Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 123
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Neuroimage ; 184: 547-554, 2019 01 01.
Article in English | MEDLINE | ID: mdl-30243958

ABSTRACT

Acute stress is known to affect the way we process rewards. For example, during, or directly after stress, activity within key brain areas of the reward circuitry is reduced when a reward is presented. Generally, the effects of stress on the brain are time-dependent, changing neural and cognitive processing in the aftermath of stress to aid recovery. Such a dynamic response to stress is important for resilience on the longer term. However, relatively little is known about reward processing during the recovery phase of stress and whether this is changed in individuals at increased risk for stress-related psychopathology. Healthy male individuals (N = 40) and unaffected siblings of schizophrenia patients (N = 40) were randomized to either an acute stress task (Trier Social Stress Test) or a no-stress task. Neural responses during reward anticipation and reward feedback (monetary gain or no gain) were examined 50 min later using an fMRI monetary incentive delay task. The ventral striatum and orbitofrontal cortex (OFC) were used as predefined hypothesis-driven regions of interest. Neural responses following stress differed between controls and siblings during reward feedback (group × stress interaction OFC p = 0.003, ventral striatum p = 0.031), showing increased ventral striatum and OFC responses following stress in healthy controls only. Exploratory analyses revealed that this effect was most pronounced during hit trials (compared to when a reward was omitted), and independent of monetary value. Stress did not affect subsequent reward processing in siblings of schizophrenia patients. We found no significant differences between controls and siblings in ventral striatum and OFC responses during reward anticipation following stress. This study shows that ventral striatum and OFC responses to positive task feedback are increased in the aftermath of stress in healthy male controls, regardless of monetary value. This indicates a dynamic shift from previously reported reduced responses in the striatum and OFC to reward feedback directly after stress to increased responses to both reward and non-reward feedback during the recovery phase of stress. These increased neural responses following stress were absent in siblings of schizophrenia patients. Together, these findings indicate that stress recovery is affected in this at-risk group, particularly in responses to positive feedback following stress.


Subject(s)
Anticipation, Psychological/physiology , Brain/physiology , Reward , Schizophrenia/physiopathology , Stress, Psychological/physiopathology , Brain Mapping , Feedback , Humans , Magnetic Resonance Imaging , Male , Motivation , Siblings
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.
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
4.
Horm Behav ; 73: 75-82, 2015 Jul.
Article in English | MEDLINE | ID: mdl-26122295

ABSTRACT

Stress is often associated with a tend-and-befriend response, a putative coping mechanism where people behave generously towards others in order to invest in social relationships to seek comfort and mutual protection. However, this increase in generosity is expected to be directed only towards a delimited number of socially close, but not distant individuals, because it would be maladaptive to befriend everyone alike. In addition, the endocrinological stress response follows a distinct temporal pattern, and it is believed that tend-and-befriend tendencies can be observed mainly under acute stress. By contrast, the aftermath (>1h after) of stress is associated with endocrinological regulatory processes that are proposed to cause increased executive control and reduced emotional reactivity, possibly eliminating the need to tend-and-befriend. In the present experiment, we set out to investigate how these changes immediately and >1h after a stressful experience affect social-distance-dependent generosity levels, a phenomenon called social discounting. We hypothesized that stress has a time-dependent effect on social discounting, with decisions made shortly after (20min), but not 90min after stress showing increased generosity particularly to close others. We found that men tested 20min after stressor onset indeed showed increased generosity towards close but not distant others compared to non-stressed men or men tested 90min after stressor onset. These findings contribute to our understanding on how stress affects prosocial behavior by highlighting the importance of social closeness and the timing of stress relative to the decision as modulating factors in this type of decision making in men.


Subject(s)
Adaptation, Psychological/physiology , Friends/psychology , Psychological Distance , Stress, Psychological/psychology , Adult , Decision Making/physiology , Humans , Male , Social Dominance , Social Marginalization/psychology , Social Support , Time Factors , Young Adult
5.
Neurobiol Learn Mem ; 112: 168-75, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24513633

ABSTRACT

The rodent stress hormone corticosterone rapidly enhances long-term potentiation in the CA1 hippocampal area, but leads to a suppression when acting in a more delayed fashion. Both actions are thought to contribute to stress effects on emotional memory. Emotional memory formation also involves the basolateral amygdala, an important target area for corticosteroid actions. We here (1) investigated the rapid effects of corticosterone on amygdalar synaptic potentiation, (2) determined to what extent these effects depend on the mouse's recent stress history or (3) on prior ß-adrenoceptor activation; earlier studies at the single cell level showed that especially a recent history of stress changes the responsiveness of basolateral amygdala neurons to corticosterone. We report that, unlike the hippocampus, stress enhances amygdalar synaptic potentiation in a slow manner. In vitro exposure to 100 nM corticosterone quickly decreases synaptic potentiation, and causes only transient potentiation in tissue from stressed mice. This transient type of potentiation is also seen when ß-adrenoceptors are blocked during stress and this is further exacerbated by subsequent in vitro administered corticosterone. We conclude that stress and corticosterone change synaptic potentiation in the basolateral amygdala in a manner opposite to that seen in the hippocampus and that renewed exposure to corticosterone only allows induction of non-persistent forms of synaptic potentiation.


Subject(s)
Basolateral Nuclear Complex/physiopathology , CA1 Region, Hippocampal/physiopathology , Corticosterone/pharmacology , Neuronal Plasticity/drug effects , Stress, Psychological/physiopathology , Adrenergic beta-Antagonists/pharmacology , Animals , Basolateral Nuclear Complex/drug effects , CA1 Region, Hippocampal/drug effects , Male , Mice , Mice, Inbred C57BL , Propranolol/pharmacology
6.
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
7.
Psychoneuroendocrinology ; 141: 105735, 2022 07.
Article in English | MEDLINE | ID: mdl-35447495

ABSTRACT

Stress initiates a cascade of (neuro)biological, physiological, and behavioral changes, allowing us to respond to a challenging environment. The human response to acute stress can be studied in detail in controlled settings, usually in a laboratory environment. To this end, many studies employ acute stress paradigms to probe stress-related outcomes in healthy and patient populations. Though valuable, these studies in themselves often have relatively limited sample sizes. We established a data-sharing and collaborative interdisciplinary initiative, the STRESS-NL database, which combines (neuro)biological, physiological, and behavioral data across many acute stress studies in order to accelerate our understanding of the human acute stress response in health and disease (www.stressdatabase.eu). Researchers in the stress field from 12 Dutch research groups of 6 Dutch universities created a database to achieve an accurate inventory of (neuro)biological, physiological, and behavioral data from laboratory-based human studies that used acute stress tests. Currently, the STRESS-NL database consists of information on 5529 individual participants (2281 females and 3348 males, age range 6-99 years, mean age 27.7 ±â€¯16 years) stemming from 57 experiments described in 42 independent studies. Studies often did not use the same stress paradigm; outcomes were different and measured at different time points. All studies currently included in the database assessed cortisol levels before, during and after experimental stress, but cortisol measurement will not be a strict requirement for future study inclusion. Here, we report on the creation of the STRESS-NL database and infrastructure to illustrate the potential of accumulating and combining existing data to allow meta-analytical, proof-of-principle analyses. The STRESS-NL database creates a framework that enables human stress research to take new avenues in explorative and hypothesis-driven data analyses with high statistical power. Future steps could be to incorporate new studies beyond the borders of the Netherlands; or build similar databases for experimental stress studies in rodents. In our view, there are major scientific benefits in initiating and maintaining such international efforts.


Subject(s)
Hydrocortisone , Databases, Factual , Female , Humans , Hydrocortisone/analysis , Male , Netherlands
8.
Nat Neurosci ; 24(4): 470-477, 2021 04.
Article in English | MEDLINE | ID: mdl-33603229

ABSTRACT

Low statistical power reduces the reliability of animal research; yet, increasing sample sizes to increase statistical power is problematic for both ethical and practical reasons. We present an alternative solution using Bayesian priors based on historical control data, which capitalizes on the observation that control groups in general are expected to be similar to each other. In a simulation study, we show that including data from control groups of previous studies could halve the minimum sample size required to reach the canonical 80% power or increase power when using the same number of animals. We validated the approach on a dataset based on seven independent rodent studies on the cognitive effects of early-life adversity. We present an open-source tool, RePAIR, that can be widely used to apply this approach and increase statistical power, thereby improving the reliability of animal experiments.


Subject(s)
Animal Experimentation , Computer Simulation , Models, Statistical , Research Design , Sample Size , Animals , Mice
10.
Science ; 245(4925): 1502-5, 1989 Sep 29.
Article in English | MEDLINE | ID: mdl-2781292

ABSTRACT

The CA1 pyramidal neurons in the hippocampus contain a high density of adrenal corticosteroid receptors. By intracellular recording, CA1 neurons in slices from adrenalectomized rats have been found to display a markedly reduced afterhyperpolarization (that is, the hyperpolarizing phase after a brief depolarizing current pulse) when compared with their sham controls. No differences were found for other tested membrane properties. Brief exposure of hippocampal slices from adrenalectomized rats to glucocorticoid agonists, 30 to 90 minutes before recording, greatly enhanced the afterhyperpolarization. In addition, glucocorticoids attenuated the norepinephrine-induced blockade of action potential accommodation in CA1 neurons. The findings indicate that glucocorticoids can reduce transmitter-evoked excitability in the hippocampus, presumably via a receptor-mediated genomic action.


Subject(s)
Glucocorticoids/pharmacology , Hippocampus/drug effects , Norepinephrine/pharmacology , Action Potentials/drug effects , Adrenalectomy , Animals , Hippocampus/cytology , In Vitro Techniques , Membrane Potentials/drug effects , Neurons/cytology , Neurons/drug effects , Rats
11.
Science ; 239(4837): 278-80, 1988 Jan 15.
Article in English | MEDLINE | ID: mdl-2892268

ABSTRACT

Immunocytochemical and electrophysiological evidence suggests that somatostatin may be a transmitter in the hippocampus. To characterize the ionic mechanisms underlying somatostatin effects, voltage-clamp and current-clamp studies on single CA1 pyramidal neurons in the hippocampal slice preparation were performed. Both somatostatin-28 and somatostatin-14 elicited a steady outward current and selectively augmented the noninactivating, voltage-dependent outward potassium current known as the M-current. Since the muscarinic cholinergic agonists carbachol and muscarine antagonized this current, these results suggest a reciprocal regulation of the M-current by somatostatin and acetylcholine.


Subject(s)
Hippocampus/physiology , Neurons/physiology , Potassium/metabolism , Somatostatin/pharmacology , Acetylcholine/pharmacology , Action Potentials/drug effects , Animals , Carbachol/pharmacology , Cesium/pharmacology , Electric Conductivity , Hippocampus/drug effects , Membrane Potentials , Muscarine/pharmacology , Neurons/drug effects , Rats , Somatostatin-28
12.
Neurosci Biobehav Rev ; 102: 299-307, 2019 07.
Article in English | MEDLINE | ID: mdl-31047892

ABSTRACT

Altered cognitive performance is considered an intermediate phenotype mediating early life adversity (ELA) effects on later-life development of mental disorders, e.g. depression. Whereas most human studies are limited to correlational conclusions, rodent studies can prospectively investigate how ELA alters cognitive performance in several domains. Despite the volume of reports, there is no consensus on i) the behavioral domains being affected by ELA and ii) the extent of these effects. To test how ELA (here: aberrant maternal care) affects specific behavioral domains, we used a 3-level mixed-effect meta-analysis, and thoroughly explored heterogeneity with MetaForest, a novel machine-learning approach. Our results are based on >400 independent experiments, involving ∼8600 animals. Especially in males, ELA promotes memory formation during stressful learning but impairs non-stressful learning. Furthermore, ELA increases anxiety-like and decreases social behavior. The ELA phenotype was strongest when i) combined with other negative experiences ("hits"); ii) in rats; iii) in ELA models of ∼10days duration. All data is easily accessible with MaBapp (https://osf.io/ra947/), allowing researchers to run tailor-made meta-analyses, thereby revealing the optimal choice of experimental protocols and study power.


Subject(s)
Anxiety , Behavior, Animal , Disease Models, Animal , Memory , Social Behavior , Stress, Psychological , Animals , Female , Male , Anxiety/physiopathology , Memory/physiology , Phenotype , Rodentia/physiology , Stress, Psychological/physiopathology
13.
Hippocampus ; 18(1): 20-8, 2008.
Article in English | MEDLINE | ID: mdl-17708551

ABSTRACT

Remodeling of synaptic networks is believed to contribute to synaptic plasticity and long-term memory performance, both of which are modulated by chronic stress. We here examined whether chronic stress modulates dendritic complexity of hippocampal CA1 pyramidal cells, under conditions of basal as well as elevated corticosteroid hormone levels. Slices were prepared from naïve, handled or chronically stressed animals and briefly treated with vehicle or corticosterone (100 nM); neurons were visualized with a fluorescent dye injected into individual CA1 pyramidal cells. We observed that 21 days of unpredictable stress did not affect hippocampal CA1 apical or basal dendritic morphology compared with naïve animals when corticosteroid levels were low. Only when slices from stressed animals were also exposed to elevated corticosteroid levels, a significant reduction in apical (but not basal) dendritic length became apparent. Unexpectedly, animals that were handled or 3 weeks showed a reduction in both apical dendritic length and number of apical branch points when compared with naïve animals. Apical dendritic length and number of branch points were restored to levels found in naïve animals several hours after in vitro treatment with 100 nM corticosterone. All effects of acute corticosterone administration could be prevented by the glucocorticoid receptor antagonist RU38486 given during the last 4 days of the stress or handling protocol. We conclude that brief exposure to high concentrations of corticosterone can differently affect apical dendritic structure, depending on the earlier history of the animal, a process that critically depends on involvement of the glucocorticoid receptor.


Subject(s)
Dendrites/pathology , Handling, Psychological , Hippocampus/pathology , Pyramidal Cells/ultrastructure , Receptors, Glucocorticoid/physiology , Stress, Psychological/pathology , Animals , Behavior, Animal , Corticosterone/administration & dosage , Dendrites/drug effects , Disease Models, Animal , Hormone Antagonists/administration & dosage , In Vitro Techniques , Male , Mifepristone/administration & dosage , Multivariate Analysis , Rats , Rats, Wistar , Stress, Psychological/prevention & control
14.
Endocr Rev ; 19(3): 269-301, 1998 Jun.
Article in English | MEDLINE | ID: mdl-9626555

ABSTRACT

In this review, we have described the function of MR and GR in hippocampal neurons. The balance in actions mediated by the two corticosteroid receptor types in these neurons appears critical for neuronal excitability, stress responsiveness, and behavioral adaptation. Dysregulation of this MR/GR balance brings neurons in a vulnerable state with consequences for regulation of the stress response and enhanced vulnerability to disease in genetically predisposed individuals. The following specific inferences can be made on the basis of the currently available facts. 1. Corticosterone binds with high affinity to MRs predominantly localized in limbic brain (hippocampus) and with a 10-fold lower affinity to GRs that are widely distributed in brain. MRs are close to saturated with low basal concentrations of corticosterone, while high corticosterone concentrations during stress occupy both MRs and GRs. 2. The neuronal effects of corticosterone, mediated by MRs and GRs, are long-lasting, site-specific, and conditional. The action depends on cellular context, which is in part determined by other signals that can activate their own transcription factors interacting with MR and GR. These interactions provide an impressive diversity and complexity to corticosteroid modulation of gene expression. 3. Conditions of predominant MR activation, i.e., at the circadian trough at rest, are associated with the maintenance of excitability so that steady excitatory inputs to the hippocampal CA1 area result in considerable excitatory hippocampal output. By contrast, additional GR activation, e.g., after acute stress, generally depresses the CA1 hippocampal output. A similar effect is seen after adrenalectomy, indicating a U-shaped dose-response dependency of these cellular responses after the exposure to corticosterone. 4. Corticosterone through GR blocks the stress-induced HPA activation in hypothalamic CRH neurons and modulates the activity of the excitatory and inhibitory neural inputs to these neurons. Limbic (e.g., hippocampal) MRs mediate the effect of corticosterone on the maintenance of basal HPA activity and are of relevance for the sensitivity or threshold of the central stress response system. How this control occurs is not known, but it probably involves a steady excitatory hippocampal output, which regulates a GABA-ergic inhibitory tone on PVN neurons. Colocalized hippocampal GRs mediate a counteracting (i.e., disinhibitory) influence. Through GRs in ascending aminergic pathways, corticosterone potentiates the effect of stressors and arousal on HPA activation. The functional interaction between these corticosteroid-responsive inputs at the level of the PVN is probably the key to understanding HPA dysregulation associated with stress-related brain disorders. 5. Fine-tuning of HPA regulation occurs through MR- and GR-mediated effects on the processing of information in higher brain structures. Under healthy conditions, hippocampal MRs are involved in processes underlying integration of sensory information, interpretation of environmental information, and execution of appropriate behavioral reactions. Activation of hippocampal GRs facilitates storage of information and promotes elimination of inadequate behavioral responses. These behavioral effects mediated by MR and GR are linked, but how they influence endocrine regulation is not well understood. 6. Dexamethasone preferentially targets the pituitary in the blockade of stress-induced HPA activation. The brain penetration of this synthetic glucocorticoid is hampered by the mdr1a P-glycoprotein in the blood-brain barrier. Administration of moderate amounts of dexamethasone partially depletes the brain of corticosterone, and this has destabilizing consequences for excitability and information processing. 7. The set points of HPA regulation and MR/GR balance are genetically programmed, but can be reset by early life experiences involving mother-infant interaction. 8. (ABSTRACT TRUNCATED)


Subject(s)
Brain Diseases/metabolism , Brain/metabolism , Health , Receptors, Steroid/metabolism , Adrenal Cortex Hormones/physiology , Aging/physiology , Brain/physiology , Humans , Stress, Physiological/physiopathology
15.
Eur J Neurosci ; 27(10): 2542-50, 2008 May.
Article in English | MEDLINE | ID: mdl-18547242

ABSTRACT

Corticosterone (100 nm) rapidly increases the frequency of miniature excitatory postsynaptic currents in mouse CA1 pyramidal neurons via membrane-located mineralocorticoid receptors (MRs). We now show that a presynaptic ERK1/2 signalling pathway mediates the nongenomic effect, as it was blocked by the MEK inhibitors U0126 (10 microm) and PD098059 (40 microm) and occluded in H-Ras(G12V)-mutant mice with constitutive activation of the ERK1/2 presynaptic pathway. Notably, the increase in mEPSC frequency was not mediated by retrograde signalling through endocannabinoids or nitric oxide, supporting presynaptic localization of the signalling pathway. Unexpectedly, corticosterone was also found to have a direct postsynaptic effect, rapidly decreasing the peak amplitude of I(A) currents. This effect takes place via postsynaptic membrane MRs coupled to a G protein-mediated pathway, as the effect of corticosterone on I(A) was effectively blocked by 0.5 mm GDP-beta-S administered via the recording pipette into the postsynaptic cell. Taken together, these results indicate that membrane MRs mediate rapid, nongenomic effects via pre- as well as postsynaptic pathways. Through these dual pathways, high corticosterone concentrations such as occur after stress could contribute to enhanced CA1 pyramidal excitability.


Subject(s)
Adrenal Cortex Hormones/metabolism , Hippocampus/metabolism , Pyramidal Cells/metabolism , Receptors, Mineralocorticoid/metabolism , Synaptic Membranes/metabolism , Synaptic Transmission/physiology , Animals , Enzyme Inhibitors/pharmacology , Excitatory Postsynaptic Potentials/drug effects , Excitatory Postsynaptic Potentials/physiology , Guanosine Diphosphate/analogs & derivatives , Guanosine Diphosphate/pharmacology , Hippocampus/ultrastructure , MAP Kinase Signaling System/drug effects , MAP Kinase Signaling System/physiology , Male , Membrane Potentials/drug effects , Membrane Potentials/genetics , Mice , Mice, Inbred C57BL , Mitogen-Activated Protein Kinase 3/drug effects , Mitogen-Activated Protein Kinase 3/metabolism , Neuronal Plasticity/drug effects , Neuronal Plasticity/physiology , Organ Culture Techniques , Patch-Clamp Techniques , Pyramidal Cells/drug effects , Pyramidal Cells/ultrastructure , Receptors, G-Protein-Coupled/drug effects , Receptors, G-Protein-Coupled/metabolism , Receptors, Mineralocorticoid/drug effects , Stress, Physiological/metabolism , Stress, Physiological/physiopathology , Synaptic Membranes/drug effects , Synaptic Transmission/drug effects , Thionucleotides/pharmacology
16.
Nat Neurosci ; 3(10): 977-8, 2000 Oct.
Article in English | MEDLINE | ID: mdl-11017168

ABSTRACT

Glucocorticoids are secreted from the adrenal gland in very high amounts after stress. In the brain, these stress hormones potently modulate ionic currents, monoaminergic transmission, synaptic plasticity and cellular viability, most notably in the hippocampus where corticosteroid receptors are highly enriched. Here we show that at least some of these actions require DNA binding of glucocorticoid receptor (GR) homodimers.


Subject(s)
Adrenal Cortex Hormones/pharmacology , Binding Sites/drug effects , DNA/metabolism , Hippocampus/drug effects , Pyramidal Cells/drug effects , Receptors, Glucocorticoid/drug effects , Adrenal Cortex Hormones/agonists , Adrenal Cortex Hormones/metabolism , Animals , Binding Sites/physiology , Calcium Channels/drug effects , Calcium Channels/metabolism , Cell Membrane/drug effects , Cell Membrane/metabolism , Culture Techniques , Hippocampus/cytology , Hippocampus/metabolism , Mice , Mice, Mutant Strains , Mutagenesis, Site-Directed/physiology , Pyramidal Cells/cytology , Pyramidal Cells/metabolism , Receptors, Glucocorticoid/chemistry , Receptors, Glucocorticoid/genetics , Serotonin/metabolism , Serotonin/pharmacology , Stress, Physiological/pathology , Stress, Physiological/physiopathology
17.
Neurosci Biobehav Rev ; 95: 1-16, 2018 12.
Article in English | MEDLINE | ID: mdl-30201218

ABSTRACT

Adverse early life events are a well-established risk factor for the precipitation of behavioral disorders characterized by anomalies in the dopaminergic system, such as schizophrenia and addiction. The correlation between early life conditions and the dopaminergic system has been causally investigated in more than 90 rodent publications. Here, we tested the validity of the hypothesis that early life stress (ELS) alters dopamine signaling by performing an extensive 3-level mixed effect meta-analysis. We included several ELS models and biochemical indicators of the dopaminergic system in a variety of brain areas, for a total of 1009 comparisons. Contrary to our expectations, only a few comparisons displayed a significant effect. Specifically, the striatal area was the most vulnerable, displaying decreased dopamine precursor and increased metabolites after ELS. To make all data openly accessible, we created MaDEapp (https://osf.io/w25m4/), a tool to explore data of the meta-analysis with the intent to guide future (pre)clinical research and allow power calculations. All in all, ELS induces a few yet robust changes on biochemical indicators of the dopaminergic system.


Subject(s)
Brain/growth & development , Brain/metabolism , Dopamine/metabolism , Stress, Psychological/metabolism , Animals , Humans , Meta-Analysis as Topic
18.
Acta Physiol (Oxf) ; 223(2): e13066, 2018 06.
Article in English | MEDLINE | ID: mdl-29575542

ABSTRACT

After stress, the brain is exposed to waves of stress mediators, including corticosterone (in rodents) and cortisol (in humans). Corticosteroid hormones affect neuronal physiology in two time-domains: rapid, non-genomic actions primarily via mineralocorticoid receptors; and delayed genomic effects via glucocorticoid receptors. In parallel, cognitive processing is affected by stress hormones. Directly after stress, emotional behaviour involving the amygdala is strongly facilitated with cognitively a strong emphasis on the "now" and "self," at the cost of higher cognitive processing. This enables the organism to quickly and adequately respond to the situation at hand. Several hours later, emotional circuits are dampened while functions related to the prefrontal cortex and hippocampus are promoted. This allows the individual to rationalize the stressful event and place it in the right context, which is beneficial in the long run. The brain's response to stress depends on an individual's genetic background in interaction with life events. Studies in rodents point to the possibility to prevent or reverse long-term consequences of early life adversity on cognitive processing, by normalizing the balance between the two receptor types for corticosteroid hormones at a critical moment just before the onset of puberty.


Subject(s)
Brain/metabolism , Corticosterone/metabolism , Receptors, Glucocorticoid/metabolism , Stress, Physiological/physiology , Animals , Corticosterone/pharmacology , Humans , Neurons/drug effects , Neurons/metabolism , Receptors, Glucocorticoid/drug effects , Stress, Physiological/drug effects , Stress, Psychological/drug therapy , Stress, Psychological/physiopathology
19.
Neuropsychopharmacology ; 43(9): 1954-1960, 2018 08.
Article in English | MEDLINE | ID: mdl-29483659

ABSTRACT

Stress is a major risk factor for almost all psychiatric disorders, however, the underlying neurobiological mechanisms remain largely elusive. In healthy individuals, a successful stress response involves an adequate neuronal adaptation to a changing environment. This adaptive response may be dysfunctional in vulnerable individuals, potentially contributing to the development of psychopathology. In the current study, we investigated brain responses to emotional stimuli following stress in healthy controls and at-risk individuals. An fMRI study was conducted in healthy male controls (N = 39) and unaffected healthy male siblings of schizophrenia patients (N = 39) who are at increased risk for the development of a broad range of psychiatric disorders. Brain responses to pictures from the International Affective Picture System (IAPS) were measured 33 min after exposure to stress induced by the validated trier social stress test (TSST) or a control condition. Stress-induced levels of cortisol, alpha-amylase, and subjective stress were comparable in both groups. Yet, stress differentially affected brain responses of schizophrenia siblings versus controls. Specifically, control subjects, but not schizophrenia siblings, showed reduced brain activity in key nodes of the default mode network (PCC/precuneus and mPFC) and salience network (anterior insula) as well as the STG, MTG, MCC, vlPFC, precentral gyrus, and cerebellar vermis in response to all pictures following stress. These results indicate that even in the absence of a psychiatric disorder, at-risk individuals display abnormal functional activation following stress, which in turn may increase their vulnerability and risk for adverse outcomes.


Subject(s)
Brain/physiopathology , Mental Disorders/physiopathology , Stress, Psychological/physiopathology , Adult , Affect/physiology , Brain/diagnostic imaging , Brain/physiology , Brain Mapping , Genetic Predisposition to Disease , Humans , Hydrocortisone/metabolism , Magnetic Resonance Imaging , Male , Mental Disorders/diagnostic imaging , Mental Disorders/genetics , Siblings , Stress, Psychological/diagnostic imaging , Visual Perception/physiology , alpha-Amylases/metabolism
20.
Brain Res ; 1150: 14-20, 2007 May 30.
Article in English | MEDLINE | ID: mdl-17383615

ABSTRACT

We previously assessed corticosterone mediated gene expression in acute explant hippocampal slices and found over 200 responsive genes 1, 3 and 5 h after glucocorticoid receptor (GR) activation by a brief corticosterone pulse. Interestingly, 1 h after GR activation all genes were downregulated, many of which are involved in hippocampal neurotransmission and plasticity. The aim of the current experiment was 1) to measure the expression of several of these neurotransmission-related genes that were corticosterone-responsive 1 h after GR-activation in an in vivo setting, 2) to elucidate in which hippocampal subregion these expression changes take place and 3) to assess the specificity of regulation by activated GRs. For this purpose, rats were subcutaneously injected with vehicle, corticosterone or corticosterone pretreated with GR-antagonist RU38486. One hour after the corticosterone injections, mRNA expression levels of 5 selected genes were measured using in situ hybridization. The mineralocorticoid receptor (MR), MAO-A, casein kinase 2 and voltage dependent potassium mRNA's, but not dynein mRNA, were rapidly downregulated in vivo after corticosterone administration in hippocampal subregions. Furthermore, RU38486 pretreatment reversed in all cases these effects, illustrating the GR-specificity of transcriptional regulation by corticosterone. The results are important for understanding the role of GR in pleiotropic control of hippocampal neurotransmission and plasticity, which is characterized by recovery of function transiently raised by excitatory input.


Subject(s)
Casein Kinase II/metabolism , Gene Expression Regulation/drug effects , Glucocorticoids/pharmacology , Hippocampus/drug effects , Monoamine Oxidase/genetics , Receptors, Mineralocorticoid/metabolism , Shaw Potassium Channels/metabolism , Analysis of Variance , Animals , Casein Kinase II/genetics , Drug Interactions , Hormone Antagonists/pharmacology , In Situ Hybridization/methods , Male , Mifepristone/pharmacology , Monoamine Oxidase/metabolism , RNA, Messenger/metabolism , Rats , Rats, Wistar , Receptors, Mineralocorticoid/genetics , Shaw Potassium Channels/genetics
SELECTION OF CITATIONS
SEARCH DETAIL