Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
Psychoneuroendocrinology ; 69: 161-71, 2016 07.
Article in English | MEDLINE | ID: mdl-27107207

ABSTRACT

Glucocorticoid challenge tests such as the dexamethasone suppression test (DST) and the combined dexamethasone/corticotropin-releasing hormone (dex-CRH) test are considered to be able to sensitively measure hypothalamic-pituitary-adrenal (HPA) axis activity in stress-related psychiatric and endocrine disorders. We used mass-spectrometry to assess the relationship of plasma dexamethasone concentrations and the outcome of these tests in two independent cohorts. Dexamethasone concentrations were measured after oral ingestion of 1.5mg dexamethasone in two cohorts that underwent a standard (dexamethasone at 23:00h) as well as modified (18:00h) DST and dex-CRH test. The first study population was a case/control cohort of 105 depressed patients and 133 controls in which peripheral blood mRNA expression was also measured. The second was a cohort of 261 depressed patients that underwent a standard dex-CRH test at baseline and after 12 weeks' treatment with cognitive-behavioral therapy or antidepressants. Dexamethasone concentrations explained significant proportions of the variance in the DST in both the first (24.6%) and the second (5.2%) cohort. Dexamethasone concentrations explained a higher proportion of the variance in the dex-CRH test readouts, with 41.9% of the cortisol area under the curve (AUC) in the first sample and 24.7% in the second sample. In contrast to these strong effects at later time points, dexamethasone concentrations did not impact cortisol or ACTH concentrations or mRNA expression 3hours after ingestion. In the second sample, dexamethasone concentrations at baseline and week 12 were highly correlated, independent of treatment type and response status. Importantly, a case/control effect in the Dex-CRH test was only apparent when controlling for dexamethasone concentrations. Our results suggest that the incorporation of plasma dexamethasone concentration or measures of earlier endocrine read-outs may help to improve the assessment of endocrine dysfunction in depression.


Subject(s)
Receptors, Glucocorticoid/drug effects , Receptors, Glucocorticoid/metabolism , Adrenocorticotropic Hormone/blood , Adult , Antidepressive Agents/therapeutic use , Case-Control Studies , Corticotropin-Releasing Hormone/metabolism , Depression/blood , Depression/metabolism , Depressive Disorder, Major/metabolism , Dexamethasone/analysis , Dexamethasone/blood , Dexamethasone/metabolism , Female , Gene Expression , Glucocorticoids/pharmacology , Humans , Hydrocortisone/analysis , Hydrocortisone/blood , Hypothalamo-Hypophyseal System/physiopathology , Male , Middle Aged , Pituitary-Adrenal System/physiopathology , Time Factors
2.
J Psychiatr Res ; 68: 261-9, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26228428

ABSTRACT

Although mental disorders as major depression are highly prevalent worldwide their underlying causes remain elusive. Despite the high heritability of depression and a clear genetic contribution to the disease, the identification of genetic risk factors for depression has been very difficult. The first published candidate to reach genome-wide significance in depression was SLC6A15, a neuronal amino acid transporter. With a reported 1,42 fold increased risk of suffering from depression associated with a single nucleotide polymorphism (SNP) in a regulatory region of SLC6A15, the polymorphism was also found to affect hippocampal morphology, integrity, and hippocampus-dependent memory. However, the function of SLC6A15 in the brain is so far largely unknown. To address this question, we investigated if alterations in SLC6A15 expression, either using a full knockout or a targeted hippocampal overexpression, affect hippocampal neurochemistry and consequently behavior. We could show that a lack of SLC6A15 reduced hippocampal tissue levels of proline and other neutral amino acids. In parallel, we observed a decreased overall availability of tissue glutamate and glutamine, while at the same time the basal tone of extracellular glutamate in the hippocampus was increased. By contrast, SLC6A15 overexpression increased glutamate/glutamine tissue concentrations. These neurochemical alterations could be linked to behavioral abnormalities in sensorimotor gating, a key translational endophenotype relevant for many psychiatric disorders. Overall, our data supports SLC6A15 as a crucial factor controlling amino acid content in the hippocampus, thereby likely interfering with glutamatergic transmission and behavior. These findings emphasize SLC6A15 as pivotal risk factor for vulnerability to psychiatric diseases.


Subject(s)
Amino Acid Transport Systems, Neutral/physiology , Behavior, Animal/physiology , Glutamic Acid/metabolism , Hippocampus/metabolism , Sensory Gating/physiology , Amino Acid Transport Systems, Neutral/genetics , Amino Acid Transport Systems, Neutral/metabolism , Animals , Hippocampus/anatomy & histology , Hippocampus/chemistry , Magnetic Resonance Spectroscopy , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Proline/metabolism , Real-Time Polymerase Chain Reaction , Signal Transduction
3.
J Neurosci ; 35(24): 9007-16, 2015 Jun 17.
Article in English | MEDLINE | ID: mdl-26085626

ABSTRACT

Anxiety-related psychiatric disorders represent one of the largest health burdens worldwide. Single nucleotide polymorphisms of the FK506 binding protein 51 (FKBP51) gene have been repeatedly associated with anxiety-related disorders and stress sensitivity. Given the intimate relationship of stress and anxiety, we hypothesized that amygdala FKBP51 may mediate anxiety-related behaviors. Mimicking the stress effect by specifically overexpressing FKBP51 in the basolateral amygdala (BLA) or central amygdala resulted in increased anxiety-related behavior, respectively. In contrast, application of a highly selective FKBP51 point mutant antagonist, following FKBP51(mut) BLA-overexpression, reduced the anxiogenic phenotype. We subsequently tested a novel FKBP51 antagonist, SAFit2, in wild-type mice via BLA microinjections, which reduced anxiety-related behavior. Remarkably, the same effect was observed following peripheral administration of SAFit2. To our knowledge, this is the first in vivo study using a specific FKBP51 antagonist, thereby unraveling the role of FKBP51 and its potential as a novel drug target for the improved treatment of anxiety-related disorders.


Subject(s)
Anti-Anxiety Agents/administration & dosage , Anxiety/metabolism , Tacrolimus Binding Proteins/antagonists & inhibitors , Tacrolimus Binding Proteins/biosynthesis , Amygdala/drug effects , Amygdala/metabolism , Animals , Anxiety/drug therapy , Anxiety/psychology , Ligands , Male , Mice , Mice, Inbred C57BL , Microinjections/methods , Risk Factors
4.
Front Behav Neurosci ; 8: 445, 2014.
Article in English | MEDLINE | ID: mdl-25610380

ABSTRACT

Major depression (MD) is one of the most common psychiatric disorders, severely affecting the quality of life of millions of people worldwide. Despite the availability of several classes of antidepressants, treatment efficacy is still very variable and many patients do not respond to the treatment. Clomipramine (CMI), a classical and widely used antidepressant, shows widespread interindividual variability of efficacy, while the environmental factors contributing to such variability remain unclear. We investigated whether chronic stress modulates the bio-distribution of CMI, and as a result the behavioral response to CMI treatment in a mouse model of chronic social defeat stress (CSDS). Our results show that stress exposure increased anxiety-like and depressive-like behaviors and altered the stress response. Chronic defeat stress furthermore significantly altered CMI bio-distribution. Interestingly, CMI bio-distribution highly correlated with anxiety-like and depressive-like behaviors only under basal conditions. Taken together, we provide first evidence demonstrating that chronic stress exposure modulates CMI bio-distribution and behavioral responses. This may contribute to CMI's broad interindividual variability, and is especially relevant in clinical practice.

5.
J Neurosci ; 30(50): 16949-58, 2010 Dec 15.
Article in English | MEDLINE | ID: mdl-21159965

ABSTRACT

Increased vulnerability to aversive experiences is one of the main risk factors for stress-related psychiatric disorders as major depression. However, the molecular bases of vulnerability, on the one hand, and stress resilience, on the other hand, are still not understood. Increasing clinical and preclinical evidence suggests a central involvement of the glutamatergic system in the pathogenesis of major depression. Using a mouse paradigm, modeling increased stress vulnerability and depression-like symptoms in a genetically diverse outbred strain, and we tested the hypothesis that differences in AMPA receptor function may be linked to individual variations in stress vulnerability. Vulnerable and resilient animals differed significantly in their dorsal hippocampal AMPA receptor expression and AMPA receptor binding. Treatment with an AMPA receptor potentiator during the stress exposure prevented the lasting effects of chronic social stress exposure on physiological, neuroendocrine, and behavioral parameters. In addition, spatial short-term memory, an AMPA receptor-dependent behavior, was found to be predictive of individual stress vulnerability and response to AMPA potentiator treatment. Finally, we provide evidence that genetic variations in the AMPA receptor subunit GluR1 are linked to the vulnerable phenotype. Therefore, we propose genetic variations in the AMPA receptor system to shape individual stress vulnerability. Those individual differences can be predicted by the assessment of short-term memory, thereby opening up the possibility for a specific treatment by enhancing AMPA receptor function.


Subject(s)
Genetic Testing/methods , Hippocampus/metabolism , Individuality , Memory, Short-Term/drug effects , Receptors, AMPA/metabolism , Stress, Psychological/metabolism , Animals , Corticosterone/blood , Depressive Disorder, Major/etiology , Disease Models, Animal , Female , Gene Expression Regulation/drug effects , Hippocampus/drug effects , Male , Mice , Phenotype , Polymorphism, Single Nucleotide , Protein Array Analysis/methods , Receptors, AMPA/agonists , Receptors, AMPA/genetics , Resilience, Psychological/drug effects , Risk Factors , Stress, Psychological/blood , Stress, Psychological/genetics , Stress, Psychological/psychology , Sulfonamides/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...