- and cue-related fear within one experimental procedure. We observed that at the end of the conditioning all mice acquired the fear-motivated response. During the first minutes of the memory test, both male and female MR(CaMKCre) mice remembered and feared the context more than the control mice. Furthermore, female MR(CaMKCre) mice were not able to extinguish this memory even on the second day of memory testing. The female mutants also could not discriminate between cue (more freezing) and context periods (less freezing). In contrast, male MR(CaMKCre) mice and the controls showed extinction and were capable to discriminate, although the MR(CaMKCre) mice needed more time before they started extinction. These findings further support the relevance of MR for behavioral flexibility and extinction of fear-motivated behavior. In conclusion, the loss of MR in the forebrain results in large differences in emotional and cognitive behaviors between female and male mice, which suggests a role of this receptor in the female prevalence of stress- and anxiety-regulated disorders.


Subject(s)
Fear/physiology , Memory/physiology , Prosencephalon/physiology , Receptors, Mineralocorticoid/physiology , Sex Characteristics , Animals , Cues , Extinction, Psychological , Female , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Prosencephalon/metabolism , Receptors, Mineralocorticoid/genetics
16.
Neurobiol Learn Mem ; 98(1): 33-40, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22543192

ABSTRACT

Corticosteroid effects on cognitive abilities during behavioral adaptation to stress are mediated by two types of receptors. While the glucocorticoid receptor (GR) is mainly involved in the consolidation of memory, the mineralocorticoid receptor (MR) mediates appraisal and initial responses to novelty. Recent findings in humans and mice suggest that under stress, the MR might be involved in the use of different learning strategies. Here, we used male mice lacking the MR in the forebrain (MR(CaMKCre)), which were subjected to 5-10 min acute restraint stress, followed 30 min later by training trials on the circular hole board. Mice had to locate an exit hole using extra- and intra-maze cues. We assessed performance and the use of spatial and stimulus-response strategies. Non-stressed MR(CaMKCre) mice showed delayed learning as compared to control littermates. Prior stress impaired performance in controls, but did not further deteriorate learning in MR(CaMKCre) mice. When stressed, 20-30% of both MR(CaMKCre) and control mice switched from a spatial to a stimulus-response strategy, which rescued performance in MR(CaMKCre) mice. Furthermore, MR(CaMKCre) mice showed increased GR mRNA expression in all CA areas of the hippocampus and an altered basal and stress-induced corticosterone secretion, which supports their role in the modulation of neuroendocrine activity. In conclusion, our data provide evidence for the critical role of MR in the fast formation of spatial memory. In the absence of forebrain MR spatial learning performance was under basal circumstances impaired, while after stress further deterioration of performance was rescued by switching behavior increasingly to a stimulus-response strategy.


Subject(s)
Adaptation, Psychological/physiology , Maze Learning/physiology , Prosencephalon/metabolism , Receptors, Mineralocorticoid/metabolism , Stress, Physiological/physiology , Stress, Psychological/metabolism , Animals , Corticosterone/blood , Cues , Hippocampus/metabolism , Male , Memory/physiology , Mice , Mice, Transgenic , Receptors, Mineralocorticoid/genetics , Stress, Psychological/genetics
17.
Physiol Behav ; 106(1): 73-80, 2012 Apr 12.
Article in English | MEDLINE | ID: mdl-21971364

ABSTRACT

Glucocorticoid hormones are secreted from the adrenal gland in hourly pulses, on top of which a surge can take place after stress. The current review describes how changes in pulse amplitude and frequency have consequences for the transcriptional responsivity of target tissues to stress-induced rises in glucocorticoids, and also how these altered pulse patterns affect neuroendocrine and behavioural responses. The mechanistic underpinning of these often rapid changes of the effects of pulsatility on stress responsivity has been greatly advanced with the discovery of membrane variants of the nuclear mineralocorticoid and glucocorticoid receptors. The new findings qualify glucocorticoid pulsatility and rapid non-genomic actions as important determinants of the allostatic state.


Subject(s)
Adrenal Cortex Hormones/physiology , Glucocorticoids/physiology , Stress, Psychological/physiopathology , Activity Cycles/physiology , Animals , Behavior/physiology , Brain/physiology , Circadian Rhythm/physiology , Humans , Hypothalamo-Hypophyseal System/physiology , Receptors, Steroid/physiology , Signal Transduction/physiology , Stress, Psychological/psychology
18.
Cell Mol Neurobiol ; 32(5): 725-35, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22113371

ABSTRACT

There are clear sex differences in incidence and onset of stress-related and other psychiatric disorders in humans. Yet, rodent models for psychiatric disorders are predominantly based on male animals. The strongest argument for not using female rodents is their estrous cycle and the fluctuating sex hormones per phase which multiplies the number of animals to be tested. Here, we will discuss studies focused on sex differences in emotionality and cognitive abilities in experimental conditions with and without stress. First, female sex hormones such as estrogens and progesterone affect emotions and cognition, contributing to sex differences in behavior. Second, females respond differently to stress than males which might be related to the phase of the estrous cycle. For example, female rats and mice express less anxiety than males in a novel environment. Proestrus females are less anxious than females in the other estrous phases. Third, males perform in spatial tasks superior to females. However, while stress impairs spatial memory in males, females improve their spatial abilities, depending on the task and kind of stressor. We conclude that the differences in emotion, cognition and responses to stress between males and females over the different phases of the estrous cycle should be used in animal models for stress-related psychiatric disorders.


Subject(s)
Cognition/physiology , Emotions/physiology , Gonadal Steroid Hormones/metabolism , Stress, Psychological/metabolism , Animals , Female , Humans , Male , Neuronal Plasticity , Receptors, Steroid/metabolism
19.
J Neuroendocrinol ; 23(4): 329-44, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21219484

ABSTRACT

We have analysed the long-term psychoneuroendocrine effects of maternal deprivation (MD) [24 h at postnatal day (PND) 9] and/or exposure to chronic unpredictable stress (CUS) during the periadolescent period (PND 28 to PND 43) in male and female Wistar rats. Animals were tested in the elevated plus maze (EPM, anxiety) at PND 44 and in two memory tests, spontaneous alternation and novel object recognition (NOT) in adulthood. The expression of hippocampal glucocorticoid (GR) and mineralocorticoid (MR) receptors, as well as of synaptophysin, neural cell adhesion molecule and brain-derived neurotrophic factor, was analysed by in situ hybridisation in selected hippocampal regions. Endocrine determinations of leptin, testosterone and oestradiol plasma levels were carried out by radioimmunoassay. Young CUS animals showed decreased anxiety behaviour in the EPM (increased percentage of time and entries in the open arms) irrespective of neonatal treatment. Memory impairments were induced by the two stressful treatments as was revealed by the NOT, with males being most clearly affected. Although each stressful procedure, when considered separately, induced different (always decrements) effects on the three synaptic molecules analysed and affected males and females differently, the combination of MD and CUS induced an unique disruptive effect on the three synaptic plasticity players. MD induced a long-term significant decrease in hippocampal GR only in males, whereas CUS tended to increase MR in males and decrease MR in females. Both neonatal MD and periadolescent CUS induced marked reductions in testosterone and oestradiol in males, whereas MD male animals also showed significantly decreased leptin levels. By contrast, in females, none of the hormones analysed was altered by any of the stressful procedures. Taking our data together in support of the 'two-hit' hypothesis, MD during neonatal life and/or exposure to CUS during the periadolescent period induced a permanent deficit in memory, which was accompanied by a decrement in markers for hippocampal plasticity. The long-term effects on body weight and hormone levels, particularly among males, might reflect sex-dependent lasting metabolic alterations as well as an impaired reproductive function.


Subject(s)
Maternal Deprivation , Stress, Physiological , Stress, Psychological , Animals , Anxiety/physiopathology , Behavior, Animal , Estradiol/blood , Female , Hippocampus/cytology , Hippocampus/metabolism , Leptin/blood , Male , Maze Learning , Neural Cell Adhesion Molecules/genetics , Neural Cell Adhesion Molecules/metabolism , Neuronal Plasticity/physiology , Neuropsychological Tests , Rats , Rats, Wistar , Receptors, Glucocorticoid/genetics , Receptors, Glucocorticoid/metabolism , Receptors, Mineralocorticoid/genetics , Receptors, Mineralocorticoid/metabolism , Synaptophysin/genetics , Synaptophysin/metabolism , Testosterone/blood
20.
Transl Psychiatry ; 1: e62, 2011 Dec 13.
Article in English | MEDLINE | ID: mdl-22832354

ABSTRACT

Mineralocorticoid (MR) and glucocorticoid receptors (GR) are abundantly expressed in the limbic brain and mediate cortisol effects on the stress-response and behavioral adaptation. Dysregulation of the stress response impairs adaptation and is a risk factor for depression, which is twice as abundant in women than in men. Because of the importance of MR for appraisal processes underlying the initial phase of the stress response we investigated whether specific MR haplotypes were associated with personality traits that predict the risk of depression. We discovered a common gene variant (haplotype 2, frequency ∼0.38) resulting in enhanced MR activity. Haplotype 2 was associated with heightened dispositional optimism in study 1 and with less hopelessness and rumination in study 2. Using data from a large genome-wide association study we then established that haplotype 2 was associated with a lower risk of depression. Interestingly, all effects were restricted to women. We propose that common functional MR haplotypes are important determinants of inter-individual variability in resilience to depression in women by differentially mediating cortisol effects on the stress system.


Subject(s)
Depressive Disorder/genetics , Haplotypes/genetics , Personality/genetics , Receptors, Mineralocorticoid/genetics , Adult , Aged , Female , Genome-Wide Association Study , Humans , Male , Middle Aged , Polymorphism, Single Nucleotide/genetics , Psychiatric Status Rating Scales , Risk , Sex Factors , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL
...